Bio


Karl Deisseroth is the D.H. Chen Professor of Bioengineering and of Psychiatry and Behavioral Sciences at Stanford University, and Investigator of the Howard Hughes Medical Institute. He received his undergraduate degree from Harvard, his PhD from Stanford, and his MD from Stanford. He also completed postdoctoral training, medical internship, and adult psychiatry residency at Stanford, and he is board-certified by the American Board of Psychiatry and Neurology. He continues as a practicing psychiatrist at Stanford with specialization in affective disorders and autism-spectrum disease, employing medications along with neural stimulation.

Over the last sixteen years, his laboratory created and developed optogenetics, hydrogel-tissue chemistry (beginning with CLARITY), and a broad range of enabling methods. He also has employed his technologies to discover the neural cell types and connections that cause adaptive and maladaptive behaviors, and has disseminated the technologies to thousands of laboratories around the world.

Among other honors, Deisseroth was the sole recipient for optogenetics of the 2010 Koetser Prize, the 2010 Nakasone Prize, the 2011 Alden Spencer Prize, the 2013 Richard Lounsbery Prize, the 2014 Dickson Prize in Science, the 2015 Keio Prize, the 2015 Lurie Prize, the 2015 Albany Prize, the 2015 Dickson Prize in Medicine, the 2017 Redelsheimer Prize, the 2017 Fresenius Prize, the 2017 NOMIS Distinguished Scientist Award, the 2018 Eisenberg Prize, the 2018 Kyoto Prize, the 2020 Heineken Prize in Medicine from the Royal Netherlands Academy of Arts and Sciences, and the 2023 Japan Prize. For his discoveries, Deisseroth has also received the Perl Prize (2012), the BRAIN prize (2013), the Pasarow Prize (2013), the Breakthrough Prize (2015) the BBVA Award (2016), the Massry Prize (2016) and the Harvey Prize from the Technion/Israel (2017). He was selected a Howard Hughes Medical Institute Investigator in 2013, and was elected to the US National Academy of Medicine in 2010, to the US National Academy of Sciences in 2012, and to the US National Academy of Engineering in 2019.

Clinical Focus


  • Psychiatry

Academic Appointments


Honors & Awards


  • Japan Prize in Life Sciences, for optogenetics (2023)
  • Luisa Gross Horwitz Prize, for optogenetics (2022)
  • Lasker Basic Medical Research Award, for development of optogenetics (2021)
  • Ariëns Kappers Award, Royal Netherlands Academy of Arts and Sciences (2020)
  • Lifetime Achievement Award in Medicine, J.E. Wallace Sterling (2020)
  • Heineken Prize in Medicine, Royal Netherlands Academy of Arts and Sciences (2020)
  • Kyoto Prize, for the discovery of optogenetics and the development of causal systems neuroscience (2018)
  • Harvey Prize in Human Health, Technion, Israel (2017)
  • Dickson Prize in Medicine, for optogenetics (2015)
  • Breakthrough Prize in Life Science, for optogenetics (2015)
  • Massry Prize, Massry Foundation (2017)
  • Warren Alpert Prize, Harvard (2019)
  • Berthold Leibinger Prize, for development of optogenetics (2018)
  • Eisenberg Prize, University of Michigan (2018)
  • Rumford Prize, AAAS (2018)
  • NOMIS Distinguished Scientist Award, NOMIS Foundation (2017)
  • BBVA Award, for optogenetics (2016)
  • Redelsheimer Award, for optogenetics and CLARITY (2016)
  • Albany Prize in Medicine, for optogenetics (2015)
  • Young investigator Award, Society for Neuroscience (2009)
  • Lurie Prize in Biomedical Sciences, Foundation for the NIH (2015)
  • Meyer Award, American Pcyhiatric Association (2015)
  • Dickson Prize in Science, for optogenetics (2014)
  • Keio Medical Science Prize, for optoenetics (2014)
  • Wilson Prize, Harvard (2014)
  • BRAIN Prize, Lundbeck Research Foundation (2013)
  • Gabbay Award, Brandeis (2013)
  • Goldman-Rakic Award, Yale (2013)
  • Pasarow Foundation Award, Pasarow Foundation (2013)
  • Premio Citta' di Firenze for Molecular Sciences, for optogenetics and CLARITY (2013)
  • Richard Lounsbery Prize, National Academy of Sciences (2013)
  • Perl Prize, UNC (2012)
  • Zuelch Prize, Max-Planck Society (2012)
  • Spencer Prize, Columbia (2011)
  • Koetser Prize, Zurich Switzerland (2010)
  • Nakasone Prize, HFSP (2010)
  • Gill YIA Award, Indiana University (2009)
  • Lawrence C. Katz Prize, Duke University (2008)
  • Schuetze Prize, Columbia (2008)
  • McKnight Foundation Scholar Award, McKnight Foundation (2007)
  • Presidential Early Career Award in Science and Engineering (PECASE), NIH (2006)
  • Director's Pioneer Award, National Institutes of Health (2005)
  • Early Career Translational Research Award, Coulter Foundation (2005)
  • Klingenstein Fellowship, Klingenstein Foundation (2005)
  • McKnight Foundation Technological Innovations in Neuroscience Award, McKnight Foundation (2005)
  • Culpeper Scholar Award, Rockefeller Brothers Fund, Goldman Philanthropic Partnerships (2004)
  • Outstanding Resident, National Institute of Mental Health (2002)

Boards, Advisory Committees, Professional Organizations


  • Member, National Academy of Medicine (2011 - Present)
  • Member, National Academy of Engineering (2019 - Present)
  • Member, National Academy of Sciences (2012 - Present)

Professional Education


  • Medical Education: Stanford University School of Medicine (2000) CA
  • Residency: Stanford University Adult Psychiatry Residency (2004) CA
  • Internship: Stanford University Adult Psychiatry Residency (2001) CA
  • Board Certification: American Board of Psychiatry and Neurology, Psychiatry (2006)
  • Ph.D., Stanford University, Neuroscience (1998)
  • M.D., Stanford University (2000)
  • A.B., Harvard, Biochemical Sciences (1992)

Current Research and Scholarly Interests


Karl Deisseroth is the D.H. Chen Professor of Bioengineering and of Psychiatry and Behavioral Sciences at Stanford University, and Investigator of the Howard Hughes Medical Institute. Over the last sixteen years, his laboratory created and developed optogenetics, hydrogel-tissue chemistry (beginning with CLARITY), and a broad range of enabling methods. He also has employed his technologies to discover the neural cell types and connections that cause adaptive and maladaptive behaviors, and has disseminated the technologies to thousands of laboratories around the world.

Stanford Advisees


All Publications


  • Human OPRM1 and murine Oprm1 promoter driven viral constructs for genetic access to μ-opioidergic cell types. Nature communications Salimando, G. J., Tremblay, S., Kimmey, B. A., Li, J., Rogers, S. A., Wojick, J. A., McCall, N. M., Wooldridge, L. M., Rodrigues, A., Borner, T., Gardiner, K. L., Jayakar, S. S., Singeç, I., Woolf, C. J., Hayes, M. R., De Jonghe, B. C., Bennett, F. C., Bennett, M. L., Blendy, J. A., Platt, M. L., Creasy, K. T., Renthal, W. R., Ramakrishnan, C., Deisseroth, K., Corder, G. 2023; 14 (1): 5632

    Abstract

    With concurrent global epidemics of chronic pain and opioid use disorders, there is a critical need to identify, target and manipulate specific cell populations expressing the mu-opioid receptor (MOR). However, available tools and transgenic models for gaining long-term genetic access to MOR+ neural cell types and circuits involved in modulating pain, analgesia and addiction across species are limited. To address this, we developed a catalog of MOR promoter (MORp) based constructs packaged into adeno-associated viral vectors that drive transgene expression in MOR+ cells. MORp constructs designed from promoter regions upstream of the mouse Oprm1 gene (mMORp) were validated for transduction efficiency and selectivity in endogenous MOR+ neurons in the brain, spinal cord, and periphery of mice, with additional studies revealing robust expression in rats, shrews, and human induced pluripotent stem cell (iPSC)-derived nociceptors. The use of mMORp for in vivo fiber photometry, behavioral chemogenetics, and intersectional genetic strategies is also demonstrated. Lastly, a human designed MORp (hMORp) efficiently transduced macaque cortical OPRM1+ cells. Together, our MORp toolkit provides researchers cell type specific genetic access to target and functionally manipulate mu-opioidergic neurons across a range of vertebrate species and translational models for pain, addiction, and neuropsychiatric disorders.

    View details for DOI 10.1038/s41467-023-41407-2

    View details for PubMedID 37704594

    View details for PubMedCentralID 6428583

  • Lifelong restructuring of 3D genome architecture in cerebellar granule cells. Science (New York, N.Y.) Tan, L., Shi, J., Moghadami, S., Parasar, B., Wright, C. P., Seo, Y., Vallejo, K., Cobos, I., Duncan, L., Chen, R., Deisseroth, K. 2023; 381 (6662): 1112-1119

    Abstract

    The cerebellum contains most of the neurons in the human brain and exhibits distinctive modes of development and aging. In this work, by developing our single-cell three-dimensional (3D) genome assay-diploid chromosome conformation capture, or Dip-C-into population-scale (Pop-C) and virus-enriched (vDip-C) modes, we resolved the first 3D genome structures of single cerebellar cells, created life-spanning 3D genome atlases for both humans and mice, and jointly measured transcriptome and chromatin accessibility during development. We found that although the transcriptome and chromatin accessibility of cerebellar granule neurons mature in early postnatal life, 3D genome architecture gradually remodels throughout life, establishing ultra-long-range intrachromosomal contacts and specific interchromosomal contacts that are rarely seen in neurons. These results reveal unexpected evolutionarily conserved molecular processes that underlie distinctive features of neural development and aging across the mammalian life span.

    View details for DOI 10.1126/science.adh3253

    View details for PubMedID 37676945

  • Structural basis for ion selectivity in potassium-selective channelrhodopsins. Cell Tajima, S., Kim, Y. S., Fukuda, M., Jo, Y., Wang, P. Y., Paggi, J. M., Inoue, M., Byrne, E. F., Kishi, K. E., Nakamura, S., Ramakrishnan, C., Takaramoto, S., Nagata, T., Konno, M., Sugiura, M., Katayama, K., Matsui, T. E., Yamashita, K., Kim, S., Ikeda, H., Kim, J., Kandori, H., Dror, R. O., Inoue, K., Deisseroth, K., Kato, H. E. 2023

    Abstract

    KCR channelrhodopsins (K+-selective light-gated ion channels) have received attention as potential inhibitory optogenetic tools but more broadly pose a fundamental mystery regarding how their K+ selectivity is achieved. Here, we present 2.5-2.7 Å cryo-electron microscopy structures of HcKCR1 and HcKCR2 and of a structure-guided mutant with enhanced K+ selectivity. Structural, electrophysiological, computational, spectroscopic, and biochemical analyses reveal a distinctive mechanism for K+ selectivity; rather than forming the symmetrical filter of canonical K+ channels achieving both selectivity and dehydration, instead, three extracellular-vestibule residues within each monomer form a flexible asymmetric selectivity gate, while a distinct dehydration pathway extends intracellularly. Structural comparisons reveal a retinal-binding pocket that induces retinal rotation (accounting for HcKCR1/HcKCR2 spectral differences), and design of corresponding KCR variants with increased K+ selectivity (KALI-1/KALI-2) provides key advantages for optogenetic inhibition in vitro and in vivo. Thus, discovery of a mechanism for ion-channel K+ selectivity also provides a framework for next-generation optogenetics.

    View details for DOI 10.1016/j.cell.2023.08.009

    View details for PubMedID 37652010

  • Cardiogenic control of affective behavioural state. Nature Hsueh, B., Chen, R., Jo, Y., Tang, D., Raffiee, M., Kim, Y. S., Inoue, M., Randles, S., Ramakrishnan, C., Patel, S., Kim, D. K., Liu, T. X., Kim, S. H., Tan, L., Mortazavi, L., Cordero, A., Shi, J., Zhao, M., Ho, T. T., Crow, A., Yoo, A. W., Raja, C., Evans, K., Bernstein, D., Zeineh, M., Goubran, M., Deisseroth, K. 2023

    Abstract

    Emotional states influence bodily physiology, as exemplified in the top-down process by which anxiety causes faster beating of the heart1-3. However, whether an increased heart rate might itself induce anxiety or fear responses is unclear3-8. Physiological theories of emotion, proposed over a century ago, have considered that in general, there could be an important and even dominant flow of information from the body to the brain9. Here, to formally test this idea, we developed a noninvasive optogenetic pacemaker for precise, cell-type-specific control of cardiac rhythms of up to 900beats per minute in freely moving mice, enabled by a wearable micro-LED harness and the systemic viral delivery of a potent pump-like channelrhodopsin. We found that optically evoked tachycardia potently enhanced anxiety-like behaviour, but crucially only in risky contexts, indicating that both central (brain) and peripheral (body) processes may be involved in the development of emotional states. To identify potential mechanisms, we used whole-brain activity screening and electrophysiology to find brain regions that wereactivated by imposed cardiac rhythms. We identified the posterior insular cortex as a potential mediator of bottom-up cardiac interoceptive processing, and found that optogenetic inhibition of this brain region attenuated the anxiety-like behaviour that was induced by optical cardiac pacing. Together, these findings reveal that cells of both the body and the brain must be considered together to understand the origins of emotional or affective states. More broadly, our results define a generalizable approach for noninvasive, temporally precise functional investigations of joint organism-wide interactions among targeted cells during behaviour.

    View details for DOI 10.1038/s41586-023-05748-8

    View details for PubMedID 36859543

  • All-optical physiology resolves a synaptic basis for behavioral timescale plasticity. Cell Fan, L. Z., Kim, D. K., Jennings, J. H., Tian, H., Wang, P. Y., Ramakrishnan, C., Randles, S., Sun, Y., Thadhani, E., Kim, Y. S., Quirin, S., Giocomo, L., Cohen, A. E., Deisseroth, K. 2023

    Abstract

    Learning has been associated with modifications of synaptic and circuit properties, but the precise changes storing information in mammals have remained largely unclear. We combined genetically targeted voltage imaging with targeted optogenetic activation and silencing of pre- and post-synaptic neurons to study the mechanisms underlying hippocampal behavioral timescale plasticity. In mice navigating a virtual-reality environment, targeted optogenetic activation of individual CA1 cells at specific places induced stable representations of these places in the targeted cells. Optical elicitation, recording, and modulation of synaptic transmission in behaving mice revealed that activity in presynaptic CA2/3 cells was required for the induction of plasticity in CA1 and, furthermore, that during induction of these place fields in single CA1 cells, synaptic input from CA2/3 onto these same cells was potentiated. These results reveal synaptic implementation of hippocampal behavioral timescale plasticity and define a methodology to resolve synaptic plasticity during learning and memory in behaving mammals.

    View details for DOI 10.1016/j.cell.2022.12.035

    View details for PubMedID 36669484

  • Video-based pooled screening yields improved far-red genetically encoded voltage indicators. Nature methods Tian, H., Davis, H. C., Wong-Campos, J. D., Park, P., Fan, L. Z., Gmeiner, B., Begum, S., Werley, C. A., Borja, G. B., Upadhyay, H., Shah, H., Jacques, J., Qi, Y., Parot, V., Deisseroth, K., Cohen, A. E. 2023

    Abstract

    Video-based screening of pooled libraries is a powerful approach for directed evolution of biosensors because it enables selection along multiple dimensions simultaneously from large libraries. Here we develop a screening platform, Photopick, which achieves precise phenotype-activated photoselection over a large field of view (2.3 × 2.3 mm, containing >103 cells, per shot). We used the Photopick platform to evolve archaerhodopsin-derived genetically encoded voltage indicators (GEVIs) with improved signal-to-noise ratio (QuasAr6a) and kinetics (QuasAr6b). These GEVIs gave improved signals in cultured neurons and in live mouse brains. By combining targeted in vivo optogenetic stimulation with high-precision voltage imaging, we characterized inhibitory synaptic coupling between individual cortical NDNF (neuron-derived neurotrophic factor) interneurons, and excitatory electrical synapses between individual hippocampal parvalbumin neurons. The QuasAr6 GEVIs are powerful tools for all-optical electrophysiology and the Photopick approach could be adapted to evolve a broad range of biosensors.

    View details for DOI 10.1038/s41592-022-01743-5

    View details for PubMedID 36624211

  • Multiregion neuronal activity: the forest and the trees. Nature reviews. Neuroscience Machado, T. A., Kauvar, I. V., Deisseroth, K. 2022

    Abstract

    The past decade has witnessed remarkable advances in the simultaneous measurement of neuronal activity across many brain regions, enabling fundamentally new explorations of the brain-spanning cellular dynamics that underlie sensation, cognition and action. These recently developed multiregion recording techniques have provided many experimental opportunities, but thoughtful consideration of methodological trade-offs is necessary, especially regarding field of view, temporal acquisition rate and ability to guarantee cellular resolution. When applied in concert with modern optogenetic and computational tools, multiregion recording has already made possible fundamental biological discoveries - in part via the unprecedented ability to perform unbiased neural activity screens for principles of brain function, spanning dozens of brain areas and from local to global scales.

    View details for DOI 10.1038/s41583-022-00634-0

    View details for PubMedID 36192596

  • Cell-type-specific population dynamics of diverse reward computations. Cell Sylwestrak, E. L., Jo, Y., Vesuna, S., Wang, X., Holcomb, B., Tien, R. H., Kim, D. K., Fenno, L., Ramakrishnan, C., Allen, W. E., Chen, R., Shenoy, K. V., Sussillo, D., Deisseroth, K. 2022; 185 (19): 3568

    Abstract

    Computational analysis of cellular activity has developed largely independently of modern transcriptomic cell typology, but integrating these approaches may be essential for full insight into cellular-level mechanisms underlying brain function and dysfunction. Applying this approach to the habenula (a structure with diverse, intermingled molecular, anatomical, and computational features), we identified encoding of reward-predictive cues and reward outcomes in distinct genetically defined neural populations, including TH+ cells and Tac1+ cells. Data from genetically targeted recordings were used to train an optimized nonlinear dynamical systems model and revealed activity dynamics consistent with a line attractor. High-density, cell-type-specific electrophysiological recordings and optogenetic perturbation provided supporting evidence for this model. Reverse-engineering predicted how Tac1+ cells might integrate reward history, which was complemented by invivo experimentation. This integrated approach describes a process by which data-driven computational models of population activity can generate and frame actionable hypotheses for cell-type-specific investigation in biological systems.

    View details for DOI 10.1016/j.cell.2022.08.019

    View details for PubMedID 36113428

  • Structural basis for channel conduction in the pump-like channelrhodopsin ChRmine. Cell Kishi, K. E., Kim, Y. S., Fukuda, M., Inoue, M., Kusakizako, T., Wang, P. Y., Ramakrishnan, C., Byrne, E. F., Thadhani, E., Paggi, J. M., Matsui, T. E., Yamashita, K., Nagata, T., Konno, M., Quirin, S., Lo, M., Benster, T., Uemura, T., Liu, K., Shibata, M., Nomura, N., Iwata, S., Nureki, O., Dror, R. O., Inoue, K., Deisseroth, K., Kato, H. E. 1800

    Abstract

    ChRmine, a recently discovered pump-like cation-conducting channelrhodopsin, exhibits puzzling properties (large photocurrents, red-shifted spectrum, and extreme light sensitivity) that have created new opportunities in optogenetics. ChRmine and its homologs function as ion channels but, by primary sequence, more closely resemble ion pump rhodopsins; mechanisms for passive channel conduction in this family have remained mysterious. Here, we present the 2.0A resolution cryo-EM structure of ChRmine, revealing architectural features atypical for channelrhodopsins: trimeric assembly, a short transmembrane-helix 3, a twisting extracellular-loop 1, large vestibules within the monomer, and an opening at the trimer interface. We applied this structure to design three proteins (rsChRmine and hsChRmine, conferring further red-shifted and high-speed properties, respectively, and frChRmine, combining faster and more red-shifted performance) suitable for fundamental neuroscience opportunities. These results illuminate the conduction and gating of pump-like channelrhodopsins and point the way toward further structure-guided creation of channelrhodopsins for applications across biology.

    View details for DOI 10.1016/j.cell.2022.01.007

    View details for PubMedID 35114111

  • From microbial membrane proteins to the mysteries of emotion. Cell Deisseroth, K. 2021

    Abstract

    On the occasion of the 2021 Lasker Basic Medical Research Award to Karl Deisseroth, Peter Hegemann, and Dieter Oesterhelt (for "the discovery of light-sensitive microbial proteins that can activate or deactivate individual brain cells-leading to the development of optogenetics and revolutionizing neuroscience"), Deisseroth reflects on this international collaboration, his basic mechanistic and structural discoveries regarding microbial channels that transduce photons into ion current, the causal exploration of brain cell function, and the pressing mysteries of psychiatry.

    View details for DOI 10.1016/j.cell.2021.08.018

    View details for PubMedID 34562367

  • A Molecular Calcium Integrator Reveals a Striatal Cell Type Driving Aversion. Cell Kim, C. K., Sanchez, M. I., Hoerbelt, P., Fenno, L. E., Malenka, R. C., Deisseroth, K., Ting, A. Y. 2020

    Abstract

    The ability to record transient cellular events in the DNA or RNA of cells would enable precise, large-scale analysis, selection, and reprogramming of heterogeneous cell populations. Here, we report a molecular technology for stable genetic tagging of cells that exhibit activity-related increases in intracellular calcium concentration (FLiCRE). We used FLiCRE to transcriptionally label activated neural ensembles in the nucleus accumbens of the mouse brain during brief stimulation of aversive inputs. Using single-cell RNA sequencing, we detected FLiCRE transcripts among the endogenous transcriptome, providing simultaneous readout of both cell-type and calcium activation history. We identified a cell type in the nucleus accumbens activated downstream of long-range excitatory projections. Taking advantage of FLiCRE's modular design, we expressed an optogenetic channel selectively in this cell type and showed that direct recruitment of this otherwise genetically inaccessible population elicits behavioral aversion. The specificity and minute resolution of FLiCRE enables molecularly informed characterization, manipulation, and reprogramming of activated cellular ensembles.

    View details for DOI 10.1016/j.cell.2020.11.015

    View details for PubMedID 33308478

  • Deep posteromedial cortical rhythm in dissociation. Nature Vesuna, S., Kauvar, I. V., Richman, E., Gore, F., Oskotsky, T., Sava-Segal, C., Luo, L., Malenka, R. C., Henderson, J. M., Nuyujukian, P., Parvizi, J., Deisseroth, K. 2020

    Abstract

    Advanced imaging methods now allow cell-type-specific recording of neural activity across the mammalian brain, potentially enabling the exploration of how brain-wide dynamical patterns give rise to complex behavioural states1-12. Dissociation is an altered behavioural state in which the integrity of experience is disrupted, resulting in reproducible cognitive phenomena including the dissociation of stimulus detection from stimulus-related affective responses. Dissociation can occur as a result of trauma, epilepsy or dissociative drug use13,14, but despite its substantial basic and clinical importance, the underlying neurophysiology of this state is unknown. Here we establish such a dissociation-like state in mice, induced by precisely-dosed administration of ketamine or phencyclidine. Large-scale imaging of neural activity revealed that these dissociative agents elicited a 1-3-Hz rhythm in layer5 neurons of the retrosplenial cortex. Electrophysiological recording with four simultaneously deployed high-density probes revealed rhythmic coupling of the retrosplenial cortex with anatomically connected components of thalamus circuitry, but uncoupling from most other brain regions was observed-including a notable inverse correlation with frontally projecting thalamic nuclei. In testing for causal significance, we found thatrhythmic optogenetic activation of retrosplenial cortex layer5 neurons recapitulated dissociation-like behavioural effects. Local retrosplenial hyperpolarization-activated cyclic-nucleotide-gated potassium channel 1 (HCN1) pacemakers were required for systemic ketamine to induce this rhythm and to elicit dissociation-like behavioural effects. In a patient with focal epilepsy, simultaneous intracranial stereoencephalography recordings from across the brain revealed a similarly localized rhythm in the homologous deep posteromedial cortex that was temporally correlated with pre-seizure self-reported dissociation, and local brief electrical stimulation of this region elicited dissociative experiences. These results identify themolecular, cellular and physiological properties of a conserved deep posteromedial cortical rhythm that underlies states of dissociation.

    View details for DOI 10.1038/s41586-020-2731-9

    View details for PubMedID 32939091

  • Multiple convergent hypothalamus-brainstem circuits drive defensive behavior. Nature neuroscience Lovett-Barron, M., Chen, R., Bradbury, S., Andalman, A. S., Wagle, M., Guo, S., Deisseroth, K. 2020

    Abstract

    The hypothalamus is composed of many neuropeptidergic cell populations and directs multiple survival behaviors, including defensive responses to threats. However, the relationship between the peptidergic identity of neurons and their roles in behavior remains unclear. Here, we address this issue by studying the function of multiple neuronal populations in the zebrafish hypothalamus during defensive responses to a variety of homeostatic threats. Cellular registration of large-scale neural activity imaging to multiplexed in situ gene expression revealed that neuronal populations encoding behavioral features encompass multiple overlapping sets of neuropeptidergic cell classes. Manipulations of different cell populations showed that multiple sets of peptidergic neurons play similar behavioral roles in this fast-timescale behavior through glutamate co-release and convergent output to spinal-projecting premotor neurons in the brainstem. Our findings demonstrate that homeostatic threats recruit neurons across multiple hypothalamic cell populations, which cooperatively drive robust defensive behaviors.

    View details for DOI 10.1038/s41593-020-0655-1

    View details for PubMedID 32572237

  • Genetically targeted chemical assembly of functional materials in living cells, tissues, and animals. Science (New York, N.Y.) Liu, J. n., Kim, Y. S., Richardson, C. E., Tom, A. n., Ramakrishnan, C. n., Birey, F. n., Katsumata, T. n., Chen, S. n., Wang, C. n., Wang, X. n., Joubert, L. M., Jiang, Y. n., Wang, H. n., Fenno, L. E., Tok, J. B., Pașca, S. P., Shen, K. n., Bao, Z. n., Deisseroth, K. n. 2020; 367 (6484): 1372–76

    Abstract

    The structural and functional complexity of multicellular biological systems, such as the brain, are beyond the reach of human design or assembly capabilities. Cells in living organisms may be recruited to construct synthetic materials or structures if treated as anatomically defined compartments for specific chemistry, harnessing biology for the assembly of complex functional structures. By integrating engineered-enzyme targeting and polymer chemistry, we genetically instructed specific living neurons to guide chemical synthesis of electrically functional (conductive or insulating) polymers at the plasma membrane. Electrophysiological and behavioral analyses confirmed that rationally designed, genetically targeted assembly of functional polymers not only preserved neuronal viability but also achieved remodeling of membrane properties and modulated cell type-specific behaviors in freely moving animals. This approach may enable the creation of diverse, complex, and functional structures and materials within living systems.

    View details for DOI 10.1126/science.aay4866

    View details for PubMedID 32193327

  • Cortical Observation by Synchronous Multifocal Optical Sampling Reveals Widespread Population Encoding of Actions. Neuron Kauvar, I. V., Machado, T. A., Yuen, E. n., Kochalka, J. n., Choi, M. n., Allen, W. E., Wetzstein, G. n., Deisseroth, K. n. 2020

    Abstract

    To advance the measurement of distributed neuronal population representations of targeted motor actions on single trials, we developed an optical method (COSMOS) for tracking neural activity in a largely uncharacterized spatiotemporal regime. COSMOS allowed simultaneous recording of neural dynamics at ∼30 Hz from over a thousand near-cellular resolution neuronal sources spread across the entire dorsal neocortex of awake, behaving mice during a three-option lick-to-target task. We identified spatially distributed neuronal population representations spanning the dorsal cortex that precisely encoded ongoing motor actions on single trials. Neuronal correlations measured at video rate using unaveraged, whole-session data had localized spatial structure, whereas trial-averaged data exhibited widespread correlations. Separable modes of neural activity encoded history-guided motor plans, with similar population dynamics in individual areas throughout cortex. These initial experiments illustrate how COSMOS enables investigation of large-scale cortical dynamics and that information about motor actions is widely shared between areas, potentially underlying distributed computations.

    View details for DOI 10.1016/j.neuron.2020.04.023

    View details for PubMedID 32433908

  • Comprehensive Dual- and Triple-Feature Intersectional Single-Vector Delivery of Diverse Functional Payloads to Cells of Behaving Mammals. Neuron Fenno, L. E., Ramakrishnan, C. n., Kim, Y. S., Evans, K. E., Lo, M. n., Vesuna, S. n., Inoue, M. n., Cheung, K. Y., Yuen, E. n., Pichamoorthy, N. n., Hong, A. S., Deisseroth, K. n. 2020

    Abstract

    The resolution and dimensionality with which biologists can characterize cell types have expanded dramatically in recent years, and intersectional consideration of such features (e.g., multiple gene expression and anatomical parameters) is increasingly understood to be essential. At the same time, genetically targeted technology for writing in and reading out activity patterns for cells in living organisms has enabled causal investigation in physiology and behavior; however, cell-type-specific delivery of these tools (including microbial opsins for optogenetics and genetically encoded Ca2+ indicators) has thus far fallen short of versatile targeting to cells jointly defined by many individually selected features. Here, we develop a comprehensive intersectional targeting toolbox including 39 novel vectors for joint-feature-targeted delivery of 13 molecular payloads (including opsins, indicators, and fluorophores), systematic approaches for development and optimization of new intersectional tools, hardware for in vivo monitoring of expression dynamics, and the first versatile single-virus tools (Triplesect) that enable targeting of triply defined cell types.

    View details for DOI 10.1016/j.neuron.2020.06.003

    View details for PubMedID 32574559

  • Deep brain optogenetics without intracranial surgery. Nature biotechnology Chen, R. n., Gore, F. n., Nguyen, Q. A., Ramakrishnan, C. n., Patel, S. n., Kim, S. H., Raffiee, M. n., Kim, Y. S., Hsueh, B. n., Krook-Magnusson, E. n., Soltesz, I. n., Deisseroth, K. n. 2020

    Abstract

    Achieving temporally precise, noninvasive control over specific neural cell types in the deep brain would advance the study of nervous system function. Here we use the potent channelrhodopsin ChRmine to achieve transcranial photoactivation of defined neural circuits, including midbrain and brainstem structures, at unprecedented depths of up to 7 mm with millisecond precision. Using systemic viral delivery of ChRmine, we demonstrate behavioral modulation without surgery, enabling implant-free deep brain optogenetics.

    View details for DOI 10.1038/s41587-020-0679-9

    View details for PubMedID 33020604

  • Neuronal Dynamics Regulating Brain and Behavioral State Transitions CELL Andalman, A. S., Burns, V. M., Lovett-Barron, M., Broxton, M., Poole, B., Yang, S. J., Grosenick, L., Lerner, T. N., Chen, R., Benster, T., Mourrain, P., Levoy, M., Rajan, K., Deisseroth, K. 2019; 177 (4): 970-+
  • Thirst regulates motivated behavior through modulation of brainwide neural population dynamics SCIENCE Allen, W. E., Chen, M. Z., Pichamoorthy, N., Tien, R. H., Pachitariu, M., Luo, L., Deisseroth, K. 2019; 364 (6437): 253-+
  • Interacting neural ensembles in orbitofrontal cortex for social and feeding behaviour NATURE Jennings, J. H., Kim, C. K., Marshel, J. H., Raffiee, M., Ye, L., Quirin, S., Pak, S., Ramakrishnan, C., Deisseroth, K. 2019; 565 (7741): 645-+
  • Cortical layer-specific critical dynamics triggering perception. Science (New York, N.Y.) Marshel, J. H., Kim, Y. S., Machado, T. A., Quirin, S. n., Benson, B. n., Kadmon, J. n., Raja, C. n., Chibukhchyan, A. n., Ramakrishnan, C. n., Inoue, M. n., Shane, J. C., McKnight, D. J., Yoshizawa, S. n., Kato, H. E., Ganguli, S. n., Deisseroth, K. n. 2019

    Abstract

    Perceptual experiences may arise from neuronal activity patterns in mammalian neocortex. We probed mouse neocortex during visual discrimination using a red-shifted channelrhodopsin (ChRmine, discovered through structure-guided genome mining) alongside multiplexed multiphoton-holography (MultiSLM), achieving control of individually-specified neurons spanning large cortical volumes with millisecond precision. Stimulating a critical number of stimulus-orientation-selective neurons drove widespread recruitment of functionally-related neurons, a process enhanced by (but not requiring) orientation-discrimination task learning. Optogenetic targeting of orientation-selective ensembles elicited correct behavioral discrimination. Cortical layer specific-dynamics were apparent, as emergent neuronal activity asymmetrically propagated from layer-2/3 to layer-5, and smaller layer-5 ensembles were as effective as larger layer-2/3 ensembles in eliciting orientation discrimination behavior. Population dynamics emerging after optogenetic stimulation both correctly predicted behavior and resembled natural neural representations of visual stimuli.

    View details for DOI 10.1126/science.aaw5202

    View details for PubMedID 31320556

  • Crystal structure of the natural anion-conducting channelrhodopsin GtACR1 NATURE Kim, Y., Kato, H. E., Yamashita, K., Ito, S., Inoue, K., Ramakrishnan, C., Fenno, L. E., Evans, K. E., Paggi, J. M., Dror, R. O., Kandori, H., Kobilka, B. K., Deisseroth, K. 2018; 561 (7723): 343-+
  • Structural mechanisms of selectivity and gating in anion channelrhodopsins NATURE Kato, H. E., Kim, Y., Paggi, J. M., Evans, K. E., Allen, W. E., Richardson, C., Inoue, K., Ito, S., Ramakrishnan, C., Fenno, L. E., Yamashita, K., Hilger, D., Lee, S., Berndt, A., Shen, K., Kandori, H., Dror, R. O., Kobilka, B. K., Deisseroth, K. 2018; 561 (7723): 349-+
  • 5-HT release in nucleus accumbens rescues social deficits in mouse autism model NATURE Walsh, J. J., Christoffel, D. J., Heifets, B. D., Ben-Dor, G. A., Selimbeyoglu, A., Hung, L. W., Deisseroth, K., Malenka, R. C. 2018; 560 (7720): 589-+
  • Three-dimensional intact-tissue sequencing of single-cell transcriptional states SCIENCE Wang, X., Allen, W. E., Wright, M. A., Sylwestrak, E. L., Samusik, N., Vesuna, S., Evans, K., Liu, C., Ramakrishnan, C., Liu, J., Nolan, G. P., Bava, F., Deisseroth, K. 2018; 361 (6400): 380-+
  • Hydrogel-Tissue Chemistry: Principles and Applications ANNUAL REVIEW OF BIOPHYSICS, VOL 47 Gradinaru, V., Treweek, J., Overton, K., Deisseroth, K., Dill, K. A. 2018; 47: 355–76

    Abstract

    Over the past five years, a rapidly developing experimental approach has enabled high-resolution and high-content information retrieval from intact multicellular animal (metazoan) systems. New chemical and physical forms are created in the hydrogel-tissue chemistry process, and the retention and retrieval of crucial phenotypic information regarding constituent cells and molecules (and their joint interrelationships) are thereby enabled. For example, rich data sets defining both single-cell-resolution gene expression and single-cell-resolution activity during behavior can now be collected while still preserving information on three-dimensional positioning and/or brain-wide wiring of those very same neurons-even within vertebrate brains. This new approach and its variants, as applied to neuroscience, are beginning to illuminate the fundamental cellular and chemical representations of sensation, cognition, and action. More generally, reimagining metazoans as metareactants-or positionally defined three-dimensional graphs of constituent chemicals made available for ongoing functionalization, transformation, and readout-is stimulating innovation across biology and medicine.

    View details for PubMedID 29792820

  • Optical and chemical discoveries recognized for impact on biology and psychiatry. EMBO reports Deisseroth, K. 2017; 18 (6): 859-860

    View details for DOI 10.15252/embr.201744405

    View details for PubMedID 28566521

    View details for PubMedCentralID PMC5452044

  • Global Representations of Goal-Directed Behavior in Distinct Cell Types of Mouse Neocortex NEURON Allen, W. E., Kauvar, I. V., Chen, M. Z., Richman, E. B., Yang, S. J., Chan, K., Gradinaru, V., Deverman, B. E., Luo, L., Deisseroth, K. 2017; 94 (4): 891-?

    Abstract

    The successful planning and execution of adaptive behaviors in mammals may require long-range coordination of neural networks throughout cerebral cortex. The neuronal implementation of signals that could orchestrate cortex-wide activity remains unclear. Here, we develop and apply methods for cortex-wide Ca(2+) imaging in mice performing decision-making behavior and identify a global cortical representation of task engagement encoded in the activity dynamics of both single cells and superficial neuropil distributed across the majority of dorsal cortex. The activity of multiple molecularly defined cell types was found to reflect this representation with type-specific dynamics. Focal optogenetic inhibition tiled across cortex revealed a crucial role for frontal cortex in triggering this cortex-wide phenomenon; local inhibition of this region blocked both the cortex-wide response to task-initiating cues and the voluntary behavior. These findings reveal cell-type-specific processes in cortex for globally representing goal-directed behavior and identify a major cortical node that gates the global broadcast of task-related information.

    View details for DOI 10.1016/j.neuron.2017.04.017

    View details for PubMedID 28521139

  • Integration of optogenetics with complementary methodologies in systems neuroscience NATURE REVIEWS NEUROSCIENCE Kim, C. K., Adhikari, A., Deisseroth, K. 2017; 18 (4): 222-235

    Abstract

    Modern optogenetics can be tuned to evoke activity that corresponds to naturally occurring local or global activity in timing, magnitude or individual-cell patterning. This outcome has been facilitated not only by the development of core features of optogenetics over the past 10 years (microbial-opsin variants, opsin-targeting strategies and light-targeting devices) but also by the recent integration of optogenetics with complementary technologies, spanning electrophysiology, activity imaging and anatomical methods for structural and molecular analysis. This integrated approach now supports optogenetic identification of the native, necessary and sufficient causal underpinnings of physiology and behaviour on acute or chronic timescales and across cellular, circuit-level or brain-wide spatial scales.

    View details for DOI 10.1038/nrn.2017.15

    View details for Web of Science ID 000396325500008

    View details for PubMedID 28303019

  • Gamma oscillations organize top-down signalling to hypothalamus and enable food seeking NATURE Carus-Cadavieco, M., Gorbati, M., Ye, L., Bender, F., van der Veldt, S., Kosse, C., Borgers, C., Lee, S. Y., Ramakrishnan, C., Hu, Y., Denisova, N., Ramm, F., Volitaki, E., Burdakov, D., Deisseroth, K., Ponomarenko, A., Korotkova, T. 2017; 542 (7640): 232-236

    Abstract

    Both humans and animals seek primary rewards in the environment, even when such rewards do not correspond to current physiological needs. An example of this is a dissociation between food-seeking behaviour and metabolic needs, a notoriously difficult-to-treat symptom of eating disorders. Feeding relies on distinct cell groups in the hypothalamus, the activity of which also changes in anticipation of feeding onset. The hypothalamus receives strong descending inputs from the lateral septum, which is connected, in turn, with cortical networks, but cognitive regulation of feeding-related behaviours is not yet understood. Cortical cognitive processing involves gamma oscillations, which support memory, attention, cognitive flexibility and sensory responses. These functions contribute crucially to feeding behaviour by unknown neural mechanisms. Here we show that coordinated gamma (30-90 Hz) oscillations in the lateral hypothalamus and upstream brain regions organize food-seeking behaviour in mice. Gamma-rhythmic input to the lateral hypothalamus from somatostatin-positive lateral septum cells evokes food approach without affecting food intake. Inhibitory inputs from the lateral septum enable separate signalling by lateral hypothalamus neurons according to their feeding-related activity, making them fire at distinct phases of the gamma oscillation. Upstream, medial prefrontal cortical projections provide gamma-rhythmic inputs to the lateral septum; these inputs are causally associated with improved performance in a food-rewarded learning task. Overall, our work identifies a top-down pathway that uses gamma synchronization to guide the activity of subcortical networks and to regulate feeding behaviour by dynamic reorganization of functional cell groups in the hypothalamus.

    View details for DOI 10.1038/nature21066

    View details for PubMedID 28146472

  • Cognitive neuroscience: In search of lost time. Nature Young, N. P., Deisseroth, K. 2017; 542 (7640): 173-174

    View details for DOI 10.1038/nature21497

    View details for PubMedID 28146476

  • The form and function of channelrhodopsin. Science (New York, N.Y.) Deisseroth, K., Hegemann, P. 2017; 357 (6356)

    Abstract

    Channelrhodopsins are light-gated ion channels that, via regulation of flagellar function, enable single-celled motile algae to seek ambient light conditions suitable for photosynthesis and survival. These plant behavioral responses were initially investigated more than 150 years ago. Recently, major principles of function for light-gated ion channels have been elucidated by creating channelrhodopsins with kinetics that are accelerated or slowed over orders of magnitude, by discovering and designing channelrhodopsins with altered spectral properties, by solving the high-resolution channelrhodopsin crystal structure, and by structural model-guided redesign of channelrhodopsins for altered ion selectivity. Each of these discoveries not only revealed basic principles governing the operation of light-gated ion channels, but also enabled the creation of new proteins for illuminating, via optogenetics, the fundamentals of brain function.

    View details for PubMedID 28912215

  • Rabies screen reveals GPe control of cocaine-triggered plasticity. Nature Beier, K. T., Kim, C. K., Hoerbelt, P. n., Hung, L. W., Heifets, B. D., DeLoach, K. E., Mosca, T. J., Neuner, S. n., Deisseroth, K. n., Luo, L. n., Malenka, R. C. 2017

    Abstract

    Identification of neural circuit changes that contribute to behavioural plasticity has routinely been conducted on candidate circuits that were preselected on the basis of previous results. Here we present an unbiased method for identifying experience-triggered circuit-level changes in neuronal ensembles in mice. Using rabies virus monosynaptic tracing, we mapped cocaine-induced global changes in inputs onto neurons in the ventral tegmental area. Cocaine increased rabies-labelled inputs from the globus pallidus externus (GPe), a basal ganglia nucleus not previously known to participate in behavioural plasticity triggered by drugs of abuse. We demonstrated that cocaine increased GPe neuron activity, which accounted for the increase in GPe labelling. Inhibition of GPe activity revealed that it contributes to two forms of cocaine-triggered behavioural plasticity, at least in part by disinhibiting dopamine neurons in the ventral tegmental area. These results suggest that rabies-based unbiased screening of changes in input populations can identify previously unappreciated circuit elements that critically support behavioural adaptations.

    View details for PubMedID 28902833

  • Pathways to clinical CLARITY: volumetric analysis of irregular, soft, and heterogeneous tissues in development and disease. Scientific reports Hsueh, B. n., Burns, V. M., Pauerstein, P. n., Holzem, K. n., Ye, L. n., Engberg, K. n., Wang, A. C., Gu, X. n., Chakravarthy, H. n., Arda, H. E., Charville, G. n., Vogel, H. n., Efimov, I. R., Kim, S. n., Deisseroth, K. n. 2017; 7 (1): 5899

    Abstract

    Three-dimensional tissue-structural relationships are not well captured by typical thin-section histology, posing challenges for the study of tissue physiology and pathology. Moreover, while recent progress has been made with intact methods for clearing, labeling, and imaging whole organs such as the mature brain, these approaches are generally unsuitable for soft, irregular, and heterogeneous tissues that account for the vast majority of clinical samples and biopsies. Here we develop a biphasic hydrogel methodology, which along with automated analysis, provides for high-throughput quantitative volumetric interrogation of spatially-irregular and friable tissue structures. We validate and apply this approach in the examination of a variety of developing and diseased tissues, with specific focus on the dynamics of normal and pathological pancreatic innervation and development, including in clinical samples. Quantitative advantages of the intact-tissue approach were demonstrated compared to conventional thin-section histology, pointing to broad applications in both research and clinical settings.

    View details for PubMedID 28724969

  • Modulation of prefrontal cortex excitation/inhibition balance rescues social behavior in CNTNAP2-deficient mice. Science translational medicine Selimbeyoglu, A. n., Kim, C. K., Inoue, M. n., Lee, S. Y., Hong, A. S., Kauvar, I. n., Ramakrishnan, C. n., Fenno, L. E., Davidson, T. J., Wright, M. n., Deisseroth, K. n. 2017; 9 (401)

    Abstract

    Alterations in the balance between neuronal excitation and inhibition (E:I balance) have been implicated in the neural circuit activity-based processes that contribute to autism phenotypes. We investigated whether acutely reducing E:I balance in mouse brain could correct deficits in social behavior. We used mice lacking the CNTNAP2 gene, which has been implicated in autism, and achieved a temporally precise reduction in E:I balance in the medial prefrontal cortex (mPFC) either by optogenetically increasing the excitability of inhibitory parvalbumin (PV) neurons or decreasing the excitability of excitatory pyramidal neurons. Surprisingly, both of these distinct, real-time, and reversible optogenetic modulations acutely rescued deficits in social behavior and hyperactivity in adult mice lacking CNTNAP2 Using fiber photometry, we discovered that native mPFC PV neuronal activity differed between CNTNAP2 knockout and wild-type mice. During social interactions with other mice, PV neuron activity increased in wild-type mice compared to interactions with a novel object, whereas this difference was not observed in CNTNAP2 knockout mice. Together, these results suggest that real-time modulation of E:I balance in the mouse prefrontal cortex can rescue social behavior deficits reminiscent of autism phenotypes.

    View details for PubMedID 28768803

  • Ancestral Circuits for the Coordinated Modulation of Brain State. Cell Lovett-Barron, M. n., Andalman, A. S., Allen, W. E., Vesuna, S. n., Kauvar, I. n., Burns, V. M., Deisseroth, K. n. 2017; 171 (6): 1411–23.e17

    Abstract

    Internal states of the brain profoundly influence behavior. Fluctuating states such as alertness can be governed by neuromodulation, but the underlying mechanisms and cell types involved are not fully understood. We developed a method to globally screen for cell types involved in behavior by integrating brain-wide activity imaging with high-content molecular phenotyping and volume registration at cellular resolution. We used this method (MultiMAP) to record from 22 neuromodulatory cell types in behaving zebrafish during a reaction-time task that reports alertness. We identified multiple monoaminergic, cholinergic, and peptidergic cell types linked to alertness and found that activity in these cell types was mutually correlated during heightened alertness. We next recorded from and controlled homologous neuromodulatory cells in mice; alertness-related cell-type dynamics exhibited striking evolutionary conservation and modulated behavior similarly. These experiments establish a method for unbiased discovery of cellular elements underlying behavior and reveal an evolutionarily conserved set of diverse neuromodulatory systems that collectively govern internal state.

    View details for PubMedID 29103613

  • Thirst-associated preoptic neurons encode an aversive motivational drive. Science (New York, N.Y.) Allen, W. E., DeNardo, L. A., Chen, M. Z., Liu, C. D., Loh, K. M., Fenno, L. E., Ramakrishnan, C. n., Deisseroth, K. n., Luo, L. n. 2017; 357 (6356): 1149–55

    Abstract

    Water deprivation produces a drive to seek and consume water. How neural activity creates this motivation remains poorly understood. We used activity-dependent genetic labeling to characterize neurons activated by water deprivation in the hypothalamic median preoptic nucleus (MnPO). Single-cell transcriptional profiling revealed that dehydration-activated MnPO neurons consist of a single excitatory cell type. After optogenetic activation of these neurons, mice drank water and performed an operant lever-pressing task for water reward with rates that scaled with stimulation frequency. This stimulation was aversive, and instrumentally pausing stimulation could reinforce lever-pressing. Activity of these neurons gradually decreased over the course of an operant session. Thus, the activity of dehydration-activated MnPO neurons establishes a scalable, persistent, and aversive internal state that dynamically controls thirst-motivated behavior.

    View details for PubMedID 28912243

  • Molecular and Circuit-Dynamical Identification of Top-Down Neural Mechanisms for Restraint of Reward Seeking. Cell Kim, C. K., Ye, L. n., Jennings, J. H., Pichamoorthy, N. n., Tang, D. D., Yoo, A. W., Ramakrishnan, C. n., Deisseroth, K. n. 2017; 170 (5): 1013–27.e14

    Abstract

    Reward-seeking behavior is fundamental to survival, but suppression of this behavior can be essential as well, even for rewards of high value. In humans and rodents, the medial prefrontal cortex (mPFC) has been implicated in suppressing reward seeking; however, despite vital significance in health and disease, the neural circuitry through which mPFC regulates reward seeking remains incompletely understood. Here, we show that a specific subset of superficial mPFC projections to a subfield of nucleus accumbens (NAc) neurons naturally encodes the decision to initiate or suppress reward seeking when faced with risk of punishment. A highly resolved subpopulation of these top-down projecting neurons, identified by 2-photon Ca(2+) imaging and activity-dependent labeling to recruit the relevant neurons, was found capable of suppressing reward seeking. This natural activity-resolved mPFC-to-NAc projection displayed unique molecular-genetic and microcircuit-level features concordant with a conserved role in the regulation of reward-seeking behavior, providing cellular and anatomical identifiers of behavioral and possible therapeutic significance.

    View details for PubMedID 28823561

  • A LOOK INSIDE THE BRAIN SCIENTIFIC AMERICAN Deisseroth, K. 2016; 315 (4): 31-37

    Abstract

    A new experimental approach at the interface of chemistry and biology lets scientists peer into the deepest reaches of the body’s master controller

    View details for Web of Science ID 000384159400023

    View details for PubMedID 27798589

    View details for PubMedCentralID PMC5846712

  • Wiring and Molecular Features of Prefrontal Ensembles Representing Distinct Experiences CELL Ye, L., Allen, W. E., Thompson, K. R., Tian, Q., Hsueh, B., Ramakrishnan, C., Wang, A., Jennings, J. H., Adhikari, A., Halpern, C. H., Witten, I. B., Barth, A. L., Luo, L., McNab, J. A., Deisseroth, K. 2016; 165 (7): 1776-1788

    Abstract

    A major challenge in understanding the cellular diversity of the brain has been linking activity during behavior with standard cellular typology. For example, it has not been possible to determine whether principal neurons in prefrontal cortex active during distinct experiences represent separable cell types, and it is not known whether these differentially active cells exert distinct causal influences on behavior. Here, we develop quantitative hydrogel-based technologies to connect activity in cells reporting on behavioral experience with measures for both brain-wide wiring and molecular phenotype. We find that positive and negative-valence experiences in prefrontal cortex are represented by cell populations that differ in their causal impact on behavior, long-range wiring, and gene expression profiles, with the major discriminant being expression of the adaptation-linked gene NPAS4. These findings illuminate cellular logic of prefrontal cortex information processing and natural adaptive behavior and may point the way to cell-type-specific understanding and treatment of disease-associated states.

    View details for DOI 10.1016/j.cell.2016.05.010

    View details for PubMedID 27238022

  • Targeting Neural Circuits CELL Rajasethupathy, P., Ferenczi, E., Deisseroth, K. 2016; 165 (3): 524-534

    Abstract

    Optogenetic methodology enables direct targeting of specific neural circuit elements for inhibition or excitation while spanning timescales from the acute (milliseconds) to the chronic (many days or more). Although the impact of this temporal versatility and cellular specificity has been greater for basic science than clinical research, it is natural to ask whether the dynamic patterns of neural circuit activity discovered to be causal in adaptive or maladaptive behaviors could become targets for treatment of neuropsychiatric diseases. Here, we consider the landscape of ideas related to therapeutic targeting of circuit dynamics. Specifically, we highlight optical, ultrasonic, and magnetic concepts for the targeted control of neural activity, preclinical/clinical discovery opportunities, and recently reported optogenetically guided clinical outcomes.

    View details for DOI 10.1016/j.cell.2016.03.047

    View details for Web of Science ID 000374636800013

    View details for PubMedID 27104976

  • Hypothalamic control of male aggression-seeking behavior NATURE NEUROSCIENCE Falkner, A. L., Grosenick, L., Davidson, T. J., Deisseroth, K., Lin, D. 2016; 19 (4): 596-?

    Abstract

    In many vertebrate species, certain individuals will seek out opportunities for aggression, even in the absence of threat-provoking cues. Although several brain areas have been implicated in the generation of attack in response to social threat, little is known about the neural mechanisms that promote self-initiated or 'voluntary' aggression-seeking when no threat is present. To explore this directly, we utilized an aggression-seeking task in which male mice self-initiated aggression trials to gain brief and repeated access to a weaker male that they could attack. In males that exhibited rapid task learning, we found that the ventrolateral part of the ventromedial hypothalamus (VMHvl), an area with a known role in attack, was essential for aggression-seeking. Using both single-unit electrophysiology and population optical recording, we found that VMHvl neurons became active during aggression-seeking and that their activity tracked changes in task learning and extinction. Inactivation of the VMHvl reduced aggression-seeking behavior, whereas optogenetic stimulation of the VMHvl accelerated moment-to-moment aggression-seeking and intensified future attack. These data demonstrate that the VMHvl can mediate both acute attack and flexible seeking actions that precede attack.

    View details for DOI 10.1038/nn.4264

    View details for Web of Science ID 000372909800017

    View details for PubMedCentralID PMC4853470

  • Simultaneous fast measurement of circuit dynamics at multiple sites across the mammalian brain. Nature methods Kim, C. K., Yang, S. J., Pichamoorthy, N., Young, N. P., Kauvar, I., Jennings, J. H., Lerner, T. N., Berndt, A., Lee, S. Y., Ramakrishnan, C., Davidson, T. J., Inoue, M., Bito, H., Deisseroth, K. 2016; 13 (4): 325-328

    Abstract

    Real-time activity measurements from multiple specific cell populations and projections are likely to be important for understanding the brain as a dynamical system. Here we developed frame-projected independent-fiber photometry (FIP), which we used to record fluorescence activity signals from many brain regions simultaneously in freely behaving mice. We explored the versatility of the FIP microscope by quantifying real-time activity relationships among many brain regions during social behavior, simultaneously recording activity along multiple axonal pathways during sensory experience, performing simultaneous two-color activity recording, and applying optical perturbation tuned to elicit dynamics that match naturally occurring patterns observed during behavior.

    View details for DOI 10.1038/nmeth.3770

    View details for PubMedID 26878381

  • Nucleus accumbens D2R cells signal prior outcomes and control risky decision-making NATURE Zalocusky, K. A., Ramakrishnan, C., Lerner, T. N., Davidson, T. J., Knutson, B., Deisseroth, K. 2016; 531 (7596): 642-?

    Abstract

    A marked bias towards risk aversion has been observed in nearly every species tested. A minority of individuals, however, instead seem to prefer risk (repeatedly choosing uncertain large rewards over certain but smaller rewards), and even risk-averse individuals sometimes opt for riskier alternatives. It is not known how neural activity underlies such important shifts in decision-making-either as a stable trait across individuals or at the level of variability within individuals. Here we describe a model of risk-preference in rats, in which stable individual differences, trial-by-trial choices, and responses to pharmacological agents all parallel human behaviour. By combining new genetic targeting strategies with optical recording of neural activity during behaviour in this model, we identify relevant temporally specific signals from a genetically and anatomically defined population of neurons. This activity occurred within dopamine receptor type-2 (D2R)-expressing cells in the nucleus accumbens (NAc), signalled unfavourable outcomes from the recent past at a time appropriate for influencing subsequent decisions, and also predicted subsequent choices made. Having uncovered this naturally occurring neural correlate of risk selection, we then mimicked the temporally specific signal with optogenetic control during decision-making and demonstrated its causal effect in driving risk-preference. Specifically, risk-preferring rats could be instantaneously converted to risk-averse rats with precisely timed phasic stimulation of NAc D2R cells. These findings suggest that individual differences in risk-preference, as well as real-time risky decision-making, can be largely explained by the encoding in D2R-expressing NAc cells of prior unfavourable outcomes during decision-making.

    View details for DOI 10.1038/nature17400

    View details for Web of Science ID 000373027400039

  • Communication in Neural Circuits: Tools, Opportunities, and Challenges CELL Lerner, T. N., Ye, L., Deisseroth, K. 2016; 164 (6): 1136-1150

    Abstract

    Communication, the effective delivery of information, is fundamental to life across all scales and species. Nervous systems (by necessity) may be most specifically adapted among biological tissues for high rate and complexity of information transmitted, and thus, the properties of neural tissue and principles of its organization into circuits may illuminate capabilities and limitations of biological communication. Here, we consider recent developments in tools for studying neural circuits with particular attention to defining neuronal cell types by input and output information streams-i.e., by how they communicate. Complementing approaches that define cell types by virtue of genetic promoter/enhancer properties, this communication-based approach to defining cell types operationally by input/output (I/O) relationships links structure and function, resolves difficulties associated with single-genetic-feature definitions, leverages technology for observing and testing significance of precisely these I/O relationships in intact brains, and maps onto processes through which behavior may be adapted during development, experience, and evolution.

    View details for DOI 10.1016/j.cell.2016.02.027

    View details for Web of Science ID 000372784900012

  • Illuminating next-generation brain therapies. Nature neuroscience Ferenczi, E., Deisseroth, K. 2016; 19 (3): 414-6

    View details for DOI 10.1038/nn.4232

    View details for PubMedID 26780510

    View details for PubMedCentralID PMC4775735

  • Multiplexed Intact-Tissue Transcriptional Analysis at Cellular Resolution CELL Sylwestrak, E. L., Rajasethupathy, P., Wright, M. A., Jaffe, A., Deisseroth, K. 2016; 164 (4): 792-804

    Abstract

    In recently developed approaches for high-resolution imaging within intact tissue, molecular characterization over large volumes has been largely restricted to labeling of proteins. But volumetric nucleic acid labeling may represent a far greater scientific and clinical opportunity, enabling detection of not only diverse coding RNA variants but also non-coding RNAs. Moreover, scaling immunohistochemical detection to large tissue volumes has limitations due to high cost, limited renewability/availability, and restricted multiplexing capability of antibody labels. With the goal of versatile, high-content, and scalable molecular phenotyping of intact tissues, we developed a method using carbodiimide-based chemistry to stably retain RNAs in clarified tissue, coupled with amplification tools for multiplexed detection. The resulting technology enables robust measurement of activity-dependent transcriptional signatures, cell-identity markers, and diverse non-coding RNAs in rodent and human tissue volumes. The growing set of validated probes is deposited in an online resource for nucleating related developments from across the scientific community.

    View details for DOI 10.1016/j.cell.2016.01.038

    View details for Web of Science ID 000369998300023

    View details for PubMedID 26871636

    View details for PubMedCentralID PMC4775740

  • Structural foundations of optogenetics: Determinants of channelrhodopsin ion selectivity PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Berndt, A., Lee, S. Y., Wietek, J., Ramakrishnan, C., Steinberg, E. E., Rashid, A. J., Kim, H., Park, S., Santoro, A., Frankland, P. W., Iyer, S. M., Pak, S., Ahrlund-Richter, S., Delp, S. L., Malenka, R. C., Josselyn, S. A., Carlen, M., Hegemann, P., Deisseroth, K. 2016; 113 (4): 822-829

    Abstract

    The structure-guided design of chloride-conducting channelrhodopsins has illuminated mechanisms underlying ion selectivity of this remarkable family of light-activated ion channels. The first generation of chloride-conducting channelrhodopsins, guided in part by development of a structure-informed electrostatic model for pore selectivity, included both the introduction of amino acids with positively charged side chains into the ion conduction pathway and the removal of residues hypothesized to support negatively charged binding sites for cations. Engineered channels indeed became chloride selective, reversing near -65 mV and enabling a new kind of optogenetic inhibition; however, these first-generation chloride-conducting channels displayed small photocurrents and were not tested for optogenetic inhibition of behavior. Here we report the validation and further development of the channelrhodopsin pore model via crystal structure-guided engineering of next-generation light-activated chloride channels (iC++) and a bistable variant (SwiChR++) with net photocurrents increased more than 15-fold under physiological conditions, reversal potential further decreased by another ∼ 15 mV, inhibition of spiking faithfully tracking chloride gradients and intrinsic cell properties, strong expression in vivo, and the initial microbial opsin channel-inhibitor-based control of freely moving behavior. We further show that inhibition by light-gated chloride channels is mediated mainly by shunting effects, which exert optogenetic control much more efficiently than the hyperpolarization induced by light-activated chloride pumps. The design and functional features of these next-generation chloride-conducting channelrhodopsins provide both chronic and acute timescale tools for reversible optogenetic inhibition, confirm fundamental predictions of the ion selectivity model, and further elucidate electrostatic and steric structure-function relationships of the light-gated pore.

    View details for DOI 10.1073/pnas.1523341113

    View details for Web of Science ID 000368617900023

    View details for PubMedCentralID PMC4743797

  • NEURAL CIRCUITS Prefrontal cortical regulation of brainwide circuit dynamics and reward-related behavior SCIENCE Ferenczi, E. A., Zalocusky, K. A., Liston, C., Grosenick, L., Warden, M. R., Amatya, D., Katovich, K., Mehta, H., Patenaude, B., Ramakrishnan, C., Kalanithi, P., Etkin, A., Knutson, B., Glover, G. H., Deisseroth, K. 2016; 351 (6268): 41-U59

    Abstract

    Motivation for reward drives adaptive behaviors, whereas impairment of reward perception and experience (anhedonia) can contribute to psychiatric diseases, including depression and schizophrenia. We sought to test the hypothesis that the medial prefrontal cortex (mPFC) controls interactions among specific subcortical regions that govern hedonic responses. By using optogenetic functional magnetic resonance imaging to locally manipulate but globally visualize neural activity in rats, we found that dopamine neuron stimulation drives striatal activity, whereas locally increased mPFC excitability reduces this striatal response and inhibits the behavioral drive for dopaminergic stimulation. This chronic mPFC overactivity also stably suppresses natural reward-motivated behaviors and induces specific new brainwide functional interactions, which predict the degree of anhedonia in individuals. These findings describe a mechanism by which mPFC modulates expression of reward-seeking behavior, by regulating the dynamical interactions between specific distant subcortical regions.

    View details for DOI 10.1126/science.aac9698

    View details for Web of Science ID 000367364200035

    View details for PubMedCentralID PMC4772156

  • Optogenetic approaches addressing extracellular modulation of neural excitability. Scientific reports Ferenczi, E. A., Vierock, J., Atsuta-Tsunoda, K., Tsunoda, S. P., Ramakrishnan, C., Gorini, C., Thompson, K., Lee, S. Y., Berndt, A., Perry, C., Minniberger, S., Vogt, A., Mattis, J., Prakash, R., Delp, S., Deisseroth, K., Hegemann, P. 2016; 6: 23947-?

    Abstract

    The extracellular ionic environment in neural tissue has the capacity to influence, and be influenced by, natural bouts of neural activity. We employed optogenetic approaches to control and investigate these interactions within and between cells, and across spatial scales. We began by developing a temporally precise means to study microdomain-scale interactions between extracellular protons and acid-sensing ion channels (ASICs). By coupling single-component proton-transporting optogenetic tools to ASICs to create two-component optogenetic constructs (TCOs), we found that acidification of the local extracellular membrane surface by a light-activated proton pump recruited a slow inward ASIC current, which required molecular proximity of the two components on the membrane. To elicit more global effects of activity modulation on 'bystander' neurons not under direct control, we used densely-expressed depolarizing (ChR2) or hyperpolarizing (eArch3.0, eNpHR3.0) tools to create a slow non-synaptic membrane current in bystander neurons, which matched the current direction seen in the directly modulated neurons. Extracellular protons played contributory role but were insufficient to explain the entire bystander effect, suggesting the recruitment of other mechanisms. Together, these findings present a new approach to the engineering of multicomponent optogenetic tools to manipulate ionic microdomains, and probe the complex neuronal-extracellular space interactions that regulate neural excitability.

    View details for DOI 10.1038/srep23947

    View details for PubMedID 27045897

  • SPED Light Sheet Microscopy: Fast Mapping of Biological System Structure and Function CELL Tomer, R., Lovett-Barron, M., Kauvar, I., Andalman, A., Burns, V. M., Sankaran, S., Grosenick, L., Broxton, M., Yang, S., Deisseroth, K. 2015; 163 (7): 1796-1806

    Abstract

    The goal of understanding living nervous systems has driven interest in high-speed and large field-of-view volumetric imaging at cellular resolution. Light sheet microscopy approaches have emerged for cellular-resolution functional brain imaging in small organisms such as larval zebrafish, but remain fundamentally limited in speed. Here, we have developed SPED light sheet microscopy, which combines large volumetric field-of-view via an extended depth of field with the optical sectioning of light sheet microscopy, thereby eliminating the need to physically scan detection objectives for volumetric imaging. SPED enables scanning of thousands of volumes-per-second, limited only by camera acquisition rate, through the harnessing of optical mechanisms that normally result in unwanted spherical aberrations. We demonstrate capabilities of SPED microscopy by performing fast sub-cellular resolution imaging of CLARITY mouse brains and cellular-resolution volumetric Ca(2+) imaging of entire zebrafish nervous systems. Together, SPED light sheet methods enable high-speed cellular-resolution volumetric mapping of biological system structure and function.

    View details for DOI 10.1016/j.cell.2015.11.061

    View details for Web of Science ID 000366854200024

    View details for PubMedID 26687363

  • Extended field-of-view and increased-signal 3D holographic illumination with time-division multiplexing OPTICS EXPRESS Yang, S. J., Allen, W. E., Kauvar, I., Andalman, A. S., Young, N. P., Kim, C. K., Marshel, J. H., Wetzstein, G., Deisseroth, K. 2015; 23 (25): 32573-32581

    Abstract

    Phase spatial light modulators (SLMs) are widely used for generating multifocal three-dimensional (3D) illumination patterns, but these are limited to a field of view constrained by the pixel count or size of the SLM. Further, with two-photon SLM-based excitation, increasing the number of focal spots penalizes the total signal linearly--requiring more laser power than is available or can be tolerated by the sample. Here we analyze and demonstrate a method of using galvanometer mirrors to time-sequentially reposition multiple 3D holograms, both extending the field of view and increasing the total time-averaged two-photon signal. We apply our approach to 3D two-photon in vivo neuronal calcium imaging.

    View details for DOI 10.1364/OE.23.032573

    View details for Web of Science ID 000366687200093

    View details for PubMedID 26699047

    View details for PubMedCentralID PMC4775739

  • Basomedial amygdala mediates top-down control of anxiety and fear. Nature Adhikari, A., Lerner, T. N., Finkelstein, J., Pak, S., Jennings, J. H., Davidson, T. J., Ferenczi, E., Gunaydin, L. A., Mirzabekov, J. J., Ye, L., Kim, S., Lei, A., Deisseroth, K. 2015; 527 (7577): 179-185

    Abstract

    Anxiety-related conditions are among the most difficult neuropsychiatric diseases to treat pharmacologically, but respond to cognitive therapies. There has therefore been interest in identifying relevant top-down pathways from cognitive control regions in medial prefrontal cortex (mPFC). Identification of such pathways could contribute to our understanding of the cognitive regulation of affect, and provide pathways for intervention. Previous studies have suggested that dorsal and ventral mPFC subregions exert opposing effects on fear, as do subregions of other structures. However, precise causal targets for top-down connections among these diverse possibilities have not been established. Here we show that the basomedial amygdala (BMA) represents the major target of ventral mPFC in amygdala in mice. Moreover, BMA neurons differentiate safe and aversive environments, and BMA activation decreases fear-related freezing and high-anxiety states. Lastly, we show that the ventral mPFC-BMA projection implements top-down control of anxiety state and learned freezing, both at baseline and in stress-induced anxiety, defining a broadly relevant new top-down behavioural regulation pathway.

    View details for DOI 10.1038/nature15698

    View details for PubMedID 26536109

  • Projections from neocortex mediate top-down control of memory retrieval. Nature Rajasethupathy, P., Sankaran, S., Marshel, J. H., Kim, C. K., Ferenczi, E., Lee, S. Y., Berndt, A., Ramakrishnan, C., Jaffe, A., Lo, M., Liston, C., Deisseroth, K. 2015; 526 (7575): 653-659

    Abstract

    Top-down prefrontal cortex inputs to the hippocampus have been hypothesized to be important in memory consolidation, retrieval, and the pathophysiology of major psychiatric diseases; however, no such direct projections have been identified and functionally described. Here we report the discovery of a monosynaptic prefrontal cortex (predominantly anterior cingulate) to hippocampus (CA3 to CA1 region) projection in mice, and find that optogenetic manipulation of this projection (here termed AC-CA) is capable of eliciting contextual memory retrieval. To explore the network mechanisms of this process, we developed and applied tools to observe cellular-resolution neural activity in the hippocampus while stimulating AC-CA projections during memory retrieval in mice behaving in virtual-reality environments. Using this approach, we found that learning drives the emergence of a sparse class of neurons in CA2/CA3 that are highly correlated with the local network and that lead synchronous population activity events; these neurons are then preferentially recruited by the AC-CA projection during memory retrieval. These findings reveal a sparsely implemented memory retrieval mechanism in the hippocampus that operates via direct top-down prefrontal input, with implications for the patterning and storage of salient memory representations.

    View details for DOI 10.1038/nature15389

    View details for PubMedID 26436451

  • Thalamic control of sensory selection in divided attention NATURE Wimmer, R. D., Schmitt, L. I., Davidson, T. J., Nakajima, M., Deisseroth, K., Halassa, M. M. 2015; 526 (7575): 705-709

    Abstract

    How the brain selects appropriate sensory inputs and suppresses distractors is unknown. Given the well-established role of the prefrontal cortex (PFC) in executive function, its interactions with sensory cortical areas during attention have been hypothesized to control sensory selection. To test this idea and, more generally, dissect the circuits underlying sensory selection, we developed a cross-modal divided-attention task in mice that allowed genetic access to this cognitive process. By optogenetically perturbing PFC function in a temporally precise window, the ability of mice to select appropriately between conflicting visual and auditory stimuli was diminished. Equivalent sensory thalamocortical manipulations showed that behaviour was causally dependent on PFC interactions with the sensory thalamus, not sensory cortex. Consistent with this notion, we found neurons of the visual thalamic reticular nucleus (visTRN) to exhibit PFC-dependent changes in firing rate predictive of the modality selected. visTRN activity was causal to performance as confirmed by bidirectional optogenetic manipulations of this subnetwork. Using a combination of electrophysiology and intracellular chloride photometry, we demonstrated that visTRN dynamically controls visual thalamic gain through feedforward inhibition. Our experiments introduce a new subcortical model of sensory selection, in which the PFC biases thalamic reticular subnetworks to control thalamic sensory gain, selecting appropriate inputs for further processing.

    View details for DOI 10.1038/nature15398

    View details for Web of Science ID 000363832100049

    View details for PubMedID 26503050

    View details for PubMedCentralID PMC4626291

  • Optogenetics: 10 years of microbial opsins in neuroscience NATURE NEUROSCIENCE Deisseroth, K. 2015; 18 (9): 1213-1225

    Abstract

    Over the past 10 years, the development and convergence of microbial opsin engineering, modular genetic methods for cell-type targeting and optical strategies for guiding light through tissue have enabled versatile optical control of defined cells in living systems, defining modern optogenetics. Despite widespread recognition of the importance of spatiotemporally precise causal control over cellular signaling, for nearly the first half (2005-2009) of this 10-year period, as optogenetics was being created, there were difficulties in implementation, few publications and limited biological findings. In contrast, the ensuing years have witnessed a substantial acceleration in the application domain, with the publication of thousands of discoveries and insights into the function of nervous systems and beyond. This Historical Commentary reflects on the scientific landscape of this decade-long transition.

    View details for Web of Science ID 000360292600008

    View details for PubMedID 26308982

  • OPTOGENETICS. Expanding the optogenetics toolkit. Science Berndt, A., Deisseroth, K. 2015; 349 (6248): 590-591

    View details for DOI 10.1126/science.aac7889

    View details for PubMedID 26250674

  • Intact-Brain Analyses Reveal Distinct Information Carried by SNc Dopamine Subcircuits CELL Lerner, T. N., Shilyansky, C., Davidson, T. J., Evans, K. E., Beier, K. T., Zalocusky, K. A., Crow, A. K., Malenka, R. C., Luo, L., Tomer, R., Deisseroth, K. 2015; 162 (3): 635-647

    Abstract

    Recent progress in understanding the diversity of midbrain dopamine neurons has highlighted the importance--and the challenges--of defining mammalian neuronal cell types. Although neurons may be best categorized using inclusive criteria spanning biophysical properties, wiring of inputs, wiring of outputs, and activity during behavior, linking all of these measurements to cell types within the intact brains of living mammals has been difficult. Here, using an array of intact-brain circuit interrogation tools, including CLARITY, COLM, optogenetics, viral tracing, and fiber photometry, we explore the diversity of dopamine neurons within the substantia nigra pars compacta (SNc). We identify two parallel nigrostriatal dopamine neuron subpopulations differing in biophysical properties, input wiring, output wiring to dorsomedial striatum (DMS) versus dorsolateral striatum (DLS), and natural activity patterns during free behavior. Our results reveal independently operating nigrostriatal information streams, with implications for understanding the logic of dopaminergic feedback circuits and the diversity of mammalian neuronal cell types.

    View details for DOI 10.1016/j.cell.2015.07.014

    View details for Web of Science ID 000358801800021

  • Optogenetics and the circuit dynamics of psychiatric disease. JAMA Deisseroth, K., Etkin, A., Malenka, R. C. 2015; 313 (20): 2019-2020

    View details for DOI 10.1001/jama.2015.2544

    View details for PubMedID 25974025

  • The BRAIN Initiative: developing technology to catalyse neuroscience discovery PHILOSOPHICAL TRANSACTIONS OF THE ROYAL SOCIETY B-BIOLOGICAL SCIENCES Jorgenson, L. A., Newsome, W. T., Anderson, D. J., Bargmann, C. I., Brown, E. N., Deisseroth, K., Donoghue, J. P., Hudson, K. L., Ling, G. S., MacLeish, P. R., Marder, E., Normann, R. A., Sanes, J. R., Schnitzer, M. J., Sejnowski, T. J., Tank, D. W., Tsien, R. Y., Ugurbil, K., Wingfield, J. C. 2015; 370 (1668): 8-19

    Abstract

    The evolution of the field of neuroscience has been propelled by the advent of novel technological capabilities, and the pace at which these capabilities are being developed has accelerated dramatically in the past decade. Capitalizing on this momentum, the United States launched the Brain Research through Advancing Innovative Neurotechnologies (BRAIN) Initiative to develop and apply new tools and technologies for revolutionizing our understanding of the brain. In this article, we review the scientific vision for this initiative set forth by the National Institutes of Health and discuss its implications for the future of neuroscience research. Particular emphasis is given to its potential impact on the mapping and study of neural circuits, and how this knowledge will transform our understanding of the complexity of the human brain and its diverse array of behaviours, perceptions, thoughts and emotions.

    View details for DOI 10.1098/rstb.2014.0164

    View details for Web of Science ID 000354071400002

    View details for PubMedID 25823863

    View details for PubMedCentralID PMC4387507

  • Closed-Loop and Activity-Guided Optogenetic Control NEURON Grosenick, L., Marshel, J. H., Deisseroth, K. 2015; 86 (1): 106-139

    Abstract

    Advances in optical manipulation and observation of neural activity have set the stage for widespread implementation of closed-loop and activity-guided optical control of neural circuit dynamics. Closing the loop optogenetically (i.e., basing optogenetic stimulation on simultaneously observed dynamics in a principled way) is a powerful strategy for causal investigation of neural circuitry. In particular, observing and feeding back the effects of circuit interventions on physiologically relevant timescales is valuable for directly testing whether inferred models of dynamics, connectivity, and causation are accurate in vivo. Here we highlight technical and theoretical foundations as well as recent advances and opportunities in this area, and we review in detail the known caveats and limitations of optogenetic experimentation in the context of addressing these challenges with closed-loop optogenetic control in behaving animals.

    View details for DOI 10.1016/j.neuron.2015.03.034

    View details for Web of Science ID 000352552900017

    View details for PubMedID 25856490

  • Simultaneous cellular-resolution optical perturbation and imaging of place cell firing fields NATURE NEUROSCIENCE Rickgauer, J. P., Deisseroth, K., Tank, D. W. 2014; 17 (12): 1816-1824

    Abstract

    Linking neural microcircuit function to emergent properties of the mammalian brain requires fine-scale manipulation and measurement of neural activity during behavior, where each neuron's coding and dynamics can be characterized. We developed an optical method for simultaneous cellular-resolution stimulation and large-scale recording of neuronal activity in behaving mice. Dual-wavelength two-photon excitation allowed largely independent functional imaging with a green fluorescent calcium sensor (GCaMP3, λ = 920 ± 6 nm) and single-neuron photostimulation with a red-shifted optogenetic probe (C1V1, λ = 1,064 ± 6 nm) in neurons coexpressing the two proteins. We manipulated task-modulated activity in individual hippocampal CA1 place cells during spatial navigation in a virtual reality environment, mimicking natural place-field activity, or 'biasing', to reveal subthreshold dynamics. Notably, manipulating single place-cell activity also affected activity in small groups of other place cells that were active around the same time in the task, suggesting a functional role for local place cell interactions in shaping firing fields.

    View details for DOI 10.1038/nn.3866

    View details for Web of Science ID 000345484000031

    View details for PubMedID 25402854

    View details for PubMedCentralID PMC4459599

  • Advanced CLARITY for rapid and high-resolution imaging of intact tissues NATURE PROTOCOLS Tomer, R., Ye, L., Hsueh, B., Deisseroth, K. 2014; 9 (7): 1682-1697

    Abstract

    CLARITY is a method for chemical transformation of intact biological tissues into a hydrogel-tissue hybrid, which becomes amenable to interrogation with light and macromolecular labels while retaining fine structure and native biological molecules. This emerging accessibility of information from large intact samples has created both new opportunities and new challenges. Here we describe protocols spanning multiple dimensions of the CLARITY workflow, ranging from simple, reliable and efficient lipid removal without electrophoretic instrumentation (passive CLARITY) to optimized objectives and integration with light-sheet optics (CLARITY-optimized light-sheet microscopy (COLM)) for accelerating data collection from clarified samples by several orders of magnitude while maintaining or increasing quality and resolution. The entire protocol takes from 7-28 d to complete for an adult mouse brain, including hydrogel embedding, full lipid removal, whole-brain antibody staining (which, if needed, accounts for 7-10 of the days), and whole-brain high-resolution imaging; timing within this window depends on the choice of lipid removal options, on the size of the tissue, and on the number and type of immunostaining rounds performed. This protocol has been successfully applied to the study of adult mouse, adult zebrafish and adult human brains, and it may find many other applications in the structural and molecular analysis of large assembled biological systems.

    View details for DOI 10.1038/nprot.2014.123

    View details for Web of Science ID 000338777400011

    View details for PubMedCentralID PMC4096681

  • Targeting cells with single vectors using multiple-feature Boolean logic. Nature methods Fenno, L. E., Mattis, J., Ramakrishnan, C., Hyun, M., Lee, S. Y., He, M., Tucciarone, J., Selimbeyoglu, A., Berndt, A., Grosenick, L., Zalocusky, K. A., Bernstein, H., Swanson, H., Perry, C., Diester, I., Boyce, F. M., Bass, C. E., Neve, R., Huang, Z. J., Deisseroth, K. 2014; 11 (7): 763-772

    Abstract

    Precisely defining the roles of specific cell types is an intriguing frontier in the study of intact biological systems and has stimulated the rapid development of genetically encoded tools for observation and control. However, targeting these tools with adequate specificity remains challenging: most cell types are best defined by the intersection of two or more features such as active promoter elements, location and connectivity. Here we have combined engineered introns with specific recombinases to achieve expression of genetically encoded tools that is conditional upon multiple cell-type features, using Boolean logical operations all governed by a single versatile vector. We used this approach to target intersectionally specified populations of inhibitory interneurons in mammalian hippocampus and neurons of the ventral tegmental area defined by both genetic and wiring properties. This flexible and modular approach may expand the application of genetically encoded interventional and observational tools for intact-systems biology.

    View details for DOI 10.1038/nmeth.2996

    View details for PubMedID 24908100

  • Natural neural projection dynamics underlying social behavior. Cell Gunaydin, L. A., Grosenick, L., Finkelstein, J. C., Kauvar, I. V., Fenno, L. E., Adhikari, A., Lammel, S., Mirzabekov, J. J., Airan, R. D., Zalocusky, K. A., Tye, K. M., Anikeeva, P., Malenka, R. C., Deisseroth, K. 2014; 157 (7): 1535-1551

    Abstract

    Social interaction is a complex behavior essential for many species and is impaired in major neuropsychiatric disorders. Pharmacological studies have implicated certain neurotransmitter systems in social behavior, but circuit-level understanding of endogenous neural activity during social interaction is lacking. We therefore developed and applied a new methodology, termed fiber photometry, to optically record natural neural activity in genetically and connectivity-defined projections to elucidate the real-time role of specified pathways in mammalian behavior. Fiber photometry revealed that activity dynamics of a ventral tegmental area (VTA)-to-nucleus accumbens (NAc) projection could encode and predict key features of social, but not novel object, interaction. Consistent with this observation, optogenetic control of cells specifically contributing to this projection was sufficient to modulate social behavior, which was mediated by type 1 dopamine receptor signaling downstream in the NAc. Direct observation of deep projection-specific activity in this way captures a fundamental and previously inaccessible dimension of mammalian circuit dynamics.

    View details for DOI 10.1016/j.cell.2014.05.017

    View details for PubMedID 24949967

    View details for PubMedCentralID PMC4123133

  • Structure-guided transformation of channelrhodopsin into a light-activated chloride channel. Science (New York, N.Y.) Berndt, A., Lee, S. Y., Ramakrishnan, C., Deisseroth, K. 2014; 344 (6182): 420-4

    Abstract

    Using light to silence electrical activity in targeted cells is a major goal of optogenetics. Available optogenetic proteins that directly move ions to achieve silencing are inefficient, pumping only a single ion per photon across the cell membrane rather than allowing many ions per photon to flow through a channel pore. Building on high-resolution crystal-structure analysis, pore vestibule modeling, and structure-guided protein engineering, we designed and characterized a class of channelrhodopsins (originally cation-conducting) converted into chloride-conducting anion channels. These tools enable fast optical inhibition of action potentials and can be engineered to display step-function kinetics for stable inhibition, outlasting light pulses and for orders-of-magnitude-greater light sensitivity of inhibited cells. The resulting family of proteins defines an approach to more physiological, efficient, and sensitive optogenetic inhibition.

    View details for DOI 10.1126/science.1252367

    View details for PubMedID 24763591

  • Circuit dynamics of adaptive and maladaptive behaviour NATURE Deisseroth, K. 2014; 505 (7483): 309-317

    Abstract

    The recent development of technologies for investigating specific components of intact biological systems has allowed elucidation of the neural circuitry underlying adaptive and maladaptive behaviours. Investigators are now able to observe and control, with high spatio-temporal resolution, structurally defined intact pathways along which electrical activity flows during and after the performance of complex behaviours. These investigations have revealed that control of projection-specific dynamics is well suited to modulating behavioural patterns that are relevant to a broad range of psychiatric diseases. Structural dynamics principles have emerged to provide diverse, unexpected and causal insights into the operation of intact and diseased nervous systems, linking form and function in the brain.

    View details for DOI 10.1038/nature12982

    View details for Web of Science ID 000329621800028

    View details for PubMedID 24429629

    View details for PubMedCentralID PMC4069282

  • Optical Neural Interfaces ANNUAL REVIEW OF BIOMEDICAL ENGINEERING, VOL 16 Warden, M. R., Cardin, J. A., Deisseroth, K. 2014; 16: 103-129

    Abstract

    Genetically encoded optical actuators and indicators have changed the landscape of neuroscience, enabling targetable control and readout of specific components of intact neural circuits in behaving animals. Here, we review the development of optical neural interfaces, focusing on hardware designed for optical control of neural activity, integrated optical control and electrical readout, and optical readout of population and single-cell neural activity in freely moving mammals.

    View details for DOI 10.1146/annurev-bioeng-071813-104733

    View details for Web of Science ID 000348433000005

    View details for PubMedID 25014785

    View details for PubMedCentralID PMC4163158

  • Causal interactions between fronto-parietal central executive and default-mode networks in humans PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Chen, A. C., Oathes, D. J., Chang, C., Bradley, T., Zhou, Z., Williams, L. M., Glover, G. H., Deisseroth, K., Etkin, A. 2013; 110 (49): 19944-19949

    Abstract

    Information processing during human cognitive and emotional operations is thought to involve the dynamic interplay of several large-scale neural networks, including the fronto-parietal central executive network (CEN), cingulo-opercular salience network (SN), and the medial prefrontal-medial parietal default mode networks (DMN). It has been theorized that there is a causal neural mechanism by which the CEN/SN negatively regulate the DMN. Support for this idea has come from correlational neuroimaging studies; however, direct evidence for this neural mechanism is lacking. Here we undertook a direct test of this mechanism by combining transcranial magnetic stimulation (TMS) with functional MRI to causally excite or inhibit TMS-accessible prefrontal nodes within the CEN or SN and determine consequent effects on the DMN. Single-pulse excitatory stimulations delivered to only the CEN node induced negative DMN connectivity with the CEN and SN, consistent with the CEN/SN's hypothesized negative regulation of the DMN. Conversely, low-frequency inhibitory repetitive TMS to the CEN node resulted in a shift of DMN signal from its normally low-frequency range to a higher frequency, suggesting disinhibition of DMN activity. Moreover, the CEN node exhibited this causal regulatory relationship primarily with the medial prefrontal portion of the DMN. These findings significantly advance our understanding of the causal mechanisms by which major brain networks normally coordinate information processing. Given that poorly regulated information processing is a hallmark of most neuropsychiatric disorders, these findings provide a foundation for ways to study network dysregulation and develop brain stimulation treatments for these disorders.

    View details for DOI 10.1073/pnas.1311772110

    View details for Web of Science ID 000327744900066

    View details for PubMedID 24248372

    View details for PubMedCentralID PMC3856839

  • Engineering Approaches to Illuminating Brain Structure and Dynamics NEURON Deisseroth, K., Schnitzer, M. J. 2013; 80 (3): 568-577

    Abstract

    Historical milestones in neuroscience have come in diverse forms, ranging from the resolution of specific biological mysteries via creative experimentation to broad technological advances allowing neuroscientists to ask new kinds of questions. The continuous development of tools is driven with a special necessity by the complexity, fragility, and inaccessibility of intact nervous systems, such that inventive technique development and application drawing upon engineering and the applied sciences has long been essential to neuroscience. Here we highlight recent technological directions in neuroscience spurred by progress in optical, electrical, mechanical, chemical, and biological engineering. These research areas are poised for rapid growth and will likely be central to the practice of neuroscience well into the future.

    View details for DOI 10.1016/j.neuron.2013.10.032

    View details for Web of Science ID 000326609900004

    View details for PubMedID 24183010

  • A causal link between prediction errors, dopamine neurons and learning NATURE NEUROSCIENCE Steinberg, E. E., Keiflin, R., Boivin, J. R., Witten, I. B., Deisseroth, K., Janak, P. H. 2013; 16 (7): 966-U248

    Abstract

    Situations in which rewards are unexpectedly obtained or withheld represent opportunities for new learning. Often, this learning includes identifying cues that predict reward availability. Unexpected rewards strongly activate midbrain dopamine neurons. This phasic signal is proposed to support learning about antecedent cues by signaling discrepancies between actual and expected outcomes, termed a reward prediction error. However, it is unknown whether dopamine neuron prediction error signaling and cue-reward learning are causally linked. To test this hypothesis, we manipulated dopamine neuron activity in rats in two behavioral procedures, associative blocking and extinction, that illustrate the essential function of prediction errors in learning. We observed that optogenetic activation of dopamine neurons concurrent with reward delivery, mimicking a prediction error, was sufficient to cause long-lasting increases in cue-elicited reward-seeking behavior. Our findings establish a causal role for temporally precise dopamine neuron signaling in cue-reward learning, bridging a critical gap between experimental evidence and influential theoretical frameworks.

    View details for DOI 10.1038/nn.3413

    View details for Web of Science ID 000321180900032

    View details for PubMedID 23708143

    View details for PubMedCentralID PMC3705924

  • Repeated Cortico-Striatal Stimulation Generates Persistent OCD-Like Behavior SCIENCE Ahmari, S. E., Spellman, T., Douglass, N. L., Kheirbek, M. A., Simpson, H. B., Deisseroth, K., Gordon, J. A., Hen, R. 2013; 340 (6137): 1234-1239

    Abstract

    Although cortico-striato-thalamo-cortical (CSTC) circuit dysregulation is correlated with obsessive compulsive disorder (OCD), causation cannot be tested in humans. We used optogenetics in mice to simulate CSTC hyperactivation observed in OCD patients. Whereas acute orbitofrontal cortex (OFC)-ventromedial striatum (VMS) stimulation did not produce repetitive behaviors, repeated hyperactivation over multiple days generated a progressive increase in grooming, a mouse behavior related to OCD. Increased grooming persisted for 2 weeks after stimulation cessation. The grooming increase was temporally coupled with a progressive increase in light-evoked firing of postsynaptic VMS cells. Both increased grooming and evoked firing were reversed by chronic fluoxetine, a first-line OCD treatment. Brief but repeated episodes of abnormal circuit activity may thus set the stage for the development of persistent psychopathology.

    View details for DOI 10.1126/science.1234733

    View details for Web of Science ID 000319972800051

    View details for PubMedID 23744948

    View details for PubMedCentralID PMC3954809

  • CLARITY for mapping the nervous system. Nature methods Chung, K., Deisseroth, K. 2013; 10 (6): 508-513

    View details for DOI 10.1038/nmeth.2481

    View details for PubMedID 23722210

  • Structural and molecular interrogation of intact biological systems. Nature Chung, K., Wallace, J., Kim, S., Kalyanasundaram, S., Andalman, A. S., Davidson, T. J., Mirzabekov, J. J., Zalocusky, K. A., Mattis, J., Denisin, A. K., Pak, S., Bernstein, H., Ramakrishnan, C., Grosenick, L., Gradinaru, V., Deisseroth, K. 2013; 497 (7449): 332-337

    Abstract

    Obtaining high-resolution information from a complex system, while maintaining the global perspective needed to understand system function, represents a key challenge in biology. Here we address this challenge with a method (termed CLARITY) for the transformation of intact tissue into a nanoporous hydrogel-hybridized form (crosslinked to a three-dimensional network of hydrophilic polymers) that is fully assembled but optically transparent and macromolecule-permeable. Using mouse brains, we show intact-tissue imaging of long-range projections, local circuit wiring, cellular relationships, subcellular structures, protein complexes, nucleic acids and neurotransmitters. CLARITY also enables intact-tissue in situ hybridization, immunohistochemistry with multiple rounds of staining and de-staining in non-sectioned tissue, and antibody labelling throughout the intact adult mouse brain. Finally, we show that CLARITY enables fine structural analysis of clinical samples, including non-sectioned human tissue from a neuropsychiatric-disease setting, establishing a path for the transmutation of human tissue into a stable, intact and accessible form suitable for probing structural and molecular underpinnings of physiological function and disease.

    View details for DOI 10.1038/nature12107

    View details for PubMedID 23575631

  • Hypothalamic Neurotensin Projections Promote Reward by Enhancing Glutamate Transmission in the VTA JOURNAL OF NEUROSCIENCE Kempadoo, K. A., Tourino, C., Cho, S. L., Magnani, F., Leinninger, G., Stuber, G. D., Zhang, F., Myers, M. G., Deisseroth, K., de Lecea, L., Bonci, A. 2013; 33 (18): 7618-?

    Abstract

    The lateral hypothalamus (LH) sends a dense glutamatergic and peptidergic projection to dopamine neurons in the ventral tegmental area (VTA), a cell group known to promote reinforcement and aspects of reward. The role of the LH to VTA projection in reward-seeking behavior can be informed by using optogenetic techniques to dissociate the actions of LH neurons from those of other descending forebrain inputs to the VTA. In the present study, we identify the effect of neurotensin (NT), one of the most abundant peptides in the LH to VTA projection, on excitatory synaptic transmission in the VTA and reward-seeking behavior. Mice displayed robust intracranial self-stimulation of LH to VTA fibers, an operant behavior mediated by NT 1 receptors (Nts1) and NMDA receptors. Whole-cell patch-clamp recordings of VTA dopamine neurons demonstrated that NT (10 nm) potentiated NMDA-mediated EPSCs via Nts1. Results suggest that NT release from the LH into the VTA activates Nts1, thereby potentiating NMDA-mediated EPSCs and promoting reward. The striking behavioral and electrophysiological effects of NT and glutamate highlight the LH to VTA pathway as an important component of reward.

    View details for DOI 10.1523/JNEUROSCI.2588-12.2013

    View details for Web of Science ID 000318420400002

    View details for PubMedID 23637156

  • Diverging neural pathways assemble a behavioural state from separable features in anxiety NATURE Kim, S., Adhikari, A., Lee, S. Y., Marshel, J. H., Kim, C. K., Mallory, C. S., Lo, M., Pak, S., Mattis, J., Lim, B. K., Malenka, R. C., Warden, M. R., Neve, R., Tye, K. M., Deisseroth, K. 2013; 496 (7444): 219-223

    Abstract

    Behavioural states in mammals, such as the anxious state, are characterized by several features that are coordinately regulated by diverse nervous system outputs, ranging from behavioural choice patterns to changes in physiology (in anxiety, exemplified respectively by risk-avoidance and respiratory rate alterations). Here we investigate if and how defined neural projections arising from a single coordinating brain region in mice could mediate diverse features of anxiety. Integrating behavioural assays, in vivo and in vitro electrophysiology, respiratory physiology and optogenetics, we identify a surprising new role for the bed nucleus of the stria terminalis (BNST) in the coordinated modulation of diverse anxiety features. First, two BNST subregions were unexpectedly found to exert opposite effects on the anxious state: oval BNST activity promoted several independent anxious state features, whereas anterodorsal BNST-associated activity exerted anxiolytic influence for the same features. Notably, we found that three distinct anterodorsal BNST efferent projections-to the lateral hypothalamus, parabrachial nucleus and ventral tegmental area-each implemented an independent feature of anxiolysis: reduced risk-avoidance, reduced respiratory rate, and increased positive valence, respectively. Furthermore, selective inhibition of corresponding circuit elements in freely moving mice showed opposing behavioural effects compared with excitation, and in vivo recordings during free behaviour showed native spiking patterns in anterodorsal BNST neurons that differentiated safe and anxiogenic environments. These results demonstrate that distinct BNST subregions exert opposite effects in modulating anxiety, establish separable anxiolytic roles for different anterodorsal BNST projections, and illustrate circuit mechanisms underlying selection of features for the assembly of the anxious state.

    View details for DOI 10.1038/nature12018

    View details for PubMedID 23515158

  • Making Waves: Initiation and Propagation of Corticothalamic Ca2+ Waves In Vivo NEURON Stroh, A., Adelsberger, H., Groh, A., Ruehlmann, C., Fischer, S., Schierloh, A., Deisseroth, K., Konnerth, A. 2013; 77 (6): 1136-1150

    Abstract

    Corticothalamic slow oscillations of neuronal activity determine internal brain states. At least in the cortex, the electrical activity is associated with large neuronal Ca(2+) transients. Here we implemented an optogenetic approach to explore causal features of the generation of slow oscillation-associated Ca(2+) waves in the in vivo mouse brain. We demonstrate that brief optogenetic stimulation (3-20 ms) of a local group of layer 5 cortical neurons is sufficient for the induction of global brain Ca(2+) waves. These Ca(2+) waves are evoked in an all-or-none manner, exhibit refractoriness during repetitive stimulation, and propagate over long distances. By local optogenetic stimulation, we demonstrate that evoked Ca(2+) waves initially invade the cortex, followed by a secondary recruitment of the thalamus. Together, our results establish that synchronous activity in a small cluster of layer 5 cortical neurons can initiate a global neuronal wave of activity suited for long-range corticothalamic integration.

    View details for DOI 10.1016/j.neuron.2013.01.031

    View details for Web of Science ID 000316645000014

  • Dopamine neurons modulate neural encoding and expression of depression-related behaviour NATURE Tye, K. M., Mirzabekov, J. J., Warden, M. R., Ferenczi, E. A., Tsai, H., Finkelstein, J., Kim, S., Adhikari, A., Thompson, K. R., Andalman, A. S., Gunaydin, L. A., Witten, I. B., Deisseroth, K. 2013; 493 (7433): 537-?

    Abstract

    Major depression is characterized by diverse debilitating symptoms that include hopelessness and anhedonia. Dopamine neurons involved in reward and motivation are among many neural populations that have been hypothesized to be relevant, and certain antidepressant treatments, including medications and brain stimulation therapies, can influence the complex dopamine system. Until now it has not been possible to test this hypothesis directly, even in animal models, as existing therapeutic interventions are unable to specifically target dopamine neurons. Here we investigated directly the causal contributions of defined dopamine neurons to multidimensional depression-like phenotypes induced by chronic mild stress, by integrating behavioural, pharmacological, optogenetic and electrophysiological methods in freely moving rodents. We found that bidirectional control (inhibition or excitation) of specified midbrain dopamine neurons immediately and bidirectionally modulates (induces or relieves) multiple independent depression symptoms caused by chronic stress. By probing the circuit implementation of these effects, we observed that optogenetic recruitment of these dopamine neurons potently alters the neural encoding of depression-related behaviours in the downstream nucleus accumbens of freely moving rodents, suggesting that processes affecting depression symptoms may involve alterations in the neural encoding of action in limbic circuitry.

    View details for DOI 10.1038/nature11740

    View details for Web of Science ID 000313871400039

    View details for PubMedID 23235822

  • Optogenetic activation of an inhibitory network enhances feedforward functional connectivity in auditory cortex. Neuron. Hamilton, L. S., Sohl-Dickstein, J., Huth, A. G., Carels, V. M., Deisseroth, K., Bao, S. 2013
  • A unique population of ventral tegmental area neurons inhibits the lateral habenula to promote reward. Neuron. Stamatakis, A. M., Jennings, J. H., Ung, R. L., Blair, G. A., Weinberg, R. J., NEve, R. L., Deisseroth, K. 2013
  • Optogenetics. PNAS. Williams, Sarah, C. P., Deisseroth, K. 2013
  • Light microscopy mapping of connections in the intact brain. Trends in Cognitive Sciences. Kim, S., Chung, K., Deisseroth, K. 2013
  • Optogenetics in the behaving rat: integration of diverse new technologies in a vital animal model. Optogenetics. Zalocusky, K., Deisseroth, K. 2013
  • A prefrontal cortex-brainstem neuronal projection that controls response to behavioural challenge NATURE Warden, M. R., Selimbeyoglu, A., Mirzabekov, J. J., Lo, M., Thompson, K. R., Kim, S., Adhikari, A., Tye, K. M., Frank, L. M., Deisseroth, K. 2012; 492 (7429): 428-432

    Abstract

    The prefrontal cortex (PFC) is thought to participate in high-level control of the generation of behaviours (including the decision to execute actions); indeed, imaging and lesion studies in human beings have revealed that PFC dysfunction can lead to either impulsive states with increased tendency to initiate action, or to amotivational states characterized by symptoms such as reduced activity, hopelessness and depressed mood. Considering the opposite valence of these two phenotypes as well as the broad complexity of other tasks attributed to PFC, we sought to elucidate the PFC circuitry that favours effortful behavioural responses to challenging situations. Here we develop and use a quantitative method for the continuous assessment and control of active response to a behavioural challenge, synchronized with single-unit electrophysiology and optogenetics in freely moving rats. In recording from the medial PFC (mPFC), we observed that many neurons were not simply movement-related in their spike-firing patterns but instead were selectively modulated from moment to moment, according to the animal's decision to act in a challenging situation. Surprisingly, we next found that direct activation of principal neurons in the mPFC had no detectable causal effect on this behaviour. We tested whether this behaviour could be causally mediated by only a subclass of mPFC cells defined by specific downstream wiring. Indeed, by leveraging optogenetic projection-targeting to control cells with specific efferent wiring patterns, we found that selective activation of those mPFC cells projecting to the brainstem dorsal raphe nucleus (DRN), a serotonergic nucleus implicated in major depressive disorder, induced a profound, rapid and reversible effect on selection of the active behavioural state. These results may be of importance in understanding the neural circuitry underlying normal and pathological patterns of action selection and motivation in behaviour.

    View details for DOI 10.1038/nature11617

    View details for Web of Science ID 000312488200056

    View details for PubMedID 23160494

  • Crystal structure of the channelrhodopsin light-gated cation channel NATURE Kato, H. E., Zhang, F., Yizhar, O., Ramakrishnan, C., Nishizawa, T., Hirata, K., Ito, J., Aita, Y., Tsukazaki, T., Hayashi, S., Hegemann, P., Maturana, A. D., Ishitani, R., Deisseroth, K., Nureki, O. 2012; 482 (7385): 369-U115

    Abstract

    Channelrhodopsins (ChRs) are light-gated cation channels derived from algae that have shown experimental utility in optogenetics; for example, neurons expressing ChRs can be optically controlled with high temporal precision within systems as complex as freely moving mammals. Although ChRs have been broadly applied to neuroscience research, little is known about the molecular mechanisms by which these unusual and powerful proteins operate. Here we present the crystal structure of a ChR (a C1C2 chimaera between ChR1 and ChR2 from Chlamydomonas reinhardtii) at 2.3 Å resolution. The structure reveals the essential molecular architecture of ChRs, including the retinal-binding pocket and cation conduction pathway. This integration of structural and electrophysiological analyses provides insight into the molecular basis for the remarkable function of ChRs, and paves the way for the precise and principled design of ChR variants with novel properties.

    View details for DOI 10.1038/nature10870

    View details for Web of Science ID 000300287100039

    View details for PubMedID 22266941

  • The Microbial Opsin Family of Optogenetic Tools CELL Zhang, F., Vierock, J., Yizhar, O., Fenno, L. E., Tsunoda, S., Kianianmomeni, A., Prigge, M., Berndt, A., Cushman, J., Polle, J., Magnuson, J., Hegemann, P., Deisseroth, K. 2011; 147 (7): 1446-1457

    Abstract

    The capture and utilization of light is an exquisitely evolved process. The single-component microbial opsins, although more limited than multicomponent cascades in processing, display unparalleled compactness and speed. Recent advances in understanding microbial opsins have been driven by molecular engineering for optogenetics and by comparative genomics. Here we provide a Primer on these light-activated ion channels and pumps, describe a group of opsins bridging prior categories, and explore the convergence of molecular engineering and genomic discovery for the utilization and understanding of these remarkable molecular machines.

    View details for DOI 10.1016/j.cell.2011.12.004

    View details for Web of Science ID 000298403400011

    View details for PubMedID 22196724

  • Neocortical excitation/inhibition balance in information processing and social dysfunction NATURE Yizhar, O., Fenno, L. E., Prigge, M., Schneider, F., Davidson, T. J., O'Shea, D. J., Sohal, V. S., Goshen, I., Finkelstein, J., Paz, J. T., Stehfest, K., Fudim, R., Ramakrishnan, C., Huguenard, J. R., Hegemann, P., Deisseroth, K. 2011; 477 (7363): 171-178

    Abstract

    Severe behavioural deficits in psychiatric diseases such as autism and schizophrenia have been hypothesized to arise from elevations in the cellular balance of excitation and inhibition (E/I balance) within neural microcircuitry. This hypothesis could unify diverse streams of pathophysiological and genetic evidence, but has not been susceptible to direct testing. Here we design and use several novel optogenetic tools to causally investigate the cellular E/I balance hypothesis in freely moving mammals, and explore the associated circuit physiology. Elevation, but not reduction, of cellular E/I balance within the mouse medial prefrontal cortex was found to elicit a profound impairment in cellular information processing, associated with specific behavioural impairments and increased high-frequency power in the 30-80 Hz range, which have both been observed in clinical conditions in humans. Consistent with the E/I balance hypothesis, compensatory elevation of inhibitory cell excitability partially rescued social deficits caused by E/I balance elevation. These results provide support for the elevated cellular E/I balance hypothesis of severe neuropsychiatric disease-related symptoms.

    View details for DOI 10.1038/nature10360

    View details for Web of Science ID 000294603900027

    View details for PubMedID 21796121

  • Optogenetics in Neural Systems NEURON Yizhar, O., Fenno, L. E., Davidson, T. J., Mogri, M., Deisseroth, K. 2011; 71 (1): 9-34

    Abstract

    Both observational and perturbational technologies are essential for advancing the understanding of brain function and dysfunction. But while observational techniques have greatly advanced in the last century, techniques for perturbation that are matched to the speed and heterogeneity of neural systems have lagged behind. The technology of optogenetics represents a step toward addressing this disparity. Reliable and targetable single-component tools (which encompass both light sensation and effector function within a single protein) have enabled versatile new classes of investigation in the study of neural systems. Here we provide a primer on the application of optogenetics in neuroscience, focusing on the single-component tools and highlighting important problems, challenges, and technical considerations.

    View details for DOI 10.1016/j.neuron.2011.06.004

    View details for PubMedID 21745635

  • Amygdala circuitry mediating reversible and bidirectional control of anxiety NATURE Tye, K. M., Prakash, R., Kim, S., Fenno, L. E., Grosenick, L., Zarabi, H., Thompson, K. R., Gradinaru, V., Ramakrishnan, C., Deisseroth, K. 2011; 471 (7338): 358-362

    Abstract

    Anxiety--a sustained state of heightened apprehension in the absence of immediate threat--becomes severely debilitating in disease states. Anxiety disorders represent the most common of psychiatric diseases (28% lifetime prevalence) and contribute to the aetiology of major depression and substance abuse. Although it has been proposed that the amygdala, a brain region important for emotional processing, has a role in anxiety, the neural mechanisms that control anxiety remain unclear. Here we explore the neural circuits underlying anxiety-related behaviours by using optogenetics with two-photon microscopy, anxiety assays in freely moving mice, and electrophysiology. With the capability of optogenetics to control not only cell types but also specific connections between cells, we observed that temporally precise optogenetic stimulation of basolateral amygdala (BLA) terminals in the central nucleus of the amygdala (CeA)--achieved by viral transduction of the BLA with a codon-optimized channelrhodopsin followed by restricted illumination in the downstream CeA--exerted an acute, reversible anxiolytic effect. Conversely, selective optogenetic inhibition of the same projection with a third-generation halorhodopsin (eNpHR3.0) increased anxiety-related behaviours. Importantly, these effects were not observed with direct optogenetic control of BLA somata, possibly owing to recruitment of antagonistic downstream structures. Together, these results implicate specific BLA-CeA projections as critical circuit elements for acute anxiety control in the mammalian brain, and demonstrate the importance of optogenetically targeting defined projections, beyond simply targeting cell types, in the study of circuit function relevant to neuropsychiatric disease.

    View details for DOI 10.1038/nature09820

    View details for Web of Science ID 000288444000041

    View details for PubMedID 21389985

    View details for PubMedCentralID PMC3154022

  • Optogenetics NATURE METHODS Deisseroth, K. 2011; 8 (1): 26-29

    View details for DOI 10.1038/NMETH.F.324

    View details for Web of Science ID 000285712000006

    View details for PubMedID 21191368

  • The Development and Application of Optogenetics ANNUAL REVIEW OF NEUROSCIENCE, VOL 34 Fenno, L., Yizhar, O., Deisseroth, K. 2011; 34: 389-412

    Abstract

    Genetically encoded, single-component optogenetic tools have made a significant impact on neuroscience, enabling specific modulation of selected cells within complex neural tissues. As the optogenetic toolbox contents grow and diversify, the opportunities for neuroscience continue to grow. In this review, we outline the development of currently available single-component optogenetic tools and summarize the application of various optogenetic tools in diverse model organisms.

    View details for DOI 10.1146/annurev-neuro-061010-113817

    View details for Web of Science ID 000293772100017

    View details for PubMedID 21692661

  • Cholinergic Interneurons Control Local Circuit Activity and Cocaine Conditioning SCIENCE Witten, I. B., Lin, S., Brodsky, M., Prakash, R., Diester, I., Anikeeva, P., Gradinaru, V., Ramakrishnan, C., Deisseroth, K. 2010; 330 (6011): 1677-1681

    Abstract

    Cholinergic neurons are widespread, and pharmacological modulation of acetylcholine receptors affects numerous brain processes, but such modulation entails side effects due to limitations in specificity for receptor type and target cell. As a result, causal roles of cholinergic neurons in circuits have been unclear. We integrated optogenetics, freely moving mammalian behavior, in vivo electrophysiology, and slice physiology to probe the cholinergic interneurons of the nucleus accumbens by direct excitation or inhibition. Despite representing less than 1% of local neurons, these cholinergic cells have dominant control roles, exerting powerful modulation of circuit activity. Furthermore, these neurons could be activated by cocaine, and silencing this drug-induced activity during cocaine exposure (despite the fact that the manipulation of the cholinergic interneurons was not aversive by itself) blocked cocaine conditioning in freely moving mammals.

    View details for DOI 10.1126/science.1193771

    View details for Web of Science ID 000285390500067

    View details for PubMedID 21164015

    View details for PubMedCentralID PMC3142356

  • Parvalbumin neurons and gamma rhythms enhance cortical circuit performance NATURE Sohal, V. S., Zhang, F., Yizhar, O., Deisseroth, K. 2009; 459 (7247): 698-702

    Abstract

    Synchronized oscillations and inhibitory interneurons have important and interconnected roles within cortical microcircuits. In particular, interneurons defined by the fast-spiking phenotype and expression of the calcium-binding protein parvalbumin have been suggested to be involved in gamma (30-80 Hz) oscillations, which are hypothesized to enhance information processing. However, because parvalbumin interneurons cannot be selectively controlled, definitive tests of their functional significance in gamma oscillations, and quantitative assessment of the impact of parvalbumin interneurons and gamma oscillations on cortical circuits, have been lacking despite potentially enormous significance (for example, abnormalities in parvalbumin interneurons may underlie altered gamma-frequency synchronization and cognition in schizophrenia and autism). Here we use a panel of optogenetic technologies in mice to selectively modulate multiple distinct circuit elements in neocortex, alone or in combination. We find that inhibiting parvalbumin interneurons suppresses gamma oscillations in vivo, whereas driving these interneurons (even by means of non-rhythmic principal cell activity) is sufficient to generate emergent gamma-frequency rhythmicity. Moreover, gamma-frequency modulation of excitatory input in turn was found to enhance signal transmission in neocortex by reducing circuit noise and amplifying circuit signals, including inputs to parvalbumin interneurons. As demonstrated here, optogenetics opens the door to a new kind of informational analysis of brain function, permitting quantitative delineation of the functional significance of individual elements in the emergent operation and function of intact neural circuitry.

    View details for DOI 10.1038/nature07991

    View details for Web of Science ID 000266608600043

    View details for PubMedID 19396159

  • Driving fast-spiking cells induces gamma rhythm and controls sensory responses NATURE Cardin, J. A., Carlen, M., Meletis, K., Knoblich, U., Zhang, F., Deisseroth, K., Tsai, L., Moore, C. I. 2009; 459 (7247): 663-U63

    Abstract

    Cortical gamma oscillations (20-80 Hz) predict increases in focused attention, and failure in gamma regulation is a hallmark of neurological and psychiatric disease. Current theory predicts that gamma oscillations are generated by synchronous activity of fast-spiking inhibitory interneurons, with the resulting rhythmic inhibition producing neural ensemble synchrony by generating a narrow window for effective excitation. We causally tested these hypotheses in barrel cortex in vivo by targeting optogenetic manipulation selectively to fast-spiking interneurons. Here we show that light-driven activation of fast-spiking interneurons at varied frequencies (8-200 Hz) selectively amplifies gamma oscillations. In contrast, pyramidal neuron activation amplifies only lower frequency oscillations, a cell-type-specific double dissociation. We found that the timing of a sensory input relative to a gamma cycle determined the amplitude and precision of evoked responses. Our data directly support the fast-spiking-gamma hypothesis and provide the first causal evidence that distinct network activity states can be induced in vivo by cell-type-specific activation.

    View details for DOI 10.1038/nature08002

    View details for Web of Science ID 000266608600035

    View details for PubMedID 19396156

    View details for PubMedCentralID PMC3655711

  • Phasic Firing in Dopaminergic Neurons Is Sufficient for Behavioral Conditioning SCIENCE Tsai, H., Zhang, F., Adamantidis, A., Stuber, G. D., Bonci, A., de Lecea, L., Deisseroth, K. 2009; 324 (5930): 1080-1084

    Abstract

    Natural rewards and drugs of abuse can alter dopamine signaling, and ventral tegmental area (VTA) dopaminergic neurons are known to fire action potentials tonically or phasically under different behavioral conditions. However, without technology to control specific neurons with appropriate temporal precision in freely behaving mammals, the causal role of these action potential patterns in driving behavioral changes has been unclear. We used optogenetic tools to selectively stimulate VTA dopaminergic neuron action potential firing in freely behaving mammals. We found that phasic activation of these neurons was sufficient to drive behavioral conditioning and elicited dopamine transients with magnitudes not achieved by longer, lower-frequency spiking. These results demonstrate that phasic dopaminergic activity is sufficient to mediate mammalian behavioral conditioning.

    View details for DOI 10.1126/science.1168878

    View details for Web of Science ID 000266246700044

    View details for PubMedID 19389999

  • Temporally precise in vivo control of intracellular signalling NATURE Airan, R. D., Thompson, K. R., Fenno, L. E., Bernstein, H., Deisseroth, K. 2009; 458 (7241): 1025-1029

    Abstract

    In the study of complex mammalian behaviours, technological limitations have prevented spatiotemporally precise control over intracellular signalling processes. Here we report the development of a versatile family of genetically encoded optical tools ('optoXRs') that leverage common structure-function relationships among G-protein-coupled receptors (GPCRs) to recruit and control, with high spatiotemporal precision, receptor-initiated biochemical signalling pathways. In particular, we have developed and characterized two optoXRs that selectively recruit distinct, targeted signalling pathways in response to light. The two optoXRs exerted opposing effects on spike firing in nucleus accumbens in vivo, and precisely timed optoXR photostimulation in nucleus accumbens by itself sufficed to drive conditioned place preference in freely moving mice. The optoXR approach allows testing of hypotheses regarding the causal impact of biochemical signalling in behaving mammals, in a targetable and temporally precise manner.

    View details for DOI 10.1038/nature07926

    View details for Web of Science ID 000265412900042

    View details for PubMedID 19295515

  • Optical Deconstruction of Parkinsonian Neural Circuitry SCIENCE Gradinaru, V., Mogri, M., Thompson, K. R., Henderson, J. M., Deisseroth, K. 2009; 324 (5925): 354-359

    Abstract

    Deep brain stimulation (DBS) is a therapeutic option for intractable neurological and psychiatric disorders, including Parkinson's disease and major depression. Because of the heterogeneity of brain tissues where electrodes are placed, it has been challenging to elucidate the relevant target cell types or underlying mechanisms of DBS. We used optogenetics and solid-state optics to systematically drive or inhibit an array of distinct circuit elements in freely moving parkinsonian rodents and found that therapeutic effects within the subthalamic nucleus can be accounted for by direct selective stimulation of afferent axons projecting to this region. In addition to providing insight into DBS mechanisms, these results demonstrate an optical approach for dissection of disease circuitry and define the technological toolbox needed for systematic deconstruction of disease circuits by selectively controlling individual components.

    View details for DOI 10.1126/science.1167093

    View details for PubMedID 19299587

  • Neural landscape diffusion resolves conflicts between needs across time. Nature Richman, E. B., Ticea, N., Allen, W. E., Deisseroth, K., Luo, L. 2023

    Abstract

    Animals perform flexible goal-directed behaviours to satisfy their basic physiological needs1-12. However, little is known about how unitary behaviours are chosen under conflicting needs. Here we reveal principles by which the brain resolves such conflicts between needs across time. We developed an experimental paradigm in which a hungry and thirsty mouse is given free choices between equidistant food and water. We found that mice collect need-appropriate rewards by structuring their choices into persistent bouts with stochastic transitions. High-density electrophysiological recordings during this behaviour revealed distributed single neuron and neuronal population correlates of a persistent internal goal state guiding future choices of the mouse. We captured these phenomena with a mathematical model describing a global need state that noisily diffuses across a shifting energy landscape. Model simulations successfully predicted behavioural and neural data, including population neural dynamics before choice transitions and in response to optogenetic thirst stimulation. These results provide a general framework for resolving conflicts between needs across time, rooted in the emergent properties of need-dependent state persistence and noise-driven shifts between behavioural goals.

    View details for DOI 10.1038/s41586-023-06715-z

    View details for PubMedID 37938783

    View details for PubMedCentralID 4306350

  • Ketamine's acute effects on negative brain states are mediated through distinct altered states of consciousness in humans. Nature communications Hack, L. M., Zhang, X., Heifets, B. D., Suppes, T., van Roessel, P. J., Yesavage, J. A., Gray, N. J., Hilton, R., Bertrand, C., Rodriguez, C. I., Deisseroth, K., Knutson, B., Williams, L. M. 2023; 14 (1): 6631

    Abstract

    Ketamine commonly and rapidly induces dissociative and other altered states of consciousness (ASCs) in humans. However, the neural mechanisms that contribute to these experiences remain unknown. We used functional neuroimaging to engage key regions of the brain's affective circuits during acute ketamine-induced ASCs within a randomized, multi-modal, placebo-controlled design examining placebo, 0.05 mg/kg ketamine, and 0.5 mg/kg ketamine in nonclinical adult participants (NCT03475277). Licensed clinicians monitored infusions for safety. Linear mixed effects models, analysis of variance, t-tests, and mediation models were used for statistical analyses. Our design enabled us to test our pre-specified primary and secondary endpoints, which were met: effects of ketamine across dose conditions on (1) emotional task-evoked brain activity, and (2) sub-components of dissociation and other ASCs. With this design, we also could disentangle which ketamine-induced affective brain states are dependent upon specific aspects of ASCs. Differently valenced ketamine-induced ASCs mediated opposing effects on right anterior insula activity. Participants experiencing relatively higher depersonalization induced by 0.5 mg/kg of ketamine showed relief from negative brain states (reduced task-evoked right anterior insula activity, 0.39 SD). In contrast, participants experiencing dissociative amnesia showed an exacerbation of insula activity (0.32 SD). These results in nonclinical participants may shed light on the mechanisms by which specific dissociative states predict response to ketamine in depressed individuals.

    View details for DOI 10.1038/s41467-023-42141-5

    View details for PubMedID 37857620

    View details for PubMedCentralID 5126726

  • Sexually dimorphic mechanisms of VGLUT-mediated protection from dopaminergic neurodegeneration. bioRxiv : the preprint server for biology Buck, S. A., Rubin, S. A., Kunkhyen, T., Treiber, C. D., Xue, X., Fenno, L. E., Mabry, S. J., Sundar, V. R., Yang, Z., Shah, D., Ketchesin, K. D., Becker-Krail, D. D., Vasylieva, I., Smith, M. C., Weisel, F. J., Wang, W., Erickson-Oberg, M. Q., O'Leary, E. I., Aravind, E., Ramakrishnan, C., Kim, Y. S., Wu, Y., Quick, M., Coleman, J. A., MacDonald, W. A., Elbakri, R., De Miranda, B. R., Palladino, M. J., McCabe, B. D., Fish, K. N., Seney, M. L., Rayport, S., Mingote, S., Deisseroth, K., Hnasko, T. S., Awatramani, R., Watson, A. M., Waddell, S., Cheetham, C. E., Logan, R. W., Freyberg, Z. 2023

    Abstract

    Parkinson's disease (PD) targets some dopamine (DA) neurons more than others. Sex differences offer insights, with females more protected from DA neurodegeneration. The mammalian vesicular glutamate transporter VGLUT2 and Drosophila ortholog dVGLUT have been implicated as modulators of DA neuron resilience. However, the mechanisms by which VGLUT2/dVGLUT protects DA neurons remain unknown. We discovered DA neuron dVGLUT knockdown increased mitochondrial reactive oxygen species in a sexually dimorphic manner in response to depolarization or paraquat-induced stress, males being especially affected. DA neuron dVGLUT also reduced ATP biosynthetic burden during depolarization. RNA sequencing of VGLUT+ DA neurons in mice and flies identified candidate genes that we functionally screened to further dissect VGLUT-mediated DA neuron resilience across PD models. We discovered transcription factors modulating dVGLUT-dependent DA neuroprotection and identified dj-1β as a regulator of sex-specific DA neuron dVGLUT expression. Overall, VGLUT protects DA neurons from PD-associated degeneration by maintaining mitochondrial health.

    View details for DOI 10.1101/2023.10.02.560584

    View details for PubMedID 37873436

    View details for PubMedCentralID PMC10592912

  • Optoα1AR activation in astrocytes modulates basal hippocampal synaptic excitation and inhibition in a stimulation-specific manner. Hippocampus Courtney, C. D., Sobieski, C., Ramakrishnan, C., Ingram, R. J., Wojnowski, N. M., DeFazio, R. A., Deisseroth, K., Christian-Hinman, C. A. 2023

    Abstract

    Astrocytes play active roles at synapses and can monitor, respond, and adapt to local synaptic activity. While there is abundant evidence that astrocytes modulate excitatory transmission in the hippocampus, evidence for astrocytic modulation of hippocampal synaptic inhibition remains more limited. Furthermore, to better investigate roles for astrocytes in modulating synaptic transmission, more tools that can selectively activate native G protein signaling pathways in astrocytes with both spatial and temporal precision are needed. Here, we utilized AAV8-GFAP-Optoα1AR-eYFP (Optoα1AR), a viral vector that enables activation of Gq signaling in astrocytes via light-sensitive α1-adrenergic receptors. To determine if stimulating astrocytic Optoα1AR modulates hippocampal synaptic transmission, recordings were made in CA1 pyramidal cells with surrounding astrocytes expressing Optoα1AR, channelrhodopsin (ChR2), or GFP. Both high-frequency (20 Hz, 45-ms light pulses, 5 mW, 5 min) and low-frequency (0.5 Hz, 1-s pulses at increasing 1, 5, and 10 mW intensities, 90 s per intensity) blue light stimulation were tested. 20 Hz Optoα1AR stimulation increased both inhibitory and excitatory postsynaptic current (IPSC and EPSC) frequency, and the effect on miniature IPSCs (mIPSCs) was largely reversible within 20 min. However, low-frequency stimulation of Optoα1AR did not modulate either IPSCs or EPSCs, suggesting that astrocytic Gq -dependent modulation of basal synaptic transmission in the hippocampus is stimulation-dependent. By contrast, low-frequency stimulation of astrocytic ChR2 was effective in increasing both synaptic excitation and inhibition. Together, these data demonstrate that Optoα1AR activation in astrocytes changes basal GABAergic and glutamatergic transmission, but only following high-frequency stimulation, highlighting the importance of temporal dynamics when using optical tools to manipulate astrocyte function.

    View details for DOI 10.1002/hipo.23580

    View details for PubMedID 37767862

  • Inhibition of dopamine neurons prevents incentive value encoding of a reward cue: With revelations from deep phenotyping. The Journal of neuroscience : the official journal of the Society for Neuroscience Iglesias, A. G., Chiu, A. S., Wong, J., Campus, P., Li, F., Liu, Z. N., Bhatti, J. K., Patel, S. A., Deisseroth, K., Akil, H., Burgess, C. R., Flagel, S. B. 2023

    Abstract

    The survival of an organism is dependent on their ability to respond to cues in the environment. Such cues can attain control over behavior as a function of the value ascribed to them. Some individuals have an inherent tendency to attribute reward-paired cues with incentive motivational value, or incentive salience. For these individuals, termed sign-trackers, a discrete cue that precedes reward delivery becomes attractive and desirable in its own right. Prior work suggests that the behavior of sign-trackers is dopamine-dependent, and cue-elicited dopamine in the nucleus accumbens is believed to encode the incentive value of reward cues. Here we exploited the temporal resolution of optogenetics to determine whether selective inhibition of ventral tegmental area (VTA) dopamine neurons during cue presentation attenuates the propensity to sign-track. Using male tyrosine hydroxylase (TH)-Cre Long Evans rats it was found that, under baseline conditions, 84% of TH-Cre rats tend to sign-track. Laser-induced inhibition of VTA dopamine neurons during cue presentation prevented the development of sign-tracking behavior, without affecting goal-tracking behavior. When laser inhibition was terminated, these same rats developed a sign-tracking response. Video analysis using DeepLabCut revealed that, relative to rats that received laser inhibition, rats in the control group spent more time near the location of the reward cue even when it was not present and were more likely to orient towards and approach the cue during its presentation. These findings demonstrate that cue-elicited dopamine release is critical for the attribution of incentive salience to reward cues.Significance StatementActivity of dopamine neurons in the ventral tegmental area (VTA) during cue presentation is necessary for the development of a sign-tracking, but not a goal-tracking, conditioned response in a Pavlovian task. We capitalized on the temporal precision of optogenetics to pair cue presentation with inhibition of VTA dopamine neurons. A detailed behavioral analysis with DeepLabCut revealed that cue-directed behaviors do not emerge without dopamine neuron activity in the VTA. Importantly, however, when optogenetic inhibition is lifted, cue-directed behaviors increase, and a sign-tracking response develops. These findings confirm the necessity of dopamine neuron activity in the VTA during cue presentation to encode the incentive value of reward cues.

    View details for DOI 10.1523/JNEUROSCI.0848-23.2023

    View details for PubMedID 37709540

  • Optogenetic stimulation probes with single-neuron resolution based on organic LEDs monolithically integrated on CMOS NATURE ELECTRONICS Taal, A. J., Uguz, I., Hillebrandt, S., Moon, C., Andino-Pavlovsky, V., Choi, J., Keum, C., Deisseroth, K., Gather, M. C., Shepard, K. L. 2023
  • Orbitofrontal cortex control of striatum leads economic decision-making. Nature neuroscience Gore, F., Hernandez, M., Ramakrishnan, C., Crow, A. K., Malenka, R. C., Deisseroth, K. 2023

    Abstract

    Animals must continually evaluate stimuli in their environment to decide which opportunities to pursue, and in many cases these decisions can be understood in fundamentally economic terms. Although several brain regions have been individually implicated in these processes, the brain-wide mechanisms relating these regions in decision-making are unclear. Using an economic decision-making task adapted for rats, we find that neural activity in both of two connected brain regions, the ventrolateral orbitofrontal cortex (OFC) and the dorsomedial striatum (DMS), was required for economic decision-making. Relevant neural activity in both brain regions was strikingly similar, dominated by the spatial features of the decision-making process. However, the neural encoding of choice direction in OFC preceded that of DMS, and this temporal relationship was strongly correlated with choice accuracy. Furthermore, activity specifically in the OFC projection to the DMS was required for appropriate economic decision-making. These results demonstrate that choice information in the OFC is relayed to the DMS to lead accurate economic decision-making.

    View details for DOI 10.1038/s41593-023-01409-1

    View details for PubMedID 37592039

  • Genetically targeted chemical assembly of polymers specifically localized extracellularly to surface membranes of living neurons. Science advances Zhang, A., Loh, K. Y., Kadur, C. S., Michalek, L., Dou, J., Ramakrishnan, C., Bao, Z., Deisseroth, K. 2023; 9 (32): eadi1870

    Abstract

    Multicellular biological systems, particularly living neural networks, exhibit highly complex organization properties that pose difficulties for building cell-specific biocompatible interfaces. We previously developed an approach to genetically program cells to assemble structures that modify electrical properties of neurons in situ, opening up the possibility of building minimally invasive cell-specific structures and interfaces. However, the efficiency and biocompatibility of this approach were challenged by limited membrane targeting of the constructed materials. Here, we design a method for highly localized expression of enzymes targeted to the plasma membrane of primary neurons, with minimal intracellular retention. Next, we show that polymers synthesized in situ by this approach form dense extracellular clusters selectively on the targeted cell membrane and that neurons remain viable after polymerization. Last, we show generalizability of this method across a range of design strategies. This platform can be readily extended to incorporate a broad diversity of materials onto specific cell membranes within tissues and may further enable next-generation biological interfaces.

    View details for DOI 10.1126/sciadv.adi1870

    View details for PubMedID 37556541

  • Unique functional responses differentially map onto genetic subtypes of dopamine neurons. Nature neuroscience Azcorra, M., Gaertner, Z., Davidson, C., He, Q., Kim, H., Nagappan, S., Hayes, C. K., Ramakrishnan, C., Fenno, L., Kim, Y. S., Deisseroth, K., Longnecker, R., Awatramani, R., Dombeck, D. A. 2023

    Abstract

    Dopamine neurons are characterized by their response to unexpected rewards, but they also fire during movement and aversive stimuli. Dopamine neuron diversity has been observed based on molecular expression profiles; however, whether different functions map onto such genetic subtypes remains unclear. In this study, we established that three genetic dopamine subtypes within the substantia nigra pars compacta, characterized by the expression of Slc17a6 (Vglut2), Calb1 and Anxa1, each have a unique set of responses to rewards, aversive stimuli and accelerations and decelerations, and these signaling patterns are highly correlated between somas and axons within subtypes. Remarkably, reward responses were almost entirely absent in the Anxa1+ subtype, which instead displayed acceleration-correlated signaling. Our findings establish a connection between functional and genetic dopamine subtypes and demonstrate that molecular expression patterns can serve as a common framework to dissect dopaminergic functions.

    View details for DOI 10.1038/s41593-023-01401-9

    View details for PubMedID 37537242

    View details for PubMedCentralID 2739096

  • Intersectional approaches for probing sex differences in dopamine neuron resilience Freyberg, Z., Buck, S., Xue, X., Fenno, L., Weisel, F., Deisseroth, K., Awatramani, R., Logan, R. WILEY. 2023: 76-77
  • Are novel treatments for brain disorders hiding in plain sight? Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology Duncan, L., Deisseroth, K. 2023

    Abstract

    Psychiatric diseases are strongly influenced by genetics, but genetically guided treatments have been slow to develop, and precise molecular mechanisms remain mysterious. Although individual locations in the genome tend to not contribute powerfully to psychiatric disease incidence, genome-wide association studies (GWAS) have now successfully linked hundreds of specific genetic loci to psychiatric disorders [1-3]. Here, building upon results from well-powered GWAS of four phenotypes relevant to psychiatry, we motivate an exploratory workflow leading from GWAS screening, through causal testing in animal models using methods such as optogenetics, to new therapies in human beings. We focus on schizophrenia and the dopamine D2 receptor (DRD2), hot flashes and the neurokinin B receptor (TACR3), cigarette smoking and receptors bound by nicotine (CHRNA5, CHRNA3, CHRNB4), and alcohol use and enzymes that help to break down alcohol (ADH1B, ADH1C, ADH7). A single genomic locus may not powerfully determine disease at the level of the population, but the same locus may nevertheless represent a potent treatment target suitable for population-wide therapeutic approaches.

    View details for DOI 10.1038/s41386-023-01636-x

    View details for PubMedID 37422511

    View details for PubMedCentralID 3257326

  • Causal evidence for the processing of bodily self in the anterior precuneus. Neuron Lyu, D., Stieger, J. R., Xin, C., Ma, E., Lusk, Z., Aparicio, M. K., Werbaneth, K., Perry, C. M., Deisseroth, K., Buch, V., Parvizi, J. 2023

    Abstract

    To probe the causal importance of the human posteromedial cortex (PMC) in processing the sense of self, we studied a rare cohort of nine patients with electrodes implanted bilaterally in the precuneus, posterior cingulate, and retrosplenial regions with a combination of neuroimaging, intracranial recordings, and direct cortical stimulations. In all participants, the stimulation of specific sites within the anterior precuneus (aPCu) caused dissociative changes in physical and spatial domains. Using single-pulse electrical stimulations and neuroimaging, we present effective and resting-state connectivity of aPCu hot zone with the rest of the brain and show that they are located outside the boundaries of the default mode network (DMN) but connected reciprocally with it. We propose that the function of this subregion of the PMC is integral to a range of cognitive processes that require the self's physical point of reference, given its location within a spatial environment.

    View details for DOI 10.1016/j.neuron.2023.05.013

    View details for PubMedID 37295420

  • CLARITY increases sensitivity and specificity of fluorescence immunostaining in long-term archived human brain tissue. BMC biology Woelfle, S., Deshpande, D., Feldengut, S., Braak, H., Del Tredici, K., Roselli, F., Deisseroth, K., Michaelis, J., Boeckers, T. M., Schön, M. 2023; 21 (1): 113

    Abstract

    Post mortem human brain tissue is an essential resource to study cell types, connectivity as well as subcellular structures down to the molecular setup of the central nervous system especially with respect to the plethora of brain diseases. A key method is immunostaining with fluorescent dyes, which allows high-resolution imaging in three dimensions of multiple structures simultaneously. Although there are large collections of formalin-fixed brains, research is often limited because several conditions arise that complicate the use of human brain tissue for high-resolution fluorescence microscopy.In this study, we developed a clearing approach for immunofluorescence-based analysis of perfusion- and immersion-fixed post mortem human brain tissue, termed human Clear Lipid-exchanged Acrylamide-hybridized Rigid Imaging / Immunostaining / In situ hybridization-compatible Tissue-hYdrogel (hCLARITY). hCLARITY is optimized for specificity by reducing off-target labeling and yields very sensitive stainings in human brain sections allowing for super-resolution microscopy with unprecedented imaging of pre- and postsynaptic compartments. Moreover, hallmarks of Alzheimer's disease were preserved with hCLARITY, and importantly classical 3,3'-diaminobenzidine (DAB) or Nissl stainings are compatible with this protocol. hCLARITY is very versatile as demonstrated by the use of more than 30 well performing antibodies and allows for de- and subsequent re-staining of the same tissue section, which is important for multi-labeling approaches, e.g., in super-resolution microscopy.Taken together, hCLARITY enables research of the human brain with high sensitivity and down to sub-diffraction resolution. It therefore has enormous potential for the investigation of local morphological changes, e.g., in neurodegenerative diseases.

    View details for DOI 10.1186/s12915-023-01582-6

    View details for PubMedID 37221592

    View details for PubMedCentralID PMC10207789

  • Rapid and precise genome engineering in a naturally short-lived vertebrate. eLife Bedbrook, C. N., Nath, R. D., Nagvekar, R., Deisseroth, K., Brunet, A. 2023; 12

    Abstract

    The African turquoise killifish is a powerful vertebrate system to study complex phenotypes at scale, including aging and age-related disease. Here, we develop a rapid and precise CRISPR/Cas9-mediated knock-in approach in the killifish. We show its efficient application to precisely insert fluorescent reporters of different sizes at various genomic loci in order to drive cell-type- and tissue-specific expression. This knock-in method should allow the establishment of humanized disease models and the development of cell-type-specific molecular probes for studying complex vertebrate biology.

    View details for DOI 10.7554/eLife.80639

    View details for PubMedID 37191291

    View details for PubMedCentralID PMC10188113

  • Inhibition of dopamine neurons prevents incentive value encoding of a reward cue: With revelations from deep phenotyping. bioRxiv : the preprint server for biology Iglesias, A. G., Chiu, A. S., Wong, J., Campus, P., Li, F., Liu, Z. N., Patel, S. A., Deisseroth, K., Akil, H., Burgess, C. R., Flagel, S. B. 2023

    Abstract

    The survival of an organism is dependent on their ability to respond to cues in the environment. Such cues can attain control over behavior as a function of the value ascribed to them. Some individuals have an inherent tendency to attribute reward-paired cues with incentive motivational value, or incentive salience. For these individuals, termed sign-trackers, a discrete cue that precedes reward delivery becomes attractive and desirable in its own right. Prior work suggests that the behavior of sign-trackers is dopamine-dependent, and cue-elicited dopamine in the nucleus accumbens is believed to encode the incentive value of reward cues. Here we exploited the temporal resolution of optogenetics to determine whether selective inhibition of ventral tegmental area (VTA) dopamine neurons during cue presentation attenuates the propensity to sign-track. Using male tyrosine hydroxylase (TH)-Cre Long Evans rats it was found that, under baseline conditions, ∼84% of TH-Cre rats tend to sign-track. Laser-induced inhibition of VTA dopamine neurons during cue presentation prevented the development of sign-tracking behavior, without affecting goal-tracking behavior. When laser inhibition was terminated, these same rats developed a sign-tracking response. Video analysis using DeepLabCut revealed that, relative to rats that received laser inhibition, rats in the control group spent more time near the location of the reward cue even when it was not present and were more likely to orient towards and approach the cue during its presentation. These findings demonstrate that cue-elicited dopamine release is critical for the attribution of incentive salience to reward cues.Activity of dopamine neurons in the ventral tegmental area (VTA) during cue presentation is necessary for the development of a sign-tracking, but not a goal-tracking, conditioned response in a Pavlovian task. We capitalized on the temporal precision of optogenetics to pair cue presentation with inhibition of VTA dopamine neurons. A detailed behavioral analysis with DeepLabCut revealed that cue-directed behaviors do not emerge without VTA dopamine. Importantly, however, when optogenetic inhibition is lifted, cue-directed behaviors increase, and a sign-tracking response develops. These findings confirm the necessity of VTA dopamine during cue presentation to encode the incentive value of reward cues.

    View details for DOI 10.1101/2023.05.03.539324

    View details for PubMedID 37205506

    View details for PubMedCentralID PMC10187226

  • Acute Effects of MDMA on Intrinsic Functional Connectomes Associated With Altered States of Consciousness and Defensiveness Zhang, X., Hack, L., Heifets, B., Suppes, T., van Roessel, P., Yesavage, J., Gray, N., Hilton, R., Rodriguez, C., Deisseroth, K., Knutson, B., Williams, L. ELSEVIER SCIENCE INC. 2023: S87-S88
  • Ketamine's Acute Effects on Negative Brain States are Mediated Through Distinct Altered States in Humans Zhang, X., Hack, L., Heifets, B., Suppes, T., Van Roessel, P., Yesavage, J., Gray, N., Hilton, R., Rodriguez, C., Deisseroth, K., Knutson, B., Williams, L. ELSEVIER SCIENCE INC. 2023: S312
  • Acute Effects of MDMA on Negative Affective Brain Circuit Function: A Randomized Controlled Mechanistic Trial Hack, L., Zhang, X., Heifets, B., Suppes, T., van Roessel, P., Yesavage, J., Gray, N., Hilton, R., Rodriguez, C., Deisseroth, K., Knutson, B., Williams, L. ELSEVIER SCIENCE INC. 2023: S88
  • Monosynaptic inputs to ventral tegmental area glutamate and GABA co-transmitting neurons. bioRxiv : the preprint server for biology Prévost, E. D., Phillips, A., Lauridsen, K., Enserro, G., Rubinstein, B., Alas, D., McGovern, D. J., Ly, A., Banks, M., McNulty, C., Kim, Y. S., Fenno, L. E., Ramakrishnan, C., Deisseroth, K., Root, D. H. 2023

    Abstract

    A unique population of ventral tegmental area (VTA) neurons co-transmits glutamate and GABA as well as functionally signals rewarding and aversive outcomes. However, the circuit inputs to VTA VGluT2+VGaT+ neurons are unknown, limiting our understanding of the functional capabilities of these neurons. To identify the inputs to VTA VGluT2+VGaT+ neurons, we coupled monosynaptic rabies tracing with intersectional genetic targeting of VTA VGluT2+VGaT+ neurons in mice. We found that VTA VGluT2+VGaT+ neurons received diverse brain-wide inputs. The largest numbers of monosynaptic inputs to VTA VGluT2+VGaT+ neurons were from superior colliculus, lateral hypothalamus, midbrain reticular nucleus, and periaqueductal gray, whereas the densest inputs relative to brain region volume were from dorsal raphe nucleus, lateral habenula, and ventral tegmental area. Based on these and prior data, we hypothesized that lateral hypothalamus and superior colliculus inputs were glutamatergic neurons. Optical activation of glutamatergic lateral hypothalamus neurons robustly activated VTA VGluT2+VGaT+ neurons regardless of stimulation frequency and resulted in flee-like ambulatory behavior. In contrast, optical activation of glutamatergic superior colliculus neurons activated VTA VGluT2+VGaT+ neurons for a brief period of time at high stimulation frequency and resulted in head rotation and arrested ambulatory behavior (freezing). For both pathways, behaviors induced by stimulation were uncorrelated with VTA VGluT2+VGaT+ neuron activity, suggesting that VGluT2+VGaT+ neurons are integrators of signals related to aversive outcomes but not of aversion-induced behavioral kinematics. We interpret these results such that VTA VGluT2+VGaT+ neurons may integrate diverse inputs related to the detection and processing of motivationally-salient outcomes.

    View details for DOI 10.1101/2023.04.06.535959

    View details for PubMedID 37066408

    View details for PubMedCentralID PMC10104150

  • Cerebellar Granule Cells Develop Non-neuronal 3D Genome Architecture over the Lifespan. bioRxiv : the preprint server for biology Tan, L., Shi, J., Moghadami, S., Wright, C. P., Parasar, B., Seo, Y., Vallejo, K., Cobos, I., Duncan, L., Chen, R., Deisseroth, K. 2023

    Abstract

    The cerebellum contains most of the neurons in the human brain, and exhibits unique modes of development, malformation, and aging. For example, granule cells-the most abundant neuron type-develop unusually late and exhibit unique nuclear morphology. Here, by developing our high-resolution single-cell 3D genome assay Dip-C into population-scale (Pop-C) and virus-enriched (vDip-C) modes, we were able to resolve the first 3D genome structures of single cerebellar cells, create life-spanning 3D genome atlases for both human and mouse, and jointly measure transcriptome and chromatin accessibility during development. We found that while the transcriptome and chromatin accessibility of human granule cells exhibit a characteristic maturation pattern within the first year of postnatal life, 3D genome architecture gradually remodels throughout life into a non-neuronal state with ultra-long-range intra-chromosomal contacts and specific inter-chromosomal contacts. This 3D genome remodeling is conserved in mice, and robust to heterozygous deletion of chromatin remodeling disease-associated genes ( Chd8 or Arid1b ). Together these results reveal unexpected and evolutionarily-conserved molecular processes underlying the unique development and aging of the mammalian cerebellum.

    View details for DOI 10.1101/2023.02.25.530020

    View details for PubMedID 36865235

  • Author Correction: Video-based pooled screening yields improved far-red genetically encoded voltage indicators. Nature methods Tian, H., Davis, H. C., Wong-Campos, J. D., Park, P., Fan, L. Z., Gmeiner, B., Begum, S., Werley, C. A., Borja, G. B., Upadhyay, H., Shah, H., Jacques, J., Qi, Y., Parot, V., Deisseroth, K., Cohen, A. E. 2023

    View details for DOI 10.1038/s41592-023-01805-2

    View details for PubMedID 36765248

  • Integrated cardio-behavioral responses to threat define defensive states. Nature neuroscience Signoret-Genest, J., Schukraft, N., L Reis, S., Segebarth, D., Deisseroth, K., Tovote, P. 2023

    Abstract

    Fear and anxiety are brain states that evolved to mediate defensive responses to threats. The defense reaction includes multiple interacting behavioral, autonomic and endocrine adjustments, but their integrative nature is poorly understood. In particular, although threat has been associated with various cardiac changes, there is no clear consensus regarding the relevance of these changes for the integrated defense reaction. Here we identify rapid microstates that are associated with specific behaviors and heart rate dynamics, which are affected by long-lasting macrostates and reflect context-dependent threat levels. In addition, we demonstrate that one of the most commonly used defensive behavioral responses-freezing as measured by immobility-is part of an integrated cardio-behavioral microstate mediated by Chx10+ neurons in the periaqueductal gray. Our framework for systematic integration of cardiac and behavioral readouts presents the basis for a better understanding of complex neural defensive states and their associated systemic functions.

    View details for DOI 10.1038/s41593-022-01252-w

    View details for PubMedID 36759559

  • Activity of a direct VTA to ventral pallidum GABA pathway encodes unconditioned reward value and sustains motivation for reward. Science advances Zhou, W., Kim, K., Ali, F., Pittenger, S. T., Calarco, C. A., Mineur, Y. S., Ramakrishnan, C., Deisseroth, K., Kwan, A. C., Picciotto, M. R. 2022; 8 (42): eabm5217

    Abstract

    Dopamine signaling from the ventral tegmental area (VTA) plays critical roles in reward-related behaviors, but less is known about the functions of neighboring VTA GABAergic neurons. We show here that a primary target of VTA GABA projection neurons is the ventral pallidum (VP). Activity of VTA-to-VP-projecting GABA neurons correlates consistently with size and palatability of the reward and does not change following cue learning, providing a direct measure of reward value. Chemogenetic stimulation of this GABA projection increased activity of a subset of VP neurons that were active while mice were seeking reward. Optogenetic stimulation of this pathway improved performance in a cue-reward task and maintained motivation to work for reward over days. This VTA GABA projection provides information about reward value directly to the VP, likely distinct from the prediction error signal carried by VTA dopamine neurons.

    View details for DOI 10.1126/sciadv.abm5217

    View details for PubMedID 36260661

  • Maturation and circuit integration of transplanted human cortical organoids. Nature Revah, O., Gore, F., Kelley, K. W., Andersen, J., Sakai, N., Chen, X., Li, M. Y., Birey, F., Yang, X., Saw, N. L., Baker, S. W., Amin, N. D., Kulkarni, S., Mudipalli, R., Cui, B., Nishino, S., Grant, G. A., Knowles, J. K., Shamloo, M., Huguenard, J. R., Deisseroth, K., Pașca, S. P. 2022; 610 (7931): 319-326

    Abstract

    Self-organizing neural organoids represent a promising in vitro platform with which to model human development and disease1-5. However, organoids lack the connectivity that exists in vivo, which limits maturation and makes integration with other circuits that control behaviour impossible. Here we show that human stem cell-derived cortical organoids transplanted into the somatosensory cortex of newborn athymic rats develop mature cell types that integrate into sensory and motivation-related circuits. MRI reveals post-transplantation organoid growth across multiple stem cell lines and animals, whereas single-nucleus profiling shows progression of corticogenesis and the emergence of activity-dependent transcriptional programs. Indeed, transplanted cortical neurons display more complex morphological, synaptic and intrinsic membrane properties than their in vitro counterparts, which enables the discovery of defects in neurons derived from individuals with Timothy syndrome. Anatomical and functional tracings show that transplanted organoids receive thalamocortical and corticocortical inputs, and in vivo recordings of neural activity demonstrate that these inputs can produce sensory responses in human cells. Finally, cortical organoids extend axons throughout the rat brain and their optogenetic activation can drive reward-seeking behaviour. Thus, transplanted human cortical neurons mature and engage host circuits that control behaviour. We anticipate that this approach will be useful for detecting circuit-level phenotypes in patient-derived cells that cannot otherwise be uncovered.

    View details for DOI 10.1038/s41586-022-05277-w

    View details for PubMedID 36224417

  • Correction to: Brain-wide perception of the emotional valence of light is regulated by distinct hypothalamic neurons. Molecular psychiatry Wagle, M., Zarei, M., Lovett-Barron, M., Poston, K. T., Xu, J., Ramey, V., Pollard, K. S., Prober, D. A., Schulkin, J., Deisseroth, K., Guo, S. 2022

    View details for DOI 10.1038/s41380-022-01647-y

    View details for PubMedID 35718796

  • Brain-wide perception of the emotional valence of light is regulated by distinct hypothalamic neurons. Molecular psychiatry Wagle, M., Zarei, M., Lovett-Barron, M., Poston, K. T., Xu, J., Ramey, V., Pollard, K. S., Prober, D. A., Schulkin, J., Deisseroth, K., Guo, S. 2022

    Abstract

    Salient sensory stimuli are perceived by the brain, which guides both the timing and outcome of behaviors in a context-dependent manner. Light is such a stimulus, which is used in treating mood disorders often associated with a dysregulated hypothalamic-pituitary-adrenal stress axis. Relationships between the emotional valence of light and the hypothalamus, and how they interact to exert brain-wide impacts remain unclear. Employing larval zebrafish with analogous hypothalamic systems to mammals, we show in free-swimming animals that hypothalamic corticotropin releasing factor (CRFHy) neurons promote dark avoidance, and such role is not shared by other hypothalamic peptidergic neurons. Single-neuron projection analyses uncover processes extended by individual CRFHy neurons to multiple targets including sensorimotor and decision-making areas. In vivo calcium imaging uncovers a complex and heterogeneous response of individual CRFHy neurons to the light or dark stimulus, with a reduced overall sum of CRF neuronal activity in the presence of light. Brain-wide calcium imaging under alternating light/dark stimuli further identifies distinct and distributed photic response neuronal types. CRFHy neuronal ablation increases an overall representation of light in the brain and broadly enhances the functional connectivity associated with an exploratory brain state. These findings delineate brain-wide photic perception, uncover a previously unknown role of CRFHy neurons in regulating the perception and emotional valence of light, and suggest that light therapy may alleviate mood disorders through reducing an overall sum of CRF neuronal activity.

    View details for DOI 10.1038/s41380-022-01567-x

    View details for PubMedID 35484242

  • Topological supramolecular network enabled high-conductivity, stretchable organic bioelectronics. Science (New York, N.Y.) Jiang, Y., Zhang, Z., Wang, Y. X., Li, D., Coen, C. T., Hwaun, E., Chen, G., Wu, H. C., Zhong, D., Niu, S., Wang, W., Saberi, A., Lai, J. C., Wu, Y., Wang, Y., Trotsyuk, A. A., Loh, K. Y., Shih, C. C., Xu, W., Liang, K., Zhang, K., Bai, Y., Gurusankar, G., Hu, W., Jia, W., Cheng, Z., Dauskardt, R. H., Gurtner, G. C., Tok, J. B., Deisseroth, K., Soltesz, I., Bao, Z. 2022; 375 (6587): 1411-1417

    Abstract

    Intrinsically stretchable bioelectronic devices based on soft and conducting organic materials have been regarded as the ideal interface for seamless and biocompatible integration with the human body. A remaining challenge is to combine high mechanical robustness with good electrical conduction, especially when patterned at small feature sizes. We develop a molecular engineering strategy based on a topological supramolecular network, which allows for the decoupling of competing effects from multiple molecular building blocks to meet complex requirements. We obtained simultaneously high conductivity and crack-onset strain in a physiological environment, with direct photopatternability down to the cellular scale. We further collected stable electromyography signals on soft and malleable octopus and performed localized neuromodulation down to single-nucleus precision for controlling organ-specific activities through the delicate brainstem.

    View details for DOI 10.1126/science.abj7564

    View details for PubMedID 35324282

  • Regulation of sensorimotor gating via Disc1/Huntingtin-mediated Bdnf transport in the cortico-striatal circuit. Molecular psychiatry Jaaro-Peled, H., Kumar, S., Hughes, D., Sumitomo, A., Kim, S., Zoubovsky, S., Hirota-Tsuyada, Y., Zala, D., Bruyere, J., Katz, B. M., Huang, B., Flores, R. 3., Narayan, S., Hou, Z., Economides, A. N., Hikida, T., Wetsel, W. C., Deisseroth, K., Mori, S., Brandon, N. J., Tanaka, M., Ishizuka, K., Houslay, M. D., Saudou, F., Dzirasa, K., Sawa, A., Tomoda, T. 2022

    Abstract

    Sensorimotor information processing underlies normal cognitive and behavioral traits and has classically been evaluated through prepulse inhibition (PPI) of a startle reflex. PPI is a behavioral dimension deregulated in several neurological and psychiatric disorders, yet the mechanisms underlying the cross-diagnostic nature of PPI deficits across these conditions remain to be understood. To identify circuitry mechanisms for PPI, we performed circuitry recording over the prefrontal cortex and striatum, two brain regions previously implicated in PPI, using wild-type (WT) mice compared to Disc1-locus-impairment (LI) mice, a model representing neuropsychiatric conditions. We demonstrated that the corticostriatal projection regulates neurophysiological responses during the PPI testing in WT, whereas these circuitry responses were disrupted in Disc1-LI mice. Because our biochemical analyses revealed attenuated brain-derived neurotrophic factor (Bdnf) transport along the corticostriatal circuit in Disc1-LI mice, we investigated the potential role of Bdnf in this circuitry for regulation of PPI. Virus-mediated delivery of Bdnf into the striatum rescued PPI deficits in Disc1-LI mice. Pharmacologically augmenting Bdnf transport by chronic lithium administration, partly via phosphorylation of Huntingtin (Htt) serine-421 and its integration into the motor machinery, restored striatal Bdnf levels and rescued PPI deficits in Disc1-LI mice. Furthermore, reducing the cortical Bdnf expression negated this rescuing effect of lithium, confirming the key role of Bdnf in lithium-mediated PPI rescuing. Collectively, the data suggest that striatal Bdnf supply, collaboratively regulated by Htt and Disc1 along the corticostriatal circuit, is involved in sensorimotor gating, highlighting the utility of dimensional approach in investigating pathophysiological mechanisms across neuropsychiatric disorders.

    View details for DOI 10.1038/s41380-021-01389-3

    View details for PubMedID 35165396

  • A functional cellular framework for sex and estrous cycle-dependent gene expression and behavior. Cell Knoedler, J. R., Inoue, S., Bayless, D. W., Yang, T., Tantry, A., Davis, C., Leung, N. Y., Parthasarathy, S., Wang, G., Alvarado, M., Rizvi, A. H., Fenno, L. E., Ramakrishnan, C., Deisseroth, K., Shah, N. M. 1800

    Abstract

    Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.

    View details for DOI 10.1016/j.cell.2021.12.031

    View details for PubMedID 35065713

  • Similar neural and perceptual masking effects of low-power optogenetic stimulation in primate V1. eLife Chen, S. C., Benvenuti, G., Chen, Y., Kumar, S., Ramakrishnan, C., Deisseroth, K., Geisler, W. S., Seidemann, E. 1800; 11

    Abstract

    Can direct stimulation of primate V1 substitute for a visual stimulus and mimic its perceptual effect? To address this question, we developed an optical-genetic toolkit to 'read' neural population responses using widefield calcium imaging, while simultaneously using optogenetics to 'write' neural responses into V1 of behaving macaques. We focused on the phenomenon of visual masking, where detection of a dim target is significantly reduced by a co-localized medium-brightness mask [1, 2]. Using our toolkit, we tested whether V1 optogenetic stimulation can recapitulate the perceptual masking effect of a visual mask. We find that, similar to a visual mask, low-power optostimulation can significantly reduce visual detection sensitivity, that a sublinear interaction between visual and optogenetic evoked V1 responses could account for this perceptual effect, and that these neural and behavioral effects are spatially selective. Our toolkit and results open the door for further exploration of perceptual substitutions by direct stimulation of sensory cortex.

    View details for DOI 10.7554/eLife.68393

    View details for PubMedID 34982033

  • Similar neural and perceptual masking effects of low-power optogenetic stimulation in primate V1 ELIFE Chen, S., Benvenuti, G., Chen, Y., Kumar, S., Ramakrishnan, C., Deisseroth, K., Geisler, W. S., Seidemann, E. 2022; 11
  • A Bright, Nontoxic, and Non-aggregating red Fluorescent Protein for Long-Term Labeling of Fine Structures in Neurons. Frontiers in cell and developmental biology Ning, L., Geng, Y., Lovett-Barron, M., Niu, X., Deng, M., Wang, L., Ataie, N., Sens, A., Ng, H., Chen, S., Deisseroth, K., Lin, M. Z., Chu, J. 2022; 10: 893468

    Abstract

    Red fluorescent proteins are useful as morphological markers in neurons, often complementing green fluorescent protein-based probes of neuronal activity. However, commonly used red fluorescent proteins show aggregation and toxicity in neurons or are dim. We report the engineering of a bright red fluorescent protein, Crimson, that enables long-term morphological labeling of neurons without aggregation or toxicity. Crimson is similar to mCherry and mKate2 in fluorescence spectra but is 100 and 28% greater in molecular brightness, respectively. We used a membrane-localized Crimson-CAAX to label thin neurites, dendritic spines and filopodia, enhancing detection of these small structures compared to cytosolic markers.

    View details for DOI 10.3389/fcell.2022.893468

    View details for PubMedID 35846353

  • Sox6 expression distinguishes dorsally and ventrally biased dopamine neurons in the substantia nigra with distinctive properties and embryonic origins. Cell reports Pereira Luppi, M., Azcorra, M., Caronia-Brown, G., Poulin, J., Gaertner, Z., Gatica, S., Moreno-Ramos, O. A., Nouri, N., Dubois, M., Ma, Y. C., Ramakrishnan, C., Fenno, L., Kim, Y. S., Deisseroth, K., Cicchetti, F., Dombeck, D. A., Awatramani, R. 2021; 37 (6): 109975

    Abstract

    Dopamine (DA) neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease, while those in the dorsal tier are relatively spared. Defining the molecular, functional, and developmental characteristics of each SNc tier is crucial to understand their distinct susceptibility. We demonstrate that Sox6 expression distinguishes ventrally and dorsally biased DA neuron populations in the SNc. The Sox6+ population in the ventral SNc includes an Aldh1a1+ subset and is enriched in gene pathways that underpin vulnerability. Sox6+ neurons project to the dorsal striatum and show activity correlated with acceleration. Sox6- neurons project to the medial, ventral, and caudal striatum and respond to rewards. Moreover, we show that this adult division is encoded early in development. Overall, our work demonstrates a dual origin of the SNc that results in DA neuron cohorts with distinct molecular profiles, projections, and functions.

    View details for DOI 10.1016/j.celrep.2021.109975

    View details for PubMedID 34758317

  • An uncommon neuronal class conveys visual signals from rods and cones to retinal ganglion cells. Proceedings of the National Academy of Sciences of the United States of America Young, B. K., Ramakrishnan, C., Ganjawala, T., Wang, P., Deisseroth, K., Tian, N. 2021; 118 (44)

    Abstract

    Neurons in the central nervous system (CNS) are distinguished by the neurotransmitter types they release, their synaptic connections, morphology, and genetic profiles. To fully understand how the CNS works, it is critical to identify all neuronal classes and reveal their synaptic connections. The retina has been extensively used to study neuronal development and circuit formation. Here, we describe a previously unidentified interneuron in mammalian retina. This interneuron shares some morphological, physiological, and molecular features with retinal bipolar cells, such as receiving input from photoreceptors and relaying visual signals to retinal ganglion cells. It also shares some features with amacrine cells (ACs), particularly Aii-ACs, such as their neurite morphology in the inner plexiform layer, the expression of some AC-specific markers, and possibly the release of the inhibitory neurotransmitter glycine. Thus, we unveil an uncommon interneuron, which may play an atypical role in vision.

    View details for DOI 10.1073/pnas.2104884118

    View details for PubMedID 34702737

  • Genetically identified amygdala-striatal circuits for valence-specific behaviors. Nature neuroscience Zhang, X., Guan, W., Yang, T., Furlan, A., Xiao, X., Yu, K., An, X., Galbavy, W., Ramakrishnan, C., Deisseroth, K., Ritola, K., Hantman, A., He, M., Josh Huang, Z., Li, B. 2021

    Abstract

    The basolateral amygdala (BLA) plays essential roles in behaviors motivated by stimuli with either positive or negative valence, but how it processes motivationally opposing information and participates in establishing valence-specific behaviors remains unclear. Here, by targeting Fezf2-expressing neurons in the BLA, we identify and characterize two functionally distinct classes in behaving mice, the negative-valence neurons and positive-valence neurons, which innately represent aversive and rewarding stimuli, respectively, and through learning acquire predictive responses that are essential for punishment avoidance or reward seeking. Notably, these two classes of neurons receive inputs from separate sets of sensory and limbic areas, and convey punishment and reward information through projections to the nucleus accumbens and olfactory tubercle, respectively, to drive negative and positive reinforcement. Thus, valence-specific BLA neurons are wired with distinctive input-output structures, forming a circuit framework that supports the roles of the BLA in encoding, learning and executing valence-specific motivated behaviors.

    View details for DOI 10.1038/s41593-021-00927-0

    View details for PubMedID 34663958

  • Neural correlates of ingroup bias for prosociality in rats. eLife Ben-Ami Bartal, I., Breton, J. M., Sheng, H., Long, K. L., Chen, S., Halliday, A., Kenney, J. W., Wheeler, A. L., Frankland, P., Shilyansky, C., Deisseroth, K., Keltner, D., Kaufer, D. 2021; 10

    Abstract

    Prosocial behavior, in particular helping others in need, occurs preferentially in response to distress of one's own group members. In order to explore the neural mechanisms promoting mammalian helping behavior, a discovery-based approach was used here to identify brain-wide activity correlated with helping behavior in rats. Demonstrating social selectivity, rats helped others of their strain ('ingroup'), but not rats of an unfamiliar strain ('outgroup'), by releasing them from a restrainer. Analysis of brain-wide neural activity via quantification of the early-immediate gene c-Fos identified a shared network, including frontal and insular cortices, that was active in the helping test irrespective of group membership. In contrast, the striatum was selectively active for ingroup members, and activity in the nucleus accumbens, a central network hub, correlated with helping. In vivo calcium imaging showed accumbens activity when rats approached a trapped ingroup member, and retrograde tracing identified a subpopulation of accumbens-projecting cells that was correlated with helping. These findings demonstrate that motivation and reward networks are associated with helping an ingroup member and provide the first description of neural correlates of ingroup bias in rodents.

    View details for DOI 10.7554/eLife.65582

    View details for PubMedID 34253289

  • CloudReg: automatic terabyte-scale cross-modal brain volume registration. Nature methods Chandrashekhar, V., Tward, D. J., Crowley, D., Crow, A. K., Wright, M. A., Hsueh, B. Y., Gore, F., Machado, T. A., Branch, A., Rosenblum, J. S., Deisseroth, K., Vogelstein, J. T. 2021

    View details for DOI 10.1038/s41592-021-01218-z

    View details for PubMedID 34253927

  • A neural circuit state change underlying skilled movements. Cell Wagner, M. J., Savall, J., Hernandez, O., Mel, G., Inan, H., Rumyantsev, O., Lecoq, J., Kim, T. H., Li, J. Z., Ramakrishnan, C., Deisseroth, K., Luo, L., Ganguli, S., Schnitzer, M. J. 2021

    Abstract

    In motor neuroscience, state changes are hypothesized to time-lock neural assemblies coordinating complex movements, but evidence for this remains slender. We tested whether a discrete change from more autonomous to coherent spiking underlies skilled movement by imaging cerebellar Purkinje neuron complex spikes in mice making targeted forelimb-reaches. As mice learned the task, millimeter-scale spatiotemporally coherent spiking emerged ipsilateral to the reaching forelimb, and consistent neural synchronization became predictive of kinematic stereotypy. Before reach onset, spiking switched from more disordered to internally time-locked concerted spiking and silence. Optogenetic manipulations of cerebellar feedback to the inferior olive bi-directionally modulated neural synchronization and reaching direction. A simple model explained the reorganization of spiking during reaching as reflecting a discrete bifurcation in olivary network dynamics. These findings argue that to prepare learned movements, olivo-cerebellar circuits enter a self-regulated, synchronized state promoting motor coordination. State changes facilitating behavioral transitions may generalize across neural systems.

    View details for DOI 10.1016/j.cell.2021.06.001

    View details for PubMedID 34214470

  • Selective filtering of excitatory inputs to nucleus accumbens by dopamine and serotonin. Proceedings of the National Academy of Sciences of the United States of America Christoffel, D. J., Walsh, J. J., Hoerbelt, P., Heifets, B. D., Llorach, P., Lopez, R. C., Ramakrishnan, C., Deisseroth, K., Malenka, R. C. 2021; 118 (24)

    Abstract

    The detailed mechanisms by which dopamine (DA) and serotonin (5-HT) act in the nucleus accumbens (NAc) to influence motivated behaviors in distinct ways remain largely unknown. Here, we examined whether DA and 5-HT selectively modulate excitatory synaptic transmission in NAc medium spiny neurons in an input-specific manner. DA reduced excitatory postsynaptic currents (EPSCs) generated by paraventricular thalamus (PVT) inputs but not by ventral hippocampus (vHip), basolateral amygdala (BLA), or medial prefrontal cortex (mPFC) inputs. In contrast, 5-HT reduced EPSCs generated by inputs from all areas except the mPFC. Release of endogenous DA and 5-HT by methamphetamine (METH) and (±)3,4-methylenedioxymethamphetamine (MDMA), respectively, recapitulated these input-specific synaptic effects. Optogenetic inhibition of PVT inputs enhanced cocaine-conditioned place preference, whereas mPFC input inhibition reduced the enhancement of sociability elicited by MDMA. These findings suggest that the distinct, input-specific filtering of excitatory inputs in the NAc by DA and 5-HT contribute to their discrete behavioral effects.

    View details for DOI 10.1073/pnas.2106648118

    View details for PubMedID 34103400

  • Reciprocal lateral hypothalamic and raphe GABAergic projections promote wakefulness. The Journal of neuroscience : the official journal of the Society for Neuroscience Gazea, M., Furdan, S., Sere, P., Oesch, L., Molnar, B., Giovanni, G. D., Fenno, L. E., Ramakrishnan, C., Mattis, J., Deisseroth, K., Dymecki, S. M., Adamantidis, A. R., Lorincz, M. L. 2021

    Abstract

    The lateral hypothalamus (LH), together with multiple neuromodulatory systems of the brain, such as the dorsal raphe nucleus (DR), is implicated in arousal, yet interactions between these systems are just beginning to be explored. Using a combination of viral tracing, circuit mapping, electrophysiological recordings from identified neurons and combinatorial optogenetics in mice, we show that GABAergic neurons in the LH selectively inhibit GABAergic neurons in the DR resulting in increased firing of a substantial fraction of its neurons that ultimately promotes arousal. These DRGABA neurons are wake active and project to multiple brain areas involved in the control of arousal including the LH, where their specific activation potently influences local network activity leading to arousal from sleep. Our results show how mutual inhibitory projections between the LH and the DR promote wakefulness and suggest a complex arousal control by intimate interactions between long-range connections and local circuit dynamics.

    View details for DOI 10.1523/JNEUROSCI.2850-20.2021

    View details for PubMedID 33888606

  • Changes in genome architecture and transcriptional dynamics progress independently of sensory experience during post-natal brain development. Cell Tan, L. n., Ma, W. n., Wu, H. n., Zheng, Y. n., Xing, D. n., Chen, R. n., Li, X. n., Daley, N. n., Deisseroth, K. n., Xie, X. S. 2021

    Abstract

    Both transcription and three-dimensional (3D) architecture of the mammalian genome play critical roles in neurodevelopment and its disorders. However, 3D genome structures of single brain cells have not been solved; little is known about the dynamics of single-cell transcriptome and 3D genome after birth. Here, we generated a transcriptome (3,517 cells) and 3D genome (3,646 cells) atlas of the developing mouse cortex and hippocampus by using our high-resolution multiple annealing and looping-based amplification cycles for digital transcriptomics (MALBAC-DT) and diploid chromatin conformation capture (Dip-C) methods and developing multi-omic analysis pipelines. In adults, 3D genome "structure types" delineate all major cell types, with high correlation between chromatin A/B compartments and gene expression. During development, both transcriptome and 3D genome are extensively transformed in the first post-natal month. In neurons, 3D genome is rewired across scales, correlated with gene expression modules, and independent of sensory experience. Finally, we examine allele-specific structure of imprinted genes, revealing local and chromosome (chr)-wide differences. These findings uncover an unknown dimension of neurodevelopment.

    View details for DOI 10.1016/j.cell.2020.12.032

    View details for PubMedID 33484631

  • Excitatory synapses and gap junctions cooperate to improve Pv neuronal burst firing and cortical social cognition in Shank2-mutant mice. Nature communications Lee, E., Lee, S., Shin, J. J., Choi, W., Chung, C., Lee, S., Kim, J., Ha, S., Kim, R., Yoo, T., Yoo, Y., Kim, J., Noh, Y. W., Rhim, I., Lee, S. Y., Kim, W., Lee, T., Shin, H., Cho, I., Deisseroth, K., Kim, S. J., Park, J. M., Jung, M. W., Paik, S., Kim, E. 2021; 12 (1): 5116

    Abstract

    NMDA receptor (NMDAR) and GABA neuronal dysfunctions are observed in animal models of autism spectrum disorders, but how these dysfunctions impair social cognition and behavior remains unclear. We report here that NMDARs in cortical parvalbumin (Pv)-positive interneurons cooperate with gap junctions to promote high-frequency (>80Hz) Pv neuronal burst firing and social cognition. Shank2-/- mice, displaying improved sociability upon NMDAR activation, show impaired cortical social representation and inhibitory neuronal burst firing. Cortical Shank2-/- Pv neurons show decreased NMDAR activity, which suppresses the cooperation between NMDARs and gap junctions (GJs) for normal burst firing. Shank2-/- Pv neurons show compensatory increases in GJ activity that are not sufficient for social rescue. However, optogenetic boosting of Pv neuronal bursts, requiring GJs, rescues cortical social cognition in Shank2-/- mice, similar to the NMDAR-dependent social rescue. Therefore, NMDARs and gap junctions cooperate to promote cortical Pv neuronal bursts and social cognition.

    View details for DOI 10.1038/s41467-021-25356-2

    View details for PubMedID 34433814

  • Transcriptional and functional divergence in lateral hypothalamic glutamate neurons projecting to the lateral habenula and ventral tegmental area. Neuron Rossi, M. A., Basiri, M. L., Liu, Y., Hashikawa, Y., Hashikawa, K., Fenno, L. E., Kim, Y. S., Ramakrishnan, C., Deisseroth, K., Stuber, G. D. 2021

    Abstract

    The lateral hypothalamic area (LHA) regulates feeding- and reward-related behavior, but because of its molecular and anatomical heterogeneity, the functions of defined neuronal populations are largely unclear. Glutamatergic neurons within the LHA (LHAVglut2) negatively regulate feeding and appetitive behavior. However, this population comprises transcriptionally distinct and functionally diverse neurons that project to diverse brain regions, including the lateral habenula (LHb) and ventral tegmental area (VTA). To resolve the function of distinct LHAVglut2 populations, we systematically compared projections to the LHb and VTA using viral tracing, single-cell sequencing, electrophysiology, and in vivo calcium imaging. LHAVglut2 neurons projecting to the LHb or VTA are anatomically, transcriptionally, electrophysiologically, and functionally distinct. While both populations encode appetitive and aversive stimuli, LHb projecting neurons are especially sensitive to satiety state and feeding hormones. These data illuminate the functional heterogeneity of LHAVglut2 neurons, suggesting that reward and aversion are differentially processed in divergent efferent pathways.

    View details for DOI 10.1016/j.neuron.2021.09.020

    View details for PubMedID 34624220

  • Supramammillary regulation of locomotion and hippocampal activity. Science (New York, N.Y.) Farrell, J. S., Lovett-Barron, M., Klein, P. M., Sparks, F. T., Gschwind, T., Ortiz, A. L., Ahanonu, B., Bradbury, S., Terada, S., Oijala, M., Hwaun, E., Dudok, B., Szabo, G., Schnitzer, M. J., Deisseroth, K., Losonczy, A., Soltesz, I. 2021; 374 (6574): 1492-1496

    Abstract

    [Figure: see text].

    View details for DOI 10.1126/science.abh4272

    View details for PubMedID 34914519

  • Dendritic calcium signals in rhesus macaque motor cortex drive an optical brain-computer interface. Nature communications Trautmann, E. M., O'Shea, D. J., Sun, X., Marshel, J. H., Crow, A., Hsueh, B., Vesuna, S., Cofer, L., Bohner, G., Allen, W., Kauvar, I., Quirin, S., MacDougall, M., Chen, Y., Whitmire, M. P., Ramakrishnan, C., Sahani, M., Seidemann, E., Ryu, S. I., Deisseroth, K., Shenoy, K. V. 2021; 12 (1): 3689

    Abstract

    Calcium imaging is a powerful tool for recording from large populations of neurons in vivo. Imaging in rhesus macaque motor cortex can enable the discovery of fundamental principles of motor cortical function and can inform the design of next generation brain-computer interfaces (BCIs). Surface two-photon imaging, however, cannot presently access somatic calcium signals of neurons from all layers of macaque motor cortex due to photon scattering. Here, we demonstrate an implant and imaging system capable of chronic, motion-stabilized two-photon imaging of neuronal calcium signals from macaques engaged in a motor task. By imaging apical dendrites, we achieved optical access to large populations of deep and superficial cortical neurons across dorsal premotor (PMd) and gyral primary motor (M1) cortices. Dendritic signals from individual neurons displayed tuning for different directions of arm movement. Combining several technical advances, we developed an optical BCI (oBCI) driven by these dendritic signalswhich successfully decoded movement direction online. By fusing two-photon functional imaging with CLARITY volumetric imaging, we verified that many imaged dendrites which contributed to oBCI decoding originated from layer 5 output neurons, including a putative Betz cell. This approach establishes newopportunities for studying motor control and designing BCIsvia two photon imaging.

    View details for DOI 10.1038/s41467-021-23884-5

    View details for PubMedID 34140486

  • Corrigendum: Proceedings of the Eighth Annual Deep Brain Stimulation Think Tank: Advances in Optogenetics, Ethical Issues Affecting DBS Research, Neuromodulatory Approaches for Depression, Adaptive Neurostimulation, and Emerging DBS Technologies. Frontiers in human neuroscience Vedam-Mai, V., Deisseroth, K., Giordano, J., Lazaro-Munoz, G., Chiong, W., Suthana, N., Langevin, J., Gill, J., Goodman, W., Provenza, N. R., Halpern, C. H., Shivacharan, R. S., Cunningham, T. N., Sheth, S. A., Pouratian, N., Scangos, K. W., Mayberg, H. S., Horn, A., Johnson, K. A., Butson, C. R., Gilron, R., de Hemptinne, C., Wilt, R., Yaroshinsky, M., Little, S., Starr, P., Worrell, G., Shirvalkar, P., Chang, E., Volkmann, J., Muthuraman, M., Groppa, S., Kuhn, A. A., Li, L., Johnson, M., Otto, K. J., Raike, R., Goetz, S., Wu, C., Silburn, P., Cheeran, B., Pathak, Y. J., Malekmohammadi, M., Gunduz, A., Wong, J. K., Cernera, S., Hu, W., Wagle Shukla, A., Ramirez-Zamora, A., Deeb, W., Patterson, A., Foote, K. D., Okun, M. S. 2021; 15: 765150

    Abstract

    [This corrects the article DOI: 10.3389/fnhum.2021.644593.].

    View details for DOI 10.3389/fnhum.2021.765150

    View details for PubMedID 34658825

  • Maximally selective single-cell target for circuit control in epilepsy models. Neuron Hadjiabadi, D., Lovett-Barron, M., Raikov, I. G., Sparks, F. T., Liao, Z., Baraban, S. C., Leskovec, J., Losonczy, A., Deisseroth, K., Soltesz, I. 2021

    Abstract

    Neurological and psychiatric disorders are associated with pathological neural dynamics. The fundamental connectivity patterns of cell-cell communication networks that enable pathological dynamics to emerge remain unknown. Here, we studied epileptic circuits using a newly developed computational pipeline that leveraged single-cell calcium imaging of larval zebrafish and chronically epileptic mice, biologically constrained effective connectivity modeling, and higher-order motif-focused network analysis. We uncovered a novel functional cell type that preferentially emerged in the preseizure state, the superhub, that was unusually richly connected to the rest of the network through feedforward motifs, critically enhancing downstream excitation. Perturbation simulations indicated that disconnecting superhubs was significantly more effective in stabilizing epileptic circuits than disconnecting hub cells that were defined traditionally by connection count. In the dentate gyrus of chronically epileptic mice, superhubs were predominately modeled adult-born granule cells. Collectively, these results predict a new maximally selective and minimally invasive cellular target for seizure control.

    View details for DOI 10.1016/j.neuron.2021.06.007

    View details for PubMedID 34197732

  • Proceedings of the Eighth Annual Deep Brain Stimulation Think Tank: Advances in Optogenetics, Ethical Issues Affecting DBS Research, Neuromodulatory Approaches for Depression, Adaptive Neurostimulation, and Emerging DBS Technologies. Frontiers in human neuroscience Vedam-Mai, V., Deisseroth, K., Giordano, J., Lazaro-Munoz, G., Chiong, W., Suthana, N., Langevin, J., Gill, J., Goodman, W., Provenza, N. R., Halpern, C. H., Shivacharan, R. S., Cunningham, T. N., Sheth, S. A., Pouratian, N., Scangos, K. W., Mayberg, H. S., Horn, A., Johnson, K. A., Butson, C. R., Gilron, R., de Hemptinne, C., Wilt, R., Yaroshinsky, M., Little, S., Starr, P., Worrell, G., Shirvalkar, P., Chang, E., Volkmann, J., Muthuraman, M., Groppa, S., Kuhn, A. A., Li, L., Johnson, M., Otto, K. J., Raike, R., Goetz, S., Wu, C., Silburn, P., Cheeran, B., Pathak, Y. J., Malekmohammadi, M., Gunduz, A., Wong, J. K., Cernera, S., Wagle Shukla, A., Ramirez-Zamora, A., Deeb, W., Patterson, A., Foote, K. D., Okun, M. S. 2021; 15: 644593

    Abstract

    We estimate that 208,000 deep brain stimulation (DBS) devices have been implanted to address neurological and neuropsychiatric disorders worldwide. DBS Think Tank presenters pooled data and determined that DBS expanded in its scope and has been applied to multiple brain disorders in an effort to modulate neural circuitry. The DBS Think Tank was founded in 2012 providing a space where clinicians, engineers, researchers from industry and academia discuss current and emerging DBS technologies and logistical and ethical issues facing the field. The emphasis is on cutting edge research and collaboration aimed to advance the DBS field. The Eighth Annual DBS Think Tank was held virtually on September 1 and 2, 2020 (Zoom Video Communications) due to restrictions related to the COVID-19 pandemic. The meeting focused on advances in: (1) optogenetics as a tool for comprehending neurobiology of diseases and on optogenetically-inspired DBS, (2) cutting edge of emerging DBS technologies, (3) ethical issues affecting DBS research and access to care, (4) neuromodulatory approaches for depression, (5) advancing novel hardware, software and imaging methodologies, (6) use of neurophysiological signals in adaptive neurostimulation, and (7) use of more advanced technologies to improve DBS clinical outcomes. There were 178 attendees who participated in a DBS Think Tank survey, which revealed the expansion of DBS into several indications such as obesity, post-traumatic stress disorder, addiction and Alzheimer's disease. This proceedings summarizes the advances discussed at the Eighth Annual DBS Think Tank.

    View details for DOI 10.3389/fnhum.2021.644593

    View details for PubMedID 33953663

  • Septohippocampal transmission from parvalbumin-positive neurons features rapid recovery from synaptic depression. Scientific reports Yi, F. n., Garrett, T. n., Deisseroth, K. n., Haario, H. n., Stone, E. n., Lawrence, J. J. 2021; 11 (1): 2117

    Abstract

    Parvalbumin-containing projection neurons of the medial-septum-diagonal band of Broca ([Formula: see text]) are essential for hippocampal rhythms and learning operations yet are poorly understood at cellular and synaptic levels. We combined electrophysiological, optogenetic, and modeling approaches to investigate [Formula: see text] neuronal properties. [Formula: see text] neurons had intrinsic membrane properties distinct from acetylcholine- and somatostatin-containing MS-DBB subtypes. Viral expression of the fast-kinetic channelrhodopsin ChETA-YFP elicited action potentials to brief (1-2 ms) 470 nm light pulses. To investigate [Formula: see text] transmission, light pulses at 5-50 Hz frequencies generated trains of inhibitory postsynaptic currents (IPSCs) in CA1 stratum oriens interneurons. Using a similar approach, optogenetic activation of local hippocampal PV ([Formula: see text]) neurons generated trains of [Formula: see text]-mediated IPSCs in CA1 pyramidal neurons. Both synapse types exhibited short-term depression (STD) of IPSCs. However, relative to [Formula: see text] synapses, [Formula: see text] synapses possessed lower initial release probability, transiently resisted STD at gamma (20-50 Hz) frequencies, and recovered more rapidly from synaptic depression. Experimentally-constrained mathematical synapse models explored mechanistic differences. Relative to the [Formula: see text] model, the [Formula: see text] model exhibited higher sensitivity to calcium accumulation, permitting a faster rate of calcium-dependent recovery from STD. In conclusion, resistance of [Formula: see text] synapses to STD during short gamma bursts enables robust long-range GABAergic transmission from MS-DBB to hippocampus.

    View details for DOI 10.1038/s41598-020-80245-w

    View details for PubMedID 33483520

  • A Genetically Defined Compartmentalized Striatal Direct Pathway for Negative Reinforcement CELL Xiao, X., Deng, H., Furlan, A., Yang, T., Zhang, X., Hwang, G., Tucciarone, J., Wu, P., He, M., Palaniswamy, R., Ramakrishnan, C., Ritola, K., Hantman, A., Deisseroth, K., Osten, P., Huang, Z., Li, B. 2020; 183 (1): 211-+
  • Distinct Signaling by Ventral Tegmental Area Glutamate, GABA, and Combinatorial Glutamate-GABA Neurons in Motivated Behavior. Cell reports Root, D. H., Barker, D. J., Estrin, D. J., Miranda-Barrientos, J. A., Liu, B., Zhang, S., Wang, H., Vautier, F., Ramakrishnan, C., Kim, Y. S., Fenno, L., Deisseroth, K., Morales, M. 2020; 32 (9): 108094

    Abstract

    Ventral tegmental area (VTA) neurons play roles in reward and aversion. We recently discovered that the VTA has neurons that co-transmit glutamate and GABA (glutamate-GABA co-transmitting neurons), transmit glutamate without GABA (glutamate-transmitting neurons), or transmit GABA without glutamate (GABA-transmitting neurons). However, the functions of these VTA cell types in motivated behavior are unclear. To identify the functions of these VTA cell types, we combine recombinase mouse lines with INTRSECT2.0 vectors to selectively target these neurons. We find that VTA cell types have unique signaling patterns for reward, aversion, and learned cues. Whereas VTA glutamate-transmitting neurons signal cues predicting reward, VTA GABA-transmitting neurons signal cues predicting the absence of reward, and glutamate-GABA co-transmitting neurons signal rewarding and aversive outcomes without signaling learned cues related to those outcomes. Thus, we demonstrate that genetically defined subclasses of VTA glutamate and GABA neurons signal different aspects of motivated behavior.

    View details for DOI 10.1016/j.celrep.2020.108094

    View details for PubMedID 32877676

  • Optogenetic manipulation of an ascending arousal system tunes cortical broadband gamma power and reveals functional deficits relevant to schizophrenia. Molecular psychiatry McNally, J. M., Aguilar, D. D., Katsuki, F., Radzik, L. K., Schiffino, F. L., Uygun, D. S., McKenna, J. T., Strecker, R. E., Deisseroth, K., Spencer, K. M., Brown, R. E. 2020

    Abstract

    Increases in broadband cortical electroencephalogram (EEG) power in the gamma band (30-80Hz) range have been observed in schizophrenia patients and in mouse models of schizophrenia. They are also seen in humans and animals treated with the psychotomimetic agent ketamine. However, the mechanisms which can result in increased broadband gamma power and the pathophysiological implications for cognition and behavior are poorly understood. Here we report that tonic optogenetic manipulation of an ascending arousal system bidirectionally tunes cortical broadband gamma power, allowing on-demand tests of the effect on cortical processing and behavior. Constant, low wattage optogenetic stimulation of basal forebrain (BF) neurons containing the calcium-binding protein parvalbumin (PV) increased broadband gamma frequency power, increased locomotor activity, and impaired novel object recognition. Concomitantly, task-associated gamma band oscillations induced by trains of auditory stimuli, or exposure to novel objects, were impaired, reminiscent of findings in schizophrenia patients. Conversely, tonic optogenetic inhibition of BF-PV neurons partially rescued the elevated broadband gamma power elicited by subanesthetic doses of ketamine. These results support the idea that increased cortical broadband gamma activity leads to impairments in cognition and behavior, and identify BF-PV activity as a modulator of this activity. As such, BF-PV neurons may represent a novel target for pharmacotherapy in disorders such as schizophrenia which involve aberrant increases in cortical broadband gamma activity.

    View details for DOI 10.1038/s41380-020-0840-3

    View details for PubMedID 32690865

  • Optical Interrogation of Memory Related Activity Across the Rodent Default Mode Network Shilyansky, C., Young, N., Ramakrishnan, C., Quirin, S., Deisseroth, K. ELSEVIER SCIENCE INC. 2020: S207
  • Identification of Parallel Functional Domains in the Paraventricular Nucleus of the Thalamus Gao, C., Leng, Y., Ma, J., Rooke, V., Rodriguez-Gonzalez, S., Ramakrishnan, C., Deisseroth, K., Penzo, M. ELSEVIER SCIENCE INC. 2020: S7
  • Activity in grafted human iPS cell-derived cortical neurons integrated in stroke-injured rat brain regulates motor behavior. Proceedings of the National Academy of Sciences of the United States of America Palma-Tortosa, S., Tornero, D., Gronning Hansen, M., Monni, E., Hajy, M., Kartsivadze, S., Aktay, S., Tsupykov, O., Parmar, M., Deisseroth, K., Skibo, G., Lindvall, O., Kokaia, Z. 2020

    Abstract

    Stem cell transplantation can improve behavioral recovery after stroke in animal models but whether stem cell-derived neurons become functionally integrated into stroke-injured brain circuitry is poorly understood. Here we show that intracortically grafted human induced pluripotent stem (iPS) cell-derived cortical neurons send widespread axonal projections to both hemispheres of rats with ischemic lesions in the cerebral cortex. Using rabies virus-based transsynaptic tracing, we find that at 6 mo after transplantation, host neurons in the contralateral somatosensory cortex receive monosynaptic inputs from grafted neurons. Immunoelectron microscopy demonstrates myelination of the graft-derived axons in the corpus callosum and that their terminals form excitatory, glutamatergic synapses on host cortical neurons. We show that the stroke-induced asymmetry in a sensorimotor (cylinder) test is reversed by transplantation. Light-induced inhibition of halorhodopsin-expressing, grafted neurons does not recreate the impairment, indicating that its reversal is not due to neuronal activity in the graft. However, we find bilateral decrease of motor performance in the cylinder test after light-induced inhibition of either grafted or endogenous halorhodopsin-expressing cortical neurons, located in the same area, and after inhibition of endogenous halorhodopsin-expressing cortical neurons by exposure of their axons to light on the contralateral side. Our data indicate that activity in the grafted neurons, probably mediated through transcallosal connections to the contralateral hemisphere, is involved in maintaining normal motor function. This is an example of functional integration of efferent projections from grafted neurons into the stroke-affected brain's neural circuitry, which raises the possibility that such repair might be achievable also in humans affected by stroke.

    View details for DOI 10.1073/pnas.2000690117

    View details for PubMedID 32253308

  • Two genetically, anatomically and functionally distinct cell types segregate across anteroposterior axis of paraventricular thalamus. Nature neuroscience Gao, C., Leng, Y., Ma, J., Rooke, V., Rodriguez-Gonzalez, S., Ramakrishnan, C., Deisseroth, K., Penzo, M. A. 2020

    Abstract

    Unlike the sensory thalamus, studies on the functional organization of the midline and intralaminar nuclei are scarce, and this has hindered the establishment of conceptual models of the function of this brain region. We investigated the functional organization of the paraventricular nucleus of the thalamus (PVT), a midline thalamic structure that is increasingly being recognized as a critical node in the control of diverse processes such as arousal, stress, emotional memory and motivation, in mice. We identify two major classes of PVT neurons-termed type I and type II-that differ in terms of gene expression, anatomy and function. In addition, we demonstrate that type II neurons belong to a previously neglected class of PVT neurons that convey arousal-related information to corticothalamic neurons of the infralimbic cortex. Our results uncover the existence of an arousal-modulated thalamo-corticothalamic loop that links the PVT and the ventromedial prefrontal cortex.

    View details for DOI 10.1038/s41593-019-0572-3

    View details for PubMedID 31932767

  • Amygdala-Midbrain Connections Modulate Appetitive and Aversive Learning. Neuron Steinberg, E. E., Gore, F. n., Heifets, B. D., Taylor, M. D., Norville, Z. C., Beier, K. T., Földy, C. n., Lerner, T. N., Luo, L. n., Deisseroth, K. n., Malenka, R. C. 2020

    Abstract

    The central amygdala (CeA) orchestrates adaptive responses to emotional events. While CeA substrates for defensive behaviors have been studied extensively, CeA circuits for appetitive behaviors and their relationship to threat-responsive circuits remain poorly defined. Here, we demonstrate that the CeA sends robust inhibitory projections to the lateral substantia nigra (SNL) that contribute to appetitive and aversive learning in mice. CeA→SNL neural responses to appetitive and aversive stimuli were modulated by expectation and magnitude consistent with a population-level salience signal, which was required for Pavlovian conditioned reward-seeking and defensive behaviors. CeA→SNL terminal activation elicited reinforcement when linked to voluntary actions but failed to support Pavlovian associations that rely on incentive value signals. Consistent with a disinhibitory mechanism, CeA inputs preferentially target SNL GABA neurons, and CeA→SNL and SNL dopamine neurons respond similarly to salient stimuli. Collectively, our results suggest that amygdala-nigra interactions represent a previously unappreciated mechanism for influencing emotional behaviors.

    View details for DOI 10.1016/j.neuron.2020.03.016

    View details for PubMedID 32294466

  • An Ultra-Sensitive Step-Function Opsin for Minimally Invasive Optogenetic Stimulation in Mice and Macaques. Neuron Gong, X. n., Mendoza-Halliday, D. n., Ting, J. T., Kaiser, T. n., Sun, X. n., Bastos, A. M., Wimmer, R. D., Guo, B. n., Chen, Q. n., Zhou, Y. n., Pruner, M. n., Wu, C. W., Park, D. n., Deisseroth, K. n., Barak, B. n., Boyden, E. S., Miller, E. K., Halassa, M. M., Fu, Z. n., Bi, G. n., Desimone, R. n., Feng, G. n. 2020

    Abstract

    Optogenetics is among the most widely employed techniques to manipulate neuronal activity. However, a major drawback is the need for invasive implantation of optical fibers. To develop a minimally invasive optogenetic method that overcomes this challenge, we engineered a new step-function opsin with ultra-high light sensitivity (SOUL). We show that SOUL can activate neurons located in deep mouse brain regions via transcranial optical stimulation and elicit behavioral changes in SOUL knock-in mice. Moreover, SOUL can be used to modulate neuronal spiking and induce oscillations reversibly in macaque cortex via optical stimulation from outside the dura. By enabling external light delivery, our new opsin offers a minimally invasive tool for manipulating neuronal activity in rodent and primate models with fewer limitations on the depth and size of target brain regions and may further facilitate the development of minimally invasive optogenetic tools for the treatment of neurological disorders.

    View details for DOI 10.1016/j.neuron.2020.03.032

    View details for PubMedID 32353253

  • A Genetically Defined Compartmentalized Striatal Direct Pathway for Negative Reinforcement. Cell Xiao, X. n., Deng, H. n., Furlan, A. n., Yang, T. n., Zhang, X. n., Hwang, G. R., Tucciarone, J. n., Wu, P. n., He, M. n., Palaniswamy, R. n., Ramakrishnan, C. n., Ritola, K. n., Hantman, A. n., Deisseroth, K. n., Osten, P. n., Huang, Z. J., Li, B. n. 2020

    Abstract

    The striosome compartment within the dorsal striatum has been implicated in reinforcement learning and regulation of motivation, but how striosomal neurons contribute to these functions remains elusive. Here, we show that a genetically identified striosomal population, which expresses the Teashirt family zinc finger 1 (Tshz1) and belongs to the direct pathway, drives negative reinforcement and is essential for aversive learning in mice. Contrasting a "conventional" striosomal direct pathway, the Tshz1 neurons cause aversion, movement suppression, and negative reinforcement once activated, and they receive a distinct set of synaptic inputs. These neurons are predominantly excited by punishment rather than reward and represent the anticipation of punishment or the motivation for avoidance. Furthermore, inhibiting these neurons impairs punishment-based learning without affecting reward learning or movement. These results establish a major role of striosomal neurons in behaviors reinforced by punishment and moreover uncover functions of the direct pathway unaccounted for in classic models.

    View details for DOI 10.1016/j.cell.2020.08.032

    View details for PubMedID 32937106

  • Basal Forebrain Parvalbumin Neurons Mediate Arousals from Sleep Induced by Hypercarbia or Auditory Stimuli. Current biology : CB McKenna, J. T., Thankachan, S. n., Uygun, D. S., Shukla, C. n., McNally, J. M., Schiffino, F. L., Cordeira, J. n., Katsuki, F. n., Zant, J. C., Gamble, M. C., Deisseroth, K. n., McCarley, R. W., Brown, R. E., Strecker, R. E., Basheer, R. n. 2020

    Abstract

    The ability to rapidly arouse from sleep is important for survival. However, increased arousals in patients with sleep apnea and other disorders prevent restful sleep and contribute to cognitive, metabolic, and physiologic dysfunction [1, 2]. Little is currently known about which neural systems mediate these brief arousals, hindering the development of treatments that restore normal sleep. The basal forebrain (BF) receives inputs from many nuclei of the ascending arousal system, including the brainstem parabrachial neurons, which promote arousal in response to elevated blood carbon dioxide levels, as seen in sleep apnea [3]. Optical inhibition of the terminals of parabrachial neurons in the BF impairs cortical arousals to hypercarbia [4], but which BF cell types mediate cortical arousals in response to hypercarbia or other sensory stimuli is unknown. Here, we tested the role of BF parvalbumin (PV) neurons in arousal using optogenetic techniques in mice. Optical stimulation of BF-PV neurons produced rapid transitions to wakefulness from non-rapid eye movement (NREM) sleep but did not affect REM-wakefulness transitions. Unlike previous studies of BF glutamatergic and cholinergic neurons, arousals induced by stimulation of BF-PV neurons were brief and only slightly increased total wake time, reminiscent of clinical findings in sleep apnea [5, 6]. Bilateral optical inhibition of BF-PV neurons increased the latency to arousal produced by exposure to hypercarbia or auditory stimuli. Thus, BF-PV neurons are an important component of the brain circuitry that generates brief arousals from sleep in response to stimuli, which may indicate physiological dysfunction or danger to the organism.

    View details for DOI 10.1016/j.cub.2020.04.029

    View details for PubMedID 32413301

  • High-speed interferometric imaging reveals dynamics of neuronal deformation during the action potential. Proceedings of the National Academy of Sciences of the United States of America Ling, T. n., Boyle, K. C., Zuckerman, V. n., Flores, T. n., Ramakrishnan, C. n., Deisseroth, K. n., Palanker, D. n. 2020

    Abstract

    Neurons undergo nanometer-scale deformations during action potentials, and the underlying mechanism has been actively debated for decades. Previous observations were limited to a single spot or the cell boundary, while movement across the entire neuron during the action potential remained unclear. Here we report full-field imaging of cellular deformations accompanying the action potential in mammalian neuron somas (-1.8 to 1.4 nm) and neurites (-0.7 to 0.9 nm), using high-speed quantitative phase imaging with a temporal resolution of 0.1 ms and an optical path length sensitivity of <4 pm per pixel. The spike-triggered average, synchronized to electrical recording, demonstrates that the time course of the optical phase changes closely matches the dynamics of the electrical signal. Utilizing the spatial and temporal correlations of the phase signals across the cell, we enhance the detection and segmentation of spiking cells compared to the shot-noise-limited performance of single pixels. Using three-dimensional (3D) cellular morphology extracted via confocal microscopy, we demonstrate that the voltage-dependent changes in the membrane tension induced by ionic repulsion can explain the magnitude, time course, and spatial features of the phase imaging. Our full-field observations of the spike-induced deformations shed light upon the electromechanical coupling mechanism in electrogenic cells and open the door to noninvasive label-free imaging of neural signaling.

    View details for DOI 10.1073/pnas.1920039117

    View details for PubMedID 32341158

  • Cerebellar nuclei evolved by repeatedly duplicating a conserved cell-type set. Science (New York, N.Y.) Kebschull, J. M., Richman, E. B., Ringach, N. n., Friedmann, D. n., Albarran, E. n., Kolluru, S. S., Jones, R. C., Allen, W. E., Wang, Y. n., Cho, S. W., Zhou, H. n., Ding, J. B., Chang, H. Y., Deisseroth, K. n., Quake, S. R., Luo, L. n. 2020; 370 (6523)

    Abstract

    How have complex brains evolved from simple circuits? Here we investigated brain region evolution at cell-type resolution in the cerebellar nuclei, the output structures of the cerebellum. Using single-nucleus RNA sequencing in mice, chickens, and humans, as well as STARmap spatial transcriptomic analysis and whole-central nervous system projection tracing, we identified a conserved cell-type set containing two region-specific excitatory neuron classes and three region-invariant inhibitory neuron classes. This set constitutes an archetypal cerebellar nucleus that was repeatedly duplicated to form new regions. The excitatory cell class that preferentially funnels information to lateral frontal cortices in mice becomes predominant in the massively expanded human lateral nucleus. Our data suggest a model of brain region evolution by duplication and divergence of entire cell-type sets.

    View details for DOI 10.1126/science.abd5059

    View details for PubMedID 33335034

  • Striosomes Mediate Value-Based Learning Vulnerable in Age and a Huntington's Disease Model. Cell Friedman, A. n., Hueske, E. n., Drammis, S. M., Toro Arana, S. E., Nelson, E. D., Carter, C. W., Delcasso, S. n., Rodriguez, R. X., Lutwak, H. n., DiMarco, K. S., Zhang, Q. n., Rakocevic, L. I., Hu, D. n., Xiong, J. K., Zhao, J. n., Gibb, L. G., Yoshida, T. n., Siciliano, C. A., Diefenbach, T. J., Ramakrishnan, C. n., Deisseroth, K. n., Graybiel, A. M. 2020

    Abstract

    Learning valence-based responses to favorable and unfavorable options requires judgments of the relative value of the options, a process necessary for species survival. We found, using engineered mice, that circuit connectivity and function of the striosome compartment of the striatum are critical for this type of learning. Calcium imaging during valence-based learning exhibited a selective correlation between learning and striosomal but not matrix signals. This striosomal activity encoded discrimination learning and was correlated with task engagement, which, in turn, could be regulated by chemogenetic excitation and inhibition. Striosomal function during discrimination learning was disturbed with aging and severely so in a mouse model of Huntington's disease. Anatomical and functional connectivity of parvalbumin-positive, putative fast-spiking interneurons (FSIs) to striatal projection neurons was enhanced in striosomes compared with matrix in mice that learned. Computational modeling of these findings suggests that FSIs can modulate the striosomal signal-to-noise ratio, crucial for discrimination and learning.

    View details for DOI 10.1016/j.cell.2020.09.060

    View details for PubMedID 33113354

  • An Open Resource for Non-human Primate Optogenetics. Neuron Tremblay, S. n., Acker, L. n., Afraz, A. n., Albaugh, D. L., Amita, H. n., Andrei, A. R., Angelucci, A. n., Aschner, A. n., Balan, P. F., Basso, M. A., Benvenuti, G. n., Bohlen, M. O., Caiola, M. J., Calcedo, R. n., Cavanaugh, J. n., Chen, Y. n., Chen, S. n., Chernov, M. M., Clark, A. M., Dai, J. n., Debes, S. R., Deisseroth, K. n., Desimone, R. n., Dragoi, V. n., Egger, S. W., Eldridge, M. A., El-Nahal, H. G., Fabbrini, F. n., Federer, F. n., Fetsch, C. R., Fortuna, M. G., Friedman, R. M., Fujii, N. n., Gail, A. n., Galvan, A. n., Ghosh, S. n., Gieselmann, M. A., Gulli, R. A., Hikosaka, O. n., Hosseini, E. A., Hu, X. n., Hüer, J. n., Inoue, K. I., Janz, R. n., Jazayeri, M. n., Jiang, R. n., Ju, N. n., Kar, K. n., Klein, C. n., Kohn, A. n., Komatsu, M. n., Maeda, K. n., Martinez-Trujillo, J. C., Matsumoto, M. n., Maunsell, J. H., Mendoza-Halliday, D. n., Monosov, I. E., Muers, R. S., Nurminen, L. n., Ortiz-Rios, M. n., O'Shea, D. J., Palfi, S. n., Petkov, C. I., Pojoga, S. n., Rajalingham, R. n., Ramakrishnan, C. n., Remington, E. D., Revsine, C. n., Roe, A. W., Sabes, P. N., Saunders, R. C., Scherberger, H. n., Schmid, M. C., Schultz, W. n., Seidemann, E. n., Senova, Y. S., Shadlen, M. N., Sheinberg, D. L., Siu, C. n., Smith, Y. n., Solomon, S. S., Sommer, M. A., Spudich, J. L., Stauffer, W. R., Takada, M. n., Tang, S. n., Thiele, A. n., Treue, S. n., Vanduffel, W. n., Vogels, R. n., Whitmire, M. P., Wichmann, T. n., Wurtz, R. H., Xu, H. n., Yazdan-Shahmorad, A. n., Shenoy, K. V., DiCarlo, J. J., Platt, M. L. 2020

    Abstract

    Optogenetics has revolutionized neuroscience in small laboratory animals, but its effect on animal models more closely related to humans, such as non-human primates (NHPs), has been mixed. To make evidence-based decisions in primate optogenetics, the scientific community would benefit from a centralized database listing all attempts, successful and unsuccessful, of using optogenetics in the primate brain. We contacted members of the community to ask for their contributions to an open science initiative. As of this writing, 45 laboratories around the world contributed more than 1,000 injection experiments, including precise details regarding their methods and outcomes. Of those entries, more than half had not been published. The resource is free for everyone to consult and contribute to on the Open Science Framework website. Here we review some of the insights from this initial release of the database and discuss methodological considerations to improve the success of optogenetic experiments in NHPs.

    View details for DOI 10.1016/j.neuron.2020.09.027

    View details for PubMedID 33080229

  • Publisher Correction: Development of an optogenetic toolkit for neural circuit dissection in squirrel monkeys. Scientific reports O'Shea, D. J., Kalanithi, P., Ferenczi, E. A., Hsueh, B., Chandrasekaran, C., Goo, W., Diester, I., Ramakrishnan, C., Kaufman, M. T., Ryu, S. I., Yeom, K. W., Deisseroth, K., Shenoy, K. V. 2019; 9 (1): 18775

    Abstract

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

    View details for DOI 10.1038/s41598-019-55025-w

    View details for PubMedID 31801956

  • Excitation of diverse classes of cholecystokinin interneurons in the basolateral amygdala facilitates fear extinction. eNeuro Rovira-Esteban, L., Gunduz-Cinar, O., Bukalo, O., Limoges, A., Brockway, E., Muller, K., Fenno, L., Kim, Y. S., Ramakrishnan, C., Andrasi, T., Deisseroth, K., Holmes, A., Hajos, N. 2019

    Abstract

    There is growing evidence that interneurons orchestrate neural activity and plasticity in corticoamygdala circuits to regulate fear behaviors. However, defining the precise role of cholecystokinin-expressing interneurons (CCK INs) remains elusive due to the technical challenge of parsing this population from CCK-expressing principal neurons (CCK PNs). Here we used an intersectional genetic strategy in CCK-Cre;Dlx5/6-Flpe double-transgenic mice to study the anatomical, molecular and electrophysiological properties of CCK INs in the basal amygdala (BA) and optogenetically manipulate these cells in fear extinction. Electrophysiological recordings confirmed that this strategy targeted GABAergic cells and that a significant proportion expressed functional cannabinoid CB1 receptors; a defining characteristic of CCK-expressing basket cells. However, immunostaining showed that subsets of the genetically-targeted cells expressed either neuropeptide Y (NPY) (29%) or parvalbumin (PV) (17%), but not somatostatin (SOM) or CaMKII-alpha. Further morphological and electrophysiological analyses showed that four interneuron types could be identified among the EYFP-expressing cells: CCK/CB1R-expressing basket cells, neurogliaform cells, PV+ basket and PV+ axo-axonic cells. At the behavioral level, in vivo optogenetic photostimulation of the targeted population during extinction acquisition led to reduced freezing on a light-free extinction retrieval test, indicating extinction memory facilitation; whereas photosilencing was without effect. Conversely, non-selective (i.e., inclusive of INs and PNs) photostimulation or photosilencing of CCK-targeted cells, using CCK-Cre single-transgenic mice, impaired extinction. These data reveal an unexpectedly high degree of phenotypic complexity in a unique population of extinction-modulating BA INs.Significance statement Distinct types of interneurons in the basolateral amygdala (BA) are known to control principal cell activity, allowing complex behaviors. Despite their importance, the role of cholecystokinin (CCK)-expressing inhibitory cells remains unknown. In this work, we could specifically alter the function of CCK-expressing interneurons in the BA by using an INTRSECT viral strategy. Using a combination of anatomical and electrophysiological methods, we found that CCK+ interneurons in the BA are comprised of CB1R-expressing basket cells, neurogliaform cells, parvalbumin-expressing basket as well as axo-axonic cells. Importantly, we provided the first direct evidence that CCK-expressing interneurons in the BA can modulate fear extinction learning. Our data thus show that CCK is expressed in functionally diverse interneuron populations, positioned to impact amygdala operation.

    View details for DOI 10.1523/ENEURO.0220-19.2019

    View details for PubMedID 31636080

  • Mapping Brain-Wide Afferent Inputs of Parvalbumin-Expressing GABAergic Neurons in Barrel Cortex Reveals Local and Long-Range Circuit Motifs. Cell reports Hafner, G., Witte, M., Guy, J., Subhashini, N., Fenno, L. E., Ramakrishnan, C., Kim, Y. S., Deisseroth, K., Callaway, E. M., Oberhuber, M., Conzelmann, K., Staiger, J. F. 2019; 28 (13): 3450

    Abstract

    Parvalbumin (PV)-expressing GABAergic neurons are the largest class of inhibitory neocortical cells. We visualize brain-wide, monosynaptic inputs to PV neurons in mouse barrel cortex. We develop intersectional rabies virus tracing to specifically target GABAergic PV cells and exclude a small fraction of excitatory PV cells from our starter population. Local inputs are mainly from layer (L) IV and excitatory cells. A small number of inhibitory inputs originate from LI neurons, which connect to LII/III PV neurons. Long-range inputs originate mainly from other sensory cortices and the thalamus. In visual cortex, most transsynaptically labeled neurons are located in LIV, which contains a molecularly mixed population of projection neurons with putative functional similarity to LIII neurons. This study expands our knowledge of the brain-wide circuits in which PV neurons are embedded and introduces intersectional rabies virus tracing as an applicable tool to dissect the circuitry of more clearly defined cell types.

    View details for DOI 10.1016/j.celrep.2019.08.064

    View details for PubMedID 31553913

  • A neuronal circuit for activating descending modulation of neuropathic pain. Nature neuroscience Huang, J., Gadotti, V. M., Chen, L., Souza, I. A., Huang, S., Wang, D., Ramakrishnan, C., Deisseroth, K., Zhang, Z., Zamponi, G. W. 2019

    Abstract

    Neuropathic pain can be a debilitating condition with both sensory and affective components, the underlying brain circuitry of which remains poorly understood. In the present study, a basolateral amygdala (BLA)-prefrontal cortex (PFC)-periaqueductal gray (PAG)-spinal cord pathway was identified that is critical for the development of mechanical and thermal hypersensitivity after peripheral nerve injury. It was shown that nerve injury strengthens synaptic input from the BLA onto inhibitory interneurons located in the prelimbic medial PFC, by virtue of reduced endocannabinoid modulation. These augmented synaptic connections mediate a feedforward inhibition of projections from the PFC to the ventrolateral PAG region and its downstream targets. Optogenetic approaches combined with in vivo pharmacology reveal that these BLA-PFC-PAG connections alter pain behaviors by reducing descending noradrenergic and serotoninergic modulation of spinal pain signals. Thus, a long-range brain circuit was identified that is crucial for pain processing and that can potentially be exploited toward targeting neuropathic pain.

    View details for DOI 10.1038/s41593-019-0481-5

    View details for PubMedID 31501573

  • Multimodal characterization of the human nucleus accumbens NEUROIMAGE Cartmell, S. D., Tian, Q., Thio, B. J., Leuze, C., Ye, L., Williams, N. R., Yang, G., Ben-Dor, G., Deisseroth, K., Grill, W. M., McNab, J. A., Halpern, C. H. 2019; 198: 137–49
  • A hypothalamus-habenula circuit controls aversion MOLECULAR PSYCHIATRY Lazaridis, I., Tzortzi, O., Weglage, M., Martin, A., Xuan, Y., Parent, M., Johansson, Y., Fuzik, J., Furth, D., Fenno, L. E., Ramakrishnan, C., Silberberg, G., Deisseroth, K., Carlen, M., Meletis, K. 2019; 24 (9): 1351–68
  • Prefrontal cortex neuronal ensembles encoding fear drive fear expression during long-term memory retrieval. Scientific reports Giannotti, G., Heinsbroek, J. A., Yue, A. J., Deisseroth, K., Peters, J. 2019; 9 (1): 10709

    Abstract

    The prefrontal cortex is an important regulator of fear expression in humans and rodents. Specifically, the rodent prelimbic (PL) prefrontal cortex drives fear expression during both encoding and retrieval of fear memory. Neuronal ensembles have been proposed to function as memory encoding units, and their re-activation is thought to be necessary for memory retrieval and expression of conditioned behavior. However, it remains unclear whether PL cortex neuronal ensembles that encode fear memory contribute to long-term fear expression during memory retrieval. To address this, we employed a viral-mediated TRAP (Targeted Recombination in Active Population) technology to target PL cortex ensembles active during fear conditioning and expressed the inhibitory Gi-DREADD in fear-encoding ensembles. Male and female rats were trained to lever press for food and subjected to Pavlovian delay fear conditioning, then 28 days later, they underwent a fear memory retrieval test. Chemogenetic inhibition of TRAPed PL cortex ensembles reduced conditioned suppression of food seeking in females, but not males. Neither context nor tone freezing behavior was altered by this manipulation during the same retrieval test. Thus, fear-encoding ensembles in PL cortex drive long-term fear expression in a sex and fear modality dependent manner.

    View details for DOI 10.1038/s41598-019-47095-7

    View details for PubMedID 31341176

  • Rational Engineering of XCaMPs, a Multicolor GECI Suite for In Vivo Imaging of Complex Brain Circuit Dynamics CELL Inoue, M., Takeuchi, A., Manita, S., Horigane, S., Sakamoto, M., Kawakami, R., Yamaguchi, K., Otomo, K., Yokoyama, H., Kim, R., Yokoyama, T., Takemoto-Kimura, S., Abe, M., Okamura, M., Kondo, Y., Quirin, S., Ramakrishnan, C., Imamura, T., Sakimura, K., Nemoto, T., Kano, M., Fujii, H., Deisseroth, K., Kitamura, K., Bito, H. 2019; 177 (5): 1346-+
  • Thirst regulates motivated behavior through modulation of brainwide neural population dynamics. Science (New York, N.Y.) Allen, W. E., Chen, M. Z., Pichamoorthy, N., Tien, R. H., Pachitariu, M., Luo, L., Deisseroth, K. 2019

    Abstract

    Physiological needs produce motivational drives, such as thirst and hunger, that regulate behaviors essential to survival. Hypothalamic neurons sense these needs and must coordinate relevant brainwide neuronal activity to produce the appropriate behavior. We studied dynamics from ~24,000 neurons in 34 brain regions during thirst-motivated choice behavior, as mice consumed water and became sated. Water-predicting sensory cues elicited activity that rapidly spread throughout the brain of thirsty animals. These dynamics were gated by a brainwide mode of population activity that encoded motivational state. Focal optogenetic activation of hypothalamic thirst-sensing neurons, after satiation, returned global activity to the pre-satiation state. Thus, motivational states specify initial conditions determining how a brainwide dynamical system transforms sensory input into behavioral output.

    View details for PubMedID 30948440

  • Dopamine Modulation of Prefrontal Cortex Activity Is Manifold and Operates at Multiple Temporal and Spatial Scales CELL REPORTS Lohani, S., Martig, A. K., Deisseroth, K., Witten, I. B., Moghaddam, B. 2019; 27 (1): 99-+
  • Dopamine Modulation of Prefrontal Cortex Activity Is Manifold and Operates at Multiple Temporal and Spatial Scales. Cell reports Lohani, S., Martig, A. K., Deisseroth, K., Witten, I. B., Moghaddam, B. 2019; 27 (1): 99

    Abstract

    Although the function of dopamine in subcortical structures is largely limited to reward and movement, dopamine neurotransmission in the prefrontal cortex (PFC) is critical to a multitude of temporally and functionally diverse processes, such as attention, working memory, behavioral flexibility, action planning, and sustained motivational and affective states. How does dopamine influence computation of these temporally complex functions? We find causative links between sustained and burst patterns of phasic dopamine neuron activation and modulation of medial PFC neuronal activity at multiple spatiotemporal scales. These include a multidirectional and weak impact on individual neuron rateactivity but a robust influence on coordinated ensemble activity, gamma oscillations, and gamma-theta coupling that persisted for minutes. In addition, PFC network responses to burst pattern of dopamine firing wereselectively strengthened in behaviorally active states. This multiplex mode of modulation by dopamine input may enable PFC to compute and generate spatiotemporally diverse and specialized outputs.

    View details for PubMedID 30943418

  • Neuronal Dynamics Regulating Brain and Behavioral State Transitions. Cell Andalman, A. S., Burns, V. M., Lovett-Barron, M., Broxton, M., Poole, B., Yang, S. J., Grosenick, L., Lerner, T. N., Chen, R., Benster, T., Mourrain, P., Levoy, M., Rajan, K., Deisseroth, K. 2019

    Abstract

    Prolonged behavioral challenges can cause animals to switch from active to passive coping strategies to manage effort-expenditure during stress; such normally adaptive behavioral state transitions can become maladaptive in psychiatric disorders such as depression. The underlying neuronal dynamics and brainwide interactions important for passive coping have remained unclear. Here, we develop a paradigm to study these behavioral state transitions at cellular-resolution across the entire vertebrate brain. Using brainwide imaging in zebrafish, we observed that the transition to passive coping is manifested by progressive activation of neurons in the ventral (lateral) habenula. Activation of these ventral-habenula neurons suppressed downstream neurons in the serotonergic raphe nucleus and caused behavioral passivity, whereas inhibition ofthese neurons prevented passivity. Data-driven recurrent neural network modeling pointed to altered intra-habenula interactions as a contributory mechanism. These results demonstrate ongoing encoding of experience features in the habenula, which guides recruitment of downstream networks and imposes a passive coping behavioral strategy.

    View details for PubMedID 31031000

  • Thalamic Reticular Nucleus Parvalbumin Neurons Regulate Sleep Spindles and Electrophysiological Aspects of Schizophrenia in Mice. Scientific reports Thankachan, S., Katsuki, F., McKenna, J. T., Yang, C., Shukla, C., Deisseroth, K., Uygun, D. S., Strecker, R. E., Brown, R. E., McNally, J. M., Basheer, R. 2019; 9 (1): 3607

    Abstract

    The thalamic reticular nucleus (TRN) is implicated in schizophrenia pathology. However, it remains unclear whether alterations of TRN activity can account for abnormal electroencephalographic activity observed in patients, namely reduced spindles (10-15Hz) during sleep and increased delta (0.5-4Hz) and gamma-band activity (30-80Hz) during wakefulness. Here, we utilized optogenetic and reverse-microdialysis approaches to modulate activity of the major subpopulation of TRN GABAergic neurons, which express the calcium-binding protein parvalbumin (PV), and are implicated in schizophrenia dysfunction. An automated algorithm with enhanced efficiency and reproducibility compared to manual detection was used for sleep spindle assessment. A novel, low power, waxing-and-waning optogenetic stimulation paradigm preferentially induced spindles that were indistinguishable from spontaneously occurring sleep spindles without altering the behavioral state, when compared to a single pulse laser stimulation used by us and others. Direct optogenetic inhibition of TRN-PV neurons was ineffective in blocking spindles but increased both wakefulness and cortical delta/gamma activity, as well as impaired the 40Hz auditory steady-state response. For the first time we demonstrate that spindle density is markedly reduced by (i) optogenetic stimulation of a major GABA/PV inhibitory input to TRN arising from basal forebrain parvalbumin neurons (BF-PV) and; (ii) localized pharmacological inhibition of low-threshold calcium channels, implicated as a genetic risk factor for schizophrenia. Together with clinical findings, our results support impaired TRN-PV neuron activity as a potential cause of schizophrenia-linked abnormalities in cortical delta, gamma, and spindle activity. Modulation of the BF-PV input to TRN may improve these neural abnormalities.

    View details for PubMedID 30837664

  • Thalamic Reticular Nucleus Parvalbumin Neurons Regulate Sleep Spindles and Electrophysiological Aspects of Schizophrenia in Mice SCIENTIFIC REPORTS Thankachan, S., Katsuki, F., McKenna, J. T., Yang, C., Shukla, C., Deisseroth, K., Uygun, D. S., Strecker, R. E., Brown, R. E., McNally, J. M., Basheer, R. 2019; 9
  • Fast near-whole-brain imaging in adult Drosophila during responses to stimuli and behavior. PLoS biology Aimon, S., Katsuki, T., Jia, T., Grosenick, L., Broxton, M., Deisseroth, K., Sejnowski, T. J., Greenspan, R. J. 2019; 17 (2): e2006732

    Abstract

    Whole-brain recordings give us a global perspective of the brain in action. In this study, we describe a method using light field microscopy to record near-whole brain calcium and voltage activity at high speed in behaving adult flies. We first obtained global activity maps for various stimuli and behaviors. Notably, we found that brain activity increased on a global scale when the fly walked but not when it groomed. This global increase with walking was particularly strong in dopamine neurons, which showed little activity otherwise. Second, we extracted maps of spatially distinct sources of activity as well as their time series using principal component analysis and independent component analysis. The characteristic shapes in the maps matched the anatomy of subneuropil regions and, in some cases, a specific neuron type. Brain structures that responded to light and odor were consistent with previous reports, confirming the new technique's validity. We also observed previously uncharacterized behavior-related activity as well as patterns of spontaneous voltage activity.

    View details for PubMedID 30768592

  • A hypothalamus-habenula circuit controls aversion. Molecular psychiatry Lazaridis, I., Tzortzi, O., Weglage, M., Martin, A., Xuan, Y., Parent, M., Johansson, Y., Fuzik, J., Furth, D., Fenno, L. E., Ramakrishnan, C., Silberberg, G., Deisseroth, K., Carlen, M., Meletis, K. 2019

    Abstract

    Encoding and predicting aversive events are critical functions of circuits that support survival and emotional well-being. Maladaptive circuit changes in emotional valence processing can underlie the pathophysiology of affective disorders. The lateral habenula (LHb) has been linked to aversion and mood regulation through modulation of the dopamine and serotonin systems. We have defined the identity and function of glutamatergic (Vglut2) control of the LHb, comparing the role of inputs originating in the globus pallidus internal segment (GPi), and lateral hypothalamic area (LHA), respectively. We found that LHb-projecting LHA neurons, and not the proposed GABA/glutamate co-releasing GPi neurons, are responsible for encoding negative value. Monosynaptic rabies tracing of the presynaptic organization revealed a predominantly limbic input onto LHA Vglut2 neurons, while sensorimotor inputs were more prominent onto GABA/glutamate co-releasing GPi neurons. We further recorded the activity of LHA Vglut2 neurons, by imaging calcium dynamics in response to appetitive versus aversive events in conditioning paradigms. LHA Vglut2 neurons formed activity clusters representing distinct reward or aversion signals, including a population that responded to mild foot shocks and predicted aversive events. We found that the LHb-projecting LHA Vglut2 neurons encode negative valence and rapidly develop a prediction signal for negative events. These findings establish the glutamatergic LHA-LHb circuit as a critical node in value processing.

    View details for PubMedID 30755721

  • Two eARCHT3.0 Lines for Optogenetic Silencing of Dopaminergic and Serotonergic Neurons FRONTIERS IN NEURAL CIRCUITS Krol, A., Lopez-Huerta, V. G., Corey, T. C., Deisseroth, K., Ting, J. T., Feng, G. 2019; 13
  • Fast near-whole-brain imaging in adult Drosophila during responses to stimuli and behavior PLOS BIOLOGY Aimon, S., Katsuki, T., Jia, T., Grosenick, L., Broxton, M., Deisseroth, K., Sejnowski, T. J., Greenspan, R. J. 2019; 17 (2)
  • Interacting neural ensembles in orbitofrontal cortex for social and feeding behaviour. Nature Jennings, J. H., Kim, C. K., Marshel, J. H., Raffiee, M., Ye, L., Quirin, S., Pak, S., Ramakrishnan, C., Deisseroth, K. 2019

    Abstract

    Categorically distinct basic drives (for example, for social versus feeding behaviour1-3) can exert potent influences on each other; such interactions are likely to have important adaptive consequences (such as appropriate regulation of feeding in the context of social hierarchies) and can become maladaptive (such as in clinical settings involving anorexia). It is known that neural systems regulating natural and adaptive caloric intake, and those regulating social behaviours, involve related circuitry4-7, butthe causal circuit mechanisms of these drive adjudications are not clear. Here we investigate the causal role in behaviour of cellular-resolution experience-specific neuronal populations in the orbitofrontal cortex, a major reward-processing hub thatcontains diverse activity-specific neuronal populations that respond differentially to various aspects of caloric intake8-13 and social stimuli14,15. We coupled genetically encoded activity imaging with thedevelopment and application of methods for optogenetic control of multiple individually defined cells, to both optically monitor and manipulate the activity of many orbitofrontal cortex neurons at the single-cell level in real time during rewarding experiences (caloric consumption and socialinteraction). We identified distinct populations within the orbitofrontal cortex that selectively responded to either caloric rewards or social stimuli, and found that activity of individually specified naturally feeding-responsive neurons was causally linked to increased feeding behaviour; this effect was selective as, by contrast, single-cell resolution activation of naturally social-responsive neurons inhibited feeding, and activation of neurons responsive to neither feeding nor social stimuli did not alter feeding behaviour. These results reveal the presence of potent cellular-level subnetworks within the orbitofrontal cortex that can be precisely engaged to bidirectionally control feeding behaviours subject to, for example, social influences.

    View details for PubMedID 30651638

  • A Neural Circuit Mechanism for Encoding Aversive Stimuli in the Mesolimbic Dopamine System NEURON de Jong, J. W., Afjei, S., Dorocic, I., Peck, J. R., Liu, C., Kim, C. K., Tian, L., Deisseroth, K., Lammel, S. 2019; 101 (1): 133-+
  • Multimodal image registration and connectivity analysis for integration of connectomic data from microscopy to MRI. Nature communications Goubran, M. n., Leuze, C. n., Hsueh, B. n., Aswendt, M. n., Ye, L. n., Tian, Q. n., Cheng, M. Y., Crow, A. n., Steinberg, G. K., McNab, J. A., Deisseroth, K. n., Zeineh, M. n. 2019; 10 (1): 5504

    Abstract

    3D histology, slice-based connectivity atlases, and diffusion MRI are common techniques to map brain wiring. While there are many modality-specific tools to process these data, there is a lack of integration across modalities. We develop an automated resource that combines histologically cleared volumes with connectivity atlases and MRI, enabling the analysis of histological features across multiple fiber tracts and networks, and their correlation with in-vivo biomarkers. We apply our pipeline in a murine stroke model, demonstrating not only strong correspondence between MRI abnormalities and CLARITY-tissue staining, but also uncovering acute cellular effects in areas connected to the ischemic core. We provide improved maps of connectivity by quantifying projection terminals from CLARITY viral injections, and integrate diffusion MRI with CLARITY viral tracing to compare connectivity maps across scales. Finally, we demonstrate tract-level histological changes of stroke through this multimodal integration. This resource can propel investigations of network alterations underlying neurological disorders.

    View details for DOI 10.1038/s41467-019-13374-0

    View details for PubMedID 31796741

  • Stretchable and Fully Degradable Semiconductors for Transient Electronics. ACS central science Tran, H. n., Feig, V. R., Liu, K. n., Wu, H. C., Chen, R. n., Xu, J. n., Deisseroth, K. n., Bao, Z. n. 2019; 5 (11): 1884–91

    Abstract

    The next materials challenge in organic stretchable electronics is the development of a fully degradable semiconductor that maintains stable electrical performance under strain. Herein, we decouple the design of stretchability and transience by harmonizing polymer physics principles and molecular design in order to demonstrate for the first time a material that simultaneously possesses three disparate attributes: semiconductivity, intrinsic stretchability, and full degradability. We show that we can design acid-labile semiconducting polymers to appropriately phase segregate within a biodegradable elastomer, yielding semiconducting nanofibers that concurrently enable controlled transience and strain-independent transistor mobilities. Along with the future development of suitable conductors and device integration advances, we anticipate that these materials could be used to build fully biodegradable diagnostic or therapeutic devices that reside inside the body temporarily, or environmental monitors that are placed in the field and break down when they are no longer needed. This fully degradable semiconductor represents a promising advance toward developing multifunctional materials for skin-inspired electronic devices that can address previously inaccessible challenges and in turn create new technologies.

    View details for DOI 10.1021/acscentsci.9b00850

    View details for PubMedID 31807690

    View details for PubMedCentralID PMC6891860

  • Investigating the feasibility of channelrhodopsin variants for nanoscale optogenetics. Neurophotonics Stahlberg, M. A., Ramakrishnan, C., Willig, K. I., Boyden, E. S., Deisseroth, K., Dean, C. 2019; 6 (1): 015007

    Abstract

    Optogenetics has revolutionized the study of circuit function in the brain, by allowing activation of specific ensembles of neurons by light. However, this technique has not yet been exploited extensively at the subcellular level. Here, we test the feasibility of a focal stimulation approach using stimulated emission depletion/reversible saturable optical fluorescence transitions-like illumination, whereby switchable light-gated channels are focally activated by a laser beam of one wavelength and deactivated by an overlapping donut-shaped beam of a different wavelength, confining activation to a center focal region. This method requires that activated channelrhodopsins are inactivated by overlapping illumination of a distinct wavelength and that photocurrents are large enough to be detected at the nanoscale. In tests of current optogenetic tools, we found that ChR2 C128A/H134R/T159C and CoChR C108S and C108S/D136A-activated with 405-nm light and inactivated by coillumination with 594-nm light-and C1V1 E122T/C167S-activated by 561-nm light and inactivated by 405-nm light-were most promising in terms of highest photocurrents and efficient inactivation with coillumination. Although further engineering of step-function channelrhodopsin variants with higher photoconductances will be required to employ this approach at the nanoscale, our findings provide a framework to guide future development of this technique.

    View details for PubMedID 30854405

  • Two eARCHT3.0 Lines for Optogenetic Silencing of Dopaminergic and Serotonergic Neurons. Frontiers in neural circuits Krol, A., Lopez-Huerta, V. G., Corey, T. E., Deisseroth, K., Ting, J. T., Feng, G. 2019; 13: 4

    Abstract

    Dopaminergic and serotonergic neurons modulate and control processes ranging from reward signaling to regulation of motor outputs. Further, dysfunction of these neurons is involved in both degenerative and psychiatric disorders. Elucidating the roles of these neurons has been greatly facilitated by bacterial artificial chromosome (BAC) transgenic mouse lines expressing channelrhodopsin to readily enable cell-type specific activation. However, corresponding lines to silence these monoaminergic neurons have been lacking. We have generated two BAC transgenic mouse lines expressing the outward proton pump, enhanced ArchT3.0 (eArchT3.0), and GFP under control of the regulatory elements of either the dopamine transporter (DAT; Jax# 031663) or the tryptophan hydroxylase 2 (TPH2; Jax# 031662) gene locus. We demonstrate highly faithful and specific expression of these lines in dopaminergic and serotonergic neurons respectively. Additionally we validate effective and sensitive eArchT3.0-mediated silencing of these neurons using slice electrophysiology as well as with a well-established behavioral assay. These new transgenic tools will help expedite the study of dopaminergic and serotonergic system function in normal behavior and disease.

    View details for PubMedID 30774584

  • Rational Engineering of XCaMPs, a Multicolor GECI Suite for In Vivo Imaging of Complex Brain Circuit Dynamics. Cell Inoue, M. n., Takeuchi, A. n., Manita, S. n., Horigane, S. I., Sakamoto, M. n., Kawakami, R. n., Yamaguchi, K. n., Otomo, K. n., Yokoyama, H. n., Kim, R. n., Yokoyama, T. n., Takemoto-Kimura, S. n., Abe, M. n., Okamura, M. n., Kondo, Y. n., Quirin, S. n., Ramakrishnan, C. n., Imamura, T. n., Sakimura, K. n., Nemoto, T. n., Kano, M. n., Fujii, H. n., Deisseroth, K. n., Kitamura, K. n., Bito, H. n. 2019

    Abstract

    To decipher dynamic brain information processing, current genetically encoded calcium indicators (GECIs) are limited in single action potential (AP) detection speed, combinatorial spectral compatibility, and two-photon imaging depth. To address this, here, we rationally engineered a next-generation quadricolor GECI suite, XCaMPs. Single AP detection was achieved within 3-10 ms of spike onset, enabling measurements of fast-spike trains in parvalbumin (PV)-positive interneurons in the barrel cortex in vivo and recording three distinct (two inhibitory and one excitatory) ensembles during pre-motion activity in freely moving mice. In vivo paired recording of pre- and postsynaptic firing revealed spatiotemporal constraints of dendritic inhibition in layer 1 in vivo, between axons of somatostatin (SST)-positive interneurons and apical tufts dendrites of excitatory pyramidal neurons. Finally, non-invasive, subcortical imaging using red XCaMP-R uncovered somatosensation-evoked persistent activity in hippocampal CA1 neurons. Thus, the XCaMPs offer a critical enhancement of solution space in studies of complex neuronal circuit dynamics. VIDEO ABSTRACT.

    View details for PubMedID 31080068

  • Neural signatures of sleep in zebrafish. Nature Leung, L. C., Wang, G. X., Madelaine, R. n., Skariah, G. n., Kawakami, K. n., Deisseroth, K. n., Urban, A. E., Mourrain, P. n. 2019; 571 (7764): 198–204

    Abstract

    Slow-wave sleep and rapid eye movement (or paradoxical) sleep have been found in mammals, birds and lizards, but it is unclear whether these neuronal signatures are found in non-amniotic vertebrates. Here we develop non-invasive fluorescence-based polysomnography for zebrafish, and show-using unbiased, brain-wide activity recording coupled with assessment of eye movement, muscle dynamics and heart rate-that there are at least two major sleep signatures in zebrafish. These signatures, which we term slow bursting sleep and propagating wave sleep, share commonalities with those of slow-wave sleep and paradoxical or rapid eye movement sleep, respectively. Further, we find that melanin-concentrating hormone signalling (which is involved in mammalian sleep) also regulates propagating wave sleep signatures and the overall amount of sleep in zebrafish, probably via activation of ependymal cells. These observations suggest that common neural signatures of sleep may have emerged in the vertebrate brain over 450 million years ago.

    View details for DOI 10.1038/s41586-019-1336-7

    View details for PubMedID 31292557

  • Functional maturation of human neural stem cells in a 3D bioengineered brain model enriched with fetal brain-derived matrix. Scientific reports Sood, D. n., Cairns, D. M., Dabbi, J. M., Ramakrishnan, C. n., Deisseroth, K. n., Black, L. D., Santaniello, S. n., Kaplan, D. L. 2019; 9 (1): 17874

    Abstract

    Brain extracellular matrix (ECM) is often overlooked in vitro brain tissue models, despite its instructive roles during development. Using developmental stage-sourced brain ECM in reproducible 3D bioengineered culture systems, we demonstrate enhanced functional differentiation of human induced neural stem cells (hiNSCs) into healthy neurons and astrocytes. Particularly, fetal brain tissue-derived ECM supported long-term maintenance of differentiated neurons, demonstrated by morphology, gene expression and secretome profiling. Astrocytes were evident within the second month of differentiation, and reactive astrogliosis was inhibited in brain ECM-enriched cultures when compared to unsupplemented cultures. Functional maturation of the differentiated hiNSCs within fetal ECM-enriched cultures was confirmed by calcium signaling and spectral/cluster analysis. Additionally, the study identified native biochemical cues in decellularized ECM with notable comparisons between fetal and adult brain-derived ECMs. The development of novel brain-specific biomaterials for generating mature in vitro brain models provides an important path forward for interrogation of neuron-glia interactions.

    View details for DOI 10.1038/s41598-019-54248-1

    View details for PubMedID 31784595

  • Investigating the feasibility of channelrhodopsin variants for nanoscale optogenetics NEUROPHOTONICS Stahlberg, M. A., Ramakrishnan, C., Willig, K., Boyden, E. S., Deisseroth, K., Dean, C. 2019; 6 (1)
  • Multimodal characterization of the human nucleus accumbens. NeuroImage Cartmell, S. C., Tian, Q. n., Thio, B. J., Leuze, C. n., Ye, L. n., Williams, N. R., Yang, G. n., Ben-Dor, G. n., Deisseroth, K. n., Grill, W. M., McNab, J. A., Halpern, C. H. 2019

    Abstract

    Dysregulation of the nucleus accumbens (NAc) is implicated in numerous neuropsychiatric disorders. Treatments targeting this area directly (e.g. deep brain stimulation) demonstrate variable efficacy, perhaps owing to non-specific targeting of a functionally heterogeneous nucleus. Here we provide support for this notion, first observing disparate behavioral effects in response to direct simulation of different locations within the NAc in a human patient. These observations motivate a segmentation of the NAc into subregions, which we produce from a diffusion-tractography based analysis of 245 young, unrelated healthy subjects. We further explore the mechanism of these stimulation-induced behavioral responses by identifying the most probable subset of axons activated using a patient-specific computational model. We validate our diffusion-based segmentation using evidence from several modalities, including MRI-based measures of function and microstructure, human post-mortem immunohistochemical staining, and cross-species comparison of cortical-NAc projections that are known to be conserved. Finally, we visualize the passage of individual axon bundles through one NAc subregion in a post-mortem human sample using CLARITY 3D histology corroborated by 7T tractography. Collectively, these findings extensively characterize human NAc subregions and provide insight into their structural and functional distinctions with implications for stereotactic treatments targeting this region.

    View details for PubMedID 31077843

  • Scale-Invariant Visual Capabilities Explained by Topographic Representations of Luminance and Texture in Primate V1 NEURON Benvenuti, G., Chen, Y., Ramakrishnan, C., Deisseroth, K., Geisler, W. S., Seidemann, E. 2018; 100 (6): 1504-+
  • A Neural Circuit Mechanism for Encoding Aversive Stimuli in the Mesolimbic Dopamine System. Neuron de Jong, J. W., Afjei, S. A., Pollak Dorocic, I., Peck, J. R., Liu, C., Kim, C. K., Tian, L., Deisseroth, K., Lammel, S. 2018

    Abstract

    Ventral tegmental area (VTA) dopamine (DA) neurons play a central role in mediating motivated behaviors, but the circuitry through which they signal positive and negative motivational stimuli is incompletely understood. Using invivo fiber photometry, we simultaneously recorded activity in DA terminals in different nucleus accumbens (NAc) subnuclei during an aversive and reward conditioning task. We find that DA terminals in the ventral NAc medial shell (vNAcMed) are excited by unexpected aversive outcomes and to cues that predict them, whereas DA terminals in other NAc subregions are persistently depressed. Excitation to reward-predictive cues dominated in the NAc lateral shell and was largely absent in the vNAcMed. Moreover, we demonstrate that glutamatergic (VGLUT2-expressing) neurons in the lateral hypothalamus represent a key afferent input for providing information about aversive outcomes to vNAcMed-projecting DA neurons. Collectively, we reveal the distinct functional contributions of separate mesolimbic DA subsystems and their afferent pathways underlying motivated behaviors.

    View details for PubMedID 30503173

  • A community-developed open-source computational ecosystem for big neuro data NATURE METHODS Vogelstein, J. T., Perlman, E., Falk, B., Baden, A., Roncal, W., Chandrashekhar, V., Collman, F., Seshamani, S., Patsolic, J. L., Lillaney, K., Kazhdan, M., Hider, R., Pryor, D., Matelsky, J., Gion, T., Manavalan, P., Wester, B., Chevillet, M., Trautman, E. T., Khairy, K., Bridgeford, E., Kleissas, D. M., Tward, D. J., Crow, A. K., Hsueh, B., Wright, M. A., Miller, M. I., Smith, S. J., Vogelstein, R., Deisseroth, K., Burns, R. 2018; 15 (11): 846–47
  • A community-developed open-source computational ecosystem for big neuro data. Nature methods Vogelstein, J. T., Perlman, E., Falk, B., Baden, A., Gray Roncal, W., Chandrashekhar, V., Collman, F., Seshamani, S., Patsolic, J. L., Lillaney, K., Kazhdan, M., Hider, R. J., Pryor, D., Matelsky, J., Gion, T., Manavalan, P., Wester, B., Chevillet, M., Trautman, E. T., Khairy, K., Bridgeford, E., Kleissas, D. M., Tward, D. J., Crow, A. K., Hsueh, B., Wright, M. A., Miller, M. I., Smith, S. J., Vogelstein, R. J., Deisseroth, K., Burns, R. 2018

    View details for PubMedID 30377345

  • Scale-Invariant Visual Capabilities Explained by Topographic Representations of Luminance and Texture in Primate V1. Neuron Benvenuti, G., Chen, Y., Ramakrishnan, C., Deisseroth, K., Geisler, W. S., Seidemann, E. 2018

    Abstract

    Humans have remarkable scale-invariant visual capabilities. For example, our orientation discrimination sensitivity is largely constant over more than two orders of magnitude of variations in stimulus spatial frequency (SF). Orientation-selective V1 neurons are likely to contribute to orientation discrimination. However, because at any V1 location neurons have a limited range of receptive field (RF) sizes, we predict that at low SFs V1 neurons will carry little orientation information. If this were the case, what could account for the high behavioral sensitivity at low SFs? Using optical imaging in behaving macaques, we show that, as predicted, V1 orientation-tuned responses drop rapidly with decreasing SF. However, we reveal a surprising coarse-scale signal that corresponds to the projection of the luminance layout of low-SF stimuli to V1's retinotopic map. This homeomorphic and distributed representation, which carries high-quality orientation information, is likely to contribute to our striking scale-invariant visual capabilities.

    View details for PubMedID 30392796

  • Uneven balance of power between hypothalamic peptidergic neurons in the control of feeding PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Wei, Q., Krolewski, D. M., Moore, S., Kumar, V., Li, F., Martin, B., Tomer, R., Murphy, G. G., Deisseroth, K., Watson, S. J., Akil, H. 2018; 115 (40): E9489–E9498

    Abstract

    Two classes of peptide-producing neurons in the arcuate nucleus (Arc) of the hypothalamus are known to exert opposing actions on feeding: the anorexigenic neurons that express proopiomelanocortin (POMC) and the orexigenic neurons that express agouti-related protein (AgRP) and neuropeptide Y (NPY). These neurons are thought to arise from a common embryonic progenitor, but our anatomical and functional understanding of the interplay of these two peptidergic systems that contribute to the control of feeding remains incomplete. The present study uses a combination of optogenetic stimulation with viral and transgenic approaches, coupled with neural activity mapping and brain transparency visualization to demonstrate the following: (i) selective activation of Arc POMC neurons inhibits food consumption rapidly in unsated animals; (ii) activation of Arc neurons arising from POMC-expressing progenitors, including POMC and a subset of AgRP neurons, triggers robust feeding behavior, even in the face of satiety signals from POMC neurons; (iii) the opposing effects on food intake are associated with distinct neuronal projection and activation patterns of adult hypothalamic POMC neurons versus Arc neurons derived from POMC-expressing lineages; and (iv) the increased food intake following the activation of orexigenic neurons derived from POMC-expressing progenitors engages an extensive neural network that involves the endogenous opioid system. Together, these findings shed further light on the dynamic balance between two peptidergic systems in the moment-to-moment regulation of feeding behavior.

    View details for PubMedID 30224492

  • Coordinated Reductions in Excitatory Input to theNucleus AccumbensUnderlie Food Consumption NEURON Reed, S. J., Lafferty, C. K., Mendoza, J. A., Yang, A. K., Davidson, T. J., Grosenick, L., Deisseroth, K., Britt, J. P. 2018; 99 (6): 1260-+

    Abstract

    Reward-seeking behavior is regulated by a diverse collection of inputs to the nucleus accumbens (NAc). The information encoded in each excitatory afferent to the NAc is unknown, in part because it is unclear when these pathways are active in relation to behavior. Here we compare the activity profiles of amygdala, hippocampal, and thalamic inputs to the NAc shell in mice performing a cued reward-seeking task using GCaMP-based fiber photometry. We find that the rostral and caudal ends of the NAc shell are innervated by distinct but intermingled populations of forebrain neurons that exhibit divergent feeding-related activity. In the rostral NAc shell, a coordinated network-wide reduction in excitatory drive correlates with feeding, and reduced input from individual pathways is sufficient to promote it. Overall, the data suggest that pathway-specific input activity at a population level may vary more across the NAc than between pathways.

    View details for PubMedID 30146308

  • Mapping projections of molecularly defined dopamine neuron subtypes using intersectional genetic approaches NATURE NEUROSCIENCE Poulin, J., Caronia, G., Hofer, C., Cui, Q., Helm, B., Ramakrishnan, C., Chan, C., Dombeck, D. A., Deisseroth, K., Awatramani, R. 2018; 21 (9): 1260-+

    Abstract

    Midbrain dopamine (DA) neurons have diverse functions that can in part be explained by their heterogeneity. Although molecularly distinct subtypes of DA neurons have been identified by single-cell gene expression profiling, fundamental features such as their projection patterns have not been elucidated. Progress in this regard has been hindered by the lack of genetic tools for studying DA neuron subtypes. Here we develop intersectional genetic labeling strategies, based on combinatorial gene expression, to map the projections of molecularly defined DA neuron subtypes. We reveal distinct genetically defined dopaminergic pathways arising from the substantia nigra pars compacta and from the ventral tegmental area that innervate specific regions of the caudate putamen, nucleus accumbens and amygdala. Together, the genetic toolbox and DA neuron subtype projections presented here constitute a resource that will accelerate the investigation of this clinically significant neurotransmitter system.

    View details for PubMedID 30104732

  • Structural mechanisms of selectivity and gating in anion channelrhodopsins. Nature Kato, H. E., Kim, Y. S., Paggi, J. M., Evans, K. E., Allen, W. E., Richardson, C., Inoue, K., Ito, S., Ramakrishnan, C., Fenno, L. E., Yamashita, K., Hilger, D., Lee, S. Y., Berndt, A., Shen, K., Kandori, H., Dror, R. O., Kobilka, B. K., Deisseroth, K. 2018

    Abstract

    Both designed and natural anion-conducting channelrhodopsins (dACRs and nACRs, respectively) have been widely applied in optogenetics (enabling selective inhibition of target-cell activity during animal behaviour studies), but each class exhibits performance limitations, underscoring trade-offs in channel structure-function relationships. Therefore, molecular and structural insights into dACRs and nACRs will be critical not only for understanding the fundamental mechanisms of these light-gated anionchannels, but also to create next-generation optogenetic tools. Here we report crystal structures of the dACR iC++, along with spectroscopic, electrophysiological and computational analyses that provide unexpected insights into pH dependence, substrate recognition, channel gating and ion selectivity of both dACRs and nACRs. These results enabled us to create an anion-conducting channelrhodopsin integrating the key features of large photocurrent and fast kinetics alongside exclusive anion selectivity.

    View details for PubMedID 30158697

  • Crystal structure of the natural anion-conducting channelrhodopsin GtACR1. Nature Kim, Y. S., Kato, H. E., Yamashita, K., Ito, S., Inoue, K., Ramakrishnan, C., Fenno, L. E., Evans, K. E., Paggi, J. M., Dror, R. O., Kandori, H., Kobilka, B. K., Deisseroth, K. 2018

    Abstract

    The naturally occurring channelrhodopsin variant anion channelrhodopsin-1 (ACR1), discovered in the cryptophyte algae Guillardia theta, exhibits large light-gated anionconductance and high anionselectivity when expressed in heterologous settings, properties that support its use as an optogenetic tool to inhibit neuronal firing with light. However, molecular insight into ACR1 is lacking owing to the absence of structural information underlying light-gated anion conductance. Here we present the crystal structure of G. theta ACR1 at 2.9A resolution. The structure reveals unusual architectural features that span the extracellular domain, retinal-binding pocket, Schiff-base region, and anion-conduction pathway. Together with electrophysiological and spectroscopic analyses, these findings reveal the fundamental molecular basis of naturally occurring light-gated anion conductance, and provide a framework for designing the next generation of optogenetic tools.

    View details for PubMedID 30158696

  • 5-HT release in nucleus accumbens rescues social deficits in mouse autism model. Nature Walsh, J. J., Christoffel, D. J., Heifets, B. D., Ben-Dor, G. A., Selimbeyoglu, A., Hung, L. W., Deisseroth, K., Malenka, R. C. 2018

    Abstract

    Dysfunction in prosocial interactions is a core symptom of autism spectrum disorder. However, the neural mechanisms that underlie sociability are poorly understood, limiting the rational development of therapies to treat social deficits. Here we show in mice that bidirectional modulation of the release of serotonin (5-HT) from dorsal raphe neurons in the nucleus accumbens bidirectionally modifies sociability. In a mouse model of a common genetic cause of autism spectrum disorder-a copy number variation on chromosome 16p11.2-genetic deletion of the syntenic region from 5-HT neurons induces deficits in social behaviour and decreases dorsal raphe 5-HT neuronal activity. These sociability deficits can be rescued by optogenetic activation of dorsal raphe 5-HT neurons, an effect requiring and mimicked by activation of 5-HT1b receptors in the nucleus accumbens. These results demonstrate an unexpected role for 5-HT action in the nucleus accumbens in social behaviours, and suggest that targeting this mechanism may prove therapeutically beneficial.

    View details for PubMedID 30089910

  • NMDA receptor activity regulates synaptic connections between retinal ganglion and bipolar cells Young, B., Sanchez, C., Ramakrishnan, C., Wang, P., Deisseroth, K., Tian, N. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2018
  • An unique subtype of BCs provides excitatory input to both ON and OFF synaptic pathways from both rods and cones in the retina Tian, N., Young, B., Ramakrishnan, C., Wang, P., Deisseroth, K., Ganjawala, T. H., Pan, Z. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2018
  • Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science (New York, N.Y.) Wang, X., Allen, W. E., Wright, M. A., Sylwestrak, E. L., Samusik, N., Vesuna, S., Evans, K., Liu, C., Ramakrishnan, C., Liu, J., Nolan, G. P., Bava, F., Deisseroth, K. 2018

    Abstract

    Retrieving high-content gene-expression information while retaining 3D positional anatomy at cellular resolution has been difficult, limiting integrative understanding of structure and function in complex biological tissues. Here we develop and apply a technology for 3D intact-tissue RNA sequencing, termed STARmap (Spatially-resolved Transcript Amplicon Readout Mapping), which integrates hydrogel-tissue chemistry, targeted signal amplification, and in situ sequencing. The capabilities of STARmap were tested by mapping 160 to 1,020 genes simultaneously in sections of mouse brain at single-cell resolution with high efficiency, accuracy and reproducibility. Moving to thick tissue blocks, we observed a molecularly-defined gradient distribution of excitatory-neuron subtypes across cubic millimeter-scale volumes (>30,000 cells), and discovered a short-range 3D self-clustering in many inhibitory-neuron subtypes that could be identified and described with 3D STARmap.

    View details for PubMedID 29930089

  • Publisher Correction: An interactive framework for whole-brain maps at cellular resolution. Nature neuroscience Fürth, D., Vaissière, T., Tzortzi, O., Xuan, Y., Märtin, A., Lazaridis, I., Spigolon, G., Fisone, G., Tomer, R., Deisseroth, K., Carlén, M., Miller, C. A., Rumbaugh, G., Meletis, K. 2018; 21 (6): 895

    Abstract

    In the version of this article initially published online, Daniel Fürth was not listed as a corresponding author. The error has been corrected in the print, PDF and HTML versions of this article.

    View details for DOI 10.1038/s41593-017-0058-0

    View details for PubMedID 29255166

  • Neuronal activity regulates neurotransmitter switching in the adult brain following light-induced stress PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Meng, D., Li, H., Deisseroth, K., Leutgeb, S., Spitzer, N. C. 2018; 115 (20): 5064–71

    Abstract

    Neurotransmitter switching in the adult mammalian brain occurs following photoperiod-induced stress, but the mechanism of regulation is unknown. Here, we demonstrate that elevated activity of dopaminergic neurons in the paraventricular nucleus of the hypothalamus (PaVN) in the adult rat is required for the loss of dopamine expression after long-day photoperiod exposure. The transmitter switch occurs exclusively in PaVN dopaminergic neurons that coexpress vesicular glutamate transporter 2 (VGLUT2), is accompanied by a loss of dopamine type 2 receptors (D2Rs) on corticotrophin-releasing factor (CRF) neurons, and can lead to increased release of CRF. Suppressing activity of all PaVN glutamatergic neurons decreases the number of inhibitory PaVN dopaminergic neurons, indicating homeostatic regulation of transmitter expression in the PaVN.

    View details for PubMedID 29686073

  • Phasic Dopamine Signals in the Nucleus Accumbens that Cause Active Avoidance Require Endocannabinoid Mobilization in the Midbrain CURRENT BIOLOGY Wenzel, J. M., Oleson, E. B., Gove, W. N., Cole, A. B., Gyawali, U., Dantrassy, H. M., Bluett, R. J., Dryanovski, D. I., Stuber, G. D., Deisseroth, K., Mathur, B. N., Patel, S., Lupica, C. R., Cheer, J. F. 2018; 28 (9): 1392-+

    Abstract

    Phasic dopamine (DA) release accompanies approach toward appetitive cues. However, a role for DA in the active avoidance of negative events remains undetermined. Warning signals informing footshock avoidance are associated with accumbal DA release, whereas depression of DA is observed with unavoidable footshock. Here, we reveal a causal role of phasic DA in active avoidance learning; specifically, optogenetic activation of DA neurons facilitates avoidance, whereas optical inhibition of these cells attenuates it. Furthermore, stimulation of DA neurons during presentation of a fear-conditioned cue accelerates the extinction of a passive defensive behavior (i.e., freezing). Dopaminergic control of avoidance requires endocannabinoids (eCBs), as perturbing eCB signaling in the midbrain disrupts avoidance, which is rescued by optical stimulation of DA neurons. Interestingly, once the avoidance task is learned, neither DA nor eCB manipulations affect performance, suggesting that once acquisition occurs, expression of this behavior is subserved by other anatomical frameworks. Our findings establish an instrumental role for DA release in learning active responses to aversive stimuli and its control by eCB signaling.

    View details for PubMedID 29681476

    View details for PubMedCentralID PMC5940536

  • A Critical Role for the Globus Pallidus in Cocaine-Triggered Plasticity Revealed Byrabies Activity Screen Beier, K., Kim, C., Hoerbelt, P., Hung, L., Heifets, B., DeLoach, K., Mosca, T., Neuner, S., Deisseroth, K., Luo, L., Malenka, R. ELSEVIER SCIENCE INC. 2018: S235–S236
  • Development of an optogenetic toolkit for neural circuit dissection in squirrel monkeys SCIENTIFIC REPORTS O'Shea, D. J., Kalanithi, P., Ferenczi, E. A., Hsueh, B., Chandrasekaran, C., Goo, W., Diester, I., Ramakrishnan, C., Kaufman, M. T., Ryu, S. I., Yeom, K. W., Deisseroth, K., Shenoy, K. V. 2018; 8: 6775

    Abstract

    Optogenetic tools have opened a rich experimental landscape for understanding neural function and disease. Here, we present the first validation of eight optogenetic constructs driven by recombinant adeno-associated virus (AAV) vectors and a WGA-Cre based dual injection strategy for projection targeting in a widely-used New World primate model, the common squirrel monkey Saimiri sciureus. We observed opsin expression around the local injection site and in axonal projections to downstream regions, as well as transduction to thalamic neurons, resembling expression patterns observed in macaques. Optical stimulation drove strong, reliable excitatory responses in local neural populations for two depolarizing opsins in anesthetized monkeys. Finally, we observed continued, healthy opsin expression for at least one year. These data suggest that optogenetic tools can be readily applied in squirrel monkeys, an important first step in enabling precise, targeted manipulation of neural circuits in these highly trainable, cognitively sophisticated animals. In conjunction with similar approaches in macaques and marmosets, optogenetic manipulation of neural circuits in squirrel monkeys will provide functional, comparative insights into neural circuits which subserve dextrous motor control as well as other adaptive behaviors across the primate lineage. Additionally, development of these tools in squirrel monkeys, a well-established model system for several human neurological diseases, can aid in identifying novel treatment strategies.

    View details for PubMedID 29712920

  • Brain-wide Electrical Spatiotemporal Dynamics Encode Depression Vulnerability CELL Hultman, R., Ulrich, K., Sachs, B. D., Blount, C., Carlson, D. E., Ndubuizu, N., Bagot, R. C., Parise, E. M., Vu, M. T., Gallagher, N. M., Wang, J., Silva, A. J., Deisseroth, K., Mague, S. D., Caron, M. G., Nestler, E. J., Carin, L., Dzirasa, K. 2018; 173 (1): 166-+

    Abstract

    Brain-wide fluctuations in local field potential oscillations reflect emergent network-level signals that mediate behavior. Cracking the code whereby these oscillations coordinate in time and space (spatiotemporal dynamics) to represent complex behaviors would provide fundamental insights into how the brain signals emotional pathology. Using machine learning, we discover a spatiotemporal dynamic network that predicts the emergence of major depressive disorder (MDD)-related behavioral dysfunction in mice subjected to chronic social defeat stress. Activity patterns in this network originate in prefrontal cortex and ventral striatum, relay through amygdala and ventral tegmental area, and converge in ventral hippocampus. This network is increased by acute threat, and it is also enhanced in three independent models of MDD vulnerability. Finally, we demonstrate that this vulnerability network is biologically distinct from the networks that encode dysfunction after stress. Thus, these findings reveal a convergent mechanism through which MDD vulnerability is mediated in the brain.

    View details for PubMedID 29502969

  • Hierarchical neural architecture underlying thirst regulation NATURE Augustine, V., Gokce, S., Lee, S., Wang, B., Davidson, T. J., Reimann, F., Gribble, F., Deisseroth, K., Lois, C., Oka, Y. 2018; 555 (7695): 204-+

    Abstract

    Neural circuits for appetites are regulated by both homeostatic perturbations and ingestive behaviour. However, the circuit organization that integrates these internal and external stimuli is unclear. Here we show in mice that excitatory neural populations in the lamina terminalis form a hierarchical circuit architecture to regulate thirst. Among them, nitric oxide synthase-expressing neurons in the median preoptic nucleus (MnPO) are essential for the integration of signals from the thirst-driving neurons of the subfornical organ (SFO). Conversely, a distinct inhibitory circuit, involving MnPO GABAergic neurons that express glucagon-like peptide 1 receptor (GLP1R), is activated immediately upon drinking and monosynaptically inhibits SFO thirst neurons. These responses are induced by the ingestion of fluids but not solids, and are time-locked to the onset and offset of drinking. Furthermore, loss-of-function manipulations of GLP1R-expressing MnPO neurons lead to a polydipsic, overdrinking phenotype. These neurons therefore facilitate rapid satiety of thirst by monitoring real-time fluid ingestion. Our study reveals dynamic thirst circuits that integrate the homeostatic-instinctive requirement for fluids and the consequent drinking behaviour to maintain internal water balance.

    View details for PubMedID 29489747

  • An interactive framework for whole-brain maps at cellular resolution NATURE NEUROSCIENCE Furth, D., Vaissiere, T., Tzortzi, O., Xuan, Y., Martin, A., Lazaridis, I., Spigolon, G., Fisone, G., Tomer, R., Deisseroth, K., Carlen, M., Miller, C. A., Rumbaugh, G., Meletis, K. 2018; 21 (1): 139-+

    Abstract

    To deconstruct the architecture and function of brain circuits, it is necessary to generate maps of neuronal connectivity and activity on a whole-brain scale. New methods now enable large-scale mapping of the mouse brain at cellular and subcellular resolution. We developed a framework to automatically annotate, analyze, visualize and easily share whole-brain data at cellular resolution, based on a scale-invariant, interactive mouse brain atlas. This framework enables connectivity and mapping projects in individual laboratories and across imaging platforms, as well as multiplexed quantitative information on the molecular identity of single neurons. As a proof of concept, we generated a comparative connectivity map of five major neuron types in the corticostriatal circuit, as well as an activity-based map to identify hubs mediating the behavioral effects of cocaine. Thus, this computational framework provides the necessary tools to generate brain maps that integrate data from connectivity, neuron identity and function.

    View details for PubMedID 29203898

  • Whole-tissue biopsy phenotyping of three-dimensional tumours reveals patterns of cancer heterogeneity (vol 1, pg 796, 2017) NATURE BIOMEDICAL ENGINEERING Tanaka, N., Kanatani, S., Tomer, R., Sahlgren, C., Kronqvist, P., Kaczynska, D., Louhivuori, L., Kis, L., Lindh, C., Mitura, P., Stepulak, A., Corvigno, S., Hartman, J., Micke, P., Mezheyeuski, A., Strell, C., Carlson, J. W., Moro, C., Dahlstrand, H., Ostman, A., Matsumoto, K., Wiklund, P., Oya, M., Miyakawa, A., Deisseroth, K., Uhlen, P. 2018; 2 (1): 48

    Abstract

    In this Article originally published, owing to a technical error, author affiliations were incorrectly assigned in the HTML version; the PDF was correct. These errors have now been corrected.

    View details for PubMedID 31015657

  • Vasopressin excites interneurons to suppress hippocampal network activity across a broad span of brain maturity at birth PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Spoljaric, A., Seja, P., Spoljaric, I., Virtanen, M. A., Lindfors, J., Uvarov, P., Summanen, M., Crow, A. K., Hsueh, B., Puskarjov, M., Ruusuvuori, E., Voipio, J., Deisseroth, K., Kaila, K. 2017; 114 (50): E10819–E10828

    Abstract

    During birth in mammals, a pronounced surge of fetal peripheral stress hormones takes place to promote survival in the transition to the extrauterine environment. However, it is not known whether the hormonal signaling involves central pathways with direct protective effects on the perinatal brain. Here, we show that arginine vasopressin specifically activates interneurons to suppress spontaneous network events in the perinatal hippocampus. Experiments done on the altricial rat and precocial guinea pig neonate demonstrated that the effect of vasopressin is not dependent on the level of maturation (depolarizing vs. hyperpolarizing) of postsynaptic GABAA receptor actions. Thus, the fetal mammalian brain is equipped with an evolutionarily conserved mechanism well-suited to suppress energetically expensive correlated network events under conditions of reduced oxygen supply at birth.

    View details for PubMedID 29183979

  • The central amygdala controls learning in the lateral amygdala NATURE NEUROSCIENCE Yu, K., Ahrens, S., Zhang, X., Schiff, H., Ramakrishnan, C., Fenno, L., Deisseroth, K., Zhao, F., Luo, M., Gong, L., He, M., Zhou, P., Paninski, L., Li, B. 2017; 20 (12): 1680-+

    Abstract

    Experience-driven synaptic plasticity in the lateral amygdala is thought to underlie the formation of associations between sensory stimuli and an ensuing threat. However, how the central amygdala participates in such a learning process remains unclear. Here we show that PKC-δ-expressing central amygdala neurons are essential for the synaptic plasticity underlying learning in the lateral amygdala, as they convey information about the unconditioned stimulus to lateral amygdala neurons during fear conditioning.

    View details for PubMedID 29184202

    View details for PubMedCentralID PMC5755715

  • Long-Range GABAergic Inputs Regulate Neural Stem Cell Quiescence and Control Adult Hippocampal Neurogenesis CELL STEM CELL Bao, H., Asrican, B., Li, W., Gu, B., Wen, Z., Lim, S., Haniff, I., Ramakrishnan, C., Deisseroth, K., Philpot, B., Song, J. 2017; 21 (5): 604-+

    Abstract

    The quiescence of adult neural stem cells (NSCs) is regulated by local parvalbumin (PV) interneurons within the dentate gyrus (DG). Little is known about how local PV interneurons communicate with distal brain regions to regulate NSCs and hippocampal neurogenesis. Here, we identify GABAergic projection neurons from the medial septum (MS) as the major afferents to dentate PV interneurons. Surprisingly, dentate PV interneurons are depolarized by GABA signaling, which is in sharp contrast to most mature neurons hyperpolarized by GABA. Functionally, these long-range GABAergic inputs are necessary and sufficient to maintain adult NSC quiescence and ablating them leads to NSC activation and subsequent depletion of the NSC pool. Taken together, these findings delineate a GABAergic network involving long-range GABAergic projection neurons and local PV interneurons that couples dynamic brain activity to the neurogenic niche in controlling NSC quiescence and hippocampal neurogenesis.

    View details for PubMedID 29100013

    View details for PubMedCentralID PMC5689456

  • Modular organization of the brainstem noradrenaline system coordinates opposing learning states NATURE NEUROSCIENCE Uematsu, A., Tan, B., Ycu, E. A., Cuevas, J., Koivumaa, J., Junyent, F., Kremer, E. J., Witten, I. B., Deisseroth, K., Johansen, J. P. 2017; 20 (11): 1602-+

    Abstract

    Noradrenaline modulates global brain states and diverse behaviors through what is traditionally believed to be a homogeneous cell population in the brainstem locus coeruleus (LC). However, it is unclear how LC coordinates disparate behavioral functions. We report a modular LC organization in rats, endowed with distinct neural projection patterns and coding properties for flexible specification of opposing behavioral learning states. LC projection mapping revealed functionally distinct cell modules with specific anatomical connectivity. An amygdala-projecting ensemble promoted aversive learning, while an independent medial prefrontal cortex-projecting ensemble extinguished aversive responses to enable flexible behavior. LC neurons displayed context-dependent inter-relationships, with moderate, discrete activation of distinct cell populations by fear or safety cues and robust, global recruitment of most cells by strong aversive stimuli. These results demonstrate a modular organization in LC in which combinatorial activation modes are coordinated with projection- and behavior-specific cell populations, enabling adaptive tuning of emotional responding and behavioral flexibility.

    View details for PubMedID 28920933

  • Brain-Derived Neurotrophic Factor in the Mesolimbic Reward Circuitry Mediates Nociception in Chronic Neuropathic Pain BIOLOGICAL PSYCHIATRY Zhang, H., Qian, Y., Li, C., Liu, D., Wang, L., Wang, X., Liu, M., Liu, H., Zhang, S., Guo, X., Yang, J., Ding, H., Koo, J., Mouzon, E., Deisseroth, K., Nestler, E. J., Zachariou, V., Han, M., Cao, J. 2017; 82 (8): 608–18

    Abstract

    The mesolimbic reward system plays a critical role in modulating nociception; however, its underlying molecular, cellular, and neural circuitry mechanisms remain unknown.Chronic constrictive injury (CCI) of the sciatic nerve was used to model neuropathic pain. Projection-specific in vitro recordings in mouse brain slices and in vivo recordings from anesthetized animals were used to measure firing of dopaminergic neurons in the ventral tegmental area (VTA). The role of VTA-nucleus accumbens (NAc) circuitry in nociceptive regulation was assessed using optogenetic and pharmacological manipulations, and the underlying molecular mechanisms were investigated by Western blotting, enzyme-linked immunosorbent assays, and conditional knockdown techniques.c-Fos expression in and firing of contralateral VTA-NAc dopaminergic neurons were elevated in CCI mice, and optogenetic inhibition of these neurons reversed CCI-induced thermal hyperalgesia. CCI increased the expression of brain-derived neurotrophic factor (BDNF) protein but not messenger RNA in the contralateral NAc. This increase was reversed by pharmacological inhibition of VTA dopaminergic neuron activity, which induced an antinociceptive effect that was neutralized by injecting exogenous BDNF into the NAc. Moreover, inhibition of BDNF synthesis in the VTA with anisomycin or selective knockdown of BDNF in the VTA-NAc pathway was antinociceptive in CCI mice.These results reveal a novel mechanism of nociceptive modulation in the mesolimbic reward circuitry and provide new insight into the neural circuits involved in the processing of nociceptive information.

    View details for PubMedID 28390647

    View details for PubMedCentralID PMC5788809

  • Whole-tissue biopsy phenotyping of three-dimensional tumours reveals patterns of cancer heterogeneity NATURE BIOMEDICAL ENGINEERING Tanaka, N., Kanatani, S., Tomer, R., Sahlgren, C., Kronqvist, P., Kaczynska, D., Louhivuori, L., Kis, L., Lindh, C., Mitura, P., Stepulak, A., Corvigno, S., Hartman, J., Micke, P., Mezheyeuski, A., Strell, C., Carlson, J. W., Moro, C., Dahlstrand, H., Ostman, A., Matsumoto, K., Wiklund, P., Oya, M., Miyakawa, A., Deisseroth, K., Uhlen, P. 2017; 1 (10): 796–806
  • Whole-tissue biopsy phenotyping of three-dimensional tumours reveals patterns of cancer heterogeneity. Nature biomedical engineering Tanaka, N., Kanatani, S., Tomer, R., Sahlgren, C., Kronqvist, P., Kaczynska, D., Louhivuori, L., Kis, L., Lindh, C., Mitura, P., Stepulak, A., Corvigno, S., Hartman, J., Micke, P., Mezheyeuski, A., Strell, C., Carlson, J. W., Fernández Moro, C., Dahlstrand, H., Östman, A., Matsumoto, K., Wiklund, P., Oya, M., Miyakawa, A., Deisseroth, K., Uhlén, P. 2017; 1 (10): 796-806

    Abstract

    Intratumoral heterogeneity is a critical factor when diagnosing and treating patients with cancer. Marked differences in the genetic and epigenetic backgrounds of cancer cells have been revealed by advances in genome sequencing, yet little is known about the phenotypic landscape and the spatial distribution of intratumoral heterogeneity within solid tumours. Here, we show that three-dimensional light-sheet microscopy of cleared solid tumours can identify unique patterns of phenotypic heterogeneity, in the epithelial-to-mesenchymal transition and in angiogenesis, at single-cell resolution in whole formalin-fixed paraffin-embedded (FFPE) biopsy samples. We also show that cleared FFPE samples can be re-embedded in paraffin after examination for future use, and that our tumour-phenotyping pipeline can determine tumour stage and stratify patient prognosis from clinical samples with higher accuracy than current diagnostic methods, thus facilitating the design of more efficient cancer therapies.

    View details for DOI 10.1038/s41551-017-0139-0

    View details for PubMedID 31015588

  • The form and function of channelrhodopsin SCIENCE Deisseroth, K., Hegemann, P. 2017; 357 (6356)
  • Place field assembly distribution encodes preferred locations. PLoS biology Mamad, O., Stumpp, L., McNamara, H. M., Ramakrishnan, C., Deisseroth, K., Reilly, R. B., Tsanov, M. 2017; 15 (9): e2002365

    Abstract

    The hippocampus is the main locus of episodic memory formation and the neurons there encode the spatial map of the environment. Hippocampal place cells represent location, but their role in the learning of preferential location remains unclear. The hippocampus may encode locations independently from the stimuli and events that are associated with these locations. We have discovered a unique population code for the experience-dependent value of the context. The degree of reward-driven navigation preference highly correlates with the spatial distribution of the place fields recorded in the CA1 region of the hippocampus. We show place field clustering towards rewarded locations. Optogenetic manipulation of the ventral tegmental area demonstrates that the experience-dependent place field assembly distribution is directed by tegmental dopaminergic activity. The ability of the place cells to remap parallels the acquisition of reward context. Our findings present key evidence that the hippocampal neurons are not merely mapping the static environment but also store the concurrent context reward value, enabling episodic memory for past experience to support future adaptive behavior.

    View details for DOI 10.1371/journal.pbio.2002365

    View details for PubMedID 28898248

    View details for PubMedCentralID PMC5609775

  • A Guide to Creating and Testing New INTRSECT Constructs. Current protocols in neuroscience Fenno, L. E., Mattis, J., Ramakrishnan, C., Deisseroth, K. 2017; 80: 4.39.1-4.39.24

    Abstract

    As the power of genetically encoded interventional and observational tools for neuroscience expands, the boundaries of experimental design are increasingly defined by limits in selectively expressing these tools in relevant cell types. Single-recombinase-dependent expression systems have been widely used as a means to restrict gene expression based on single features by combining recombinase-dependent viruses with recombinase-expressing transgenic animals. This protocol details how to create INTRSECT constructs and use multiple recombinases to achieve targeting of a desired gene to subsets of neurons that are defined by multiple genetic and/or topological features. This method includes the design and utilization of both viruses and transgenic animals: these tools are inherently flexible and modular and may be used in different combinations to achieve the desired gene expression pattern. © 2017 by John Wiley & Sons, Inc.

    View details for DOI 10.1002/cpns.30

    View details for PubMedID 28678399

  • In Vivo Fiber Photometry Reveals Signature of Future Stress Susceptibility in Nucleus Accumbens. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology Muir, J., Lorsch, Z. S., Ramakrishnan, C., Deisseroth, K., Nestler, E. J., Calipari, E. S., Bagot, R. C. 2017

    Abstract

    Recognizing why chronic stress causes only a subset of individuals to become depressed is critical to understanding depression on a basic level and, also to developing treatments that increase resilience. Stress-induced alterations in the activity of reward-related brain regions, such as the nucleus accumbens (NAc), are linked to the pathophysiology of depression. However, it has been difficult to determine if differences in stress susceptibility are pre-existing or merely an effect of chronic stress. The NAc consists largely of medium spiny neurons (MSNs), distinguished by their predominant expression of either D1 or D2 dopamine receptors. Mice that develop depressive-like symptoms after chronic social defeat stress (CSDS) show distinct changes in the activity of these two cell subtypes. Until now it has not been possible to determine if such effects are merely a consequence of stress or in fact precede stress and, thus, have utility in pre-identifying stress-susceptible individuals. The goal of this study was to define a cell-type specific signature of stress susceptibility and resilience. Using fiber photometry calcium imaging, we recorded calcium transients in NAc D1- and D2-MSNs in awake behaving mice and found that D1-MSN activity is a predictive marker of depression susceptibility: prior to stress, mice that will later become resilient had increased baseline D1- MSN activity, and increased calcium transients specific to social interaction. Differences in D2- MSN activity were not specific to social interaction. Our findings identify a pre-existing mechanism of stress-induced susceptibility, creating the potential to target preventative interventions to the most relevant populations.Neuropsychopharmacology accepted article preview online, 07 June 2017. doi:10.1038/npp.2017.122.

    View details for DOI 10.1038/npp.2017.122

    View details for PubMedID 28589967

  • Distinct Thalamic Reticular Cell Types Differentially Modulate Normal and Pathological Cortical Rhythms. Cell reports Clemente-Perez, A., Makinson, S. R., Higashikubo, B., Brovarney, S., Cho, F. S., Urry, A., Holden, S. S., Wimer, M., Dávid, C., Fenno, L. E., Acsády, L., Deisseroth, K., Paz, J. T. 2017; 19 (10): 2130-2142

    Abstract

    Integrative brain functions depend on widely distributed, rhythmically coordinated computations. Through its long-ranging connections with cortex and most senses, the thalamus orchestrates the flow of cognitive and sensory information. Essential in this process, the nucleus reticularis thalami (nRT) gates different information streams through its extensive inhibition onto other thalamic nuclei, however, we lack an understanding of how different inhibitory neuron subpopulations in nRT function as gatekeepers. We dissociated the connectivity, physiology, and circuit functions of neurons within rodent nRT, based on parvalbumin (PV) and somatostatin (SOM) expression, and validated the existence of such populations in human nRT. We found that PV, but not SOM, cells are rhythmogenic, and that PV and SOM neurons are connected to and modulate distinct thalamocortical circuits. Notably, PV, but not SOM, neurons modulate somatosensory behavior and disrupt seizures. These results provide a conceptual framework for how nRT may gate incoming information to modulate brain-wide rhythms.

    View details for DOI 10.1016/j.celrep.2017.05.044

    View details for PubMedID 28591583

  • Next-generation probes, particles, and proteins for neural interfacing SCIENCE ADVANCES Rivnay, J., Wang, H., Fenno, L., Deisseroth, K., Malliaras, G. G. 2017; 3 (6): e1601649

    Abstract

    Bidirectional interfacing with the nervous system enables neuroscience research, diagnosis, and therapy. This two-way communication allows us to monitor the state of the brain and its composite networks and cells as well as to influence them to treat disease or repair/restore sensory or motor function. To provide the most stable and effective interface, the tools of the trade must bridge the soft, ion-rich, and evolving nature of neural tissue with the largely rigid, static realm of microelectronics and medical instruments that allow for readout, analysis, and/or control. In this Review, we describe how the understanding of neural signaling and material-tissue interactions has fueled the expansion of the available tool set. New probe architectures and materials, nanoparticles, dyes, and designer genetically encoded proteins push the limits of recording and stimulation lifetime, localization, and specificity, blurring the boundary between living tissue and engineered tools. Understanding these approaches, their modality, and the role of cross-disciplinary development will support new neurotherapies and prostheses and provide neuroscientists and neurologists with unprecedented access to the brain.

    View details for PubMedID 28630894

  • Estrous Cycle-Dependent Alterations in Cocaine Affinity at the Dopamine Transporter Underlie Enhanced Cocaine Reward in Females Calipari, E., Juarez, B., Morel, C., Walker, D., Cahill, M., Ramakrishnan, C., Deisseroth, K., Han, M., Nestler, E. ELSEVIER SCIENCE INC. 2017: S276
  • Role of BDNF in Regulating Sensitive Periods for Fear Regulation Pattwell, S., Liston, C., Giza, J., Deisseroth, K., Lee, F. ELSEVIER SCIENCE INC. 2017: S9-S10
  • The separate effects of lipids and proteins on brain MRI contrast revealed through tissue clearing. NeuroImage Leuze, C., Aswendt, M., Ferenczi, E., Liu, C. W., Hsueh, B., Goubran, M., Tian, Q., Steinberg, G., Zeineh, M. M., Deisseroth, K., McNab, J. A. 2017

    Abstract

    Despite the widespread use of magnetic resonance imaging (MRI) of the brain, the relative contribution of different biological components (e.g. lipids and proteins) to structural MRI contrasts (e.g., T1, T2, T2*, proton density, diffusion) remains incompletely understood. This limitation can undermine the interpretation of clinical MRI and hinder the development of new contrast mechanisms. Here, we determine the respective contribution of lipids and proteins to MRI contrast by removing lipids and preserving proteins in mouse brains using CLARITY. We monitor the temporal dynamics of tissue clearance via NMR spectroscopy, protein assays and optical emission spectroscopy. MRI of cleared brain tissue showed: 1) minimal contrast on standard MRI sequences; 2) increased relaxation times; and 3) diffusion rates close to free water. We conclude that lipids, present in myelin and membranes, are a dominant source of MRI contrast in brain tissue.

    View details for DOI 10.1016/j.neuroimage.2017.04.021

    View details for PubMedID 28411157

  • The separate effects of lipids and proteins on brain MRI contrast revealed through tissue clearing. NeuroImage Leuze, C., Aswendt, M., Ferenczi, E., Liu, C. W., Hsueh, B., Goubran, M., Tian, Q., Steinberg, G., Zeineh, M. M., Deisseroth, K., McNab, J. A. 2017

    Abstract

    Despite the widespread use of magnetic resonance imaging (MRI) of the brain, the relative contribution of different biological components (e.g. lipids and proteins) to structural MRI contrasts (e.g., T1, T2, T2*, proton density, diffusion) remains incompletely understood. This limitation can undermine the interpretation of clinical MRI and hinder the development of new contrast mechanisms. Here, we determine the respective contribution of lipids and proteins to MRI contrast by removing lipids and preserving proteins in mouse brains using CLARITY. We monitor the temporal dynamics of tissue clearance via NMR spectroscopy, protein assays and optical emission spectroscopy. MRI of cleared brain tissue showed: 1) minimal contrast on standard MRI sequences; 2) increased relaxation times; and 3) diffusion rates close to free water. We conclude that lipids, present in myelin and membranes, are a dominant source of MRI contrast in brain tissue.

    View details for DOI 10.1016/j.neuroimage.2017.04.021

    View details for PubMedID 28411157

  • Estrous cycle-dependent alterations in cocaine affinity at the dopamine transporter underlie enhanced cocaine reward in females Calipari, E. S., Juarez, B., Morel, C., Walker, D. M., Cahill, M., Riberio, E., Deisseroth, K., Han, M., Neslter, E. J. FEDERATION AMER SOC EXP BIOL. 2017
  • CLARITY reveals dynamics of ovarian follicular architecture and vasculature in three-dimensions. Scientific reports Feng, Y., Cui, P., Lu, X., Hsueh, B., Möller Billig, F., Zarnescu Yanez, L., Tomer, R., Boerboom, D., Carmeliet, P., Deisseroth, K., Hsueh, A. J. 2017; 7: 44810-?

    Abstract

    Optimal distribution of heterogeneous organelles and cell types within an organ is essential for physiological processes. Unique for the ovary, hormonally regulated folliculogenesis, ovulation, luteal formation/regression and associated vasculature changes lead to tissue remodeling during each reproductive cycle. Using the CLARITY approach and marker immunostaining, we identified individual follicles and corpora lutea in intact ovaries. Monitoring lifetime changes in follicle populations showed age-dependent decreases in total follicles and percentages of advanced follicles. Follicle development from primordial to preovulatory stage was characterized by 3 × 10(5)-fold increases in volume, decreases in roundness, and decreased clustering of same stage follicles. Construction of follicle-vasculature relationship maps indicated age- and gonadotropin-dependent increases in vasculature and branching surrounding follicles. Heterozygous mutant mice with deletion of hypoxia-response element in the vascular endothelial growth factor A (VEGFA) promoter showed defective ovarian vasculature and decreased ovulatory responses. Unilateral intrabursal injection of axitinib, an inhibitor of VEGF receptors, retarded neo-angiogenesis that was associated with defective ovulation in treated ovaries. Our approach uncovers unique features of ovarian architecture and essential roles of vasculature in organizing follicles to allow future studies on normal and diseased human ovaries. Similar approaches could also reveal roles of neo-angiogenesis during embryonic development and tumorigenesis.

    View details for DOI 10.1038/srep44810

    View details for PubMedID 28333125

  • A Brainstem-Spinal Cord Inhibitory Circuit for Mechanical Pain Modulation by GABA and Enkephalins. Neuron François, A., Low, S. A., Sypek, E. I., Christensen, A. J., Sotoudeh, C., Beier, K. T., Ramakrishnan, C., Ritola, K. D., Sharif-Naeini, R., Deisseroth, K., Delp, S. L., Malenka, R. C., Luo, L., Hantman, A. W., Scherrer, G. 2017; 93 (4): 822-839 e6

    Abstract

    Pain thresholds are, in part, set as a function of emotional and internal states by descending modulation of nociceptive transmission in the spinal cord. Neurons of the rostral ventromedial medulla (RVM) are thought to critically contribute to this process; however, the neural circuits and synaptic mechanisms by which distinct populations of RVM neurons facilitate or diminish pain remain elusive. Here we used in vivo opto/chemogenetic manipulations and trans-synaptic tracing of genetically identified dorsal horn and RVM neurons to uncover an RVM-spinal cord-primary afferent circuit controlling pain thresholds. Unexpectedly, we found that RVM GABAergic neurons facilitate mechanical pain by inhibiting dorsal horn enkephalinergic/GABAergic interneurons. We further demonstrate that these interneurons gate sensory inputs and control pain through temporally coordinated enkephalin- and GABA-mediated presynaptic inhibition of somatosensory neurons. Our results uncover a descending disynaptic inhibitory circuit that facilitates mechanical pain, is engaged during stress, and could be targeted to establish higher pain thresholds. VIDEO ABSTRACT.

    View details for DOI 10.1016/j.neuron.2017.01.008

    View details for PubMedID 28162807

  • Coordination of Brain-Wide Activity Dynamics by Dopaminergic Neurons NEUROPSYCHOPHARMACOLOGY Decot, H. K., Namboodiri, V. M., Gao, W., McHenry, J. A., Jennings, J. H., Lee, S., Kantak, P. A., Kao, Y. J., Das, M., Witten, I. B., Deisseroth, K., Shih, Y. I., Stuber, G. D. 2017; 42 (3): 615-627

    Abstract

    Several neuropsychiatric conditions, such as addiction and schizophrenia, may arise in part from dysregulated activity of ventral tegmental area dopaminergic (TH(VTA)) neurons, as well as from more global maladaptation in neurocircuit function. However, whether TH(VTA) activity affects large-scale brain-wide function remains unknown. Here we selectively activated TH(VTA) neurons in transgenic rats and measured resulting changes in whole-brain activity using stimulus-evoked functional magnetic resonance imaging. Applying a standard generalized linear model analysis approach, our results indicate that selective optogenetic stimulation of TH(VTA) neurons enhanced cerebral blood volume signals in striatal target regions in a dopamine receptor-dependent manner. However, brain-wide voxel-based principal component analysis of the same data set revealed that dopaminergic modulation activates several additional anatomically distinct regions throughout the brain, not typically associated with dopamine release events. Furthermore, explicit pairing of TH(VTA) neuronal activation with a forepaw stimulus of a particular frequency expanded the sensory representation of that stimulus, not exclusively within the somatosensory cortices, but brain-wide. These data suggest that modulation of TH(VTA) neurons can impact brain dynamics across many distributed anatomically distinct regions, even those that receive little to no direct TH(VTA) input.

    View details for DOI 10.1038/npp.2016.15

    View details for Web of Science ID 000392166400006

  • Molecular interrogation of hypothalamic organization reveals distinct dopamine neuronal subtypes NATURE NEUROSCIENCE Romanov, R. A., Zeisel, A., Bakker, J., Girach, F., Hellysaz, A., Tomer, R., Alpar, A., Mulder, J., Clotman, F., Keimpema, E., Hsueh, B., Crow, A. K., Martens, H., Schwindling, C., Calvigioni, D., Bains, J. S., Mate, Z., Szabo, G., Yanagawa, Y., Zhang, M., Rendeiro, A., Farlik, M., Uhlen, M., Wulff, P., Bock, C., Broberger, C., Deisseroth, K., Hokfelt, T., Linnarsson, S., Horvath, T. L., Harkany, T. 2017; 20 (2): 176-188

    Abstract

    The hypothalamus contains the highest diversity of neurons in the brain. Many of these neurons can co-release neurotransmitters and neuropeptides in a use-dependent manner. Investigators have hitherto relied on candidate protein-based tools to correlate behavioral, endocrine and gender traits with hypothalamic neuron identity. Here we map neuronal identities in the hypothalamus by single-cell RNA sequencing. We distinguished 62 neuronal subtypes producing glutamatergic, dopaminergic or GABAergic markers for synaptic neurotransmission and harboring the ability to engage in task-dependent neurotransmitter switching. We identified dopamine neurons that uniquely coexpress the Onecut3 and Nmur2 genes, and placed these in the periventricular nucleus with many synaptic afferents arising from neuromedin S(+) neurons of the suprachiasmatic nucleus. These neuroendocrine dopamine cells may contribute to the dopaminergic inhibition of prolactin secretion diurnally, as their neuromedin S(+) inputs originate from neurons expressing Per2 and Per3 and their tyrosine hydroxylase phosphorylation is regulated in a circadian fashion. Overall, our catalog of neuronal subclasses provides new understanding of hypothalamic organization and function.

    View details for DOI 10.1038/nn.4462

    View details for Web of Science ID 000393271000012

    View details for PubMedID 27991900

  • Dopaminergic dynamics underlying sex-specific cocaine reward NATURE COMMUNICATIONS Calipari, E. S., Juarez, B., Morel, C., Walker, D. M., Cahill, M. E., Ribeiro, E., Roman-Ortiz, C., Ramakrishnan, C., Deisseroth, K., Han, M., Nestler, E. J. 2017; 8

    Abstract

    Although both males and females become addicted to cocaine, females transition to addiction faster and experience greater difficulties remaining abstinent. We demonstrate an oestrous cycle-dependent mechanism controlling increased cocaine reward in females. During oestrus, ventral tegmental area (VTA) dopamine neuron activity is enhanced and drives post translational modifications at the dopamine transporter (DAT) to increase the ability of cocaine to inhibit its function, an effect mediated by estradiol. Female mice conditioned to associate cocaine with contextual cues during oestrus have enhanced mesolimbic responses to these cues in the absence of drug. Using chemogenetic approaches, we increase VTA activity to mechanistically link oestrous cycle-dependent enhancement of VTA firing to enhanced cocaine affinity at DAT and subsequent reward processing. These data have implications for sexual dimorphism in addiction vulnerability and define a mechanism by which cellular activity results in protein alterations that contribute to dysfunctional learning and reward processing.

    View details for DOI 10.1038/ncomms13877

    View details for Web of Science ID 000391544800001

    View details for PubMedID 28072417

    View details for PubMedCentralID PMC5234081

  • Bidirectional Control of Generalized Epilepsy Networks via Rapid Real-Time Switching of Firing Mode. Neuron Sorokin, J. M., Davidson, T. J., Frechette, E., Abramian, A. M., Deisseroth, K., Huguenard, J. R., Paz, J. T. 2017; 93 (1): 194-210

    Abstract

    Thalamic relay neurons have well-characterized dual firing modes: bursting and tonic spiking. Studies in brain slices have led to a model in which rhythmic synchronized spiking (phasic firing) in a population of relay neurons leads to hyper-synchronous oscillatory cortico-thalamo-cortical rhythms that result in absence seizures. This model suggests that blocking thalamocortical phasic firing would treat absence seizures. However, recent in vivo studies in anesthetized animals have questioned this simple model. Here we resolve this issue by developing a real-time, mode-switching approach to drive thalamocortical neurons into or out of a phasic firing mode in two freely behaving genetic rodent models of absence epilepsy. Toggling between phasic and tonic firing in thalamocortical neurons launched and aborted absence seizures, respectively. Thus, a synchronous thalamocortical phasic firing state is required for absence seizures, and switching to tonic firing rapidly halts absences. This approach should be useful for modulating other networks that have mode-dependent behaviors.

    View details for DOI 10.1016/j.neuron.2016.11.026

    View details for PubMedID 27989462

    View details for PubMedCentralID PMC5268077

  • The need for calcium imaging in nonhuman primates: New motor neuroscience and brain-machine interfaces EXPERIMENTAL NEUROLOGY O'Shea, D. J., Tiautmann, E., Chandrasekaran, C., Stavisky, S., Kao, J. C., Sahani, M., Ryu, S., Deisseroth, K., Shenoy, K. V. 2017; 287: 437-451

    Abstract

    A central goal of neuroscience is to understand how populations of neurons coordinate and cooperate in order to give rise to perception, cognition, and action. Nonhuman primates (NHPs) are an attractive model with which to understand these mechanisms in humans, primarily due to the strong homology of their brains and the cognitively sophisticated behaviors they can be trained to perform. Using electrode recordings, the activity of one to a few hundred individual neurons may be measured electrically, which has enabled many scientific findings and the development of brain-machine interfaces. Despite these successes, electrophysiology samples sparsely from neural populations and provides little information about the genetic identity and spatial micro-organization of recorded neurons. These limitations have spurred the development of all-optical methods for neural circuit interrogation. Fluorescent calcium signals serve as a reporter of neuronal responses, and when combined with post-mortem optical clearing techniques such as CLARITY, provide dense recordings of neuronal populations, spatially organized and annotated with genetic and anatomical information. Here, we advocate that this methodology, which has been of tremendous utility in smaller animal models, can and should be developed for use with NHPs. We review here several of the key opportunities and challenges for calcium-based optical imaging in NHPs. We focus on motor neuroscience and brain-machine interface design as representative domains of opportunity within the larger field of NHP neuroscience.

    View details for DOI 10.1016/j.expneurol.2016.08.003

    View details for Web of Science ID 000391158800002

    View details for PubMedCentralID PMC5154795

  • Gating of social reward by oxytocin in the ventral tegmental area. Science (New York, N.Y.) Hung, L. W., Neuner, S. n., Polepalli, J. S., Beier, K. T., Wright, M. n., Walsh, J. J., Lewis, E. M., Luo, L. n., Deisseroth, K. n., Dölen, G. n., Malenka, R. C. 2017; 357 (6358): 1406–11

    Abstract

    The reward generated by social interactions is critical for promoting prosocial behaviors. Here we present evidence that oxytocin (OXT) release in the ventral tegmental area (VTA), a key node of the brain's reward circuitry, is necessary to elicit social reward. During social interactions, activity in paraventricular nucleus (PVN) OXT neurons increased. Direct activation of these neurons in the PVN or their terminals in the VTA enhanced prosocial behaviors. Conversely, inhibition of PVN OXT axon terminals in the VTA decreased social interactions. OXT increased excitatory drive onto reward-specific VTA dopamine (DA) neurons. These results demonstrate that OXT promotes prosocial behavior through direct effects on VTA DA neurons, thus providing mechanistic insight into how social interactions can generate rewarding experiences.

    View details for PubMedID 28963257

  • Developmental Dysfunction of VIP Interneurons Impairs Cortical Circuits. Neuron Batista-Brito, R. n., Vinck, M. n., Ferguson, K. A., Chang, J. T., Laubender, D. n., Lur, G. n., Mossner, J. M., Hernandez, V. G., Ramakrishnan, C. n., Deisseroth, K. n., Higley, M. J., Cardin, J. A. 2017; 95 (4): 884–95.e9

    Abstract

    GABAergic interneurons play important roles in cortical circuit development. However, there are multiple populations of interneurons and their respective developmental contributions remain poorly explored. Neuregulin 1 (NRG1) and its interneuron-specific receptor ERBB4 are critical genes for interneuron maturation. Using a conditional ErbB4 deletion, we tested the role of vasoactive intestinal peptide (VIP)-expressing interneurons in the postnatal maturation of cortical circuits in vivo. ErbB4 removal from VIP interneurons during development leads to changes in their activity, along with severe dysregulation of cortical temporal organization and state dependence. These alterations emerge during adolescence, and mature animals in which VIP interneurons lack ErbB4 exhibit reduced cortical responses to sensory stimuli and impaired sensory learning. Our data support a key role for VIP interneurons in cortical circuit development and suggest a possible contribution to pathophysiology in neurodevelopmental disorders. These findings provide a new perspective on the role of GABAergic interneuron diversity in cortical development. VIDEO ABSTRACT.

    View details for PubMedID 28817803

  • A radial axis defined by semaphorin-to-neuropilin signaling controls pancreatic islet morphogenesis. Development (Cambridge, England) Pauerstein, P. T., Tellez, K. n., Willmarth, K. B., Park, K. M., Hsueh, B. n., Efsun Arda, H. n., Gu, X. n., Aghajanian, H. n., Deisseroth, K. n., Epstein, J. A., Kim, S. K. 2017; 144 (20): 3744–54

    Abstract

    The islets of Langerhans are endocrine organs characteristically dispersed throughout the pancreas. During development, endocrine progenitors delaminate, migrate radially and cluster to form islets. Despite the distinctive distribution of islets, spatially localized signals that control islet morphogenesis have not been discovered. Here, we identify a radial signaling axis that instructs developing islet cells to disperse throughout the pancreas. A screen of pancreatic extracellular signals identified factors that stimulated islet cell development. These included semaphorin 3a, a guidance cue in neural development without known functions in the pancreas. In the fetal pancreas, peripheral mesenchymal cells expressed Sema3a, while central nascent islet cells produced the semaphorin receptor neuropilin 2 (Nrp2). Nrp2 mutant islet cells developed in proper numbers, but had defects in migration and were unresponsive to purified Sema3a. Mutant Nrp2 islets aggregated centrally and failed to disperse radially. Thus, Sema3a-Nrp2 signaling along an unrecognized pancreatic developmental axis constitutes a chemoattractant system essential for generating the hallmark morphogenetic properties of pancreatic islets. Unexpectedly, Sema3a- and Nrp2-mediated control of islet morphogenesis is strikingly homologous to mechanisms that regulate radial neuronal migration and cortical lamination in the developing mammalian brain.

    View details for PubMedID 28893946

  • Patterned photostimulation via visible-wavelength photonic probes for deep brain optogenetics. Neurophotonics Segev, E., Reimer, J., Moreaux, L. C., Fowler, T. M., Chi, D., Sacher, W. D., Lo, M., Deisseroth, K., Tolias, A. S., Faraon, A., Roukes, M. L. 2017; 4 (1): 011002-?

    Abstract

    Optogenetic methods developed over the past decade enable unprecedented optical activation and silencing of specific neuronal cell types. However, light scattering in neural tissue precludes illuminating areas deep within the brain via free-space optics; this has impeded employing optogenetics universally. Here, we report an approach surmounting this significant limitation. We realize implantable, ultranarrow, silicon-based photonic probes enabling the delivery of complex illumination patterns deep within brain tissue. Our approach combines methods from integrated nanophotonics and microelectromechanical systems, to yield photonic probes that are robust, scalable, and readily producible en masse. Their minute cross sections minimize tissue displacement upon probe implantation. We functionally validate one probe design in vivo with mice expressing channelrhodopsin-2. Highly local optogenetic neural activation is demonstrated by recording the induced response-both by extracellular electrical recordings in the hippocampus and by two-photon functional imaging in the cortex of mice coexpressing GCaMP6.

    View details for PubMedID 27990451

  • Basomedial Amygdala Mediates Top-Down Control of Anxiety and Fear Adhikari, A., Lerner, T., Finkelstein, J., Pak, S., Jennings, J., Davidson, T., Ferenczi, E., Gunaydin, L., Mirzabekov, J., Ye, L., Kim, S., Lei, A., Deisseroth, K. NATURE PUBLISHING GROUP. 2016: S293-S294
  • In Vivo Interrogation of Spinal Mechanosensory Circuits. Cell reports Christensen, A. J., Iyer, S. M., François, A., Vyas, S., Ramakrishnan, C., Vesuna, S., Deisseroth, K., Scherrer, G., Delp, S. L. 2016; 17 (6): 1699-1710

    Abstract

    Spinal dorsal horn circuits receive, process, and transmit somatosensory information. To understand how specific components of these circuits contribute to behavior, it is critical to be able to directly modulate their activity in unanesthetized in vivo conditions. Here, we develop experimental tools that enable optogenetic control of spinal circuitry in freely moving mice using commonly available materials. We use these tools to examine mechanosensory processing in the spinal cord and observe that optogenetic activation of somatostatin-positive interneurons facilitates both mechanosensory and itch-related behavior, while reversible chemogenetic inhibition of these neurons suppresses mechanosensation. These results extend recent findings regarding the processing of mechanosensory information in the spinal cord and indicate the potential for activity-induced release of the somatostatin neuropeptide to affect processing of itch. The spinal implant approach we describe here is likely to enable a wide range of studies to elucidate spinal circuits underlying pain, touch, itch, and movement.

    View details for DOI 10.1016/j.celrep.2016.10.010

    View details for PubMedID 27806306

  • Locus coeruleus and dopaminergic consolidation of everyday memory NATURE Takeuchi, T., Duszkiewicz, A. J., Sonneborn, A., Spooner, P. A., Yamasaki, M., Watanabe, M., Smith, C. C., Fernandez, G., Deisseroth, K., Greene, R. W., Morris, R. G. 2016; 537 (7620): 357-?

    Abstract

    The retention of episodic-like memory is enhanced, in humans and animals, when something novel happens shortly before or after encoding. Using an everyday memory task in mice, we sought the neurons mediating this dopamine-dependent novelty effect, previously thought to originate exclusively from the tyrosine-hydroxylase-expressing (TH(+)) neurons in the ventral tegmental area. Here we report that neuronal firing in the locus coeruleus is especially sensitive to environmental novelty, locus coeruleus TH(+) neurons project more profusely than ventral tegmental area TH(+) neurons to the hippocampus, optogenetic activation of locus coeruleus TH(+) neurons mimics the novelty effect, and this novelty-associated memory enhancement is unaffected by ventral tegmental area inactivation. Surprisingly, two effects of locus coeruleus TH(+) photoactivation are sensitive to hippocampal D1/D5 receptor blockade and resistant to adrenoceptor blockade: memory enhancement and long-lasting potentiation of synaptic transmission in CA1 ex vivo. Thus, locus coeruleus TH(+) neurons can mediate post-encoding memory enhancement in a manner consistent with possible co-release of dopamine in the hippocampus.

    View details for DOI 10.1038/nature19325

    View details for Web of Science ID 000383098000045

    View details for PubMedID 27602521

    View details for PubMedCentralID PMC5161591

  • Coordination of Brain-Wide Activity Dynamics by Dopaminergic Neurons. Neuropsychopharmacology Decot, H. K., Namboodiri, V. M., Gao, W., McHenry, J. A., Jennings, J. H., Lee, S., Kantak, P. A., Jill Kao, Y., Das, M., Witten, I. B., Deisseroth, K., Shih, Y. I., Stuber, G. D. 2016

    Abstract

    Several neuropsychiatric conditions, such as addiction and schizophrenia, may arise in part from dysregulated activity of ventral tegmental area dopaminergic (TH(VTA)) neurons, as well as from more global maladaptation in neurocircuit function. However, whether TH(VTA) activity affects large-scale brain-wide function remains unknown. Here we selectively activated TH(VTA) neurons in transgenic rats and measured resulting changes in whole-brain activity using stimulus-evoked functional magnetic resonance imaging. Applying a standard generalized linear model analysis approach, our results indicate that selective optogenetic stimulation of TH(VTA) neurons enhanced cerebral blood volume signals in striatal target regions in a dopamine receptor-dependent manner. However, brain-wide voxel-based principal component analysis of the same data set revealed that dopaminergic modulation activates several additional anatomically distinct regions throughout the brain, not typically associated with dopamine release events. Furthermore, explicit pairing of TH(VTA) neuronal activation with a forepaw stimulus of a particular frequency expanded the sensory representation of that stimulus, not exclusively within the somatosensory cortices, but brain-wide. These data suggest that modulation of TH(VTA) neurons can impact brain dynamics across many distributed anatomically distinct regions, even those that receive little to no direct TH(VTA) input.

    View details for DOI 10.1038/npp.2016.151

    View details for PubMedID 27515791

  • Pontomesencephalic Tegmental Afferents to VTA Non-dopamine Neurons Are Necessary for Appetitive Pavlovian Learning. Cell reports Yau, H., Wang, D. V., Tsou, J., Chuang, Y., Chen, B. T., Deisseroth, K., Ikemoto, S., Bonci, A. 2016; 16 (10): 2699-2710

    Abstract

    The ventral tegmental area (VTA) receives phenotypically distinct innervations from the pedunculopontine tegmental nucleus (PPTg). While PPTg-to-VTA inputs are thought to play a critical role in stimulus-reward learning, direct evidence linking PPTg-to-VTA phenotypically distinct inputs in the learning process remains lacking. Here, we used optogenetic approaches to investigate the functional contribution of PPTg excitatory and inhibitory inputs to the VTA in appetitive Pavlovian conditioning. We show that photoinhibition of PPTg-to-VTA cholinergic or glutamatergic inputs during cue presentation dampens the development of anticipatory approach responding to the food receptacle during the cue. Furthermore, we employed in vivo optetrode recordings to show that photoinhibition of PPTg cholinergic or glutamatergic inputs significantly decreases VTA non-dopamine (non-DA) neural activity. Consistently, photoinhibition of VTA non-DA neurons disrupts the development of cue-elicited anticipatory approach responding. Taken together, our study reveals a crucial regulatory mechanism by PPTg excitatory inputs onto VTA non-DA neurons during appetitive Pavlovian conditioning.

    View details for DOI 10.1016/j.celrep.2016.08.007

    View details for PubMedID 27568569

  • Serotonin engages an anxiety and fear-promoting circuit in the extended amygdala NATURE Marcinkiewcz, C. A., Mazzone, C. M., D'Agostino, G., Halladay, L. R., Hardaway, J. A., DiBerto, J. F., Navarro, M., Burnham, N., Cristiano, C., Dorrier, C. E., Tipton, G. J., Ramakrishnan, C., Kozicz, T., Deisseroth, K., Thiele, T. E., McElligott, Z. A., Holmes, A., Heisler, L. K., Kash, T. L. 2016; 537 (7618): 97-101

    Abstract

    Serotonin (also known as 5-hydroxytryptamine (5-HT)) is a neurotransmitter that has an essential role in the regulation of emotion. However, the precise circuits have not yet been defined through which aversive states are orchestrated by 5-HT. Here we show that 5-HT from the dorsal raphe nucleus (5-HTDRN) enhances fear and anxiety and activates a subpopulation of corticotropin-releasing factor (CRF) neurons in the bed nucleus of the stria terminalis (CRFBNST) in mice. Specifically, 5-HTDRN projections to the BNST, via actions at 5-HT2C receptors (5-HT2CRs), engage a CRFBNST inhibitory microcircuit that silences anxiolytic BNST outputs to the ventral tegmental area and lateral hypothalamus. Furthermore, we demonstrate that this CRFBNST inhibitory circuit underlies aversive behaviour following acute exposure to selective serotonin reuptake inhibitors (SSRIs). This early aversive effect is mediated via the corticotrophin-releasing factor type 1 receptor (CRF1R, also known as CRHR1), given that CRF1R antagonism is sufficient to prevent acute SSRI-induced enhancements in aversive learning. These results reveal an essential 5-HTDRN→CRFBNST circuit governing fear and anxiety, and provide a potential mechanistic explanation for the clinical observation of early adverse events to SSRI treatment in some patients with anxiety disorders.

    View details for DOI 10.1038/nature19318

    View details for Web of Science ID 000382426900046

    View details for PubMedCentralID PMC5124365

  • From the retina to the brain: retinal ganglion cell subtype specific visual circuits Young, B., Wang, P., Ramakrishnan, C., Deisseroth, K., Tian, N. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2016
  • Retinal ganglion cell subtype specific circuits in retina Tian, N., Young, B., Huang, K., Wang, P., Ramakrishnan, C., Deisseroth, K. ASSOC RESEARCH VISION OPHTHALMOLOGY INC. 2016
  • Serotonin engages an anxiety and fear-promoting circuit in the extended amygdala. Nature Marcinkiewcz, C. A., Mazzone, C. M., D'Agostino, G., Halladay, L. R., Hardaway, J. A., DiBerto, J. F., Navarro, M., Burnham, N., Cristiano, C., Dorrier, C. E., Tipton, G. J., Ramakrishnan, C., Kozicz, T., Deisseroth, K., Thiele, T. E., McElligott, Z. A., Holmes, A., Heisler, L. K., Kash, T. L. 2016; 537 (7618): 97-101

    Abstract

    Serotonin (also known as 5-hydroxytryptamine (5-HT)) is a neurotransmitter that has an essential role in the regulation of emotion. However, the precise circuits have not yet been defined through which aversive states are orchestrated by 5-HT. Here we show that 5-HT from the dorsal raphe nucleus (5-HTDRN) enhances fear and anxiety and activates a subpopulation of corticotropin-releasing factor (CRF) neurons in the bed nucleus of the stria terminalis (CRFBNST) in mice. Specifically, 5-HTDRN projections to the BNST, via actions at 5-HT2C receptors (5-HT2CRs), engage a CRFBNST inhibitory microcircuit that silences anxiolytic BNST outputs to the ventral tegmental area and lateral hypothalamus. Furthermore, we demonstrate that this CRFBNST inhibitory circuit underlies aversive behaviour following acute exposure to selective serotonin reuptake inhibitors (SSRIs). This early aversive effect is mediated via the corticotrophin-releasing factor type 1 receptor (CRF1R, also known as CRHR1), given that CRF1R antagonism is sufficient to prevent acute SSRI-induced enhancements in aversive learning. These results reveal an essential 5-HTDRN→CRFBNST circuit governing fear and anxiety, and provide a potential mechanistic explanation for the clinical observation of early adverse events to SSRI treatment in some patients with anxiety disorders.

    View details for DOI 10.1038/nature19318

    View details for PubMedID 27556938

    View details for PubMedCentralID PMC5124365

  • Sustained Attentional States Require Distinct Temporal Involvement of the Dorsal and Ventral Medial Prefrontal Cortex FRONTIERS IN NEURAL CIRCUITS Luchicchi, A., Mnie-Filali, O., Terra, H., Bruinsma, B., de Kloet, S. F., Obermayer, J., Heistek, T. S., de Haan, R., De Kock, C. P., Deisseroth, K., Pattij, T., Mansvelder, H. D. 2016; 10

    Abstract

    Attending the sensory environment for cue detection is a cognitive operation that occurs on a time scale of seconds. The dorsal and ventral medial prefrontal cortex (mPFC) contribute to separate aspects of attentional processing. Pyramidal neurons in different parts of the mPFC are active during cognitive behavior, yet whether this activity is causally underlying attentional processing is not known. We aimed to determine the precise temporal requirements for activation of the mPFC subregions during the seconds prior to cue detection. To test this, we used optogenetic silencing of dorsal or ventral mPFC pyramidal neurons at defined time windows during a sustained attentional state. We find that the requirement of ventral mPFC pyramidal neuron activity is strictly time-locked to stimulus detection. Inhibiting the ventral mPFC 2 s before or during cue presentation reduces response accuracy and hampers behavioral inhibition. The requirement for dorsal mPFC activity on the other hand is temporally more loosely related to a preparatory attentional state, and short lapses in pyramidal neuron activity in dorsal mPFC do not affect performance. This only occurs when the dorsal mPFC is inhibited during the entire preparatory period. Together, our results reveal that a dissociable temporal recruitment of ventral and dorsal mPFC is required during attentional processing.

    View details for DOI 10.3389/fncir.2016.00070

    View details for Web of Science ID 000382301300001

    View details for PubMedID 27630545

    View details for PubMedCentralID PMC5005373

  • The need for calcium imaging in nonhuman primates: New motor neuroscience and brain-machine interfaces. Experimental neurology O'Shea, D. J., Trautmann, E., Chandrasekaran, C., Stavisky, S., Kao, J. C., Sahani, M., Ryu, S., Deisseroth, K., Shenoy, K. V. 2016

    Abstract

    A central goal of neuroscience is to understand how populations of neurons coordinate and cooperate in order to give rise to perception, cognition, and action. Nonhuman primates (NHPs) are an attractive model with which to understand these mechanisms in humans, primarily due to the strong homology of their brains and the cognitively sophisticated behaviors they can be trained to perform. Using electrode recordings, the activity of one to a few hundred individual neurons may be measured electrically, which has enabled many scientific findings and the development of brain-machine interfaces. Despite these successes, electrophysiology samples sparsely from neural populations and provides little information about the genetic identity and spatial micro-organization of recorded neurons. These limitations have spurred the development of all-optical methods for neural circuit interrogation. Fluorescent calcium signals serve as a reporter of neuronal responses, and when combined with post-mortem optical clearing techniques such as CLARITY, provide dense recordings of neuronal populations, spatially organized and annotated with genetic and anatomical information. Here, we advocate that this methodology, which has been of tremendous utility in smaller animal models, can and should be developed for use with NHPs. We review here several of the key opportunities and challenges for calcium-based optical imaging in NHPs. We focus on motor neuroscience and brain-machine interface design as representative domains of opportunity within the larger field of NHP neuroscience.

    View details for DOI 10.1016/j.expneurol.2016.08.003

    View details for PubMedID 27511294

    View details for PubMedCentralID PMC5154795

  • Segregated cholinergic transmission modulates dopamine neurons integrated in distinct functional circuits NATURE NEUROSCIENCE Dautan, D., Souza, A. S., Huerta-Ocampo, I., Valencia, M., Assous, M., Witten, I. B., Deisseroth, K., Tepper, J. M., Bolam, J. P., Gerdjikov, T. V., Mena-Segovia, J. 2016; 19 (8): 1025-?

    Abstract

    Dopamine neurons in the ventral tegmental area (VTA) receive cholinergic innervation from brainstem structures that are associated with either movement or reward. Whereas cholinergic neurons of the pedunculopontine nucleus (PPN) carry an associative/motor signal, those of the laterodorsal tegmental nucleus (LDT) convey limbic information. We used optogenetics and in vivo juxtacellular recording and labeling to examine the influence of brainstem cholinergic innervation of distinct neuronal subpopulations in the VTA. We found that LDT cholinergic axons selectively enhanced the bursting activity of mesolimbic dopamine neurons that were excited by aversive stimulation. In contrast, PPN cholinergic axons activated and changed the discharge properties of VTA neurons that were integrated in distinct functional circuits and were inhibited by aversive stimulation. Although both structures conveyed a reinforcing signal, they had opposite roles in locomotion. Our results demonstrate that two modes of cholinergic transmission operate in the VTA and segregate the neurons involved in different reward circuits.

    View details for DOI 10.1038/nn.4335

    View details for Web of Science ID 000380773200011

    View details for PubMedID 27348215

  • Competition between engrams influences fear memory formation and recall SCIENCE Rashid, A. J., Yan, C., Mercaldo, V., Hsiang, H. (., Park, S., Cole, C. J., De Cristofaro, A., Yu, J., Ramakrishnan, C., Lee, S. Y., Deisseroth, K., Frankland, P. W., Josselyn, S. A. 2016; 353 (6297): 383-387

    Abstract

    Collections of cells called engrams are thought to represent memories. Although there has been progress in identifying and manipulating single engrams, little is known about how multiple engrams interact to influence memory. In lateral amygdala (LA), neurons with increased excitability during training outcompete their neighbors for allocation to an engram. We examined whether competition based on neuronal excitability also governs the interaction between engrams. Mice received two distinct fear conditioning events separated by different intervals. LA neuron excitability was optogenetically manipulated and revealed a transient competitive process that integrates memories for events occurring closely in time (coallocating overlapping populations of neurons to both engrams) and separates memories for events occurring at distal times (disallocating nonoverlapping populations to each engram).

    View details for DOI 10.1126/science.aaf0594

    View details for PubMedID 27463673

  • Dysregulation of Prefrontal Cortex-Mediated Slow-Evolving Limbic Dynamics Drives Stress-Induced Emotional Pathology NEURON Hultman, R., Mague, S. D., Li, Q., Katz, B. M., Michel, N., Lin, L., Wang, J., David, L. K., Blount, C., Chandy, R., Carlson, D., Ulrich, K., Carin, L., Dunson, D., Kumar, S., Deisseroth, K., Moore, S. D., Dzirasa, K. 2016; 91 (2): 439-452

    Abstract

    Circuits distributed across cortico-limbic brain regions compose the networks that mediate emotional behavior. The prefrontal cortex (PFC) regulates ultraslow (<1 Hz) dynamics across these networks, and PFC dysfunction is implicated in stress-related illnesses including major depressive disorder (MDD). To uncover the mechanism whereby stress-induced changes in PFC circuitry alter emotional networks to yield pathology, we used a multi-disciplinary approach including in vivo recordings in mice and chronic social defeat stress. Our network model, inferred using machine learning, linked stress-induced behavioral pathology to the capacity of PFC to synchronize amygdala and VTA activity. Direct stimulation of PFC-amygdala circuitry with DREADDs normalized PFC-dependent limbic synchrony in stress-susceptible animals and restored normal behavior. In addition to providing insights into MDD mechanisms, our findings demonstrate an interdisciplinary approach that can be used to identify the large-scale network changes that underlie complex emotional pathologies and the specific network nodes that can be used to develop targeted interventions.

    View details for DOI 10.1016/j.neuron.2016.05.038

    View details for PubMedID 27346529

  • Phototactic guidance of a tissue-engineered soft-robotic ray SCIENCE Park, S., Gazzola, M., Park, K. S., Park, S., Di Santo, V., Blevins, E. L., Lind, J. U., Campbell, P. H., Dauth, S., Capulli, A. K., Pasqualini, F. S., Ahn, S., Cho, A., Yuan, H., Maoz, B. M., Vijaykumar, R., Choi, J., Deisseroth, K., Lauder, G. V., Mahadevan, L., Parker, K. K. 2016; 353 (6295): 158-162

    Abstract

    Inspired by the relatively simple morphological blueprint provided by batoid fish such as stingrays and skates, we created a biohybrid system that enables an artificial animal--a tissue-engineered ray--to swim and phototactically follow a light cue. By patterning dissociated rat cardiomyocytes on an elastomeric body enclosing a microfabricated gold skeleton, we replicated fish morphology at 1/10 scale and captured basic fin deflection patterns of batoid fish. Optogenetics allows for phototactic guidance, steering, and turning maneuvers. Optical stimulation induced sequential muscle activation via serpentine-patterned muscle circuits, leading to coordinated undulatory swimming. The speed and direction of the ray was controlled by modulating light frequency and by independently eliciting right and left fins, allowing the biohybrid machine to maneuver through an obstacle course.

    View details for DOI 10.1126/science.aaf4292

    View details for Web of Science ID 000379208400036

    View details for PubMedID 27387948

  • LSPS/Optogenetics to Improve Synaptic Connectivity Mapping: Unmasking the Role of Basket Cell-Mediated Feedforward Inhibition. eNeuro Brill, J., Mattis, J., Deisseroth, K., Huguenard, J. R. 2016; 3 (4)

    Abstract

    Neocortical pyramidal cells (PYRs) receive synaptic inputs from many types of GABAergic interneurons. Connections between parvalbumin (PV)-positive, fast-spiking interneurons ("PV cells") and PYRs are characterized by perisomatic synapses and high-amplitude, short-latency IPSCs. Here, we present novel methods to study the functional influence of PV cells on layer 5 PYRs using optogenetics combined with laser-scanning photostimulation (LSPS). First, we examined the strength and spatial distribution of PV-to-PYR inputs. To that end, the fast channelrhodopsin variant AAV5-EF1α-DIO-hChR2(E123T)-eYFP (ChETA) was expressed in PV cells in somatosensory cortex of mice using an adeno-associated virus-based viral construct. Focal blue illumination (100-150 µm half-width) was directed through the microscope objective to excite PV cells along a spatial grid covering layers 2-6, while IPSCs were recorded in layer 5 PYRs. The resulting optogenetic input maps showed evoked PV cell inputs originating from an ∼500-μm-diameter area surrounding the recorded PYR. Evoked IPSCs had the short-latency/high-amplitude characteristic of PV cell inputs. Second, we investigated how PV cell activity modulates PYR output in response to synaptic excitation. We expressed halorhodopsin (eNpHR3.0) in PV cells using the same strategy as for ChETA. Yellow illumination hyperpolarized eNpHR3.0-expressing PV cells, effectively preventing action potential generation and thus decreasing the inhibition of downstream targets. Synaptic input maps onto layer 5 PYRs were acquired using standard glutamate-photolysis LSPS either with or without full-field yellow illumination to silence PV cells. The resulting IPSC input maps selectively lacked short-latency perisomatic inputs, while EPSC input maps showed increased connectivity, particularly from upper layers. This indicates that glutamate uncaging LSPS-based excitatory synaptic maps will consistently underestimate connectivity.

    View details for DOI 10.1523/ENEURO.0142-15.2016

    View details for PubMedID 27517089

    View details for PubMedCentralID PMC4976301

  • Endocannabinoid Modulation of Orbitostriatal Circuits Gates Habit Formation NEURON Gremel, C. M., Chancey, J. H., Atwood, B. K., Luo, G., Neve, R., Ramakrishnan, C., Deisseroth, K., Lovinger, D. M., Costa, R. M. 2016; 90 (6): 1312-1324

    Abstract

    Everyday function demands efficient and flexible decision-making that allows for habitual and goal-directed action control. An inability to shift has been implicated in disorders with impaired decision-making, including obsessive-compulsive disorder and addiction. Despite this, our understanding of the specific molecular mechanisms and circuitry involved in shifting action control remains limited. Here we identify an endogenous molecular mechanism in a specific cortical-striatal pathway that mediates the transition between goal-directed and habitual action strategies. Deletion of cannabinoid type 1 (CB1) receptors from cortical projections originating in the orbital frontal cortex (OFC) prevents mice from shifting from goal-directed to habitual instrumental lever pressing. Activity of OFC neurons projecting to dorsal striatum (OFC-DS) and, specifically, activity of OFC-DS terminals is necessary for goal-directed action control. Lastly, CB1 deletion from OFC-DS neurons prevents the shift from goal-directed to habitual action control. These data suggest that the emergence of habits depends on endocannabinoid-mediated attenuation of a competing circuit controlling goal-directed behaviors.

    View details for DOI 10.1016/j.neuron.2016.04.043

    View details for Web of Science ID 000378527600018

    View details for PubMedID 27238866

    View details for PubMedCentralID PMC4911264

  • Midbrain circuits for defensive behaviour NATURE Tovote, P., Esposito, M. S., Botta, P., Haudun, F. C., Fadok, J. P., Markovic, M., Wolff, S. B., Ramakrishnan, C., Fenno, L., Deisseroth, K., Herry, C., Arber, S., Luthi, A. 2016; 534 (7606): 206-?

    Abstract

    Survival in threatening situations depends on the selection and rapid execution of an appropriate active or passive defensive response, yet the underlying brain circuitry is not understood. Here we use circuit-based optogenetic, in vivo and in vitro electrophysiological, and neuroanatomical tracing methods to define midbrain periaqueductal grey circuits for specific defensive behaviours. We identify an inhibitory pathway from the central nucleus of the amygdala to the ventrolateral periaqueductal grey that produces freezing by disinhibition of ventrolateral periaqueductal grey excitatory outputs to pre-motor targets in the magnocellular nucleus of the medulla. In addition, we provide evidence for anatomical and functional interaction of this freezing pathway with long-range and local circuits mediating flight. Our data define the neuronal circuitry underlying the execution of freezing, an evolutionarily conserved defensive behaviour, which is expressed by many species including fish, rodents and primates. In humans, dysregulation of this 'survival circuit' has been implicated in anxiety-related disorders.

    View details for DOI 10.1038/nature17996

    View details for Web of Science ID 000377475100032

    View details for PubMedID 27279213

  • Hilar somatostatin interneuron loss reduces dentate gyrus inhibition in a mouse model of temporal lobe epilepsy EPILEPSIA Hofmann, G., Balgooyen, L., Mattis, J., Deisseroth, K., Buckmaster, P. S. 2016; 57 (6): 977-983

    Abstract

    In patients with temporal lobe epilepsy, seizures usually start in the hippocampus, and dentate granule cells are hyperexcitable. Somatostatin interneurons are a major subpopulation of inhibitory neurons in the dentate gyrus, and many are lost in patients and animal models. However, surviving somatostatin interneurons sprout axon collaterals and form new synapses, so the net effect on granule cell inhibition remains unclear.The present study uses optogenetics to activate hilar somatostatin interneurons and measure the inhibitory effect on dentate gyrus perforant path-evoked local field potential responses in a mouse model of temporal lobe epilepsy.In controls, light activation of hilar somatostatin interneurons inhibited evoked responses up to 40%. Epileptic pilocarpine-treated mice exhibited loss of hilar somatostatin interneurons and less light-induced inhibition of evoked responses.These findings suggest that severe epilepsy-related loss of hilar somatostatin interneurons can overwhelm the surviving interneurons' capacity to compensate by sprouting axon collaterals.

    View details for DOI 10.1111/epi.13376

    View details for Web of Science ID 000380151100017

    View details for PubMedID 27030321

    View details for PubMedCentralID PMC4903908

  • Integrated device for combined optical neuromodulation and electrical recording for chronic in vivo applications (vol 9, 016001, 2012) JOURNAL OF NEURAL ENGINEERING Wang, J., Wagner, F., Diagne, M., Borton, D. A., Zhang, J., Ozden, I., Burwell, R. D., Nurmikko, A. V., van Wagenen, R., Diester, I., Deisseroth, K. 2016; 13 (3)
  • Astrocyte Intermediaries of Septal Cholinergic Modulation in the Hippocampus NEURON Pabst, M., Braganza, O., Dannenberg, H., Hu, W., Pothmann, L., Rosen, J., Mody, I., van Loo, K., Deisseroth, K., Becker, A. J., Schoch, S., Beck, H. 2016; 90 (4): 853-865

    Abstract

    The neurotransmitter acetylcholine, derived from the medial septum/diagonal band of Broca complex, has been accorded an important role in hippocampal learning and memory processes. However, the precise mechanisms whereby acetylcholine released from septohippocampal cholinergic neurons acts to modulate hippocampal microcircuits remain unknown. Here, we show that acetylcholine release from cholinergic septohippocampal projections causes a long-lasting GABAergic inhibition of hippocampal dentate granule cells in vivo and in vitro. This inhibition is caused by cholinergic activation of hilar astrocytes, which provide glutamatergic excitation of hilar inhibitory interneurons. These results demonstrate that acetylcholine release can cause slow inhibition of principal neuronal activity via astrocyte intermediaries.

    View details for DOI 10.1016/j.neuron.2016.04.003

    View details for Web of Science ID 000376255600017

    View details for PubMedID 27161528

  • Beyond the brain: Optogenetic control in the spinal cord and peripheral nervous system SCIENCE TRANSLATIONAL MEDICINE Montgomery, K. L., Iyer, S. M., Christensen, A. J., Deisseroth, K., Delp, S. L. 2016; 8 (337)

    Abstract

    Optogenetics offers promise for dissecting the complex neural circuits of the spinal cord and peripheral nervous system and has therapeutic potential for addressing unmet clinical needs. Much progress has been made to enable optogenetic control in normal and disease states, both in proof-of-concept and mechanistic studies in rodent models. In this Review, we discuss challenges in using optogenetics to study the mammalian spinal cord and peripheral nervous system, synthesize common features that unite the work done thus far, and describe a route forward for the successful application of optogenetics to translational research beyond the brain.

    View details for DOI 10.1126/scitranslmed.aad7577

    View details for Web of Science ID 000375802500004

    View details for PubMedID 27147590

  • Dynamic changes in neural circuitry during adolescence are associated with persistent attenuation of fear memories NATURE COMMUNICATIONS Pattwell, S. S., Liston, C., Jing, D., Ninan, I., Yang, R. R., Witztum, J., Murdock, M. H., Dincheva, I., Bath, K. G., Casey, B. J., Deisseroth, K., Lee, F. S. 2016; 7

    Abstract

    Fear can be highly adaptive in promoting survival, yet it can also be detrimental when it persists long after a threat has passed. Flexibility of the fear response may be most advantageous during adolescence when animals are prone to explore novel, potentially threatening environments. Two opposing adolescent fear-related behaviours-diminished extinction of cued fear and suppressed expression of contextual fear-may serve this purpose, but the neural basis underlying these changes is unknown. Using microprisms to image prefrontal cortical spine maturation across development, we identify dynamic BLA-hippocampal-mPFC circuit reorganization associated with these behavioural shifts. Exploiting this sensitive period of neural development, we modified existing behavioural interventions in an age-specific manner to attenuate adolescent fear memories persistently into adulthood. These findings identify novel strategies that leverage dynamic neurodevelopmental changes during adolescence with the potential to extinguish pathological fears implicated in anxiety and stress-related disorders.

    View details for DOI 10.1038/ncomms11475

    View details for Web of Science ID 000376614700001

    View details for PubMedID 27215672

    View details for PubMedCentralID PMC4890178

  • Optogenetic approaches addressing extracellular modulation of neural excitability SCIENTIFIC REPORTS Ferenczi, E. A., Vierock, J., Atsuta-Tsunoda, K., Tsunoda, S. P., Ramakrishnan, C., Gorini, C., Thompson, K., Lee, S. Y., Berndt, A., Perry, C., Minniberger, S., Vogt, A., Mattis, J., Prakash, R., Delp, S., Deisseroth, K., Hegemann, P. 2016; 6

    Abstract

    The extracellular ionic environment in neural tissue has the capacity to influence, and be influenced by, natural bouts of neural activity. We employed optogenetic approaches to control and investigate these interactions within and between cells, and across spatial scales. We began by developing a temporally precise means to study microdomain-scale interactions between extracellular protons and acid-sensing ion channels (ASICs). By coupling single-component proton-transporting optogenetic tools to ASICs to create two-component optogenetic constructs (TCOs), we found that acidification of the local extracellular membrane surface by a light-activated proton pump recruited a slow inward ASIC current, which required molecular proximity of the two components on the membrane. To elicit more global effects of activity modulation on 'bystander' neurons not under direct control, we used densely-expressed depolarizing (ChR2) or hyperpolarizing (eArch3.0, eNpHR3.0) tools to create a slow non-synaptic membrane current in bystander neurons, which matched the current direction seen in the directly modulated neurons. Extracellular protons played contributory role but were insufficient to explain the entire bystander effect, suggesting the recruitment of other mechanisms. Together, these findings present a new approach to the engineering of multicomponent optogenetic tools to manipulate ionic microdomains, and probe the complex neuronal-extracellular space interactions that regulate neural excitability.

    View details for DOI 10.1038/srep23947

    View details for Web of Science ID 000373399700001

    View details for PubMedCentralID PMC4820717

  • Preclinical Model of Parkinson's Disease Using Circuit Specific Optogenetics Patel, N., Iyer, V., Venkiteswaran, K., Handly, E., White, C., Iqbal, N., Sridhar, P., Thiagarajan, K., Liu, Z., Ramakrishnan, C., Deisseroth, K., Subramanian, T. LIPPINCOTT WILLIAMS & WILKINS. 2016
  • Hypothalamic control of male aggression-seeking behavior. Nature neuroscience Falkner, A. L., Grosenick, L., Davidson, T. J., Deisseroth, K., Lin, D. 2016; 19 (4): 596-604

    Abstract

    In many vertebrate species, certain individuals will seek out opportunities for aggression, even in the absence of threat-provoking cues. Although several brain areas have been implicated in the generation of attack in response to social threat, little is known about the neural mechanisms that promote self-initiated or 'voluntary' aggression-seeking when no threat is present. To explore this directly, we utilized an aggression-seeking task in which male mice self-initiated aggression trials to gain brief and repeated access to a weaker male that they could attack. In males that exhibited rapid task learning, we found that the ventrolateral part of the ventromedial hypothalamus (VMHvl), an area with a known role in attack, was essential for aggression-seeking. Using both single-unit electrophysiology and population optical recording, we found that VMHvl neurons became active during aggression-seeking and that their activity tracked changes in task learning and extinction. Inactivation of the VMHvl reduced aggression-seeking behavior, whereas optogenetic stimulation of the VMHvl accelerated moment-to-moment aggression-seeking and intensified future attack. These data demonstrate that the VMHvl can mediate both acute attack and flexible seeking actions that precede attack.

    View details for DOI 10.1038/nn.4264

    View details for PubMedID 26950005

    View details for PubMedCentralID PMC4853470

  • Chronic cocaine exposure alters D1 medium spiny neuron activity to promote relapse Calipari, E. S., Bagot, R. C., Purushothaman, I., Pirpinias, S., Davidson, T. J., Deisseroth, K., Nestler, E. J. FEDERATION AMER SOC EXP BIOL. 2016
  • Nucleus accumbens D2R cells signal prior outcomes and control risky decision-making. Nature Zalocusky, K. A., Ramakrishnan, C., Lerner, T. N., Davidson, T. J., Knutson, B., Deisseroth, K. 2016; 531 (7596): 642-646

    Abstract

    A marked bias towards risk aversion has been observed in nearly every species tested. A minority of individuals, however, instead seem to prefer risk (repeatedly choosing uncertain large rewards over certain but smaller rewards), and even risk-averse individuals sometimes opt for riskier alternatives. It is not known how neural activity underlies such important shifts in decision-making-either as a stable trait across individuals or at the level of variability within individuals. Here we describe a model of risk-preference in rats, in which stable individual differences, trial-by-trial choices, and responses to pharmacological agents all parallel human behaviour. By combining new genetic targeting strategies with optical recording of neural activity during behaviour in this model, we identify relevant temporally specific signals from a genetically and anatomically defined population of neurons. This activity occurred within dopamine receptor type-2 (D2R)-expressing cells in the nucleus accumbens (NAc), signalled unfavourable outcomes from the recent past at a time appropriate for influencing subsequent decisions, and also predicted subsequent choices made. Having uncovered this naturally occurring neural correlate of risk selection, we then mimicked the temporally specific signal with optogenetic control during decision-making and demonstrated its causal effect in driving risk-preference. Specifically, risk-preferring rats could be instantaneously converted to risk-averse rats with precisely timed phasic stimulation of NAc D2R cells. These findings suggest that individual differences in risk-preference, as well as real-time risky decision-making, can be largely explained by the encoding in D2R-expressing NAc cells of prior unfavourable outcomes during decision-making.

    View details for DOI 10.1038/nature17400

    View details for PubMedID 27007845

  • Communication in Neural Circuits: Tools, Opportunities, and Challenges. Cell Lerner, T. N., Ye, L., Deisseroth, K. 2016; 164 (6): 1136-50

    Abstract

    Communication, the effective delivery of information, is fundamental to life across all scales and species. Nervous systems (by necessity) may be most specifically adapted among biological tissues for high rate and complexity of information transmitted, and thus, the properties of neural tissue and principles of its organization into circuits may illuminate capabilities and limitations of biological communication. Here, we consider recent developments in tools for studying neural circuits with particular attention to defining neuronal cell types by input and output information streams-i.e., by how they communicate. Complementing approaches that define cell types by virtue of genetic promoter/enhancer properties, this communication-based approach to defining cell types operationally by input/output (I/O) relationships links structure and function, resolves difficulties associated with single-genetic-feature definitions, leverages technology for observing and testing significance of precisely these I/O relationships in intact brains, and maps onto processes through which behavior may be adapted during development, experience, and evolution.

    View details for DOI 10.1016/j.cell.2016.02.027

    View details for PubMedID 26967281

  • In vivo imaging identifies temporal signature of D1 and D2 medium spiny neurons in cocaine reward PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Calipari, E. S., Bagot, R. C., Purushothaman, I., Davidson, T. J., Yorgason, J. T., Pena, C. J., Walker, D. M., Pirpinias, S. T., Guise, K. G., Ramakrishnan, C., Deisseroth, K., Nestler, E. J. 2016; 113 (10): 2726-2731

    Abstract

    The reinforcing and rewarding properties of cocaine are attributed to its ability to increase dopaminergic transmission in nucleus accumbens (NAc). This action reinforces drug taking and seeking and leads to potent and long-lasting associations between the rewarding effects of the drug and the cues associated with its availability. The inability to extinguish these associations is a key factor contributing to relapse. Dopamine produces these effects by controlling the activity of two subpopulations of NAc medium spiny neurons (MSNs) that are defined by their predominant expression of either dopamine D1 or D2 receptors. Previous work has demonstrated that optogenetically stimulating D1 MSNs promotes reward, whereas stimulating D2 MSNs produces aversion. However, we still lack a clear understanding of how the endogenous activity of these cell types is affected by cocaine and encodes information that drives drug-associated behaviors. Using fiber photometry calcium imaging we define D1 MSNs as the specific population of cells in NAc that encodes information about drug associations and elucidate the temporal profile with which D1 activity is increased to drive drug seeking in response to contextual cues. Chronic cocaine exposure dysregulates these D1 signals to both prevent extinction and facilitate reinstatement of drug seeking to drive relapse. Directly manipulating these D1 signals using designer receptors exclusively activated by designer drugs prevents contextual associations. Together, these data elucidate the responses of D1- and D2-type MSNs in NAc to acute cocaine and during the formation of context-reward associations and define how prior cocaine exposure selectively dysregulates D1 signaling to drive relapse.

    View details for DOI 10.1073/pnas.1521238113

    View details for PubMedID 26831103

  • Whole Brain Screening of Cellular and Molecular Changes After Stroke Aswendt, M., Hsueh, B., Ishizaka, S., Sun Guohua, Cheng, M., Deisseroth, K., Steinberg, G. K. LIPPINCOTT WILLIAMS & WILKINS. 2016
  • Structural foundations of optogenetics: Determinants of channelrhodopsin ion selectivity. Proceedings of the National Academy of Sciences of the United States of America Berndt, A., Lee, S. Y., Wietek, J., Ramakrishnan, C., Steinberg, E. E., Rashid, A. J., Kim, H., Park, S., Santoro, A., Frankland, P. W., Iyer, S. M., Pak, S., Ährlund-Richter, S., Delp, S. L., Malenka, R. C., Josselyn, S. A., Carlén, M., Hegemann, P., Deisseroth, K. 2016; 113 (4): 822-9

    Abstract

    The structure-guided design of chloride-conducting channelrhodopsins has illuminated mechanisms underlying ion selectivity of this remarkable family of light-activated ion channels. The first generation of chloride-conducting channelrhodopsins, guided in part by development of a structure-informed electrostatic model for pore selectivity, included both the introduction of amino acids with positively charged side chains into the ion conduction pathway and the removal of residues hypothesized to support negatively charged binding sites for cations. Engineered channels indeed became chloride selective, reversing near -65 mV and enabling a new kind of optogenetic inhibition; however, these first-generation chloride-conducting channels displayed small photocurrents and were not tested for optogenetic inhibition of behavior. Here we report the validation and further development of the channelrhodopsin pore model via crystal structure-guided engineering of next-generation light-activated chloride channels (iC++) and a bistable variant (SwiChR++) with net photocurrents increased more than 15-fold under physiological conditions, reversal potential further decreased by another ∼ 15 mV, inhibition of spiking faithfully tracking chloride gradients and intrinsic cell properties, strong expression in vivo, and the initial microbial opsin channel-inhibitor-based control of freely moving behavior. We further show that inhibition by light-gated chloride channels is mediated mainly by shunting effects, which exert optogenetic control much more efficiently than the hyperpolarization induced by light-activated chloride pumps. The design and functional features of these next-generation chloride-conducting channelrhodopsins provide both chronic and acute timescale tools for reversible optogenetic inhibition, confirm fundamental predictions of the ion selectivity model, and further elucidate electrostatic and steric structure-function relationships of the light-gated pore.

    View details for DOI 10.1073/pnas.1523341113

    View details for PubMedID 26699459

    View details for PubMedCentralID PMC4743797

  • Prefrontal Parvalbumin Neurons in Control of Attention CELL Kim, H., Ahrlund-Richter, S., Wang, X., Deisseroth, K., Carlen, M. 2016; 164 (1-2): 208-218

    Abstract

    While signatures of attention have been extensively studied in sensory systems, the neural sources and computations responsible for top-down control of attention are largely unknown. Using chronic recordings in mice, we found that fast-spiking parvalbumin (FS-PV) interneurons in medial prefrontal cortex (mPFC) uniformly show increased and sustained firing during goal-driven attentional processing, correlating to the level of attention. Elevated activity of FS-PV neurons on the timescale of seconds predicted successful execution of behavior. Successful allocation of attention was characterized by strong synchronization of FS-PV neurons, increased gamma oscillations, and phase locking of pyramidal firing. Phase-locked pyramidal neurons showed gamma-phase-dependent rate modulation during successful attentional processing. Optogenetic silencing of FS-PV neurons deteriorated attentional processing, while optogenetic synchronization of FS-PV neurons at gamma frequencies had pro-cognitive effects and improved goal-directed behavior. FS-PV neurons thus act as a functional unit coordinating the activity in the local mPFC circuit during goal-driven attentional processing.

    View details for DOI 10.1016/j.cell.2015.11.038

    View details for PubMedID 26771492

  • Prefrontal cortical regulation of brainwide circuit dynamics and reward-related behavior. Science Ferenczi, E. A., Zalocusky, K. A., Liston, C., Grosenick, L., Warden, M. R., Amatya, D., Katovich, K., Mehta, H., Patenaude, B., Ramakrishnan, C., Kalanithi, P., Etkin, A., Knutson, B., Glover, G. H., Deisseroth, K. 2016; 351 (6268)

    Abstract

    Motivation for reward drives adaptive behaviors, whereas impairment of reward perception and experience (anhedonia) can contribute to psychiatric diseases, including depression and schizophrenia. We sought to test the hypothesis that the medial prefrontal cortex (mPFC) controls interactions among specific subcortical regions that govern hedonic responses. By using optogenetic functional magnetic resonance imaging to locally manipulate but globally visualize neural activity in rats, we found that dopamine neuron stimulation drives striatal activity, whereas locally increased mPFC excitability reduces this striatal response and inhibits the behavioral drive for dopaminergic stimulation. This chronic mPFC overactivity also stably suppresses natural reward-motivated behaviors and induces specific new brainwide functional interactions, which predict the degree of anhedonia in individuals. These findings describe a mechanism by which mPFC modulates expression of reward-seeking behavior, by regulating the dynamical interactions between specific distant subcortical regions.

    View details for DOI 10.1126/science.aac9698

    View details for PubMedID 26722001

  • Form Meets Function in the Brain: Observing the Activity and Structure of Specific Neural Connections MICRO-, MESO- AND MACRO- CONNECTOMICS OF THE BRAIN Deisseroth, K., Kennedy, H., VanEssen, D. C., Christen, Y. 2016: 19-29
  • Highly Multiplexed Nanophotonic Probes With Independently Controllable Emitters for Optogenetic Brain Stimulation Segev, E., Moreaux, L. C., Reimer, J., Fowler, T. M., Chi, D., Sacher, W. D., Lo, M., Deisseroth, K., Tolias, A. S., Faraon, A., Roukes, M. L., IEEE IEEE. 2016
  • Deformably Registering and Annotating Whole CLARITY Brains to an Atlas via Masked LDDMM Kutten, K. S., Vogelstein, J. T., Charon, N., Ye, L., Deisseroth, K., Miller, M. I., Schelkens, P., Ebrahimi, T., Cristobal, G., Truchetet, F., Saarikko, P. SPIE-INT SOC OPTICAL ENGINEERING. 2016

    View details for DOI 10.1117/12.2227444

    View details for Web of Science ID 000385792600033

  • Molecular and Cellular Mechanisms for Trapping and Activating Emotional Memories. PloS one Rogerson, T., Jayaprakash, B., Cai, D. J., Sano, Y., Lee, Y., Zhou, Y., Bekal, P., Deisseroth, K., Silva, A. J. 2016; 11 (8)

    Abstract

    Recent findings suggest that memory allocation to specific neurons (i.e., neuronal allocation) in the amygdala is not random, but rather the transcription factor cAMP-response element binding protein (CREB) modulates this process, perhaps by regulating the transcription of channels that control neuronal excitability. Here, optogenetic studies in the mouse lateral amygdala (LA) were used to demonstrate that CREB and neuronal excitability regulate which neurons encode an emotional memory. To test the role of CREB in memory allocation, we overexpressed CREB in the lateral amygdala to recruit the encoding of an auditory-fear conditioning (AFC) memory to a subset of neurons. Then, post-training activation of these neurons with Channelrhodopsin-2 was sufficient to trigger recall of the memory for AFC, suggesting that CREB regulates memory allocation. To test the role of neuronal excitability in memory allocation, we used a step function opsin (SFO) to transiently increase neuronal excitability in a subset of LA neurons during AFC. Post-training activation of these neurons with Volvox Channelrhodopsin-1 was able to trigger recall of that memory. Importantly, our studies show that activation of the SFO did not affect AFC by either increasing anxiety or by strengthening the unconditioned stimulus. Our findings strongly support the hypothesis that CREB regulates memory allocation by modulating neuronal excitability.

    View details for DOI 10.1371/journal.pone.0161655

    View details for PubMedID 27579481

  • Optogenetic Stimulation of Neural Grafts Enhances Neurotransmission and Downregulates the Inflammatory Response in Experimental Stroke Model. Cell transplantation Daadi, M. M., Klausner, J. Q., Bajar, B., Goshen, I., Lee-Messer, C., Lee, S. Y., Winge, M. C., Ramakrishnan, C., Lo, M., Sun, G., Deisseroth, K., Steinberg, G. K. 2016; 25 (7): 1371-1380

    Abstract

    Compelling evidence suggests that transplantation of neural stem cells (NSCs) from multiple sources ameliorates motor deficits after stroke. However, it is currently unknown to what extent the electrophysiological activity of grafted NSC progeny participates in the improvement of motor deficits and whether excitatory phenotypes of the grafted cells are beneficial or deleterious to sensorimotor performances. To address this question, we used optogenetic tools to drive the excitatory outputs of the grafted NSCs and assess the impact on local circuitry and sensorimotor performance. We genetically engineered NSCs to express the Channelrhodopsin-2 (ChR2), a light-gated cation channel that evokes neuronal depolarization and initiation of action potentials with precise temporal control to light stimulation. To test the function of these cells in a stroke model, rats were subjected to an ischemic stroke and grafted with ChR2-NSCs. The grafted NSCs identified with a human-specific nuclear marker survived in the peri-infarct tissue and coexpressed the ChR2 transgene with the neuronal markers TuJ1 and NeuN. Gene expression analysis in stimulated versus vehicle-treated animals showed a differential upregulation of transcripts involved in neurotransmission, neuronal differentiation, regeneration, axonal guidance, and synaptic plasticity. Interestingly, genes involved in the inflammatory response were significantly downregulated. Behavioral analysis demonstrated that chronic optogenetic stimulation of the ChR2-NSCs enhanced forelimb use on the stroke-affected side and motor activity in an open field test. Together these data suggest that excitatory stimulation of grafted NSCs elicits beneficial effects in experimental stroke model through cell replacement and non-cell replacement, anti-inflammatory/neurotrophic effects.

    View details for DOI 10.3727/096368915X688533

    View details for PubMedID 26132738

  • Optogenetic and chemogenetic strategies for sustained inhibition of pain. Scientific reports Iyer, S. M., Vesuna, S., Ramakrishnan, C., Huynh, K., Young, S., Berndt, A., Lee, S. Y., Gorini, C. J., Deisseroth, K., Delp, S. L. 2016; 6: 30570-?

    Abstract

    Spatially targeted, genetically-specific strategies for sustained inhibition of nociceptors may help transform pain science and clinical management. Previous optogenetic strategies to inhibit pain have required constant illumination, and chemogenetic approaches in the periphery have not been shown to inhibit pain. Here, we show that the step-function inhibitory channelrhodopsin, SwiChR, can be used to persistently inhibit pain for long periods of time through infrequent transdermally delivered light pulses, reducing required light exposure by >98% and resolving a long-standing limitation in optogenetic inhibition. We demonstrate that the viral expression of the hM4D receptor in small-diameter primary afferent nociceptor enables chemogenetic inhibition of mechanical and thermal nociception thresholds. Finally, we develop optoPAIN, an optogenetic platform to non-invasively assess changes in pain sensitivity, and use this technique to examine pharmacological and chemogenetic inhibition of pain.

    View details for DOI 10.1038/srep30570

    View details for PubMedID 27484850

    View details for PubMedCentralID PMC4971509

  • Optogenetic stimulation of cholinergic brainstem neurons during focal limbic seizures: Effects on cortical physiology EPILEPSIA Furman, M., Zhan, Q., McCafferty, C., Lerner, B. A., Motelow, J. E., Meng, J., Ma, C., Buchanan, G. F., Witten, I. B., Deisseroth, K., Cardin, J. A., Blumenfeld, H. 2015; 56 (12): E198-E202

    Abstract

    Focal temporal lobe seizures often cause impaired cortical function and loss of consciousness. Recent work suggests that the mechanism for depressed cortical function during focal seizures may depend on decreased subcortical cholinergic arousal, which leads to a sleep-like state of cortical slow-wave activity. To test this hypothesis, we sought to directly activate subcortical cholinergic neurons during focal limbic seizures to determine the effects on cortical function. Here we used an optogenetic approach to selectively stimulate cholinergic brainstem neurons in the pedunculopontine tegmental nucleus during focal limbic seizures induced in a lightly anesthetized rat model. We found an increase in cortical gamma activity and a decrease in delta activity in response to cholinergic stimulation. These findings support the mechanistic role of reduced subcortical cholinergic arousal in causing cortical dysfunction during seizures. Through further work, electrical or optogenetic stimulation of subcortical arousal networks may ultimately lead to new treatments aimed at preventing cortical dysfunction during seizures.

    View details for DOI 10.1111/epi.13220

    View details for PubMedID 26530287

    View details for PubMedCentralID PMC4679683

  • Basomedial amygdala mediates top-down control of anxiety and fear NATURE Adhikari, A., Lerner, T. N., Finkelstein, J., Pak, S., Jennings, J. H., Davidson, T. J., Ferenczi, E., Gunaydin, L. A., Irzabekov, J. J., Ye, L., Kim, S., Lei, A., Deisseroth, K. 2015; 527 (7577): 179-?

    Abstract

    Anxiety-related conditions are among the most difficult neuropsychiatric diseases to treat pharmacologically, but respond to cognitive therapies. There has therefore been interest in identifying relevant top-down pathways from cognitive control regions in medial prefrontal cortex (mPFC). Identification of such pathways could contribute to our understanding of the cognitive regulation of affect, and provide pathways for intervention. Previous studies have suggested that dorsal and ventral mPFC subregions exert opposing effects on fear, as do subregions of other structures. However, precise causal targets for top-down connections among these diverse possibilities have not been established. Here we show that the basomedial amygdala (BMA) represents the major target of ventral mPFC in amygdala in mice. Moreover, BMA neurons differentiate safe and aversive environments, and BMA activation decreases fear-related freezing and high-anxiety states. Lastly, we show that the ventral mPFC-BMA projection implements top-down control of anxiety state and learned freezing, both at baseline and in stress-induced anxiety, defining a broadly relevant new top-down behavioural regulation pathway.

    View details for DOI 10.1038/nature15698

    View details for Web of Science ID 000364396700034

    View details for PubMedID 26536109

  • Daytime spikes in dopaminergic activity drive rapid mood-cycling in mice MOLECULAR PSYCHIATRY Sidor, M. M., Spencer, S. M., Dzirasa, K., PAREKH, P. K., Tye, K. M., WARDEN, M. R., Arey, R. N., Enwright, J. F., Jacobsen, J. P., Kumar, S., Remillard, E. M., Caron, M. G., Deisseroth, K., McClung, C. A. 2015; 20 (11): 1406-1419

    Abstract

    Disruptions in circadian rhythms and dopaminergic activity are involved in the pathophysiology of bipolar disorder, though their interaction remains unclear. Moreover, a lack of animal models that display spontaneous cycling between mood states has hindered our mechanistic understanding of mood switching. Here, we find that mice with a mutation in the circadian Clock gene (ClockΔ19) exhibit rapid mood-cycling, with a profound manic-like phenotype emerging during the day following a period of euthymia at night. Mood-cycling coincides with abnormal daytime spikes in ventral tegmental area (VTA) dopaminergic activity, tyrosine hydroxylase (TH) levels and dopamine synthesis. To determine the significance of daytime increases in VTA dopamine activity to manic behaviors, we developed a novel optogenetic stimulation paradigm that produces a sustained increase in dopamine neuronal activity and find that this induces a manic-like behavioral state. Time-dependent dampening of TH activity during the day reverses manic-related behaviors in ClockΔ19 mice. Finally, we show that CLOCK acts as a negative regulator of TH transcription, revealing a novel molecular mechanism underlying cyclic changes in mood-related behavior. Taken together, these studies have identified a mechanistic connection between circadian gene disruption and the precipitation of manic episodes in bipolar disorder.

    View details for DOI 10.1038/mp.2014.167

    View details for Web of Science ID 000363465300015

    View details for PubMedID 25560763

    View details for PubMedCentralID PMC4492925

  • Striatal Cholinergic Interneurons Control Motor Behavior and Basal Ganglia Function in Experimental Parkinsonism. Cell reports Maurice, N., Liberge, M., Jaouen, F., Ztaou, S., Hanini, M., Camon, J., Deisseroth, K., Amalric, M., Kerkerian-Le Goff, L., Beurrier, C. 2015; 13 (4): 657-666

    Abstract

    Despite evidence showing that anticholinergic drugs are of clinical relevance in Parkinson's disease (PD), the causal role of striatal cholinergic interneurons (CINs) in PD pathophysiology remains elusive. Here, we show that optogenetic inhibition of CINs alleviates motor deficits in PD mouse models, providing direct demonstration for their implication in parkinsonian motor dysfunctions. As neural correlates, CIN inhibition in parkinsonian mice differentially impacts the excitability of striatal D1 and D2 medium spiny neurons, normalizes pathological bursting activity in the main basal ganglia output structure, and increases the functional weight of the direct striatonigral pathway in cortical information processing. By contrast, CIN inhibition in non-lesioned mice does not affect locomotor activity, equally modulates medium spiny neuron excitability, and does not modify spontaneous or cortically driven activity in the basal ganglia output, suggesting that the role of these interneurons in motor function is highly dependent on dopamine tone.

    View details for DOI 10.1016/j.celrep.2015.09.034

    View details for PubMedID 26489458

  • Cortical and Subcortical Contributions to Short-Term Memory for Orienting Movements. Neuron Kopec, C. D., Erlich, J. C., Brunton, B. W., Deisseroth, K., Brody, C. D. 2015; 88 (2): 367-77

    Abstract

    Neural activity in frontal cortical areas has been causally linked to short-term memory (STM), but whether this activity is necessary for forming, maintaining, or reading out STM remains unclear. In rats performing a memory-guided orienting task, the frontal orienting fields in cortex (FOF) are considered critical for STM maintenance, and during each trial display a monotonically increasing neural encoding for STM. Here, we transiently inactivated either the FOF or the superior colliculus and found that the resulting impairments in memory-guided orienting performance followed a monotonically decreasing time course, surprisingly opposite to the neural encoding. A dynamical attractor model in which STM relies equally on cortical and subcortical regions reconciled the encoding and inactivation data. We confirmed key predictions of the model, including a time-dependent relationship between trial difficulty and perturbability, and substantial, supralinear, impairment following simultaneous inactivation of the FOF and superior colliculus during memory maintenance.

    View details for DOI 10.1016/j.neuron.2015.08.033

    View details for PubMedID 26439529

    View details for PubMedCentralID PMC5521275

  • A skin-inspired organic digital mechanoreceptor SCIENCE Tee, B. C., Chortos, A., Berndt, A., Nguyen, A. K., Tom, A., McGuire, A., Lin, Z. C., Tien, K., Bae, W., Wang, H., Mei, P., Chou, H., Cui, B., Deisseroth, K., Ng, T. N., Bao, Z. 2015; 350 (6258): 313-?

    Abstract

    Human skin relies on cutaneous receptors that output digital signals for tactile sensing in which the intensity of stimulation is converted to a series of voltage pulses. We present a power-efficient skin-inspired mechanoreceptor with a flexible organic transistor circuit that transduces pressure into digital frequency signals directly. The output frequency ranges between 0 and 200 hertz, with a sublinear response to increasing force stimuli that mimics slow-adapting skin mechanoreceptors. The output of the sensors was further used to stimulate optogenetically engineered mouse somatosensory neurons of mouse cortex in vitro, achieving stimulated pulses in accordance with pressure levels. This work represents a step toward the design and use of large-area organic electronic skins with neural-integrated touch feedback for replacement limbs.

    View details for DOI 10.1126/science.aaa9306

    View details for Web of Science ID 000362838700039

    View details for PubMedID 26472906

  • All-Optical Interrogation of Neural Circuits JOURNAL OF NEUROSCIENCE Emiliani, V., Cohen, A. E., Deisseroth, K., Haeusser, M. 2015; 35 (41): 13917-13926

    Abstract

    There have been two recent revolutionary advances in neuroscience: First, genetically encoded activity sensors have brought the goal of optical detection of single action potentials in vivo within reach. Second, optogenetic actuators now allow the activity of neurons to be controlled with millisecond precision. These revolutions have now been combined, together with advanced microscopies, to allow "all-optical" readout and manipulation of activity in neural circuits with single-spike and single-neuron precision. This is a transformational advance that will open new frontiers in neuroscience research. Harnessing the power of light in the all-optical approach requires coexpression of genetically encoded activity sensors and optogenetic probes in the same neurons, as well as the ability to simultaneously target and record the light from the selected neurons. It has recently become possible to combine sensors and optical strategies that are sufficiently sensitive and cross talk free to enable single-action-potential sensitivity and precision for both readout and manipulation in the intact brain. The combination of simultaneous readout and manipulation from the same genetically defined cells will enable a wide range of new experiments as well as inspire new technologies for interacting with the brain. The advances described in this review herald a future where the traditional tools used for generations by physiologists to study and interact with the brain-stimulation and recording electrodes-can largely be replaced by light. We outline potential future developments in this field and discuss how the all-optical strategy can be applied to solve fundamental problems in neuroscience.This review describes the nexus of dramatic recent developments in optogenetic probes, genetically encoded activity sensors, and novel microscopies, which together allow the activity of neural circuits to be recorded and manipulated entirely using light. The optical and protein engineering strategies that form the basis of this "all-optical" approach are now sufficiently advanced to enable single-neuron and single-action potential precision for simultaneous readout and manipulation from the same functionally defined neurons in the intact brain. These advances promise to illuminate many fundamental challenges in neuroscience, including transforming our search for the neural code and the links between neural circuit activity and behavior.

    View details for DOI 10.1523/JNEUROSCI.2916-15.2015

    View details for Web of Science ID 000366051800013

    View details for PubMedCentralID PMC4604230

  • All-Optical Interrogation of Neural Circuits. journal of neuroscience Emiliani, V., Cohen, A. E., Deisseroth, K., Häusser, M. 2015; 35 (41): 13917-13926

    Abstract

    There have been two recent revolutionary advances in neuroscience: First, genetically encoded activity sensors have brought the goal of optical detection of single action potentials in vivo within reach. Second, optogenetic actuators now allow the activity of neurons to be controlled with millisecond precision. These revolutions have now been combined, together with advanced microscopies, to allow "all-optical" readout and manipulation of activity in neural circuits with single-spike and single-neuron precision. This is a transformational advance that will open new frontiers in neuroscience research. Harnessing the power of light in the all-optical approach requires coexpression of genetically encoded activity sensors and optogenetic probes in the same neurons, as well as the ability to simultaneously target and record the light from the selected neurons. It has recently become possible to combine sensors and optical strategies that are sufficiently sensitive and cross talk free to enable single-action-potential sensitivity and precision for both readout and manipulation in the intact brain. The combination of simultaneous readout and manipulation from the same genetically defined cells will enable a wide range of new experiments as well as inspire new technologies for interacting with the brain. The advances described in this review herald a future where the traditional tools used for generations by physiologists to study and interact with the brain-stimulation and recording electrodes-can largely be replaced by light. We outline potential future developments in this field and discuss how the all-optical strategy can be applied to solve fundamental problems in neuroscience.This review describes the nexus of dramatic recent developments in optogenetic probes, genetically encoded activity sensors, and novel microscopies, which together allow the activity of neural circuits to be recorded and manipulated entirely using light. The optical and protein engineering strategies that form the basis of this "all-optical" approach are now sufficiently advanced to enable single-neuron and single-action potential precision for simultaneous readout and manipulation from the same functionally defined neurons in the intact brain. These advances promise to illuminate many fundamental challenges in neuroscience, including transforming our search for the neural code and the links between neural circuit activity and behavior.

    View details for DOI 10.1523/JNEUROSCI.2916-15.2015

    View details for PubMedID 26468193

    View details for PubMedCentralID PMC4604230

  • Wirelessly powered, fully internal optogenetics for brain, spinal and peripheral circuits in mice. Nature methods Montgomery, K. L., Yeh, A. J., Ho, J. S., Tsao, V., Mohan Iyer, S., Grosenick, L., Ferenczi, E. A., Tanabe, Y., Deisseroth, K., Delp, S. L., Poon, A. S. 2015; 12 (10): 969-974

    Abstract

    To enable sophisticated optogenetic manipulation of neural circuits throughout the nervous system with limited disruption of animal behavior, light-delivery systems beyond fiber optic tethering and large, head-mounted wireless receivers are desirable. We report the development of an easy-to-construct, implantable wireless optogenetic device. Our smallest version (20 mg, 10 mm(3)) is two orders of magnitude smaller than previously reported wireless optogenetic systems, allowing the entire device to be implanted subcutaneously. With a radio-frequency (RF) power source and controller, this implant produces sufficient light power for optogenetic stimulation with minimal tissue heating (<1 °C). We show how three adaptations of the implant allow for untethered optogenetic control throughout the nervous system (brain, spinal cord and peripheral nerve endings) of behaving mice. This technology opens the door for optogenetic experiments in which animals are able to behave naturally with optogenetic manipulation of both central and peripheral targets.

    View details for DOI 10.1038/nmeth.3536

    View details for PubMedID 26280330

  • Hybrid Periportal Hepatocytes Regenerate the Injured Liver without Giving Rise to Cancer CELL Font-Burgada, J., Shalapour, S., Ramaswamy, S., Hsueh, B., Rossell, D., Umemura, A., Taniguchi, K., Nakagawa, H., Valasek, M. A., Ye, L., Kopp, J. L., Sander, M., Carter, H., Deisseroth, K., Verma, I. M., Karin, M. 2015; 162 (4): 766-779

    Abstract

    Compensatory proliferation triggered by hepatocyte loss is required for liver regeneration and maintenance but also promotes development of hepatocellular carcinoma (HCC). Despite extensive investigation, the cells responsible for hepatocyte restoration or HCC development remain poorly characterized. We used genetic lineage tracing to identify cells responsible for hepatocyte replenishment following chronic liver injury and queried their roles in three distinct HCC models. We found that a pre-existing population of periportal hepatocytes, located in the portal triads of healthy livers and expressing low amounts of Sox9 and other bile-duct-enriched genes, undergo extensive proliferation and replenish liver mass after chronic hepatocyte-depleting injuries. Despite their high regenerative potential, these so-called hybrid hepatocytes do not give rise to HCC in chronically injured livers and thus represent a unique way to restore tissue function and avoid tumorigenesis. This specialized set of pre-existing differentiated cells may be highly suitable for cell-based therapy of chronic hepatocyte-depleting disorders.

    View details for DOI 10.1016/j.cell.2015.07.026

    View details for Web of Science ID 000359741400011

    View details for PubMedID 26276631

    View details for PubMedCentralID PMC4545590

  • Self-Tracking Energy Transfer for Neural Stimulation in Untethered Mice PHYSICAL REVIEW APPLIED Ho, J. S., Tanabe, Y., Iyer, S. M., Christensen, A. J., Grosenick, L., Deisseroth, K., Delp, S. L., Poon, A. S. 2015; 4 (2)
  • Intact-Brain Analyses Reveal Distinct Information Carried by SNc Dopamine Subcircuits. Cell Lerner, T. N., Shilyansky, C., Davidson, T. J., Evans, K. E., Beier, K. T., Zalocusky, K. A., Crow, A. K., Malenka, R. C., Luo, L., Tomer, R., Deisseroth, K. 2015; 162 (3): 635-647

    Abstract

    Recent progress in understanding the diversity of midbrain dopamine neurons has highlighted the importance--and the challenges--of defining mammalian neuronal cell types. Although neurons may be best categorized using inclusive criteria spanning biophysical properties, wiring of inputs, wiring of outputs, and activity during behavior, linking all of these measurements to cell types within the intact brains of living mammals has been difficult. Here, using an array of intact-brain circuit interrogation tools, including CLARITY, COLM, optogenetics, viral tracing, and fiber photometry, we explore the diversity of dopamine neurons within the substantia nigra pars compacta (SNc). We identify two parallel nigrostriatal dopamine neuron subpopulations differing in biophysical properties, input wiring, output wiring to dorsomedial striatum (DMS) versus dorsolateral striatum (DLS), and natural activity patterns during free behavior. Our results reveal independently operating nigrostriatal information streams, with implications for understanding the logic of dopaminergic feedback circuits and the diversity of mammalian neuronal cell types.

    View details for DOI 10.1016/j.cell.2015.07.014

    View details for PubMedID 26232229

  • Ca(V)3.2 calcium channels control NMDA receptor-mediated transmission: a new mechanism for absence epilepsy GENES & DEVELOPMENT Wang, G., Bochorishvili, G., Chen, Y., Salvati, K. A., Zhang, P., Dubel, S. J., Perez-Reyes, E., Snutch, T. P., Stornetta, R. L., Deisseroth, K., Erisir, A., Todorovic, S. M., Luo, J., Kapur, J., Beenhakker, M. P., Zhu, J. J. 2015; 29 (14): 1535-1551

    Abstract

    CaV3.2 T-type calcium channels, encoded by CACNA1H, are expressed throughout the brain, yet their general function remains unclear. We discovered that CaV3.2 channels control NMDA-sensitive glutamatergic receptor (NMDA-R)-mediated transmission and subsequent NMDA-R-dependent plasticity of AMPA-R-mediated transmission at rat central synapses. Interestingly, functional CaV3.2 channels primarily incorporate into synapses, replace existing CaV3.2 channels, and can induce local calcium influx to control NMDA transmission strength in an activity-dependent manner. Moreover, human childhood absence epilepsy (CAE)-linked hCaV3.2(C456S) mutant channels have a higher channel open probability, induce more calcium influx, and enhance glutamatergic transmission. Remarkably, cortical expression of hCaV3.2(C456S) channels in rats induces 2- to 4-Hz spike and wave discharges and absence-like epilepsy characteristic of CAE patients, which can be suppressed by AMPA-R and NMDA-R antagonists but not T-type calcium channel antagonists. These results reveal an unexpected role of CaV3.2 channels in regulating NMDA-R-mediated transmission and a novel epileptogenic mechanism for human CAE.

    View details for DOI 10.1101/gad.260869.115

    View details for Web of Science ID 000358596300007

    View details for PubMedCentralID PMC4526737

  • CaV3.2 calcium channels control NMDA receptor-mediated transmission: a new mechanism for absence epilepsy. Genes & development Wang, G., Bochorishvili, G., Chen, Y., Salvati, K. A., Zhang, P., Dubel, S. J., Perez-Reyes, E., Snutch, T. P., Stornetta, R. L., Deisseroth, K., Erisir, A., Todorovic, S. M., Luo, J. H., Kapur, J., Beenhakker, M. P., Zhu, J. J. 2015; 29 (14): 1535-51

    Abstract

    CaV3.2 T-type calcium channels, encoded by CACNA1H, are expressed throughout the brain, yet their general function remains unclear. We discovered that CaV3.2 channels control NMDA-sensitive glutamatergic receptor (NMDA-R)-mediated transmission and subsequent NMDA-R-dependent plasticity of AMPA-R-mediated transmission at rat central synapses. Interestingly, functional CaV3.2 channels primarily incorporate into synapses, replace existing CaV3.2 channels, and can induce local calcium influx to control NMDA transmission strength in an activity-dependent manner. Moreover, human childhood absence epilepsy (CAE)-linked hCaV3.2(C456S) mutant channels have a higher channel open probability, induce more calcium influx, and enhance glutamatergic transmission. Remarkably, cortical expression of hCaV3.2(C456S) channels in rats induces 2- to 4-Hz spike and wave discharges and absence-like epilepsy characteristic of CAE patients, which can be suppressed by AMPA-R and NMDA-R antagonists but not T-type calcium channel antagonists. These results reveal an unexpected role of CaV3.2 channels in regulating NMDA-R-mediated transmission and a novel epileptogenic mechanism for human CAE.

    View details for DOI 10.1101/gad.260869.115

    View details for PubMedID 26220996

    View details for PubMedCentralID PMC4526737

  • Excitatory transmission at thalamo-striatal synapses mediates susceptibility to social stress. Nature neuroscience Christoffel, D. J., Golden, S. A., Walsh, J. J., Guise, K. G., Heshmati, M., Friedman, A. K., Dey, A., Smith, M., Rebusi, N., Pfau, M., Ables, J. L., Aleyasin, H., Khibnik, L. A., Hodes, G. E., Ben-Dor, G. A., Deisseroth, K., Shapiro, M. L., Malenka, R. C., Ibanez-Tallon, I., Han, M., Russo, S. J. 2015; 18 (7): 962-964

    Abstract

    Postsynaptic remodeling of glutamatergic synapses on ventral striatum (vSTR) medium spiny neurons (MSNs) is critical for shaping stress responses. However, it is unclear which presynaptic inputs are involved. Susceptible mice exhibited increased synaptic strength at intralaminar thalamus (ILT), but not prefrontal cortex (PFC), inputs to vSTR MSNs following chronic social stress. Modulation of ILT-vSTR versus PFC-vSTR neuronal activity differentially regulated dendritic spine plasticity and social avoidance.

    View details for DOI 10.1038/nn.4034

    View details for PubMedID 26030846

    View details for PubMedCentralID PMC4482771

  • Excitatory transmission at thalamo-striatal synapses mediates susceptibility to social stress NATURE NEUROSCIENCE Christoffel, D. J., Golden, S. A., Walsh, J. J., Guise, K. G., Heshmati, M., Friedman, A. K., Dey, A., Smith, M., Rebusi, N., Pfau, M., Ables, J. L., Aleyasin, H., Khibnik, L. A., Hodes, G. E., Ben-Dor, G. A., Deisseroth, K., Shapiro, M. L., Malenka, R. C., Ibanez-Tallon, I., Han, M., Russo, S. J. 2015; 18 (7): 962-?

    Abstract

    Postsynaptic remodeling of glutamatergic synapses on ventral striatum (vSTR) medium spiny neurons (MSNs) is critical for shaping stress responses. However, it is unclear which presynaptic inputs are involved. Susceptible mice exhibited increased synaptic strength at intralaminar thalamus (ILT), but not prefrontal cortex (PFC), inputs to vSTR MSNs following chronic social stress. Modulation of ILT-vSTR versus PFC-vSTR neuronal activity differentially regulated dendritic spine plasticity and social avoidance.

    View details for DOI 10.1038/nn.4034

    View details for Web of Science ID 000356866200011

    View details for PubMedID 26030846

    View details for PubMedCentralID PMC4482771

  • Molecular Dynamics of Channelrhodopsin at the Early Stages of Channel Opening PLOS ONE Takemoto, M., Kato, H. E., Koyama, M., Ito, J., Kamiya, M., Hayashi, S., Maturana, A. D., Deisseroth, K., Ishitani, R., Nureki, O. 2015; 10 (6)

    Abstract

    Channelrhodopsin (ChR) is a light-gated cation channel that responds to blue light. Since ChR can be readily expressed in specific neurons to precisely control their activities by light, it has become a powerful tool in neuroscience. Although the recently solved crystal structure of a chimeric ChR, C1C2, provided the structural basis for ChR, our understanding of the molecular mechanism of ChR still remains limited. Here we performed electrophysiological analyses and all-atom molecular dynamics (MD) simulations, to investigate the importance of the intracellular and central constrictions of the ion conducting pore observed in the crystal structure of C1C2. Our electrophysiological analysis revealed that two glutamate residues, Glu122 and Glu129, in the intracellular and central constrictions, respectively, should be deprotonated in the photocycle. The simulation results suggested that the deprotonation of Glu129 in the central constriction leads to ion leakage in the ground state, and implied that the protonation of Glu129 is important for preventing ion leakage in the ground state. Moreover, we modeled the 13-cis retinal bound; i.e., activated C1C2, and performed MD simulations to investigate the conformational changes in the early stage of the photocycle. Our simulations suggested that retinal photoisomerization induces the conformational change toward channel opening, including the movements of TM6, TM7 and TM2. These insights into the dynamics of the ground states and the early photocycle stages enhance our understanding of the channel function of ChR.

    View details for DOI 10.1371/journal.pone.0131094

    View details for Web of Science ID 000358147500127

    View details for PubMedCentralID PMC4482709

  • Basolateral amygdala bidirectionally modulates stress-induced hippocampal learning and memory deficits through a p25/Cdk5-dependent pathway PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Rei, D., Mason, X., Seo, J., Graeff, J., Rudenko, A., Wang, J., Rueda, R., Siegert, S., Cho, S., Canter, R. G., Mungenast, A. E., Deisseroth, K., Tsai, L. 2015; 112 (23): 7291-7296

    Abstract

    Repeated stress has been suggested to underlie learning and memory deficits via the basolateral amygdala (BLA) and the hippocampus; however, the functional contribution of BLA inputs to the hippocampus and their molecular repercussions are not well understood. Here we show that repeated stress is accompanied by generation of the Cdk5 (cyclin-dependent kinase 5)-activator p25, up-regulation and phosphorylation of glucocorticoid receptors, increased HDAC2 expression, and reduced expression of memory-related genes in the hippocampus. A combination of optogenetic and pharmacosynthetic approaches shows that BLA activation is both necessary and sufficient for stress-associated molecular changes and memory impairments. Furthermore, we show that this effect relies on direct glutamatergic projections from the BLA to the dorsal hippocampus. Finally, we show that p25 generation is necessary for the stress-induced memory dysfunction. Taken together, our data provide a neural circuit model for stress-induced hippocampal memory deficits through BLA activity-dependent p25 generation.

    View details for DOI 10.1073/pnas.1415845112

    View details for Web of Science ID 000355823200056

    View details for PubMedCentralID PMC4466741

  • Basolateral amygdala bidirectionally modulates stress-induced hippocampal learning and memory deficits through a p25/Cdk5-dependent pathway. Proceedings of the National Academy of Sciences of the United States of America Rei, D., Mason, X., Seo, J., Gräff, J., Rudenko, A., Wang, J., Rueda, R., Siegert, S., Cho, S., Canter, R. G., Mungenast, A. E., Deisseroth, K., Tsai, L. 2015; 112 (23): 7291-7296

    Abstract

    Repeated stress has been suggested to underlie learning and memory deficits via the basolateral amygdala (BLA) and the hippocampus; however, the functional contribution of BLA inputs to the hippocampus and their molecular repercussions are not well understood. Here we show that repeated stress is accompanied by generation of the Cdk5 (cyclin-dependent kinase 5)-activator p25, up-regulation and phosphorylation of glucocorticoid receptors, increased HDAC2 expression, and reduced expression of memory-related genes in the hippocampus. A combination of optogenetic and pharmacosynthetic approaches shows that BLA activation is both necessary and sufficient for stress-associated molecular changes and memory impairments. Furthermore, we show that this effect relies on direct glutamatergic projections from the BLA to the dorsal hippocampus. Finally, we show that p25 generation is necessary for the stress-induced memory dysfunction. Taken together, our data provide a neural circuit model for stress-induced hippocampal memory deficits through BLA activity-dependent p25 generation.

    View details for DOI 10.1073/pnas.1415845112

    View details for PubMedID 25995364

    View details for PubMedCentralID PMC4466741

  • The contribution of raised intraneuronal chloride to epileptic network activity. The Journal of neuroscience : the official journal of the Society for Neuroscience Alfonsa, H., Merricks, E. M., Codadu, N. K., Cunningham, M. O., Deisseroth, K., Racca, C., Trevelyan, A. J. 2015; 35 (20): 7715-26

    Abstract

    Altered inhibitory function is an important facet of epileptic pathology. A key concept is that GABAergic activity can become excitatory if intraneuronal chloride rises. However, it has proved difficult to separate the role of raised chloride from other contributory factors in complex network phenomena, such as epileptic pathology. Therefore, we asked what patterns of activity are associated with chloride dysregulation by making novel use of Halorhodopsin to load clusters of mouse pyramidal cells artificially with Cl(-). Brief (1-10 s) activation of Halorhodopsin caused substantial positive shifts in the GABAergic reversal potential that were proportional to the charge transfer during the illumination and in adult neocortical pyramidal neurons decayed with a time constant of τ = 8.0 ± 2.8s. At the network level, these positive shifts in EGABA produced a transient rise in network excitability, with many distinctive features of epileptic foci, including high-frequency oscillations with evidence of out-of-phase firing (Ibarz et al., 2010). We show how such firing patterns can arise from quite small shifts in the mean intracellular Cl(-) level, within heterogeneous neuronal populations. Notably, however, chloride loading by itself did not trigger full ictal events, even with additional electrical stimulation to the underlying white matter. In contrast, when performed in combination with low, subepileptic levels of 4-aminopyridine, Halorhodopsin activation rapidly induced full ictal activity. These results suggest that chloride loading has at most an adjunctive role in ictogenesis. Our simulations also show how chloride loading can affect the jitter of action potential timing associated with imminent recruitment to an ictal event (Netoff and Schiff, 2002).

    View details for DOI 10.1523/JNEUROSCI.4105-14.2015

    View details for PubMedID 25995461

    View details for PubMedCentralID PMC4438123

  • Mesolimbic Dopamine Dynamically Tracks, and Is Causally Linked to, Discrete Aspects of Value-Based Decision Making BIOLOGICAL PSYCHIATRY Saddoris, M. P., Sugam, J. A., Stuber, G. D., Witten, I. B., Deisseroth, K., Carelli, R. M. 2015; 77 (10): 903-911

    Abstract

    To make appropriate choices, organisms must weigh the costs and benefits of potential valuable outcomes, a process known to involve the nucleus accumbens (NAc) and its dopaminergic input. However, it is currently unknown if dopamine dynamically tracks alterations in expected reward value online as behavioral preferences change and if so, if it is causally linked to specific components of value such as reward magnitude and/or delay to reinforcement.Electrochemical methods were used to measure subsecond NAc dopamine release during a delay discounting task where magnitude was fixed but delay varied across blocks (n = 7 rats). Next, to assess whether this dopamine signaling was causally related to specific components of choice behavior, we employed selective optogenetic stimulation of dopamine terminals in the NAc using a modified delay discounting task in which both delay and magnitude varied independently (n = 23 rats).Cues predictive of available choices evoked dopamine release that scaled with the rat's preferred choices and dynamically shifted as delay to reinforcement for the large reward increased. In the second experiment, dopamine signaling was causally related to features of decision making, as optogenetically enhanced dopamine release within the NAc during predictive cue presentation was sufficient to alter subsequent value-related choices. Importantly, this dopamine-mediated shift in choice was limited to delay-based, but not magnitude-based, decisions.These findings indicate that NAc dopamine dynamically tracks delay discounting and establishes a causal role for this signaling in a subset of value-based associative strategies.

    View details for DOI 10.1016/j.biopsych.2014.10.024

    View details for Web of Science ID 000353559600010

    View details for PubMedID 25541492

    View details for PubMedCentralID PMC4416981

  • Chronic Optogenetic Activation Augments A beta Pathology in a Mouse Model of Alzheimer Disease CELL REPORTS Yamamoto, K., Tanei, Z., Hashimoto, T., Wakabayashi, T., Okuno, H., Naka, Y., Yizhar, O., Fenno, L. E., Fukayama, M., Bito, H., Cirrito, J. R., Holtzman, D. M., Deisseroth, K., Iwatsubo, T. 2015; 11 (6): 859-865

    Abstract

    In vivo experimental evidence indicates that acute neuronal activation increases Aβ release from presynaptic terminals, whereas long-term effects of chronic synaptic activation on Aβ pathology remain unclear. To address this issue, we adopted optogenetics and transduced stabilized step-function opsin, a channelrhodopsin engineered to elicit a long-lasting neuronal hyperexcitability, into the hippocampal perforant pathway of APP transgenic mice. In vivo microdialysis revealed a ∼24% increase in the hippocampal interstitial fluid Aβ42 levels immediately after acute light activation. Five months of chronic optogenetic stimulation increased Aβ burden specifically in the projection area of the perforant pathway (i.e., outer molecular layer of the dentate gyrus) of the stimulated side by ∼2.5-fold compared with that in the contralateral side. Epileptic seizures were observed during the course of chronic stimulation, which might have partly contributed to the Aβ pathology. These findings implicate functional abnormalities of specific neuronal circuitry in Aβ pathology and Alzheimer disease.

    View details for DOI 10.1016/j.celrep.2015.04.017

    View details for Web of Science ID 000354406900002

    View details for PubMedID 25937280

  • Ventral hippocampal afferents to the nucleus accumbens regulate susceptibility to depression NATURE COMMUNICATIONS Bagot, R. C., Parise, E. M., Pena, C. J., Zhang, H., Maze, I., Chaudhury, D., Persaud, B., Cachope, R., Bolanos-Guzman, C. A., Cheer, J., Deisseroth, K., Han, M., Nestler, E. J. 2015; 6

    Abstract

    Enhanced glutamatergic transmission in the nucleus accumbens (NAc), a region critical for reward and motivation, has been implicated in the pathophysiology of depression; however, the afferent source of this increased glutamate tone is not known. The NAc receives glutamatergic inputs from the medial prefrontal cortex (mPFC), ventral hippocampus (vHIP) and basolateral amygdala (AMY). Here, we demonstrate that glutamatergic vHIP afferents to NAc regulate susceptibility to chronic social defeat stress (CSDS). We observe reduced activity in vHIP in mice resilient to CSDS. Furthermore, attenuation of vHIP-NAc transmission by optogenetic induction of long-term depression is pro-resilient, whereas acute enhancement of this input is pro-susceptible. This effect is specific to vHIP afferents to the NAc, as optogenetic stimulation of either mPFC or AMY afferents to the NAc is pro-resilient. These data indicate that vHIP afferents to NAc uniquely regulate susceptibility to CSDS, highlighting an important, novel circuit-specific mechanism in depression.

    View details for DOI 10.1038/ncomms8062

    View details for Web of Science ID 000355530900013

    View details for PubMedID 25952660

    View details for PubMedCentralID PMC4430111

  • Optical Tools for Probing Intact Biological Systems Deisseroth, K. ELSEVIER SCIENCE INC. 2015: 1S-2S
  • Optimization of CLARITY for Clearing Whole-Brain and Other Intact Organs(1,2,3). eNeuro Epp, J. R., Niibori, Y., Liz Hsiang, H., Mercaldo, V., Deisseroth, K., Josselyn, S. A., Frankland, P. W. 2015; 2 (3)

    Abstract

    The development, refinement, and use of techniques that allow high-throughput imaging of whole brains with cellular resolution will help us understand the complex functions of the brain. Such techniques are crucial for the analysis of complete neuronal morphology-anatomical and functional-connectivity, and repeated molecular phenotyping. CLARITY is a recently introduced technique that produces structurally intact, yet optically transparent tissue, which may be labeled and imaged without sectioning. However, the utility of this technique depends on several procedural variables during the process in which the light-scattering lipids in a tissue are replaced by a transparent hydrogel matrix. Here, we systematically varied a number of factors (including temperature, hydrogel composition, and polymerization conditions) to provide an optimized, highly replicable CLARITY procedure for clearing mouse brains. We found that for these preparations optimal tissue clearing requires electrophoresis (and cannot be achieved with passive clearing alone) for 5 d with a combination of 37 and 55°C temperature. Although this protocol is optimized for brains, we also show that it can be used to clear and analyze a variety of organs. Brain or other tissue prepared using this protocol is suitable for high-throughput imaging with confocal or single-plane illumination microscopy.

    View details for DOI 10.1523/ENEURO.0022-15.2015

    View details for PubMedID 26464982

    View details for PubMedCentralID PMC4586927

  • Activation of Corticostriatal Circuitry Relieves Chronic Neuropathic Pain JOURNAL OF NEUROSCIENCE Lee, M., Manders, T. R., Eberle, S. E., Su, C., D'amour, J., Yang, R., Lin, H. Y., Deisseroth, K., Froemke, R. C., Wang, J. 2015; 35 (13): 5247-5259

    Abstract

    Neural circuits that determine the perception and modulation of pain remain poorly understood. The prefrontal cortex (PFC) provides top-down control of sensory and affective processes. While animal and human imaging studies have shown that the PFC is involved in pain regulation, its exact role in pain states remains incompletely understood. A key output target for the PFC is the nucleus accumbens (NAc), an important component of the reward circuitry. Interestingly, recent human imaging studies suggest that the projection from the PFC to the NAc is altered in chronic pain. The function of this corticostriatal projection in pain states, however, is not known. Here we show that optogenetic activation of the PFC produces strong antinociceptive effects in a rat model (spared nerve injury model) of persistent neuropathic pain. PFC activation also reduces the affective symptoms of pain. Furthermore, we show that this pain-relieving function of the PFC is likely mediated by projections to the NAc. Thus, our results support a novel role for corticostriatal circuitry in pain regulation.

    View details for DOI 10.1523/JNEUROSCI.3494-14.2015

    View details for PubMedID 25834050

  • Cortically projecting basal forebrain parvalbumin neurons regulate cortical gamma band oscillations PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Kim, T., Thankachan, S., McKenna, J. T., McNally, J. M., Yang, C., Choi, J. H., Chen, L., Kocsis, B., Deisseroth, K., Strecker, R. E., Basheer, R., Brown, R. E., McCarley, R. W. 2015; 112 (11): 3535-3540

    Abstract

    Cortical gamma band oscillations (GBO, 30-80 Hz, typically ∼40 Hz) are involved in higher cognitive functions such as feature binding, attention, and working memory. GBO abnormalities are a feature of several neuropsychiatric disorders associated with dysfunction of cortical fast-spiking interneurons containing the calcium-binding protein parvalbumin (PV). GBO vary according to the state of arousal, are modulated by attention, and are correlated with conscious awareness. However, the subcortical cell types underlying the state-dependent control of GBO are not well understood. Here we tested the role of one cell type in the wakefulness-promoting basal forebrain (BF) region, cortically projecting GABAergic neurons containing PV, whose virally transduced fibers we found apposed cortical PV interneurons involved in generating GBO. Optogenetic stimulation of BF PV neurons in mice preferentially increased cortical GBO power by entraining a cortical oscillator with a resonant frequency of ∼40 Hz, as revealed by analysis of both rhythmic and nonrhythmic BF PV stimulation. Selective saporin lesions of BF cholinergic neurons did not alter the enhancement of cortical GBO power induced by BF PV stimulation. Importantly, bilateral optogenetic inhibition of BF PV neurons decreased the power of the 40-Hz auditory steady-state response, a read-out of the ability of the cortex to generate GBO used in clinical studies. Our results are surprising and novel in indicating that this presumptively inhibitory BF PV input controls cortical GBO, likely by synchronizing the activity of cortical PV interneurons. BF PV neurons may represent a previously unidentified therapeutic target to treat disorders involving abnormal GBO, such as schizophrenia.

    View details for DOI 10.1073/pnas.1413625112

    View details for Web of Science ID 000351060000095

    View details for PubMedID 25733878

    View details for PubMedCentralID PMC4371918

  • Illuminating circuitry relevant to psychiatric disorders with optogenetics CURRENT OPINION IN NEUROBIOLOGY Steinberg, E. E., Christoffel, D. J., Deisseroth, K., Malenka, R. C. 2015; 30: 9-16

    Abstract

    The brain's remarkable capacity to generate cognition and behavior is mediated by an extraordinarily complex set of neural interactions that remain largely mysterious. This complexity poses a significant challenge in developing therapeutic interventions to ameliorate psychiatric disease. Accordingly, few new classes of drugs have been made available for patients with mental illness since the 1950s. Optogenetics offers the ability to selectively manipulate individual neural circuit elements that underlie disease-relevant behaviors and is currently accelerating the pace of preclinical research into neurobiological mechanisms of disease. In this review, we highlight recent findings from studies that employ optogenetic approaches to gain insight into normal and aberrant brain function relevant to mental illness. Emerging data from these efforts offers an exquisitely detailed picture of disease-relevant neural circuits in action, and hints at the potential of optogenetics to open up entirely new avenues in the treatment of psychiatric disorders.

    View details for DOI 10.1016/j.conb.2014.08.004

    View details for Web of Science ID 000348337600002

    View details for PubMedID 25215625

  • Optogenetics enables functional analysis of human embryonic stem cell-derived grafts in a Parkinson's disease model. Nature biotechnology Steinbeck, J. A., Choi, S. J., Mrejeru, A., Ganat, Y., Deisseroth, K., Sulzer, D., Mosharov, E. V., Studer, L. 2015; 33 (2): 204-9

    Abstract

    Recent studies have shown evidence of behavioral recovery after transplantation of human pluripotent stem cell (PSC)-derived neural cells in animal models of neurological disease. However, little is known about the mechanisms underlying graft function. Here we use optogenetics to modulate in real time electrophysiological and neurochemical properties of mesencephalic dopaminergic (mesDA) neurons derived from human embryonic stem cells (hESCs). In mice that had recovered from lesion-induced Parkinsonian motor deficits, light-induced selective silencing of graft activity rapidly and reversibly re-introduced the motor deficits. The re-introduction of motor deficits was prevented by the dopamine agonist apomorphine. These results suggest that functionality depends on graft neuronal activity and dopamine release. Combining optogenetics, slice electrophysiology and pharmacological approaches, we further show that mesDA-rich grafts modulate host glutamatergic synaptic transmission onto striatal medium spiny neurons in a manner reminiscent of endogenous mesDA neurons. Thus, application of optogenetics in cell therapy can link transplantation, animal behavior and postmortem analysis to enable the identification of mechanisms that drive recovery.

    View details for DOI 10.1038/nbt.3124

    View details for PubMedID 25580598

    View details for PubMedCentralID PMC5117952

  • Muscarinic excitation of parvalbumin-positive interneurons contributes to the severity of pilocarpine-induced seizures EPILEPSIA Yi, F., DeCan, E., Stoll, K., Marceau, E., Deisseroth, K., Lawrence, J. J. 2015; 56 (2): 297-309

    Abstract

    A common rodent model in epilepsy research employs the muscarinic acetylcholine receptor (mAChR) agonist pilocarpine, yet the mechanisms underlying the induction of pilocarpine-induced seizures (PISs) remain unclear. Global M1 mAChR (M1 R) knockout mice are resistant to PISs, implying that M1 R activation disrupts excitation/inhibition balance. Parvalbumin-positive (PV) inhibitory neurons express M1 Rs, participate in cholinergically induced oscillations, and can enter a state of depolarization block (DB) during epileptiform activity. Here, we test the hypothesis that pilocarpine activation of M1 Rs expressed on PV cells contributes to PISs.CA1 PV cells in PV-CRE mice were visualized with a floxed YFP or hM3Dq-mCherry adeno-associated virus, or by crossing PV-CRE mice with the RosaYFP reporter line. To eliminate M1 Rs from PV cells, we generated PV-M1 knockout (KO) mice by crossing PV-CRE and floxed M1 mice. Action potential (AP) frequency was monitored during application of pilocarpine (200 μm). In behavioral experiments, locomotion and seizure symptoms were recorded in wild-type (WT) or PV-M1 KO mice during PISs.Pilocarpine significantly increased AP frequency in CA1 PV cells into the gamma range. In the continued presence of pilocarpine, a subset (5/7) of PV cells progressed to DB, which was mimicked by hM3Dq activation of Gq-receptor signaling. Pilocarpine-induced depolarization, AP firing at gamma frequency, and progression to DB were prevented in CA1 PV cells of PV-M1 KO mice. Finally, compared to WT mice, PV-M1 KO mice were associated with reduced severity of PISs.Pilocarpine can directly depolarize PV+ cells via M1 R activation, but a subset of these cells progress to DB. Our electrophysiologic and behavioral results suggest that this mechanism is active during PISs, contributing to a collapse of PV-mediated γ-aminobutyric acid (GABA)ergic inhibition, dysregulation of excitation/inhibition balance, and increased susceptibility to PISs.

    View details for DOI 10.1111/epi.12883

    View details for Web of Science ID 000350146300028

    View details for PubMedID 25495999

    View details for PubMedCentralID PMC4339470

  • Visualizing hypothalamic network dynamics for appetitive and consummatory behaviors. Cell Jennings, J. H., Ung, R. L., Resendez, S. L., Stamatakis, A. M., Taylor, J. G., Huang, J., Veleta, K., Kantak, P. A., Aita, M., Shilling-Scrivo, K., Ramakrishnan, C., Deisseroth, K., Otte, S., Stuber, G. D. 2015; 160 (3): 516-527

    Abstract

    Optimally orchestrating complex behavioral states, such as the pursuit and consumption of food, is critical for an organism's survival. The lateral hypothalamus (LH) is a neuroanatomical region essential for appetitive and consummatory behaviors, but whether individual neurons within the LH differentially contribute to these interconnected processes is unknown. Here, we show that selective optogenetic stimulation of a molecularly defined subset of LH GABAergic (Vgat-expressing) neurons enhances both appetitive and consummatory behaviors, whereas genetic ablation of these neurons reduced these phenotypes. Furthermore, this targeted LH subpopulation is distinct from cells containing the feeding-related neuropeptides, melanin-concentrating hormone (MCH), and orexin (Orx). Employing in vivo calcium imaging in freely behaving mice to record activity dynamics from hundreds of cells, we identified individual LH GABAergic neurons that preferentially encode aspects of either appetitive or consummatory behaviors, but rarely both. These tightly regulated, yet highly intertwined, behavioral processes are thus dissociable at the cellular level.

    View details for DOI 10.1016/j.cell.2014.12.026

    View details for PubMedID 25635459

    View details for PubMedCentralID PMC4312416

  • Mapping Anatomy to Behavior in Thy1:18 ChR2-YFP Transgenic Mice Using Optogenetics. Cold Spring Harbor protocols Fenno, L. E., Gunaydin, L. A., Deisseroth, K. 2015; 2015 (6): pdb prot075598-?

    Abstract

    Linking the activity of defined neural populations with behavior is a key goal of neuroscience. In the context of controlling behavior, electrical stimulation affords researchers precision in the temporal domain with gross regional specificity, whereas pharmacology allows for more specific manipulation of cell types, but in the absence of temporal precision. The use of microbial opsins-light activated, genetically encoded ion channels and pumps-to control mammalian neurons now allows researchers to "sensitize" genetically and/or topologically defined populations of neurons to light to induce either depolarization or hyperpolarization in both a cell-type-specific and temporally precise manner not achievable with previous techniques. Here, we describe the use of transgenic mice expressing the blue-light gated cation channel Channelrhodopsin-2 (ChR2) under control of the Thy1 promoter for the purpose of linking neuronal activity to behavior through restricted delivery of light to an anatomic region of interest. The surgical procedure for implanting a fiber-optic light delivery guide into the mouse brain, the process of optically stimulating the brain in a behaving animal, and post hoc evaluation are given, along with necessary reagents and discussion of common technical problems and their solutions.

    View details for DOI 10.1101/pdb.prot075598

    View details for PubMedID 26034299

  • Optogenetic Dissection of Neural Circuit Function in Behaving Animals NEURAL TRACING METHODS: TRACING NEURONS AND THEIR CONNECTIONS Herrera, C., Adamantidis, A., Zhang, F., Deisseroth, K., de Lecea, L., Arenkiel, B. R. 2015; 92: 143–60
  • Hippocampal "cholinergic interneurons" visualized with the choline acetyltransferase promoter: anatomical distribution, intrinsic membrane properties, neurochemical characteristics, and capacity for cholinergic modulation. Frontiers in synaptic neuroscience Yi, F., Catudio-Garrett, E., Gábriel, R., Wilhelm, M., Erdelyi, F., Szabo, G., Deisseroth, K., Lawrence, J. 2015; 7: 4-?

    Abstract

    Release of acetylcholine (ACh) in the hippocampus (HC) occurs during exploration, arousal, and learning. Although the medial septum-diagonal band of Broca (MS-DBB) is the major extrinsic source of cholinergic input to the HC, cholinergic neurons intrinsic to the HC also exist but remain poorly understood. Here, ChAT-tauGFP and ChAT-CRE/Rosa26YFP (ChAT-Rosa) mice were examined in HC. The HC of ChAT-tauGFP mice was densely innervated with GFP-positive axons, often accompanied by large GFP-positive structures, some of which were Neurotrace/DAPI-negative and likely represent large axon terminals. In the HC of ChAT-Rosa mice, ChAT-YFP cells were Neurotrace-positive and more abundant in CA3 and dentate gyrus than CA1 with partial overlap with calretinin/VIP. Moreover, an anti-ChAT antibody consistently showed ChAT immunoreactivity in ChAT-YFP cells from MS-DBB but rarely from HC. Furthermore, ChAT-YFP cells from CA1 stratum radiatum/stratum lacunosum moleculare (SR/SLM) exhibited a stuttering firing phenotype but a delayed firing phenotype in stratum pyramidale (SP) of CA3. Input resistance and capacitance were also different between CA1 SR/LM and CA3 SP ChAT-YFP cells. Bath application of ACh increased firing frequency in all ChAT-YFP cells; however, cholinergic modulation was larger in CA1 SR/SLM than CA3 SP ChAT-YFP cells. Finally, CA3 SP ChAT-YFP cells exhibited a wider AP half-width and weaker cholinergic modulation than YFP-negative CA3 pyramidal cells. Consistent with CRE expression in a subpopulation of principal cells, optogenetic stimulation evoked glutamatergic postsynaptic currents in CA1 SR/SLM interneurons. In conclusion, the presence of fluorescently labeled hippocampal cells common to both ChAT-tauGFP and ChAT-Rosa mice are in good agreement with previous reports on the existence of cholinergic interneurons, but both transgenic mouse lines exhibited unexpected anatomical features that departed considerably from earlier observations.

    View details for DOI 10.3389/fnsyn.2015.00004

    View details for PubMedID 25798106

  • Molecular Dynamics of Channelrhodopsin at the Early Stages of Channel Opening. PloS one Takemoto, M., Kato, H. E., Koyama, M., Ito, J., Kamiya, M., Hayashi, S., Maturana, A. D., Deisseroth, K., Ishitani, R., Nureki, O. 2015; 10 (6)

    Abstract

    Channelrhodopsin (ChR) is a light-gated cation channel that responds to blue light. Since ChR can be readily expressed in specific neurons to precisely control their activities by light, it has become a powerful tool in neuroscience. Although the recently solved crystal structure of a chimeric ChR, C1C2, provided the structural basis for ChR, our understanding of the molecular mechanism of ChR still remains limited. Here we performed electrophysiological analyses and all-atom molecular dynamics (MD) simulations, to investigate the importance of the intracellular and central constrictions of the ion conducting pore observed in the crystal structure of C1C2. Our electrophysiological analysis revealed that two glutamate residues, Glu122 and Glu129, in the intracellular and central constrictions, respectively, should be deprotonated in the photocycle. The simulation results suggested that the deprotonation of Glu129 in the central constriction leads to ion leakage in the ground state, and implied that the protonation of Glu129 is important for preventing ion leakage in the ground state. Moreover, we modeled the 13-cis retinal bound; i.e., activated C1C2, and performed MD simulations to investigate the conformational changes in the early stage of the photocycle. Our simulations suggested that retinal photoisomerization induces the conformational change toward channel opening, including the movements of TM6, TM7 and TM2. These insights into the dynamics of the ground states and the early photocycle stages enhance our understanding of the channel function of ChR.

    View details for DOI 10.1371/journal.pone.0131094

    View details for PubMedID 26114863

    View details for PubMedCentralID PMC4482709

  • Optogenetics in Freely Moving Mammals: Dopamine and Reward. Cold Spring Harbor protocols Zhang, F., Tsai, H., Airan, R. D., Stuber, G. D., Adamantidis, A. R., de Lecea, L., Bonci, A., Deisseroth, K. 2015; 2015 (8): pdb top086330-?

    Abstract

    Brain reward systems play a central role in the cognitive and hedonic behaviors of mammals. Multiple neuron types and brain regions are involved in reward processing, posing fascinating scientific questions, and major experimental challenges. Using diverse approaches including genetics, electrophysiology, imaging, and behavioral analysis, a large body of research has focused on both normal functioning of the reward circuitry and on its potential significance in neuropsychiatric diseases. In this introduction, we illustrate a real-world application of optogenetics to mammalian behavior and physiology, delineating procedures and technologies for optogenetic control of individual components of the reward circuitry. We describe the experimental setup and protocol for integrating optogenetic modulation of dopamine neurons with fast-scan cyclic voltammetry, conditioned place preference, and operant conditioning to assess the causal role of well-defined electrical and biochemical signals in reward-related behavior.

    View details for DOI 10.1101/pdb.top086330

    View details for PubMedID 26240415

  • Ventral hippocampal afferents to the nucleus accumbens regulate susceptibility to depression. Nature communications Bagot, R. C., Parise, E. M., Peña, C. J., Zhang, H., Maze, I., Chaudhury, D., Persaud, B., Cachope, R., Bolaños-Guzmán, C. A., Cheer, J. F., Deisseroth, K., Han, M., Nestler, E. J. 2015; 6: 7062-?

    Abstract

    Enhanced glutamatergic transmission in the nucleus accumbens (NAc), a region critical for reward and motivation, has been implicated in the pathophysiology of depression; however, the afferent source of this increased glutamate tone is not known. The NAc receives glutamatergic inputs from the medial prefrontal cortex (mPFC), ventral hippocampus (vHIP) and basolateral amygdala (AMY). Here, we demonstrate that glutamatergic vHIP afferents to NAc regulate susceptibility to chronic social defeat stress (CSDS). We observe reduced activity in vHIP in mice resilient to CSDS. Furthermore, attenuation of vHIP-NAc transmission by optogenetic induction of long-term depression is pro-resilient, whereas acute enhancement of this input is pro-susceptible. This effect is specific to vHIP afferents to the NAc, as optogenetic stimulation of either mPFC or AMY afferents to the NAc is pro-resilient. These data indicate that vHIP afferents to NAc uniquely regulate susceptibility to CSDS, highlighting an important, novel circuit-specific mechanism in depression.

    View details for DOI 10.1038/ncomms8062

    View details for PubMedID 25952660

    View details for PubMedCentralID PMC4430111

  • Hebbian and neuromodulatory mechanisms interact to trigger associative memory formation PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Johansen, J. P., Diaz-Mataix, L., Hamanaka, H., Ozawa, T., Ycu, E., Koivumaa, J., Kumar, A., Hou, M., Deisseroth, K., Boyden, E. S., LeDoux, J. E. 2014; 111 (51): E5584-E5592

    Abstract

    A long-standing hypothesis termed "Hebbian plasticity" suggests that memories are formed through strengthening of synaptic connections between neurons with correlated activity. In contrast, other theories propose that coactivation of Hebbian and neuromodulatory processes produce the synaptic strengthening that underlies memory formation. Using optogenetics we directly tested whether Hebbian plasticity alone is both necessary and sufficient to produce physiological changes mediating actual memory formation in behaving animals. Our previous work with this method suggested that Hebbian mechanisms are sufficient to produce aversive associative learning under artificial conditions involving strong, iterative training. Here we systematically tested whether Hebbian mechanisms are necessary and sufficient to produce associative learning under more moderate training conditions that are similar to those that occur in daily life. We measured neural plasticity in the lateral amygdala, a brain region important for associative memory storage about danger. Our findings provide evidence that Hebbian mechanisms are necessary to produce neural plasticity in the lateral amygdala and behavioral memory formation. However, under these conditions Hebbian mechanisms alone were not sufficient to produce these physiological and behavioral effects unless neuromodulatory systems were coactivated. These results provide insight into how aversive experiences trigger memories and suggest that combined Hebbian and neuromodulatory processes interact to engage associative aversive learning.

    View details for DOI 10.1073/pnas.1421304111

    View details for Web of Science ID 000346767200015

    View details for PubMedID 25489081

    View details for PubMedCentralID PMC4280619

  • Optogenetics Reveal Delayed Afferent Synaptogenesis on Grafted Human-Induced Pluripotent Stem Cell-Derived Neural Progenitors STEM CELLS Avaliani, N., Sorensen, A. T., Ledri, M., Bengzon, J., Koch, P., Bruestle, O., Deisseroth, K., Andersson, M., Kokaia, M. 2014; 32 (12): 3088-3098

    Abstract

    Reprogramming of somatic cells into pluripotency stem cell state has opened new opportunities in cell replacement therapy and disease modeling in a number of neurological disorders. It still remains unknown, however, to what degree the grafted human-induced pluripotent stem cells (hiPSCs) differentiate into a functional neuronal phenotype and if they integrate into the host circuitry. Here, we present a detailed characterization of the functional properties and synaptic integration of hiPSC-derived neurons grafted in an in vitro model of hyperexcitable epileptic tissue, namely organotypic hippocampal slice cultures (OHSCs), and in adult rats in vivo. The hiPSCs were first differentiated into long-term self-renewing neuroepithelial stem (lt-NES) cells, which are known to form primarily GABAergic neurons. When differentiated in OHSCs for 6 weeks, lt-NES cell-derived neurons displayed neuronal properties such as tetrodotoxin-sensitive sodium currents and action potentials (APs), as well as both spontaneous and evoked postsynaptic currents, indicating functional afferent synaptic inputs. The grafted cells had a distinct electrophysiological profile compared to host cells in the OHSCs with higher input resistance, lower resting membrane potential, and APs with lower amplitude and longer duration. To investigate the origin of synaptic afferents to the grafted lt-NES cell-derived neurons, the host neurons were transduced with Channelrhodopsin-2 (ChR2) and optogenetically activated by blue light. Simultaneous recordings of synaptic currents in grafted lt-NES cell-derived neurons using whole-cell patch-clamp technique at 6 weeks after grafting revealed limited synaptic connections from host neurons. Longer differentiation times, up to 24 weeks after grafting in vivo, revealed more mature intrinsic properties and extensive synaptic afferents from host neurons to the lt-NES cell-derived neurons, suggesting that these cells require extended time for differentiation/maturation and synaptogenesis. However, even at this later time point, the grafted cells maintained a higher input resistance. These data indicate that grafted lt-NES cell-derived neurons receive ample afferent input from the host brain. Since the lt-NES cells used in this study show a strong propensity for GABAergic differentiation, the host-to-graft synaptic afferents may facilitate inhibitory neurotransmitter release, and normalize hyperexcitable neuronal networks in brain diseases, for example, such as epilepsy.

    View details for DOI 10.1002/stem.1823

    View details for Web of Science ID 000345593700006

    View details for PubMedID 25183299

  • Depression: the best way forward. Nature Monteggia, L. M., Malenka, R. C., Deisseroth, K. 2014; 515 (7526): 200-201

    View details for DOI 10.1038/515200a

    View details for PubMedID 25391955

  • Fix faulty circuits NATURE Malenka, R. C., Deisseroth, K. 2014; 515 (7526): 200-201
  • Left-right dissociation of hippocampal memory processes in mice PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Shipton, O. A., El-Gaby, M., Apergis-Schoute, J., Deisseroth, K., Bannerman, D. M., Paulsen, O., Kohl, M. M. 2014; 111 (42): 15238-15243

    Abstract

    Left-right asymmetries have likely evolved to make optimal use of bilaterian nervous systems; however, little is known about the synaptic and circuit mechanisms that support divergence of function between equivalent structures in each hemisphere. Here we examined whether lateralized hippocampal memory processing is present in mice, where hemispheric asymmetry at the CA3-CA1 pyramidal neuron synapse has recently been demonstrated, with different spine morphology, glutamate receptor content, and synaptic plasticity, depending on whether afferents originate in the left or right CA3. To address this question, we used optogenetics to acutely silence CA3 pyramidal neurons in either the left or right dorsal hippocampus while mice performed hippocampus-dependent memory tasks. We found that unilateral silencing of either the left or right CA3 was sufficient to impair short-term memory. However, a striking asymmetry emerged in long-term memory, wherein only left CA3 silencing impaired performance on an associative spatial long-term memory task, whereas right CA3 silencing had no effect. To explore whether synaptic properties intrinsic to the hippocampus might contribute to this left-right behavioral asymmetry, we investigated the expression of hippocampal long-term potentiation. Following the induction of long-term potentiation by high-frequency electrical stimulation, synapses between CA3 and CA1 pyramidal neurons were strengthened only when presynaptic input originated in the left CA3, confirming an asymmetry in synaptic properties. The dissociation of hippocampal long-term memory function between hemispheres suggests that memory is routed via distinct left-right pathways within the mouse hippocampus, and provides a promising approach to help elucidate the synaptic basis of long-term memory.

    View details for DOI 10.1073/pnas.1405648111

    View details for Web of Science ID 000343302600069

    View details for PubMedID 25246561

    View details for PubMedCentralID PMC4210314

  • Manipulating a "Cocaine Engram" in Mice JOURNAL OF NEUROSCIENCE Hsiang, H. (., Epp, J. R., Van den Oever, M. C., Yan, C., Rashid, A. J., Insel, N., Ye, L., Niibori, Y., Deisseroth, K., Frankland, P. W., Josselyn, S. A. 2014; 34 (42): 14115-14127

    Abstract

    Experience with drugs of abuse (such as cocaine) produces powerful, long-lasting memories that may be important in the development and persistence of drug addiction. The neural mechanisms that mediate how and where these cocaine memories are encoded, consolidated and stored are unknown. Here we used conditioned place preference in mice to examine the precise neural circuits that support the memory of a cocaine-cue association (the "cocaine memory trace" or "cocaine engram"). We found that a small population of neurons (∼10%) in the lateral nucleus of amygdala (LA) were recruited at the time of cocaine-conditioning to become part of this cocaine engram. Neurons with increased levels of the transcription factor CREB were preferentially recruited or allocated to the cocaine engram. Ablating or silencing neurons overexpressing CREB (but not a similar number of random LA neurons) before testing disrupted the expression of a previously acquired cocaine memory, suggesting that neurons overexpressing CREB become a critical hub in what is likely a larger cocaine memory engram. Consistent with theories that coordinated postencoding reactivation of neurons within an engram or cell assembly is crucial for memory consolidation (Marr, 1971; Buzsáki, 1989; Wilson and McNaughton, 1994; McClelland et al., 1995; Girardeau et al., 2009; Dupret et al., 2010; Carr et al., 2011), we also found that post-training suppression, or nondiscriminate activation, of CREB overexpressing neurons impaired consolidation of the cocaine memory. These findings reveal mechanisms underlying how and where drug memories are encoded and stored in the brain and may also inform the development of treatments for drug addiction.

    View details for DOI 10.1523/JNEUROSCI.3327-14.2014

    View details for Web of Science ID 000343142800026

    View details for PubMedID 25319707

  • Enhancing the performance of the light field microscope using wavefront coding OPTICS EXPRESS Cohen, N., Yang, S., Andalman, A., Broxton, M., Grosenick, L., Deisseroth, K., Horowitz, M., Levoy, M. 2014; 22 (20): 24817-24839

    Abstract

    Light field microscopy has been proposed as a new high-speed volumetric computational imaging method that enables reconstruction of 3-D volumes from captured projections of the 4-D light field. Recently, a detailed physical optics model of the light field microscope has been derived, which led to the development of a deconvolution algorithm that reconstructs 3-D volumes with high spatial resolution. However, the spatial resolution of the reconstructions has been shown to be non-uniform across depth, with some z planes showing high resolution and others, particularly at the center of the imaged volume, showing very low resolution. In this paper, we enhance the performance of the light field microscope using wavefront coding techniques. By including phase masks in the optical path of the microscope we are able to address this non-uniform resolution limitation. We have also found that superior control over the performance of the light field microscope can be achieved by using two phase masks rather than one, placed at the objective's back focal plane and at the microscope's native image plane. We present an extended optical model for our wavefront coded light field microscope and develop a performance metric based on Fisher information, which we use to choose adequate phase masks parameters. We validate our approach using both simulated data and experimental resolution measurements of a USAF 1951 resolution target; and demonstrate the utility for biological applications with in vivo volumetric calcium imaging of larval zebrafish brain.

    View details for DOI 10.1364/OE.22.024817

    View details for Web of Science ID 000342757000104

    View details for PubMedCentralID PMC4247191

  • A fourth generation of neuroanatomical tracing techniques: Exploiting the offspring of genetic engineering JOURNAL OF NEUROSCIENCE METHODS Wouterlood, F. G., Bloem, B., Mansvelder, H. D., Luchicchi, A., Deisseroth, K. 2014; 235: 331-348

    Abstract

    The first three generations of neuroanatomical tract-tracing methods include, respectively, techniques exploiting degeneration, retrograde cellular transport and anterograde cellular transport. This paper reviews the most recent development in third-generation tracing, i.e., neurochemical fingerprinting based on BDA tracing, and continues with an emerging tracing technique called here 'selective fluorescent protein expression' that in our view belongs to an entirely new 'fourth-generation' class. Tracing techniques in this class lean on gene expression technology designed to 'label' projections exclusively originating from neurons expressing a very specific molecular phenotype. Genetically engineered mice that express cre-recombinase in a neurochemically specific neuronal population receive into a brain locus of interest an injection of an adeno-associated virus (AAV) carrying a double-floxed promoter-eYFP DNA sequence. After transfection this sequence is expressed only in neurons metabolizing recombinase protein. These particular neurons promptly start manufacturing the fluorescent protein which then accumulates and labels to full detail all the neuronal processes, including fibers and terminal arborizations. All other neurons remain optically 'dark'. The AAV is not replicated by the neurons, prohibiting intracerebral spread of 'infection'. The essence is that the fiber projections of discrete subpopulations of neurochemically specific neurons can be traced in full detail. One condition is that the transgenic mouse strain is recombinase-perfect. We illustrate selective fluorescent protein expression in parvalbumin-cre (PV-cre) mice and choline acetyltransferase-cre (ChAT-cre) mice. In addition we compare this novel tracing technique with observations in brains of native PV mice and ChAT-GFP mice. We include a note on tracing techniques using viruses.

    View details for DOI 10.1016/j.jneumeth.2014.07.021

    View details for Web of Science ID 000342269400035

    View details for PubMedID 25107853

  • Optical suppression of drug-evoked phasic dopamine release FRONTIERS IN NEURAL CIRCUITS McCutcheon, J. E., Cone, J. J., Sinon, C. G., Fortin, S. M., Kantak, P. A., Witten, I. B., Deisseroth, K., Stuber, G. D., Roitman, M. F. 2014; 8

    Abstract

    Brief fluctuations in dopamine concentration (dopamine transients) play a key role in behavior towards rewards, including drugs of abuse. Drug-evoked dopamine transients may result from actions at both dopamine cell bodies and dopamine terminals. Inhibitory opsins can be targeted to dopamine neurons permitting their firing activity to be suppressed. However, as dopamine transients can become uncoupled from firing, it is unknown whether optogenetic hyperpolarization at the level of the soma is able to suppress dopamine transients. Here, we used in vivo fast-scan cyclic voltammetry to record transients evoked by cocaine and raclopride in nucleus accumbens (NAc) of urethane-anesthetized rats. We targeted halorhodopsin (NpHR) specifically to dopamine cells by injecting Cre-inducible virus into ventral tegmental area (VTA) of transgenic rats that expressed Cre recombinase under control of the tyrosine hydroxylase promoter (TH-Cre(+) rats). Consistent with previous work, co-administration of cocaine and raclopride led to the generation of dopamine transients in NAc shell. Illumination of VTA with laser strongly suppressed the frequency of transients in NpHR-expressing rats, but not in control rats. Laser did not have any effect on amplitude of transients. Thus, optogenetics can effectively reduce the occurrence of drug-evoked transients and is therefore a suitable approach for studying the functional role of such transients in drug-associated behavior.

    View details for DOI 10.3389/fncir.2014.00114

    View details for Web of Science ID 000341954100001

    View details for PubMedID 25278845

    View details for PubMedCentralID PMC4166314

  • Optogenetic neuronal stimulation promotes functional recovery after stroke PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Cheng, M. Y., Wang, E. H., Woodson, W. J., Wang, S., Sun, G., Lee, A. G., Arac, A., Fenno, L. E., Deisseroth, K., Steinberg, G. K. 2014; 111 (35): 12913-12918

    Abstract

    Clinical and research efforts have focused on promoting functional recovery after stroke. Brain stimulation strategies are particularly promising because they allow direct manipulation of the target area's excitability. However, elucidating the cell type and mechanisms mediating recovery has been difficult because existing stimulation techniques nonspecifically target all cell types near the stimulated site. To circumvent these barriers, we used optogenetics to selectively activate neurons that express channelrhodopsin 2 and demonstrated that selective neuronal stimulations in the ipsilesional primary motor cortex (iM1) can promote functional recovery. Stroke mice that received repeated neuronal stimulations exhibited significant improvement in cerebral blood flow and the neurovascular coupling response, as well as increased expression of activity-dependent neurotrophins in the contralesional cortex, including brain-derived neurotrophic factor, nerve growth factor, and neurotrophin 3. Western analysis also indicated that stimulated mice exhibited a significant increase in the expression of a plasticity marker growth-associated protein 43. Moreover, iM1 neuronal stimulations promoted functional recovery, as stimulated stroke mice showed faster weight gain and performed significantly better in sensory-motor behavior tests. Interestingly, stimulations in normal nonstroke mice did not alter motor behavior or neurotrophin expression, suggesting that the prorecovery effect of selective neuronal stimulations is dependent on the poststroke environment. These results demonstrate that stimulation of neurons in the stroke hemisphere is sufficient to promote recovery.

    View details for DOI 10.1073/pnas.1404109111

    View details for Web of Science ID 000341230800084

    View details for PubMedCentralID PMC4156770

  • Optogenetic neuronal stimulation promotes functional recovery after stroke. Proceedings of the National Academy of Sciences of the United States of America Cheng, M. Y., Wang, E. H., Woodson, W. J., Wang, S., Sun, G., Lee, A. G., Arac, A., Fenno, L. E., Deisseroth, K., Steinberg, G. K. 2014; 111 (35): 12913-12918

    Abstract

    Clinical and research efforts have focused on promoting functional recovery after stroke. Brain stimulation strategies are particularly promising because they allow direct manipulation of the target area's excitability. However, elucidating the cell type and mechanisms mediating recovery has been difficult because existing stimulation techniques nonspecifically target all cell types near the stimulated site. To circumvent these barriers, we used optogenetics to selectively activate neurons that express channelrhodopsin 2 and demonstrated that selective neuronal stimulations in the ipsilesional primary motor cortex (iM1) can promote functional recovery. Stroke mice that received repeated neuronal stimulations exhibited significant improvement in cerebral blood flow and the neurovascular coupling response, as well as increased expression of activity-dependent neurotrophins in the contralesional cortex, including brain-derived neurotrophic factor, nerve growth factor, and neurotrophin 3. Western analysis also indicated that stimulated mice exhibited a significant increase in the expression of a plasticity marker growth-associated protein 43. Moreover, iM1 neuronal stimulations promoted functional recovery, as stimulated stroke mice showed faster weight gain and performed significantly better in sensory-motor behavior tests. Interestingly, stimulations in normal nonstroke mice did not alter motor behavior or neurotrophin expression, suggesting that the prorecovery effect of selective neuronal stimulations is dependent on the poststroke environment. These results demonstrate that stimulation of neurons in the stroke hemisphere is sufficient to promote recovery.

    View details for DOI 10.1073/pnas.1404109111

    View details for PubMedID 25136109

  • Frequency-dependent, cell type-divergent signaling in the hippocamposeptal projection. journal of neuroscience Mattis, J., Brill, J., Evans, S., Lerner, T. N., Davidson, T. J., Hyun, M., Ramakrishnan, C., Deisseroth, K., Huguenard, J. R. 2014; 34 (35): 11769-11780

    Abstract

    Hippocampal oscillations are critical for information processing, and are strongly influenced by inputs from the medial septum. Hippocamposeptal neurons provide direct inhibitory feedback from the hippocampus onto septal cells, and are therefore likely to also play an important role in the circuit; these neurons fire at either low or high frequency, reflecting hippocampal network activity during theta oscillations or ripple events, respectively. Here, we optogenetically target the long-range GABAergic projection from the hippocampus to the medial septum in rats, and thereby simulate hippocampal input onto downstream septal cells in an acute slice preparation. In response to optogenetic activation of hippocamposeptal fibers at theta and ripple frequencies, we elicit postsynaptic GABAergic responses in a subset (24%) of septal cells, most predominantly in fast-spiking cells. In addition, in another subset of septal cells (19%) corresponding primarily to cholinergic cells, we observe a slow hyperpolarization of the resting membrane potential and a decrease in input resistance, particularly in response to prolonged high-frequency (ripple range) stimulation. This slow response is partially sensitive to GIRK channel and D2 dopamine receptor block. Our results suggest that two independent populations of septal cells distinctly encode hippocampal feedback, enabling the septum to monitor ongoing patterns of activity in the hippocampus.

    View details for DOI 10.1523/JNEUROSCI.5188-13.2014

    View details for PubMedID 25164672

    View details for PubMedCentralID PMC4145178

  • Direct excitation of parvalbumin-positive interneurons by M-1 muscarinic acetylcholine receptors: roles in cellular excitability, inhibitory transmission and cognition JOURNAL OF PHYSIOLOGY-LONDON Yi, F., Ball, J., Stoll, K. E., Satpute, V. C., Mitchell, S. M., Pauli, J. L., Holloway, B. B., Johnston, A. D., Nathanson, N. M., Deisseroth, K., Gerber, D. J., Tonegawa, S., Lawrence, J. J. 2014; 592 (16): 3463-3494

    Abstract

    Parvalbumin-containing (PV) neurons, a major class of GABAergic interneurons, are essential circuit elements of learning networks. As levels of acetylcholine rise during active learning tasks, PV neurons become increasingly engaged in network dynamics. Conversely, impairment of either cholinergic or PV interneuron function induces learning deficits. Here, we examined PV interneurons in hippocampus (HC) and prefrontal cortex (PFC) and their modulation by muscarinic acetylcholine receptors (mAChRs). HC PV cells, visualized by crossing PV-CRE mice with Rosa26YFP mice, were anatomically identified as basket cells and PV bistratified cells in the stratum pyramidale; in stratum oriens, HC PV cells were electrophysiologically distinct from somatostatin-containing cells. With glutamatergic transmission pharmacologically blocked, mAChR activation enhanced PV cell excitability in both CA1 HC and PFC; however, CA1 HC PV cells exhibited a stronger postsynaptic depolarization than PFC PV cells. To delete M1 mAChRs genetically from PV interneurons, we created PV-M1 knockout mice by crossing PV-CRE and floxed M1 mice. The elimination of M1 mAChRs from PV cells diminished M1 mAChR immunoreactivity and muscarinic excitation of HC PV cells. Selective cholinergic activation of HC PV interneurons using Designer Receptors Exclusively Activated by Designer Drugs technology enhanced the frequency and amplitude of inhibitory synaptic currents in CA1 pyramidal cells. Finally, relative to wild-type controls, PV-M1 knockout mice exhibited impaired novel object recognition and, to a lesser extent, impaired spatial working memory, but reference memory remained intact. Therefore, the direct activation of M1 mAChRs on PV cells contributes to some forms of learning and memory.

    View details for DOI 10.1113/jphysiol.2014.275453

    View details for Web of Science ID 000340599200014

    View details for PubMedID 24879872

    View details for PubMedCentralID PMC4229343

  • Nucleus Accumbens-Specific Interventions in RGS9-2 Activity Modulate Responses to Morphine. Neuropsychopharmacology Gaspari, S., Papachatzaki, M. M., Koo, J. W., Carr, F. B., Tsimpanouli, M., Stergiou, E., Bagot, R. C., Ferguson, D., Mouzon, E., Chakravarty, S., Deisseroth, K., Lobo, M. K., Zachariou, V. 2014; 39 (8): 1968-1977

    Abstract

    Regulator of G protein signalling 9-2 (Rgs9-2) modulates the actions of a wide range of CNS-acting drugs by controlling signal transduction of several GPCRs in the striatum. RGS9-2 acts via a complex mechanism that involves interactions with Gα subunits, the Gβ5 protein, and the adaptor protein R7BP. Our recent work identified Rgs9-2 complexes in the striatum associated with acute or chronic exposures to mu opioid receptor (MOR) agonists. In this study we use several new genetic tools that allow manipulations of Rgs9-2 activity in particular brain regions of adult mice in order to better understand the mechanism via which this protein modulates opiate addiction and analgesia. We used adeno-associated viruses (AAVs) to express forms of Rgs9-2 in the dorsal and ventral striatum (nucleus accumbens, NAc) in order to examine the influence of this protein in morphine actions. Consistent with earlier behavioural findings from constitutive Rgs9 knockout mice, we show that Rgs9-2 actions in the NAc modulate morphine reward and dependence. Notably, Rgs9-2 in the NAc affects the analgesic actions of morphine as well as the development of analgesic tolerance. Using optogenetics we demonstrate that activation of Channelrhodopsin2 in Rgs9-2-expressing neurons, or in D1 dopamine receptor (Drd1)-enriched medium spiny neurons, accelerates the development of morphine tolerance, whereas activation of D2 dopamine receptor (Drd2)-enriched neurons does not significantly affect the development of tolerance. Together, these data provide new information on the signal transduction mechanisms underlying opiate actions in the NAc.

    View details for DOI 10.1038/npp.2014.45

    View details for PubMedID 24561386

    View details for PubMedCentralID PMC4059906

  • Advanced CLARITY for rapid and high-resolution imaging of intact tissues. Nature protocols Tomer, R., Ye, L., Hsueh, B., Deisseroth, K. 2014; 9 (7): 1682-1697

    Abstract

    CLARITY is a method for chemical transformation of intact biological tissues into a hydrogel-tissue hybrid, which becomes amenable to interrogation with light and macromolecular labels while retaining fine structure and native biological molecules. This emerging accessibility of information from large intact samples has created both new opportunities and new challenges. Here we describe protocols spanning multiple dimensions of the CLARITY workflow, ranging from simple, reliable and efficient lipid removal without electrophoretic instrumentation (passive CLARITY) to optimized objectives and integration with light-sheet optics (CLARITY-optimized light-sheet microscopy (COLM)) for accelerating data collection from clarified samples by several orders of magnitude while maintaining or increasing quality and resolution. The entire protocol takes from 7-28 d to complete for an adult mouse brain, including hydrogel embedding, full lipid removal, whole-brain antibody staining (which, if needed, accounts for 7-10 of the days), and whole-brain high-resolution imaging; timing within this window depends on the choice of lipid removal options, on the size of the tissue, and on the number and type of immunostaining rounds performed. This protocol has been successfully applied to the study of adult mouse, adult zebrafish and adult human brains, and it may find many other applications in the structural and molecular analysis of large assembled biological systems.

    View details for DOI 10.1038/nprot.2014.123

    View details for PubMedID 24945384

  • Targeting cells with single vectors using multiple-feature Boolean logic NATURE METHODS Fenno, L. E., Mattis, J., Ramakrishnan, C., Hyun, M., Lee, S. Y., He, M., Tucciarone, J., Selimbeyoglu, A., Berndt, A., Grosenick, L., Zalocusky, K. A., Bernstein, H., Swanson, H., Perry, C., Diester, I., Boyce, F. M., Bass, C. E., Neve, R., Huang, Z. J., Deisseroth, K. 2014; 11 (7): 763-U116

    Abstract

    Precisely defining the roles of specific cell types is an intriguing frontier in the study of intact biological systems and has stimulated the rapid development of genetically encoded tools for observation and control. However, targeting these tools with adequate specificity remains challenging: most cell types are best defined by the intersection of two or more features such as active promoter elements, location and connectivity. Here we have combined engineered introns with specific recombinases to achieve expression of genetically encoded tools that is conditional upon multiple cell-type features, using Boolean logical operations all governed by a single versatile vector. We used this approach to target intersectionally specified populations of inhibitory interneurons in mammalian hippocampus and neurons of the ventral tegmental area defined by both genetic and wiring properties. This flexible and modular approach may expand the application of genetically encoded interventional and observational tools for intact-systems biology.

    View details for DOI 10.1038/NMETH.2996

    View details for Web of Science ID 000338321400022

    View details for PubMedCentralID PMC4085277

  • Optogenetic inhibition of chemically induced hypersynchronized bursting in mice NEUROBIOLOGY OF DISEASE Berglind, F., Ledri, M., Sorensen, A. T., Nikitidou, L., Melis, M., Bielefeld, P., Kirik, D., Deisseroth, K., Andersson, M., Kokaia, M. 2014; 65: 133-141

    Abstract

    Synchronized activity is common during various physiological operations but can culminate in seizures and consequently in epilepsy in pathological hyperexcitable conditions in the brain. Many types of seizures are not possible to control and impose significant disability for patients with epilepsy. Such intractable epilepsy cases are often associated with degeneration of inhibitory interneurons in the cortical areas resulting in impaired inhibitory drive onto the principal neurons. Recently emerging optogenetic technique has been proposed as an alternative approach to control such seizures but whether it may be effective in situations where inhibitory processes in the brain are compromised has not been addressed. Here we used pharmacological and optogenetic techniques to block inhibitory neurotransmission and induce epileptiform activity in vitro and in vivo. We demonstrate that NpHR-based optogenetic hyperpolarization and thereby inactivation of a principal neuronal population in the hippocampus is effectively attenuating seizure activity caused by disconnected network inhibition both in vitro and in vivo. Our data suggest that epileptiform activity in the hippocampus caused by impaired inhibition may be controlled by optogenetic silencing of principal neurons and potentially can be developed as an alternative treatment for epilepsy.

    View details for DOI 10.1016/j.nbd.2014.01.015

    View details for Web of Science ID 000333546300013

    View details for PubMedID 24491965

  • Prefrontal Cortex-amygdalar Circuit Dynamics Predict Stress Susceptibility Dzirasa, K., Kumar, S., Hultman, R., Lin, L., Li, Q., Hughes, D., Mague, S., Michel, N., Katz, B., Moore, S., Deisseroth, K., Roth, B., Dunson, D. ELSEVIER SCIENCE INC. 2014: 267S–268S
  • Structure-Guided Transformation of Channelrhodopsin into a Light-Activated Chloride Channel SCIENCE Berndt, A., Lee, S. Y., Ramakrishnan, C., Deisseroth, K. 2014; 344 (6182): 420-424

    Abstract

    Using light to silence electrical activity in targeted cells is a major goal of optogenetics. Available optogenetic proteins that directly move ions to achieve silencing are inefficient, pumping only a single ion per photon across the cell membrane rather than allowing many ions per photon to flow through a channel pore. Building on high-resolution crystal-structure analysis, pore vestibule modeling, and structure-guided protein engineering, we designed and characterized a class of channelrhodopsins (originally cation-conducting) converted into chloride-conducting anion channels. These tools enable fast optical inhibition of action potentials and can be engineered to display step-function kinetics for stable inhibition, outlasting light pulses and for orders-of-magnitude-greater light sensitivity of inhibited cells. The resulting family of proteins defines an approach to more physiological, efficient, and sensitive optogenetic inhibition.

    View details for DOI 10.1126/science.1252367

    View details for Web of Science ID 000334867800043

  • Positive Reinforcement Mediated by Midbrain Dopamine Neurons Requires D1 and D2 Receptor Activation in the Nucleus Accumbens PLOS ONE Steinberg, E. E., Boivin, J. R., Saunders, B. T., Witten, I. B., Deisseroth, K., Janak, P. H. 2014; 9 (4)

    Abstract

    The neural basis of positive reinforcement is often studied in the laboratory using intracranial self-stimulation (ICSS), a simple behavioral model in which subjects perform an action in order to obtain exogenous stimulation of a specific brain area. Recently we showed that activation of ventral tegmental area (VTA) dopamine neurons supports ICSS behavior, consistent with proposed roles of this neural population in reinforcement learning. However, VTA dopamine neurons make connections with diverse brain regions, and the specific efferent target(s) that mediate the ability of dopamine neuron activation to support ICSS have not been definitively demonstrated. Here, we examine in transgenic rats whether dopamine neuron-specific ICSS relies on the connection between the VTA and the nucleus accumbens (NAc), a brain region also implicated in positive reinforcement. We find that optogenetic activation of dopaminergic terminals innervating the NAc is sufficient to drive ICSS, and that ICSS driven by optical activation of dopamine neuron somata in the VTA is significantly attenuated by intra-NAc injections of D1 or D2 receptor antagonists. These data demonstrate that the NAc is a critical efferent target sustaining dopamine neuron-specific ICSS, identify receptor subtypes through which dopamine acts to promote this behavior, and ultimately help to refine our understanding of the neural circuitry mediating positive reinforcement.

    View details for DOI 10.1371/journal.pone.0094771

    View details for Web of Science ID 000336970400098

    View details for PubMedCentralID PMC3986242

  • Designer receptors show role for ventral pallidum input to ventral tegmental area in cocaine seeking NATURE NEUROSCIENCE Mahler, S. V., Vazey, E. M., Beckley, J. T., Keistler, C. R., McGlinchey, E. M., Kaufling, J., Wilson, S. P., Deisseroth, K., Woodward, J. J., Aston-Jones, G. 2014; 17 (4): 577-U136

    Abstract

    The ventral pallidum is centrally positioned within mesocorticolimbic reward circuits, and its dense projection to the ventral tegmental area (VTA) regulates neuronal activity there. However, the ventral pallidum is a heterogeneous structure, and how this complexity affects its role within wider reward circuits is unclear. We found that projections to VTA from the rostral ventral pallidum (RVP), but not the caudal ventral pallidum (CVP), were robustly Fos activated during cue-induced reinstatement of cocaine seeking--a rat model of relapse in addiction. Moreover, designer receptor-mediated transient inactivation of RVP neurons, their terminals in VTA or functional connectivity between RVP and VTA dopamine neurons blocked the ability of drug-associated cues (but not a cocaine prime) to reinstate cocaine seeking. In contrast, CVP neuronal inhibition blocked cocaine-primed, but not cue-induced, reinstatement. This double dissociation in ventral pallidum subregional roles in drug seeking is likely to be important for understanding the mesocorticolimbic circuits underlying reward seeking and addiction.

    View details for DOI 10.1038/nn.3664

    View details for Web of Science ID 000333405300018

    View details for PubMedID 24584054

    View details for PubMedCentralID PMC3973180

  • A Major External Source of Cholinergic Innervation of the Striatum and Nucleus Accumbens Originates in the Brainstem JOURNAL OF NEUROSCIENCE Dautan, D., Huerta-Ocampo, I., Witten, I. B., Deisseroth, K., Bolam, J. P., Gerdjikov, T., Mena-Segovia, J. 2014; 34 (13): 4509-4518

    Abstract

    Cholinergic transmission in the striatal complex is critical for the modulation of the activity of local microcircuits and dopamine release. Release of acetylcholine has been considered to originate exclusively from a subtype of striatal interneuron that provides widespread innervation of the striatum. Cholinergic neurons of the pedunculopontine (PPN) and laterodorsal tegmental (LDT) nuclei indirectly influence the activity of the dorsal striatum and nucleus accumbens through their innervation of dopamine and thalamic neurons, which in turn converge at the same striatal levels. Here we show that cholinergic neurons in the brainstem also provide a direct innervation of the striatal complex. By the expression of fluorescent proteins in choline acetyltransferase (ChAT)::Cre(+) transgenic rats, we selectively labeled cholinergic neurons in the rostral PPN, caudal PPN, and LDT. We show that cholinergic neurons topographically innervate wide areas of the striatal complex: rostral PPN preferentially innervates the dorsolateral striatum, and LDT preferentially innervates the medial striatum and nucleus accumbens core in which they principally form asymmetric synapses. Retrograde labeling combined with immunohistochemistry in wild-type rats confirmed the topography and cholinergic nature of the projection. Furthermore, transynaptic gene activation and conventional double retrograde labeling suggest that LDT neurons that innervate the nucleus accumbens also send collaterals to the thalamus and the dopaminergic midbrain, thus providing both direct and indirect projections, to the striatal complex. The differential activity of cholinergic interneurons and cholinergic neurons of the brainstem during reward-related paradigms suggest that the two systems play different but complementary roles in the processing of information in the striatum.

    View details for DOI 10.1523/JNEUROSCI.5071-13.2014

    View details for Web of Science ID 000333674200007

    View details for PubMedID 24671996

    View details for PubMedCentralID PMC3965779

  • Dendritic inhibition provided by interneuron-specific cells controls the firing rate and timing of the hippocampal feedback inhibitory circuitry. The Journal of neuroscience : the official journal of the Society for Neuroscience Tyan, L., Chamberland, S., Magnin, E., Camiré, O., Francavilla, R., David, L. S., Deisseroth, K., Topolnik, L. 2014; 34 (13): 4534-47

    Abstract

    In cortical networks, different types of inhibitory interneurons control the activity of glutamatergic principal cells and GABAergic interneurons. Principal neurons represent the major postsynaptic target of most interneurons; however, a population of interneurons that is dedicated to the selective innervation of GABAergic cells exists in the CA1 area of the hippocampus. The physiological properties of these cells and their functional relevance for network computations remain unknown. Here, we used a combination of dual simultaneous patch-clamp recordings and targeted optogenetic stimulation in acute mouse hippocampal slices to examine how one class of interneuron-specific (IS) cells controls the activity of its GABAergic targets. We found that type 3 IS (IS3) cells that coexpress the vasoactive intestinal polypeptide (VIP) and calretinin contact several distinct types of interneurons within the hippocampal CA1 stratum oriens/alveus (O/A), with preferential innervation of oriens-lacunosum moleculare cells (OLMs) through dendritic synapses. In contrast, VIP-positive basket cells provided perisomatic inhibition to CA1 pyramidal neurons with the asynchronous GABA release and were not connected with O/A interneurons. Furthermore, unitary IPSCs recorded at IS3-OLM synapses had a small amplitude and low release probability but summated efficiently during high-frequency firing of IS3 interneurons. Moreover, the synchronous generation of a single spike in several IS cells that converged onto a single OLM controlled the firing rate and timing of OLM interneurons. Therefore, dendritic inhibition originating from IS cells is needed for the flexible activity-dependent recruitment of OLM interneurons for feedback inhibition.

    View details for DOI 10.1523/JNEUROSCI.3813-13.2014

    View details for PubMedID 24671999

  • Neuronal calcium-binding proteins 1/2 localize to dorsal root ganglia and excitatory spinal neurons and are regulated by nerve injury PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Zhang, M., Tortoriello, G., Hsueh, B., Tomer, R., Ye, L., Mitsios, N., Borgius, L., Grant, G., Kiehn, O., Watanabe, M., Uhlen, M., Mulder, J., Deisseroth, K., Harkany, T., Hokfelt, T. G. 2014; 111 (12): E1149-E1158

    Abstract

    Neuronal calcium (Ca(2+))-binding proteins 1 and 2 (NECAB1/2) are members of the phylogenetically conserved EF-hand Ca(2+)-binding protein superfamily. To date, NECABs have been explored only to a limited extent and, so far, not at all at the spinal level. Here, we describe the distribution, phenotype, and nerve injury-induced regulation of NECAB1/NECAB2 in mouse dorsal root ganglia (DRGs) and spinal cord. In DRGs, NECAB1/2 are expressed in around 70% of mainly small- and medium-sized neurons. Many colocalize with calcitonin gene-related peptide and isolectin B4, and thus represent nociceptors. NECAB1/2 neurons are much more abundant in DRGs than the Ca(2+)-binding proteins (parvalbumin, calbindin, calretinin, and secretagogin) studied to date. In the spinal cord, the NECAB1/2 distribution is mainly complementary. NECAB1 labels interneurons and a plexus of processes in superficial layers of the dorsal horn, commissural neurons in the intermediate area, and motor neurons in the ventral horn. Using CLARITY, a novel, bilaterally connected neuronal system with dendrites that embrace the dorsal columns like palisades is observed. NECAB2 is present in cell bodies and presynaptic boutons across the spinal cord. In the dorsal horn, most NECAB1/2 neurons are glutamatergic. Both NECAB1/2 are transported into dorsal roots and peripheral nerves. Peripheral nerve injury reduces NECAB2, but not NECAB1, expression in DRG neurons. Our study identifies NECAB1/2 as abundant Ca(2+)-binding proteins in pain-related DRG neurons and a variety of spinal systems, providing molecular markers for known and unknown neuron populations of mechanosensory and pain circuits in the spinal cord.

    View details for DOI 10.1073/pnas.1402318111

    View details for Web of Science ID 000333341100013

    View details for PubMedID 24616509

    View details for PubMedCentralID PMC3970515

  • Virally mediated optogenetic excitation and inhibition of pain in freely moving nontransgenic mice NATURE BIOTECHNOLOGY Iyer, S. M., Montgomery, K. L., Towne, C., Lee, S. Y., Ramakrishnan, C., Deisseroth, K., Delp, S. L. 2014; 32 (3): 274-278

    Abstract

    Primary nociceptors are the first neurons involved in the complex processing system that regulates normal and pathological pain. Because of constraints on pharmacological and electrical stimulation, noninvasive excitation and inhibition of these neurons in freely moving nontransgenic animals has not been possible. Here we use an optogenetic strategy to bidirectionally control nociceptors of nontransgenic mice. Intrasciatic nerve injection of adeno-associated viruses encoding an excitatory opsin enabled light-inducible stimulation of acute pain, place aversion and optogenetically mediated reductions in withdrawal thresholds to mechanical and thermal stimuli. In contrast, viral delivery of an inhibitory opsin enabled light-inducible inhibition of acute pain perception, and reversed mechanical allodynia and thermal hyperalgesia in a model of neuropathic pain. Light was delivered transdermally, allowing these behaviors to be induced in freely moving animals. This approach may have utility in basic and translational pain research, and enable rapid drug screening and testing of newly engineered opsins.

    View details for DOI 10.1038/nbt.2834

    View details for Web of Science ID 000332819800026

    View details for PubMedID 24531797

  • Medial prefrontal D1 dopamine neurons control food intake NATURE NEUROSCIENCE Land, B. B., Narayanan, N. S., Liu, R., Gianessi, C. A., Brayton, C. E., Grimaldi, D. M., Sarhan, M., Guarnieri, D. J., Deisseroth, K., Aghajanian, G. K., DiLeone, R. J. 2014; 17 (2): 248-253

    Abstract

    Although the prefrontal cortex influences motivated behavior, its role in food intake remains unclear. Here, we demonstrate a role for D1-type dopamine receptor-expressing neurons in the medial prefrontal cortex (mPFC) in the regulation of feeding. Food intake increases activity in D1 neurons of the mPFC in mice, and optogenetic photostimulation of D1 neurons increases feeding. Conversely, inhibition of D1 neurons decreases intake. Stimulation-based mapping of prefrontal D1 neuron projections implicates the medial basolateral amygdala (mBLA) as a downstream target of these afferents. mBLA neurons activated by prefrontal D1 stimulation are CaMKII positive and closely juxtaposed to prefrontal D1 axon terminals. Finally, photostimulating these axons in the mBLA is sufficient to increase feeding, recapitulating the effects of mPFC D1 stimulation. These data describe a new circuit for top-down control of food intake.

    View details for DOI 10.1038/nn.3625

    View details for Web of Science ID 000330910000019

    View details for PubMedID 24441680

    View details for PubMedCentralID PMC3968853

  • Human pluripotent stem cell tools for cardiac optogenetics. Conference proceedings : ... Annual International Conference of the IEEE Engineering in Medicine and Biology Society. IEEE Engineering in Medicine and Biology Society. Annual Conference Zhuge, Y., Patlolla, B., Ramakrishnan, C., Beygui, R. E., Zarins, C. K., Deisseroth, K., Kuhl, E., Abilez, O. J. 2014; 2014: 6171-6174

    Abstract

    It is likely that arrhythmias should be avoided for therapies based on human pluripotent stem cell (hPSC)-derived cardiomyocytes (CM) to be effective. Towards achieving this goal, we introduced light-activated channelrhodopsin-2 (ChR2), a cation channel activated with 480 nm light, into human embryonic stem cells (hESC). By using in vitro approaches, hESC-CM are able to be activated with light. ChR2 is stably transduced into undifferentiated hESC via a lentiviral vector. Via directed differentiation, hESC(ChR2)-CM are produced and subjected to optical stimulation. hESC(ChR2)-CM respond to traditional electrical stimulation and produce similar contractility features as their wild-type counterparts but only hESC(ChR2)-CM can be activated by optical stimulation. Here it is shown that a light sensitive protein can enable in vitro optical control of hESC-CM and that this activation occurs optimally above specific light stimulation intensity and pulse width thresholds. For future therapy, in vivo optical stimulation along with optical inhibition could allow for acute synchronization of implanted hPSC-CM with patient cardiac rhythms.

    View details for DOI 10.1109/EMBC.2014.6945038

    View details for PubMedID 25571406

  • Optogenetic Tools for Control of Neural Activity OPTICAL IMAGING OF NEOCORTICAL DYNAMICS Fenno, L. E., Deisseroth, K., Weber, B., Helmchen, F. 2014; 85: 73-86
  • Neocortical Circuit Interrogation with Optogenetics OPTICAL IMAGING OF NEOCORTICAL DYNAMICS Fenno, L. E., Deisseroth, K., Weber, B., Helmchen, F. 2014; 85: 175-188
  • Establishing a fiber-optic-based optical neural interface. Cold Spring Harbor protocols Adamantidis, A. R., Zhang, F., de Lecea, L., Deisseroth, K. 2014; 2014 (8): pdb prot083337-?

    Abstract

    Selective expression of opsins in genetically defined neurons makes it possible to control a subset of neurons without affecting nearby cells and processes in the intact brain, but light must still be delivered to the target brain structure. Light scattering limits the delivery of light from the surface of the brain. For this reason, we have developed a fiber-optic-based optical neural interface (ONI), which allows optical access to any brain structure in freely moving mammals. The ONI system is constructed by modifying the small animal cannula system from PlasticsOne. The system for bilateral stimulation consists of a bilateral cannula guide that has been stereotactically implanted over the target brain region, a screw cap for securing the optical fiber to the animal's head, a fiber guard modified from the internal cannula adapter, and a bare fiber whose length is customized based on the depth of the target region. For unilateral stimulation, a single-fiber system can be constructed using unilateral cannula parts from PlasticsOne. We describe here the preparation of the bilateral ONI system and its use in optical stimulation of the mouse or rat brain. Delivery of opsin-expressing virus and implantation of the ONI may be conducted in the same surgical session; alternatively, with a transgenic animal no opsin virus is delivered during the surgery. Similar procedures are useful for deep or superficial injections (even for neocortical targets, although in some cases surface light-emitting diodes or cortex-apposed fibers can be used for the most superficial cortical targets).

    View details for DOI 10.1101/pdb.prot083337

    View details for PubMedID 25086020

  • Positive reinforcement mediated by midbrain dopamine neurons requires D1 and D2 receptor activation in the nucleus accumbens. PloS one Steinberg, E. E., Boivin, J. R., Saunders, B. T., Witten, I. B., Deisseroth, K., Janak, P. H. 2014; 9 (4)

    Abstract

    The neural basis of positive reinforcement is often studied in the laboratory using intracranial self-stimulation (ICSS), a simple behavioral model in which subjects perform an action in order to obtain exogenous stimulation of a specific brain area. Recently we showed that activation of ventral tegmental area (VTA) dopamine neurons supports ICSS behavior, consistent with proposed roles of this neural population in reinforcement learning. However, VTA dopamine neurons make connections with diverse brain regions, and the specific efferent target(s) that mediate the ability of dopamine neuron activation to support ICSS have not been definitively demonstrated. Here, we examine in transgenic rats whether dopamine neuron-specific ICSS relies on the connection between the VTA and the nucleus accumbens (NAc), a brain region also implicated in positive reinforcement. We find that optogenetic activation of dopaminergic terminals innervating the NAc is sufficient to drive ICSS, and that ICSS driven by optical activation of dopamine neuron somata in the VTA is significantly attenuated by intra-NAc injections of D1 or D2 receptor antagonists. These data demonstrate that the NAc is a critical efferent target sustaining dopamine neuron-specific ICSS, identify receptor subtypes through which dopamine acts to promote this behavior, and ultimately help to refine our understanding of the neural circuitry mediating positive reinforcement.

    View details for DOI 10.1371/journal.pone.0094771

    View details for PubMedID 24733061

    View details for PubMedCentralID PMC3986242

  • Dopaminergic Dynamics Contributing to Social Behavior. Cold Spring Harbor symposia on quantitative biology Gunaydin, L. A., Deisseroth, K. 2014; 79: 221-227

    Abstract

    Social interaction is a complex behavior that is essential for the survival of many species, and it is impaired in a broad range of neuropsychiatric disorders. Several cortical and subcortical brain regions have been implicated in a variety of sociosexual behaviors, with pharmacological studies pointing to a key role of the neurotransmitter dopamine. However, little is understood about the real-time circuit dynamics causally underlying social interaction. Here, we consider current knowledge on the role of brain reward circuitry in same-sex social behavior and describe findings from new methods for probing how this circuitry governs social motivation in health and disease.

    View details for DOI 10.1101/sqb.2014.79.024711

    View details for PubMedID 25943769

  • Synaptic encoding of fear extinction in mPFC-amygdala circuits. Neuron Cho, J. H., Deisseroth, K., Bolshakov, V. Y. 2013; 80 (6): 1491-507

    Abstract

    Retrieval of fear extinction memory is associated with increased firing of neurons in the medial prefrontal cortex (mPFC). It is unknown, however, how extinction learning-induced changes in mPFC activity are relayed to target structures in the amygdala, resulting in diminished fear responses. Here, we show that fear extinction decreases the efficacy of excitatory synaptic transmission in projections from the mPFC to the basolateral nucleus of the amygdala (BLA), whereas inhibitory responses are not altered. In contrast, synaptic strength at direct mPFC inputs to intercalated neurons remains unchanged after extinction. Moreover, priming stimulation of mPFC projections induced heterosynaptic inhibition in auditory cortical inputs to the BLA. These synaptic mechanisms could contribute to the encoding of extinction memory by diminishing the ability of projections from the mPFC to drive BLA activity while retaining the ability of intercalated neurons to inhibit the output nuclei of the amygdala.

    View details for DOI 10.1016/j.neuron.2013.09.025

    View details for PubMedID 24290204

    View details for PubMedCentralID PMC3872173

  • Cerebellar Purkinje cell activity drives motor learning. Nature neuroscience Nguyen-Vu, T. D., Kimpo, R. R., Rinaldi, J. M., Kohli, A., Zeng, H., Deisseroth, K., Raymond, J. L. 2013; 16 (12): 1734-1736

    Abstract

    The climbing fiber input to the cerebellar cortex is thought to provide instructive signals that drive the induction of motor skill learning. We found that optogenetic activation of Purkinje cells, the sole output neurons of the cerebellar cortex, can also drive motor learning in mice. This dual control over the induction of learning by climbing fibers and Purkinje cells can expand the learning capacity of motor circuits.

    View details for DOI 10.1038/nn.3576

    View details for PubMedID 24162651

  • Light microscopy mapping of connections in the intact brain TRENDS IN COGNITIVE SCIENCES Kim, S., Chung, K., Deisseroth, K. 2013; 17 (12): 596-599

    Abstract

    Mapping of neural connectivity across the mammalian brain is a daunting and exciting prospect. Current approaches can be divided into three classes: macroscale, focusing on coarse inter-regional connectivity; mesoscale, involving a finer focus on neurons and projections; and microscale, reconstructing full details of all synaptic contacts. It remains to be determined how to bridge the datasets or insights from the different levels of study. Here we review recent light-microscopy-based approaches that may help in integration across scales.

    View details for DOI 10.1016/j.tics.2013.10.005

    View details for Web of Science ID 000328719600002

    View details for PubMedID 24210964

  • Cerebellar Purkinje cell activity drives motor learning. Nature neuroscience Nguyen-Vu, T. D., Kimpo, R. R., Rinaldi, J. M., Kohli, A., Zeng, H., Deisseroth, K., Raymond, J. L. 2013; 16 (12): 1734-1736

    Abstract

    The climbing fiber input to the cerebellar cortex is thought to provide instructive signals that drive the induction of motor skill learning. We found that optogenetic activation of Purkinje cells, the sole output neurons of the cerebellar cortex, can also drive motor learning in mice. This dual control over the induction of learning by climbing fibers and Purkinje cells can expand the learning capacity of motor circuits.

    View details for DOI 10.1038/nn.3576

    View details for PubMedID 24162651

  • Next-generation transgenic mice for optogenetic analysis of neural circuits analysis of neural circuits FRONTIERS IN NEURAL CIRCUITS Asrican, B., Augustine, G. J., Berglund, K., Chen, S., Chow, N., Deisseroth, K., Feng, G., Gloss, B., Hira, R., Hoffmann, C., Kasai, H., Katarya, M., Kim, J., Kudolo, J., Lee, L. M., Lo, S. Q., Mancuso, J., Matsuzaki, M., Nakajima, R., Qiu, L., Tan, G., Tang, Y., Ting, J. T., Tsuda, S., Wen, L., Zhang, X., Zhao, S. 2013; 7

    Abstract

    Here we characterize several new lines of transgenic mice useful for optogenetic analysis of brain circuit function. These mice express optogenetic probes, such as enhanced halorhodopsin or several different versions of channelrhodopsins, behind various neuron-specific promoters. These mice permit photoinhibition or photostimulation both in vitro and in vivo. Our results also reveal the important influence of fluorescent tags on optogenetic probe expression and function in transgenic mice.

    View details for DOI 10.3389/fncir.2013.00160

    View details for Web of Science ID 000328276300001

    View details for PubMedCentralID PMC3840435

  • A Unique Population of Ventral Tegmental Area Neurons Inhibits the Lateral Habenula to Promote Reward NEURON Stamatakis, A. M., Jennings, J. H., Ung, R. L., Blair, G. A., Weinberg, R. J., Neve, R. L., Boyce, F., Mattis, J., Ramakrishnan, C., Deisseroth, K., Stuber, G. D. 2013; 80 (4): 1039-1053

    Abstract

    Lateral habenula (LHb) neurons convey aversive and negative reward conditions through potent indirect inhibition of ventral tegmental area (VTA) dopaminergic neurons. Although VTA dopaminergic neurons reciprocally project to the LHb, the electrophysiological properties and the behavioral consequences associated with selective manipulations of this circuit are unknown. Here, we identify an inhibitory input to the LHb arising from a unique population of VTA neurons expressing dopaminergic markers. Optogenetic activation of this circuit resulted in no detectable dopamine release in LHb brain slices. Instead, stimulation produced GABA-mediated inhibitory synaptic transmission, which suppressed the firing of postsynaptic LHb neurons in brain slices and increased the spontaneous firing rate of VTA dopaminergic neurons in vivo. Furthermore, in vivo activation of this pathway produced reward-related phenotypes that were dependent on intra-LHb GABAA receptor signaling. These results suggest that noncanonical inhibitory signaling by these hybrid dopaminergic-GABAergic neurons act to suppress LHb output under rewarding conditions.

    View details for DOI 10.1016/j.neuron.2013.08.023

    View details for Web of Science ID 000327281200019

    View details for PubMedID 24267654

    View details for PubMedCentralID PMC3873746

  • Optogenetic Activation of an Inhibitory Network Enhances Feedforward Functional Connectivity in Auditory Cortex NEURON Hamilton, L. S., Sohl-Dickstein, J., Huth, A. G., Carels, V. M., Deisseroth, K., Bao, S. 2013; 80 (4): 1066-1076

    Abstract

    The mammalian neocortex is a highly interconnected network of different types of neurons organized into both layers and columns. Overlaid on this structural organization is a pattern of functional connectivity that can be rapidly and flexibly altered during behavior. Parvalbumin-positive (PV+) inhibitory neurons, which are implicated in cortical oscillations and can change neuronal selectivity, may play a pivotal role in these dynamic changes. We found that optogenetic activation of PV+ neurons in the auditory cortex enhanced feedforward functional connectivity in the putative thalamorecipient circuit and in cortical columnar circuits. In contrast, stimulation of PV+ neurons induced no change in connectivity between sites in the same layers. The activity of PV+ neurons may thus serve as a gating mechanism to enhance feedforward, but not lateral or feedback, information flow in cortical circuits. Functionally, it may preferentially enhance the contribution of bottom-up sensory inputs to perception.

    View details for DOI 10.1016/j.neuron.2013.08.017

    View details for Web of Science ID 000327281200021

    View details for PubMedID 24267655

    View details for PubMedCentralID PMC3841078

  • Ventromedial Prefrontal Cortex Pyramidal Cells Have a Temporal Dynamic Role in Recall and Extinction of Cocaine-Associated Memory JOURNAL OF NEUROSCIENCE Van den Oever, M. C., Rotaru, D. C., Heinsbroek, J. A., Gouwenberg, Y., Deisseroth, K., Stuber, G. D., Mansvelder, H. D., Smit, A. B. 2013; 33 (46): 18225-18233

    Abstract

    In addicts, associative memories related to the rewarding effects of drugs of abuse can evoke powerful craving and drug seeking urges, but effective treatment to suppress these memories is not available. Detailed insight into the neural circuitry that mediates expression of drug-associated memory is therefore of crucial importance. Substantial evidence from rodent models of addictive behavior points to the involvement of the ventromedial prefrontal cortex (vmPFC) in conditioned drug seeking, but specific knowledge of the temporal role of vmPFC pyramidal cells is lacking. To this end, we used an optogenetics approach to probe the involvement of vmPFC pyramidal cells in expression of a recent and remote conditioned cocaine memory. In mice, we expressed Channelrhodopsin-2 (ChR2) or Halorhodopsin (eNpHR3.0) in pyramidal cells of the vmPFC and studied the effect of activation or inhibition of these cells during expression of a cocaine-contextual memory on days 1-2 (recent) and ∼3 weeks (remote) after conditioning. Whereas optical activation of pyramidal cells facilitated extinction of remote memory, without affecting recent memory, inhibition of pyramidal cells acutely impaired recall of recent cocaine memory, without affecting recall of remote memory. In addition, we found that silencing pyramidal cells blocked extinction learning at the remote memory time-point. We provide causal evidence of a critical time-dependent switch in the contribution of vmPFC pyramidal cells to recall and extinction of cocaine-associated memory, indicating that the circuitry that controls expression of cocaine memories reorganizes over time.

    View details for DOI 10.1523/JNEUROSCI.2412-13.2013

    View details for Web of Science ID 000327020600024

    View details for PubMedID 24227731

    View details for PubMedCentralID PMC3828471

  • Genetically encoded voltage sensor goes live. Nature biotechnology Marshel, J. H., Deisseroth, K. 2013; 31 (11): 994-995

    View details for DOI 10.1038/nbt.2738

    View details for PubMedID 24213775

  • Wave optics theory and 3-D deconvolution for the light field microscope OPTICS EXPRESS Broxton, M., Grosenick, L., Yang, S., Cohen, N., Andalman, A., Deisseroth, K., Levoy, M. 2013; 21 (21): 25418-25439

    Abstract

    Light field microscopy is a new technique for high-speed volumetric imaging of weakly scattering or fluorescent specimens. It employs an array of microlenses to trade off spatial resolution against angular resolution, thereby allowing a 4-D light field to be captured using a single photographic exposure without the need for scanning. The recorded light field can then be used to computationally reconstruct a full volume. In this paper, we present an optical model for light field microscopy based on wave optics, instead of previously reported ray optics models. We also present a 3-D deconvolution method for light field microscopy that is able to reconstruct volumes at higher spatial resolution, and with better optical sectioning, than previously reported. To accomplish this, we take advantage of the dense spatio-angular sampling provided by a microlens array at axial positions away from the native object plane. This dense sampling permits us to decode aliasing present in the light field to reconstruct high-frequency information. We formulate our method as an inverse problem for reconstructing the 3-D volume, which we solve using a GPU-accelerated iterative algorithm. Theoretical limits on the depth-dependent lateral resolution of the reconstructed volumes are derived. We show that these limits are in good agreement with experimental results on a standard USAF 1951 resolution target. Finally, we present 3-D reconstructions of pollen grains that demonstrate the improvements in fidelity made possible by our method.

    View details for DOI 10.1364/OE.21.025418

    View details for Web of Science ID 000326085600097

    View details for PubMedID 24150383

    View details for PubMedCentralID PMC3867103

  • Optogenetics. Proceedings of the National Academy of Sciences of the United States of America Williams, S. C., Deisseroth, K. 2013; 110 (41): 16287-?

    View details for DOI 10.1073/pnas.1317033110

    View details for PubMedID 24106299

  • VENTROMEDIAL PREFRONTAL CORTEX PYRAMIDAL CELLS HAVE A TEMPORAL DYNAMIC ROLE IN RECALL AND EXTINCTION OF COCAINE-ASSOCIATED MEMORY van den Oever, M., Rotaru, D., Heinsbroek, J., Gouwenberga, Y., Deisseroth, K., Mansvelder, H., Smit, A. LIPPINCOTT WILLIAMS & WILKINS. 2013: E33
  • A coaxial optrode as multifunction write-read probe for optogenetic studies in non-human primates. Journal of neuroscience methods Ozden, I., Wang, J., Lu, Y., May, T., Lee, J., Goo, W., O'Shea, D. J., Kalanithi, P., Diester, I., Diagne, M., Deisseroth, K., Shenoy, K. V., Nurmikko, A. V. 2013; 219 (1): 142-154

    Abstract

    Advances in optogenetics have led to first reports of expression of light-gated ion-channels in non-human primates (NHPs). However, a major obstacle preventing effective application of optogenetics in NHPs and translation to optogenetic therapeutics is the absence of compatible multifunction optoelectronic probes for (1) precision light delivery, (2) low-interference electrophysiology, (3) protein fluorescence detection, and (4) repeated insertion with minimal brain trauma.Here we describe a novel brain probe device, a "coaxial optrode", designed to minimize brain tissue damage while microfabricated to perform simultaneous electrophysiology, light delivery and fluorescence measurements in the NHP brain. The device consists of a tapered, gold-coated optical fiber inserted in a polyamide tube. A portion of the gold coating is exposed at the fiber tip to allow electrophysiological recordings in addition to light delivery/collection at the tip.Coaxial optrode performance was demonstrated by experiments in rodents and NHPs, and characterized by computational models. The device mapped opsin expression in the brain and achieved precisely targeted optical stimulation and electrophysiology with minimal cortical damage.Overall, combined electrical, optical and mechanical features of the coaxial optrode allowed a performance for NHP studies which was not possible with previously existing devices.Coaxial optrode is currently being used in two NHP laboratories as a major tool to study brain function by inducing light modulated neural activity and behavior. By virtue of its design, the coaxial optrode can be extended for use as a chronic implant and multisite neural stimulation/recording.

    View details for DOI 10.1016/j.jneumeth.2013.06.011

    View details for PubMedID 23867081

    View details for PubMedCentralID PMC3789534

  • GABAergic projection neurons route selective olfactory inputs to specific higher-order neurons. Neuron Liang, L., Li, Y., Potter, C. J., Yizhar, O., Deisseroth, K., Tsien, R. W., Luo, L. 2013; 79 (5): 917-931

    Abstract

    We characterize an inhibitory circuit motif in the Drosophila olfactory system, parallel inhibition, which differs from feedforward or feedback inhibition. Excitatory and GABAergic inhibitory projection neurons (ePNs and iPNs) each receive input from antennal lobe glomeruli and send parallel output to the lateral horn, a higher center implicated in regulating innate olfactory behavior. Ca(2+) imaging of specific lateral horn neurons as an olfactory readout revealed that iPNs selectively suppressed food-related odor responses, but spared signal transmission from pheromone channels. Coapplying food odorant did not affect pheromone signal transmission, suggesting that the differential effects likely result from connection specificity of iPNs, rather than a generalized inhibitory tone. Ca(2+) responses in the ePN axon terminals show no detectable suppression by iPNs, arguing against presynaptic inhibition as a primary mechanism. The parallel inhibition motif may provide specificity in inhibition to funnel specific olfactory information, such as food and pheromone, into distinct downstream circuits.

    View details for DOI 10.1016/j.neuron.2013.06.014

    View details for PubMedID 24012005

  • Arc/Arg3.1 Is a Postsynaptic Mediator of Activity-Dependent Synapse Elimination in the Developing Cerebellum NEURON Mikuni, T., Uesaka, N., Okuno, H., Hirai, H., Deisseroth, K., Bito, H., Kano, M. 2013; 78 (6): 1024-1035

    Abstract

    Neural circuits are shaped by activity-dependent elimination of redundant synapses during postnatal development. In many systems, postsynaptic activity is known to be crucial, but the precise mechanisms remain elusive. Here, we report that the immediate early gene Arc/Arg3.1 mediates elimination of surplus climbing fiber (CF) to Purkinje cell (PC) synapses in the developing cerebellum. CF synapse elimination was accelerated when activity of channelrhodopsin-2-expressing PCs was elevated by 2-day photostimulation. This acceleration was suppressed by PC-specific knockdown of either the P/Q-type voltage-dependent Ca(2+) channels (VDCCs) or Arc. PC-specific Arc knockdown had no appreciable effect until around postnatal day 11 but significantly impaired CF synapse elimination thereafter, leaving redundant CF terminals on PC somata. The effect of Arc knockdown was occluded by simultaneous knockdown of P/Q-type VDCCs in PCs. We conclude that Arc mediates the final stage of CF synapse elimination downstream of P/Q-type VDCCs by removing CF synapses from PC somata.

    View details for DOI 10.1016/j.neuron.2013.04.036

    View details for Web of Science ID 000321026900009

    View details for PubMedID 23791196

    View details for PubMedCentralID PMC3773328

  • Optical inhibition of motor nerve and muscle activity in vivo. Muscle & nerve Liske, H., Towne, C., Anikeeva, P., Zhao, S., Feng, G., Deisseroth, K., Delp, S. 2013; 47 (6): 916-921

    Abstract

    There is no therapeutic approach that provides precise and rapidly reversible inhibition of motor nerve and muscle activity for treatment of spastic hypertonia.We used optogenetics to demonstrate precise and rapidly reversible light-mediated inhibition of motor nerve and muscle activity in vivo in transgenic Thy1::eNpHR2.0 mice.We found optical inhibition of motor nerve and muscle activity to be effective at all muscle force amplitudes and determined that muscle activity can be modulated by changing light pulse duration and light power density.This demonstration of optical inhibition of motor nerves is an important advancement toward novel optogenetics-based therapies for spastic hypertonia.

    View details for DOI 10.1002/mus.23696

    View details for PubMedID 23629741

  • Recent advances in optogenetics and pharmacogenetics BRAIN RESEARCH Aston-Jones, G., Deisseroth, K. 2013; 1511: 1-5

    Abstract

    Optogenetics with microbial opsin genes, and pharmacogenetics with designer receptors, represent potent and versatile experimental modalities that can be integrated with each other as well as with a rich diversity of synergistic methods to provide fundamental opportunities in neuroscience research. The 7th Annual Brain Research Meeting in New Orleans in October 2012, Optogenetics and Pharmacogenetics in Neuronal Function and Dysfunction, brought together leading researchers that have developed and used these tools to explore a wide range of questions in nervous system function and dysfunction. This special issue of Brain Research includes articles by speakers in this meeting and others, which together synthesize and summarize the state of the art for optogenetics and designer receptors. This article is part of a Special Issue entitled Optogenetics (7th BRES).

    View details for DOI 10.1016/j.brainres.2013.01.026

    View details for Web of Science ID 000320087000001

    View details for PubMedID 23422677

    View details for PubMedCentralID PMC3663045

  • Structural and molecular interrogation of intact biological systems. Nature Chung, K., Wallace, J., Kim, S., Kalyanasundaram, S., Andalman, A. S., Davidson, T. J., Mirzabekov, J. J., Zalocusky, K. A., Mattis, J., Denisin, A. K., Pak, S., Bernstein, H., Ramakrishnan, C., Grosenick, L., Gradinaru, V., Deisseroth, K. 2013; 497 (7449): 332-337

    Abstract

    Obtaining high-resolution information from a complex system, while maintaining the global perspective needed to understand system function, represents a key challenge in biology. Here we address this challenge with a method (termed CLARITY) for the transformation of intact tissue into a nanoporous hydrogel-hybridized form (crosslinked to a three-dimensional network of hydrophilic polymers) that is fully assembled but optically transparent and macromolecule-permeable. Using mouse brains, we show intact-tissue imaging of long-range projections, local circuit wiring, cellular relationships, subcellular structures, protein complexes, nucleic acids and neurotransmitters. CLARITY also enables intact-tissue in situ hybridization, immunohistochemistry with multiple rounds of staining and de-staining in non-sectioned tissue, and antibody labelling throughout the intact adult mouse brain. Finally, we show that CLARITY enables fine structural analysis of clinical samples, including non-sectioned human tissue from a neuropsychiatric-disease setting, establishing a path for the transmutation of human tissue into a stable, intact and accessible form suitable for probing structural and molecular underpinnings of physiological function and disease.

    View details for DOI 10.1038/nature12107

    View details for PubMedID 23575631

    View details for PubMedCentralID PMC4092167

  • Multiple Sources of Striatal Inhibition Are Differentially Affected in Huntington's Disease Mouse Models. journal of neuroscience Cepeda, C., Galvan, L., Holley, S. M., Rao, S. P., André, V. M., Botelho, E. P., Chen, J. Y., Watson, J. B., Deisseroth, K., Levine, M. S. 2013; 33 (17): 7393-7406

    Abstract

    In Huntington's disease (HD) mouse models, spontaneous inhibitory synaptic activity is enhanced in a subpopulation of medium-sized spiny neurons (MSNs), which could dampen striatal output. We examined the potential source(s) of increased inhibition using electrophysiological and optogenetic methods to assess feedback and feedforward inhibition in two transgenic mouse models of HD. Single whole-cell patch-clamp recordings demonstrated that increased GABA synaptic activity impinges principally on indirect pathway MSNs. Dual patch recordings between MSNs demonstrated reduced connectivity between MSNs in HD mice. However, while connectivity was strictly unidirectional in controls, in HD mice bidirectional connectivity occurred. Other sources of increased GABA activity in MSNs also were identified. Dual patch recordings from fast spiking (FS) interneuron-MSN pairs demonstrated greater but variable amplitude responses in MSNs. In agreement, selective optogenetic stimulation of parvalbumin-expressing, FS interneurons induced significantly larger amplitude MSN responses in HD compared with control mice. While there were no differences in responses of MSNs evoked by activating single persistent low-threshold spiking (PLTS) interneurons in recorded pairs, these interneurons fired more action potentials in both HD models, providing another source for increased frequency of spontaneous GABA synaptic activity in MSNs. Selective optogenetic stimulation of somatostatin-expressing, PLTS interneurons did not reveal any significant differences in responses of MSNs in HD mice. These findings provide strong evidence that both feedforward and to a lesser extent feedback inhibition to MSNs in HD can potentially be sources for the increased GABA synaptic activity of indirect pathway MSNs.

    View details for DOI 10.1523/JNEUROSCI.2137-12.2013

    View details for PubMedID 23616545

    View details for PubMedCentralID PMC3686572

  • Optogenetic Delay of Status Epilepticus Onset in an In Vivo Rodent Epilepsy Model PLOS ONE Sukhotinsky, I., Chan, A. M., Ahmed, O. J., Rao, V. R., Gradinaru, V., Ramakrishnan, C., Deisseroth, K., Majewska, A. K., Cash, S. S. 2013; 8 (4)

    Abstract

    Epilepsy is a devastating disease, currently treated with medications, surgery or electrical stimulation. None of these approaches is totally effective and our ability to control seizures remains limited and complicated by frequent side effects. The emerging revolutionary technique of optogenetics enables manipulation of the activity of specific neuronal populations in vivo with exquisite spatiotemporal resolution using light. We used optogenetic approaches to test the role of hippocampal excitatory neurons in the lithium-pilocarpine model of acute elicited seizures in awake behaving rats. Hippocampal pyramidal neurons were transduced in vivo with a virus carrying an enhanced halorhodopsin (eNpHR), a yellow light activated chloride pump, and acute seizure progression was then monitored behaviorally and electrophysiologically in the presence and absence of illumination delivered via an optical fiber. Inhibition of those neurons with illumination prior to seizure onset significantly delayed electrographic and behavioral initiation of status epilepticus, and altered the dynamics of ictal activity development. These results reveal an essential role of hippocampal excitatory neurons in this model of ictogenesis and illustrate the power of optogenetic approaches for elucidation of seizure mechanisms. This early success in controlling seizures also suggests future therapeutic avenues.

    View details for DOI 10.1371/journal.pone.0062013

    View details for Web of Science ID 000318340400068

    View details for PubMedID 23637949

    View details for PubMedCentralID PMC3634849

  • Making waves: initiation and propagation of corticothalamic Ca2+ waves in vivo. Neuron Stroh, A., Adelsberger, H., Groh, A., Rühlmann, C., Fischer, S., Schierloh, A., Deisseroth, K., Konnerth, A. 2013; 77 (6): 1136-1150

    Abstract

    Corticothalamic slow oscillations of neuronal activity determine internal brain states. At least in the cortex, the electrical activity is associated with large neuronal Ca(2+) transients. Here we implemented an optogenetic approach to explore causal features of the generation of slow oscillation-associated Ca(2+) waves in the in vivo mouse brain. We demonstrate that brief optogenetic stimulation (3-20 ms) of a local group of layer 5 cortical neurons is sufficient for the induction of global brain Ca(2+) waves. These Ca(2+) waves are evoked in an all-or-none manner, exhibit refractoriness during repetitive stimulation, and propagate over long distances. By local optogenetic stimulation, we demonstrate that evoked Ca(2+) waves initially invade the cortex, followed by a secondary recruitment of the thalamus. Together, our results establish that synchronous activity in a small cluster of layer 5 cortical neurons can initiate a global neuronal wave of activity suited for long-range corticothalamic integration.

    View details for DOI 10.1016/j.neuron.2013.01.031

    View details for PubMedID 23522048

  • The Brain Activity Map SCIENCE Alivisatos, A. P., Chun, M., Church, G. M., Deisseroth, K., Donoghue, J. P., Greenspan, R. J., McEuen, P. L., Roukes, M. L., Sejnowski, T. J., Weiss, P. S., Yuste, R. 2013; 339 (6125): 1284-1285
  • Neuroscience. The brain activity map. Science Alivisatos, A. P., Chun, M., Church, G. M., Deisseroth, K., Donoghue, J. P., Greenspan, R. J., McEuen, P. L., Roukes, M. L., Sejnowski, T. J., Weiss, P. S., Yuste, R. 2013; 339 (6125): 1284-1285

    View details for DOI 10.1126/science.1236939

    View details for PubMedID 23470729

  • Nanotools for Neuroscience and Brain Activity Mapping ACS NANO Alivisatos, A. P., Andrews, A. M., Boyden, E. S., Chun, M., Church, G. M., Deisseroth, K., Donoghue, J. P., Fraser, S. E., Lippincott-Schwartz, J., Looger, L. L., Masmanidis, S., McEuen, P. L., Nurmikko, A. V., Park, H., Peterka, D. S., Reid, C., Roukes, M. L., Scherer, A., Schnitzer, M., Sejnowski, T. J., Shepard, K. L., Tsao, D., Turrigiano, G., Weiss, P. S., Xu, C., Yuste, R., Zhuang, X. 2013; 7 (3): 1850-1866

    Abstract

    Neuroscience is at a crossroads. Great effort is being invested into deciphering specific neural interactions and circuits. At the same time, there exist few general theories or principles that explain brain function. We attribute this disparity, in part, to limitations in current methodologies. Traditional neurophysiological approaches record the activities of one neuron or a few neurons at a time. Neurochemical approaches focus on single neurotransmitters. Yet, there is an increasing realization that neural circuits operate at emergent levels, where the interactions between hundreds or thousands of neurons, utilizing multiple chemical transmitters, generate functional states. Brains function at the nanoscale, so tools to study brains must ultimately operate at this scale, as well. Nanoscience and nanotechnology are poised to provide a rich toolkit of novel methods to explore brain function by enabling simultaneous measurement and manipulation of activity of thousands or even millions of neurons. We and others refer to this goal as the Brain Activity Mapping Project. In this Nano Focus, we discuss how recent developments in nanoscale analysis tools and in the design and synthesis of nanomaterials have generated optical, electrical, and chemical methods that can readily be adapted for use in neuroscience. These approaches represent exciting areas of technical development and research. Moreover, unique opportunities exist for nanoscientists, nanotechnologists, and other physical scientists and engineers to contribute to tackling the challenging problems involved in understanding the fundamentals of brain function.

    View details for DOI 10.1021/nn4012847

    View details for Web of Science ID 000316846700005

    View details for PubMedID 23514423

    View details for PubMedCentralID PMC3665747

  • Posttraining optogenetic manipulations of basolateral amygdala activity modulate consolidation of inhibitory avoidance memory in rats PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Huff, M. L., Miller, R. L., Deisseroth, K., Moorman, D. E., LaLumiere, R. T. 2013; 110 (9): 3597-3602

    Abstract

    Memory consolidation studies, including those examining the role of the basolateral amygdala (BLA), have traditionally used techniques limited in their temporal and spatial precision. The development of optogenetics provides increased precision in the control of neuronal activity that can be used to address the temporal nature of the modulation of memory consolidation. The present experiments, therefore, investigated whether optogenetically stimulating and inhibiting BLA activity immediately after training on an inhibitory avoidance task enhances and impairs retention, respectively. The BLA of male Sprague-Dawley rats was transduced to express either ChR2(E123A) or archaerhodopsin-3 from the Halorubrum sodomense strain TP009 (ArchT). Immediately after inhibitory avoidance training, rats received optical stimulation or inhibition of the BLA, and 2 d later, rats' retention was tested. Stimulation of ChR2(E123A)-expressing neurons in the BLA using trains of 40-Hz light pulses enhanced retention, consistent with recording studies suggesting the importance of BLA activity at this frequency. Light pulses alone given to control rats had no effect on retention. Inhibition of ArchT-expressing neurons in the BLA for 15 min, but not 1 min, significantly impaired retention. Again, illumination alone given to control rats had no effect on retention, and BLA inhibition 3 h after training had no effect. These findings provide critical evidence of the importance of specific frequency patterns of activity in the BLA during consolidation and indicate that optogenetic manipulations can be used to alter activity after a learning event to investigate the processes underlying memory consolidation.

    View details for DOI 10.1073/pnas.1219593110

    View details for Web of Science ID 000315841900078

    View details for PubMedID 23401523

    View details for PubMedCentralID PMC3587230

  • Optogenetic Stimulation of Human Neural Stem Cell Grafts In Ischemic Brain Daadi, M., Lee, S., Bajar, B., Lo, M., Ramakrishnan, C., Sun, G., Deisseroth, K., Steinberg, G. LIPPINCOTT WILLIAMS & WILKINS. 2013
  • Optogenetic Stimulation Of Motor Cortex Neurons Promotes Recovery After Stroke Cheng, M. Y., Woodson, W. J., Wang, E. H., Wang, S., Sun, G., Lee, A. G., Arac, A., Fenno, L., Deisseroth, K., Steinberg, G. K. LIPPINCOTT WILLIAMS & WILKINS. 2013
  • Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons NATURE Chaudhury, D., Walsh, J. J., Friedman, A. K., Juarez, B., Ku, S. M., Koo, J. W., Ferguson, D., Tsai, H., Pomeranz, L., Christoffel, D. J., Nectow, A. R., Ekstrand, M., Domingos, A., Mazei-Robison, M. S., Mouzon, E., Lobo, M. K., Neve, R. L., Friedman, J. M., Russo, S. J., Deisseroth, K., Nestler, E. J., Han, M. 2013; 493 (7433): 532-?

    Abstract

    Ventral tegmental area (VTA) dopamine neurons in the brain's reward circuit have a crucial role in mediating stress responses, including determining susceptibility versus resilience to social-stress-induced behavioural abnormalities. VTA dopamine neurons show two in vivo patterns of firing: low frequency tonic firing and high frequency phasic firing. Phasic firing of the neurons, which is well known to encode reward signals, is upregulated by repeated social-defeat stress, a highly validated mouse model of depression. Surprisingly, this pathophysiological effect is seen in susceptible mice only, with no apparent change in firing rate in resilient individuals. However, direct evidence--in real time--linking dopamine neuron phasic firing in promoting the susceptible (depression-like) phenotype is lacking. Here we took advantage of the temporal precision and cell-type and projection-pathway specificity of optogenetics to show that enhanced phasic firing of these neurons mediates susceptibility to social-defeat stress in freely behaving mice. We show that optogenetic induction of phasic, but not tonic, firing in VTA dopamine neurons of mice undergoing a subthreshold social-defeat paradigm rapidly induced a susceptible phenotype as measured by social avoidance and decreased sucrose preference. Optogenetic phasic stimulation of these neurons also quickly induced a susceptible phenotype in previously resilient mice that had been subjected to repeated social-defeat stress. Furthermore, we show differences in projection-pathway specificity in promoting stress susceptibility: phasic activation of VTA neurons projecting to the nucleus accumbens (NAc), but not to the medial prefrontal cortex (mPFC), induced susceptibility to social-defeat stress. Conversely, optogenetic inhibition of the VTA-NAc projection induced resilience, whereas inhibition of the VTA-mPFC projection promoted susceptibility. Overall, these studies reveal novel firing-pattern- and neural-circuit-specific mechanisms of depression.

    View details for DOI 10.1038/nature11713

    View details for Web of Science ID 000313871400038

    View details for PubMedID 23235832

    View details for PubMedCentralID PMC3554860

  • Glutamatergic Neurotransmission between the C1 Neurons and the Parasympathetic Preganglionic Neurons of the Dorsal Motor Nucleus of the Vagus JOURNAL OF NEUROSCIENCE DePuy, S. D., Stornetta, R. L., Bochorishvili, G., Deisseroth, K., Witten, I., Coates, M., Guyenet, P. G. 2013; 33 (4): 1486-1497

    Abstract

    The C1 neurons are a nodal point for blood pressure control and other autonomic responses. Here we test whether these rostral ventrolateral medullary catecholaminergic (RVLM-CA) neurons use glutamate as a transmitter in the dorsal motor nucleus of the vagus (DMV). After injecting Cre-dependent adeno-associated virus (AAV2) DIO-Ef1α-channelrhodopsin2(ChR2)-mCherry (AAV2) into the RVLM of dopamine-β-hydroxylase Cre transgenic mice (DβH(Cre/0)), mCherry was detected exclusively in RVLM-CA neurons. Within the DMV >95% mCherry-immunoreactive(ir) axonal varicosities were tyrosine hydroxylase (TH)-ir and the same proportion were vesicular glutamate transporter 2 (VGLUT2)-ir. VGLUT2-mCherry colocalization was virtually absent when AAV2 was injected into the RVLM of DβH(Cre/0);VGLUT2(flox/flox) mice, into the caudal VLM (A1 noradrenergic neuron-rich region) of DβH(Cre/0) mice or into the raphe of ePet(Cre/0) mice. Following injection of AAV2 into RVLM of TH-Cre rats, phenylethanolamine N-methyl transferase and VGLUT2 immunoreactivities were highly colocalized in DMV within EYFP-positive or EYFP-negative axonal varicosities. Ultrastructurally, mCherry terminals from RVLM-CA neurons in DβH(Cre/0) mice made predominantly asymmetric synapses with choline acetyl-transferase-ir DMV neurons. Photostimulation of ChR2-positive axons in DβH(Cre/0) mouse brain slices produced EPSCs in 71% of tested DMV preganglionic neurons (PGNs) but no IPSCs. Photostimulation (20 Hz) activated PGNs up to 8 spikes/s (current-clamp). EPSCs were eliminated by tetrodotoxin, reinstated by 4-aminopyridine, and blocked by ionotropic glutamate receptor blockers. In conclusion, VGLUT2 is expressed by RVLM-CA (C1) neurons in rats and mice regardless of the presence of AAV2, the C1 neurons activate DMV parasympathetic PGNs monosynaptically and this connection uses glutamate as an ionotropic transmitter.

    View details for DOI 10.1523/JNEUROSCI.4269-12.2013

    View details for Web of Science ID 000313956900020

    View details for PubMedID 23345223

  • Optogenetic Inhibition of Dorsal Medial Prefrontal Cortex Attenuates Stress-Induced Reinstatement of Palatable Food Seeking in Female Rats JOURNAL OF NEUROSCIENCE Calu, D. J., Kawa, A. B., Marchant, N. J., Navarre, B. M., Henderson, M. J., Chen, B., Yau, H., Bossert, J. M., Schoenbaum, G., Deisseroth, K., Harvey, B. K., Hope, B. T., Shaham, Y. 2013; 33 (1): 214-U626

    Abstract

    Relapse to maladaptive eating habits during dieting is often provoked by stress. Recently, we identified a role of dorsal medial prefrontal cortex (mPFC) neurons in stress-induced reinstatement of palatable food seeking in male rats. It is unknown whether endogenous neural activity in dorsal mPFC drives stress-induced reinstatement in female rats. Here, we used an optogenetic approach, in which female rats received bilateral dorsal mPFC microinjections of viral constructs coding light-sensitive eNpHR3.0-eYFP or control eYFP protein and intracranial fiber optic implants. Rats were food restricted and trained to lever press for palatable food pellets. Subsequently, pellets were removed, and lever pressing was extinguished; then the effect of bilateral dorsal mPFC light delivery on reinstatement of food seeking was assessed after injections of the pharmacological stressor yohimbine (an α-2 andrenoceptor antagonist) or pellet priming, a manipulation known to provoke food seeking in hungry rats. Dorsal mPFC light delivery attenuated yohimbine-induced reinstatement of food seeking in eNpHR3.0-injected but not eYFP-injected rats. This optical manipulation had no effect on pellet-priming-induced reinstatement or ongoing food-reinforced responding. Dorsal mPFC light delivery attenuated yohimbine-induced Fos immunoreactivity and disrupted neural activity during in vivo electrophysiological recording in awake rats. Optical stimulation caused significant outward currents and blocked electrically evoked action potentials in eNpHR3.0-injected but not eYFP-injected mPFC hemispheres. Light delivery alone caused no significant inflammatory response in mPFC. These findings indicate that intracranial light delivery in eNpHR3.0 rats disrupts endogenous dorsal mPFC neural activity that plays a role in stress-induced relapse to food seeking in female rats.

    View details for DOI 10.1523/JNEUROSCI.2016-12.2013

    View details for Web of Science ID 000313046500021

    View details for PubMedID 23283335

  • Closed-loop optogenetic control of thalamus as a tool for interrupting seizures after cortical injury NATURE NEUROSCIENCE Paz, J. T., Davidson, T. J., Frechette, E. S., Delord, B., Parada, I., Peng, K., Deisseroth, K., Huguenard, J. R. 2013; 16 (1): 64-U98

    Abstract

    Cerebrocortical injuries such as stroke are a major source of disability. Maladaptive consequences can result from post-injury local reorganization of cortical circuits. For example, epilepsy is a common sequela of cortical stroke, but the mechanisms responsible for seizures following cortical injuries remain unknown. In addition to local reorganization, long-range, extra-cortical connections might be critical for seizure maintenance. In rats, we found that the thalamus, a structure that is remote from, but connected to, the injured cortex, was required to maintain cortical seizures. Thalamocortical neurons connected to the injured epileptic cortex underwent changes in HCN channel expression and became hyperexcitable. Targeting these neurons with a closed-loop optogenetic strategy revealed that reducing their activity in real-time was sufficient to immediately interrupt electrographic and behavioral seizures. This approach is of therapeutic interest for intractable epilepsy, as it spares cortical function between seizures, in contrast with existing treatments, such as surgical lesioning or drugs.

    View details for DOI 10.1038/nn.3269

    View details for Web of Science ID 000312633900014

    View details for PubMedID 23143518

  • Optogenetic control of targeted peripheral axons in freely moving animals. PloS one Towne, C., Montgomery, K. L., Iyer, S. M., Deisseroth, K., Delp, S. L. 2013; 8 (8)

    Abstract

    Optogenetic control of the peripheral nervous system (PNS) would enable novel studies of motor control, somatosensory transduction, and pain processing. Such control requires the development of methods to deliver opsins and light to targeted sub-populations of neurons within peripheral nerves. We report here methods to deliver opsins and light to targeted peripheral neurons and robust optogenetic modulation of motor neuron activity in freely moving, non-transgenic mammals. We show that intramuscular injection of adeno-associated virus serotype 6 enables expression of channelrhodopsin (ChR2) in motor neurons innervating the injected muscle. Illumination of nerves containing mixed populations of axons from these targeted neurons and from neurons innervating other muscles produces ChR2-mediated optogenetic activation restricted to the injected muscle. We demonstrate that an implanted optical nerve cuff is well-tolerated, delivers light to the sciatic nerve, and optically stimulates muscle in freely moving rats. These methods can be broadly applied to study PNS disorders and lay the groundwork for future therapeutic application of optogenetics.

    View details for DOI 10.1371/journal.pone.0072691

    View details for PubMedID 23991144

  • The Open Connectome Project Data Cluster: Scalable Analysis and Vision for High-Throughput Neuroscience. Scientific and statistical database management : International Conference, SSDBM ... : proceedings. International Conference on Scientific and Statistical Database Management Burns, R., Roncal, W. G., Kleissas, D., Lillaney, K., Manavalan, P., Perlman, E., Berger, D. R., Bock, D. D., Chung, K., Grosenick, L., Kasthuri, N., Weiler, N. C., Deisseroth, K., Kazhdan, M., Lichtman, J., Reid, R. C., Smith, S. J., Szalay, A. S., Vogelstein, J. T., Vogelstein, R. J. 2013

    Abstract

    We describe a scalable database cluster for the spatial analysis and annotation of high-throughput brain imaging data, initially for 3-d electron microscopy image stacks, but for time-series and multi-channel data as well. The system was designed primarily for workloads that build connectomes- neural connectivity maps of the brain-using the parallel execution of computer vision algorithms on high-performance compute clusters. These services and open-science data sets are publicly available at openconnecto.me. The system design inherits much from NoSQL scale-out and data-intensive computing architectures. We distribute data to cluster nodes by partitioning a spatial index. We direct I/O to different systems-reads to parallel disk arrays and writes to solid-state storage-to avoid I/O interference and maximize throughput. All programming interfaces are RESTful Web services, which are simple and stateless, improving scalability and usability. We include a performance evaluation of the production system, highlighting the effec-tiveness of spatial data organization.

    View details for PubMedID 24401992

  • Next-generation transgenic mice for optogenetic analysis of neural circuits. Frontiers in neural circuits Asrican, B., Augustine, G. J., Berglund, K., Chen, S., Chow, N., Deisseroth, K., Feng, G., Gloss, B., Hira, R., Hoffmann, C., Kasai, H., Katarya, M., Kim, J., Kudolo, J., Lee, L. M., Lo, S. Q., Mancuso, J., Matsuzaki, M., Nakajima, R., Qiu, L., Tan, G., Tang, Y., Ting, J. T., Tsuda, S., Wen, L., Zhang, X., Zhao, S. 2013; 7: 160-?

    Abstract

    Here we characterize several new lines of transgenic mice useful for optogenetic analysis of brain circuit function. These mice express optogenetic probes, such as enhanced halorhodopsin or several different versions of channelrhodopsins, behind various neuron-specific promoters. These mice permit photoinhibition or photostimulation both in vitro and in vivo. Our results also reveal the important influence of fluorescent tags on optogenetic probe expression and function in transgenic mice.

    View details for DOI 10.3389/fncir.2013.00160

    View details for PubMedID 24324405

    View details for PubMedCentralID PMC3840435

  • A precise and minimally invasive approach to optogenetics in the awake primate Conference on Optogenetics - Optical Methods for Cellular Control Nassi, J. J., Cetin, A. H., Roe, A. W., Callaway, E. M., Deisseroth, K., Reynolds, J. H. SPIE-INT SOC OPTICAL ENGINEERING. 2013

    View details for DOI 10.1117/12.2007609

    View details for Web of Science ID 000322901400004

  • Optical control of neuronal excitation and inhibition using a single opsin protein, ChR2. Scientific reports Liske, H., Qian, X., Anikeeva, P., Deisseroth, K., Delp, S. 2013; 3: 3110-?

    Abstract

    The effect of electrical stimulation on neuronal membrane potential is frequency dependent. Low frequency electrical stimulation can evoke action potentials, whereas high frequency stimulation can inhibit action potential transmission. Optical stimulation of channelrhodopsin-2 (ChR2) expressed in neuronal membranes can also excite action potentials. However, it is unknown whether optical stimulation of ChR2-expressing neurons produces a transition from excitation to inhibition with increasing light pulse frequencies. Here we report optical inhibition of motor neuron and muscle activity in vivo in the cooled sciatic nerves of Thy1-ChR2-EYFP mice. We also demonstrate all-optical single-wavelength control of neuronal excitation and inhibition without co-expression of inhibitory and excitatory opsins. This all-optical system is free from stimulation-induced electrical artifacts and thus provides a new approach to investigate mechanisms of high frequency inhibition in neuronal circuits in vivo and in vitro.

    View details for DOI 10.1038/srep03110

    View details for PubMedID 24173561

    View details for PubMedCentralID PMC3813941

  • Optogenetic control of targeted peripheral axons in freely moving animals. PloS one Towne, C., Montgomery, K. L., Iyer, S. M., Deisseroth, K., Delp, S. L. 2013; 8 (8): e72691

    Abstract

    Optogenetic control of the peripheral nervous system (PNS) would enable novel studies of motor control, somatosensory transduction, and pain processing. Such control requires the development of methods to deliver opsins and light to targeted sub-populations of neurons within peripheral nerves. We report here methods to deliver opsins and light to targeted peripheral neurons and robust optogenetic modulation of motor neuron activity in freely moving, non-transgenic mammals. We show that intramuscular injection of adeno-associated virus serotype 6 enables expression of channelrhodopsin (ChR2) in motor neurons innervating the injected muscle. Illumination of nerves containing mixed populations of axons from these targeted neurons and from neurons innervating other muscles produces ChR2-mediated optogenetic activation restricted to the injected muscle. We demonstrate that an implanted optical nerve cuff is well-tolerated, delivers light to the sciatic nerve, and optically stimulates muscle in freely moving rats. These methods can be broadly applied to study PNS disorders and lay the groundwork for future therapeutic application of optogenetics.

    View details for DOI 10.1371/journal.pone.0072691

    View details for PubMedID 23991144

    View details for PubMedCentralID PMC3749160

  • Cortico-Striatal Stimulation Generates Persistent OCD-Like Behavior. Science. Ahmari, S. E., Spellman, T., Douglass, N. L., Kheirbek, M. A., Simpson, H. B., Deisseroth, K. 2013; 340: 1234-9
  • A causal link between prediction errors, dopamine neurons and learning Nature Neuroscience. Advance Online Pulbication Steinberg, E. E., Keiflin, R., Boivin, J. R., Witten, I. B., Deisseroth, K., Janak, P, H. 2013
  • Engineering approaches to illuminating brain structure and dynamics. Neuron. Deisseroth, K., Schnitzer, Mark, J. 2013
  • Causal interactions between fronto-parietal central executive and default-mode networks in humans. PNAS. Chen, Ashley, C., Oathes, Desmond, J., Chang, C., Bradley, T., Zhou, Z., Williams, Leanne, M., Deisseroth, K. 2013
  • Optogenetic inhibition of cocaine seeking in rats ADDICTION BIOLOGY Stefanik, M. T., Moussawi, K., Kupchik, Y. M., Smith, K. C., Miller, R. L., Huff, M. L., Deisseroth, K., Kalivas, P. W., LaLumiere, R. T. 2013; 18 (1): 50-53

    Abstract

    Inhibitory optogenetics was used to examine the roles of the prelimbic cortex (PL), the nucleus accumbens core (NAcore) and the PL projections to the NAcore in the reinstatement of cocaine seeking. Rats were microinjected into the PL or NAcore with an adeno-associated virus containing halorhodopsin or archaerhodopsin. After 12 days of cocaine self-administration, followed by extinction training, animals underwent reinstatement testing along with the presence/absence of optically induced inhibition via laser light. Bilateral optical inhibition of the PL, NAcore or the PL fibers in the NAcore inhibited the reinstatement of cocaine seeking.

    View details for DOI 10.1111/j.1369-1600.2012.00479.x

    View details for Web of Science ID 000312740500006

    View details for PubMedID 22823160

    View details for PubMedCentralID PMC3578202

  • Prefrontal D1 dopamine signaling is required for temporal control PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Narayanan, N. S., Land, B. B., Solder, J. E., Deisseroth, K., DiLeone, R. J. 2012; 109 (50): 20726-20731

    Abstract

    Temporal control, or how organisms guide movements in time to achieve behavioral goals, depends on dopamine signaling. The medial prefrontal cortex controls many goal-directed behaviors and receives dopaminergic input primarily from the midbrain ventral tegmental area. However, this system has never been linked with temporal control. Here, we test the hypothesis that dopaminergic projections from the ventral tegmental area to the prefrontal cortex influence temporal control. Rodents were trained to perform a fixed-interval timing task with an interval of 20 s. We report several results: first, that decreasing dopaminergic neurotransmission using virally mediated RNA interference of tyrosine hydroxylase impaired temporal control, and second that pharmacological disruption of prefrontal D1 dopamine receptors, but not D2 dopamine receptors, impaired temporal control. We then used optogenetics to specifically and selectively manipulate prefrontal neurons expressing D1 dopamine receptors during fixed-interval timing performance. Selective inhibition of D1-expressing prefrontal neurons impaired fixed-interval timing, whereas stimulation made animals more efficient during task performance. These data provide evidence that ventral tegmental dopaminergic projections to the prefrontal cortex influence temporal control via D1 receptors. The results identify a critical circuit for temporal control of behavior that could serve as a target for the treatment of dopaminergic diseases.

    View details for DOI 10.1073/pnas.1211258109

    View details for Web of Science ID 000312605600114

    View details for PubMedID 23185016

    View details for PubMedCentralID PMC3528521

  • Two-photon optogenetic toolbox for fast inhibition, excitation and bistable modulation NATURE METHODS Prakash, R., Yizhar, O., Grewe, B., Ramakrishnan, C., Wang, N., Goshen, I., Packer, A. M., Peterka, D. S., Yuste, R., Schnitzer, M. J., Deisseroth, K. 2012; 9 (12): 1171-U132

    Abstract

    Optogenetics with microbial opsin genes has enabled high-speed control of genetically specified cell populations in intact tissue. However, it remains a challenge to independently control subsets of cells within the genetically targeted population. Although spatially precise excitation of target molecules can be achieved using two-photon laser-scanning microscopy (TPLSM) hardware, the integration of two-photon excitation with optogenetics has thus far required specialized equipment or scanning and has not yet been widely adopted. Here we take a complementary approach, developing opsins with custom kinetic, expression and spectral properties uniquely suited to scan times typical of the raster approach that is ubiquitous in TPLSMlaboratories. We use a range of culture, slice and mammalian in vivo preparations to demonstrate the versatility of this toolbox, and we quantitatively map parameter space for fast excitation, inhibition and bistable control. Together these advances may help enable broad adoption of integrated optogenetic and TPLSMtechnologies across experimental fields and systems.

    View details for DOI 10.1038/NMETH.2215

    View details for PubMedID 23169303

  • Two-photon optogenetics of dendritic spines and neural circuits NATURE METHODS Packer, A. M., Peterka, D. S., Hirtz, J. J., Prakash, R., Deisseroth, K., Yuste, R. 2012; 9 (12): 1202-U103

    Abstract

    We demonstrate a two-photon optogenetic method that generates action potentials in neurons with single-cell precision, using the red-shifted opsin C1V1(T). We applied the method to optically map synaptic circuits in mouse neocortical brain slices and to activate small dendritic regions and individual spines. Using a spatial light modulator, we split the laser beam onto several neurons and performed simultaneous optogenetic activation of selected neurons in three dimensions.

    View details for DOI 10.1038/NMETH.2249

    View details for Web of Science ID 000312093500025

    View details for PubMedID 23142873

    View details for PubMedCentralID PMC3518588

  • Optogenetic and Potassium Channel Gene Therapy in a Rodent Model of Focal Neocortical Epilepsy SCIENCE TRANSLATIONAL MEDICINE Wykes, R. C., Heeroma, J. H., Mantoan, L., Zheng, K., Macdonald, D. C., Deisseroth, K., Hashemi, K. S., Walker, M. C., Schorge, S., Kullmann, D. M. 2012; 4 (161)

    Abstract

    Neocortical epilepsy is frequently drug-resistant. Surgery to remove the epileptogenic zone is only feasible in a minority of cases, leaving many patients without an effective treatment. We report the potential efficacy of gene therapy in focal neocortical epilepsy using a rodent model in which epilepsy is induced by tetanus toxin injection in the motor cortex. By applying several complementary methods that use continuous wireless electroencephalographic monitoring to quantify epileptic activity, we observed increases in high frequency activity and in the occurrence of epileptiform events. Pyramidal neurons in the epileptic focus showed enhanced intrinsic excitability consistent with seizure generation. Optogenetic inhibition of a subset of principal neurons transduced with halorhodopsin targeted to the epileptic focus by lentiviral delivery was sufficient to attenuate electroencephalographic seizures. Local lentiviral overexpression of the potassium channel Kv1.1 reduced the intrinsic excitability of transduced pyramidal neurons. Coinjection of this Kv1.1 lentivirus with tetanus toxin fully prevented the occurrence of electroencephalographic seizures. Finally, administration of the Kv1.1 lentivirus to an established epileptic focus progressively suppressed epileptic activity over several weeks without detectable behavioral side effects. Thus, gene therapy in a rodent model can be used to suppress seizures acutely, prevent their occurrence after an epileptogenic stimulus, and successfully treat established focal epilepsy.

    View details for DOI 10.1126/scitranslmed.3004190

    View details for Web of Science ID 000311426900005

    View details for PubMedID 23147003

    View details for PubMedCentralID PMC3605784

  • Reversible online control of habitual behavior by optogenetic perturbation of medial prefrontal cortex PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Smith, K. S., Virkud, A., Deisseroth, K., Graybiel, A. M. 2012; 109 (46): 18932-18937

    Abstract

    Habits tend to form slowly but, once formed, can have great stability. We probed these temporal characteristics of habitual behaviors by intervening optogenetically in forebrain habit circuits as rats performed well-ingrained habitual runs in a T-maze. We trained rats to perform a maze habit, confirmed the habitual behavior by devaluation tests, and then, during the maze runs (ca. 3 s), we disrupted population activity in a small region in the medial prefrontal cortex, the infralimbic cortex. In accordance with evidence that this region is necessary for the expression of habits, we found that this cortical disruption blocked habitual behavior. Notably, however, this blockade of habitual performance occurred on line, within an average of three trials (ca. 9 s of inhibition), and as soon as during the first trial (<3 s). During subsequent weeks of training, the rats acquired a new behavioral pattern. When we again imposed the same cortical perturbation, the rats regained the suppressed maze-running that typified the original habit, and, simultaneously, the more recently acquired habit was blocked. These online changes occurred within an average of two trials (ca. 6 s of infralimbic inhibition). Measured changes in generalized performance ability and motivation to consume reward were unaffected. This immediate toggling between breaking old habits and returning to them demonstrates that even semiautomatic behaviors are under cortical control and that this control occurs online, second by second. These temporal characteristics define a framework for uncovering cellular transitions between fixed and flexible behaviors, and corresponding disturbances in pathologies.

    View details for DOI 10.1073/pnas.1216264109

    View details for Web of Science ID 000311576300064

    View details for PubMedID 23112197

    View details for PubMedCentralID PMC3503190

  • Input-specific control of reward and aversion in the ventral tegmental area NATURE Lammel, S., Lim, B. K., Ran, C., Huang, K. W., Betley, M. J., Tye, K. M., Deisseroth, K., Malenka, R. C. 2012; 491 (7423): 212-?

    Abstract

    Ventral tegmental area (VTA) dopamine neurons have important roles in adaptive and pathological brain functions related to reward and motivation. However, it is unknown whether subpopulations of VTA dopamine neurons participate in distinct circuits that encode different motivational signatures, and whether inputs to the VTA differentially modulate such circuits. Here we show that, because of differences in synaptic connectivity, activation of inputs to the VTA from the laterodorsal tegmentum and the lateral habenula elicit reward and aversion in mice, respectively. Laterodorsal tegmentum neurons preferentially synapse on dopamine neurons projecting to the nucleus accumbens lateral shell, whereas lateral habenula neurons synapse primarily on dopamine neurons projecting to the medial prefrontal cortex as well as on GABAergic (γ-aminobutyric-acid-containing) neurons in the rostromedial tegmental nucleus. These results establish that distinct VTA circuits generate reward and aversion, and thereby provide a new framework for understanding the circuit basis of adaptive and pathological motivated behaviours.

    View details for DOI 10.1038/nature11527

    View details for Web of Science ID 000310774300035

    View details for PubMedID 23064228

    View details for PubMedCentralID PMC3493743

  • High-Frequency Hippocampal Oscillations Activated by Optogenetic Stimulation of Transplanted Human ESC-Derived Neurons JOURNAL OF NEUROSCIENCE Pina-Crespo, J. C., Talantova, M., Cho, E., Soussou, W., Dolatabadi, N., Ryan, S. D., Ambasudhan, R., McKercher, S., Deisseroth, K., Lipton, S. A. 2012; 32 (45): 15837-15842

    Abstract

    After transplantation, individual stem cell-derived neurons can functionally integrate into the host CNS; however, evidence that neurons derived from transplanted human embryonic stem cells (hESCs) can drive endogenous neuronal network activity in CNS tissue is still lacking. Here, using multielectrode array recordings, we report activation of high-frequency oscillations in the β and γ ranges (10-100 Hz) in the host hippocampal network via targeted optogenetic stimulation of transplanted hESC-derived neurons.

    View details for DOI 10.1523/JNEUROSCI.3735-12.2012

    View details for Web of Science ID 000310842000018

    View details for PubMedID 23136422

    View details for PubMedCentralID PMC3513396

  • Color-tuned Channelrhodopsins for Multiwavelength Optogenetics JOURNAL OF BIOLOGICAL CHEMISTRY Prigge, M., Schneider, F., Tsunoda, S. P., Shilyansky, C., Wietek, J., Deisseroth, K., Hegemann, P. 2012; 287 (38): 31804-31812

    Abstract

    Channelrhodopsin-2 is a light-gated ion channel and a major tool of optogenetics. It is used to control neuronal activity via blue light. Here we describe the construction of color-tuned high efficiency channelrhodopsins (ChRs), based on chimeras of Chlamydomonas channelrhodopsin-1 and Volvox channelrhodopsin-1. These variants show superb expression and plasma membrane integration, resulting in 3-fold larger photocurrents in HEK cells compared with channelrhodopsin-2. Further molecular engineering gave rise to chimeric variants with absorption maxima ranging from 526 to 545 nm, dovetailing well with maxima of channelrhodopsin-2 derivatives ranging from 461 to 492 nm. Additional kinetic fine-tuning led to derivatives in which the lifetimes of the open state range from 19 ms to 5 s. Finally, combining green- with blue-absorbing variants allowed independent activation of two distinct neural cell populations at 560 and 405 nm. This novel panel of channelrhodopsin variants may serve as an important toolkit element for dual-color cell stimulation in neural circuits.

    View details for DOI 10.1074/jbc.M112.391185

    View details for Web of Science ID 000309059400021

    View details for PubMedID 22843694

    View details for PubMedCentralID PMC3442514

  • Neuronal circuitry mechanism regulating adult quiescent neural stem-cell fate decision NATURE Song, J., Zhong, C., Bonaguidi, M. A., Sun, G. J., Hsu, D., Gu, Y., Meletis, K., Huang, Z. J., Ge, S., Enikolopov, G., Deisseroth, K., Luscher, B., Christian, K. M., Ming, G., Song, H. 2012; 489 (7414): 150-U216

    Abstract

    Adult neurogenesis arises from neural stem cells within specialized niches. Neuronal activity and experience, presumably acting on this local niche, regulate multiple stages of adult neurogenesis, from neural progenitor proliferation to new neuron maturation, synaptic integration and survival. It is unknown whether local neuronal circuitry has a direct impact on adult neural stem cells. Here we show that, in the adult mouse hippocampus, nestin-expressing radial glia-like quiescent neural stem cells (RGLs) respond tonically to the neurotransmitter γ-aminobutyric acid (GABA) by means of γ2-subunit-containing GABAA receptors. Clonal analysis of individual RGLs revealed a rapid exit from quiescence and enhanced symmetrical self-renewal after conditional deletion of γ2. RGLs are in close proximity to terminals expressing 67-kDa glutamic acid decarboxylase (GAD67) of parvalbumin-expressing (PV+) interneurons and respond tonically to GABA released from these neurons. Functionally, optogenetic control of the activity of dentate PV+ interneurons, but not that of somatostatin-expressing or vasoactive intestinal polypeptide (VIP)-expressing interneurons, can dictate the RGL choice between quiescence and activation. Furthermore, PV+ interneuron activation restores RGL quiescence after social isolation, an experience that induces RGL activation and symmetrical division. Our study identifies a niche cell–signal–receptor trio and a local circuitry mechanism that control the activation and self-renewal mode of quiescent adult neural stem cells in response to neuronal activity and experience.

    View details for DOI 10.1038/nature11306

    View details for Web of Science ID 000308347000053

    View details for PubMedID 22842902

    View details for PubMedCentralID PMC3438284

  • Photothermal Genetic Engineering ACS NANO Anikeeva, P., Deisseroth, K. 2012; 6 (9): 7548-7552

    Abstract

    Optical methods for manipulation of cellular function have enabled deconstruction of genetic and neural circuits in vitro and in vivo. Plasmonic gold nanomaterials provide an alternative platform for external optical manipulation of genetic circuits. The tunable absorption of gold nanoparticles in the infrared spectral region and straightforward surface functionalization has led to applications in intracellular delivery and photorelease of short RNAs, recently enabling bidirectional photothermal modulation of specific genes via RNA interference (RNAi). We discuss recent advances in optical gene circuit engineering and plasmonic nanomaterials, as well as future research opportunities and challenges in photothermal gene manipulation.

    View details for DOI 10.1021/nn3039287

    View details for Web of Science ID 000309040600002

    View details for PubMedID 22954475

  • Activation of specific interneurons improves V1 feature selectivity and visual perception NATURE Lee, S., Kwan, A. C., Zhang, S., Phoumthipphavong, V., Flannery, J. G., Masmanidis, S. C., Taniguchi, H., Huang, Z. J., Zhang, F., Boyden, E. S., Deisseroth, K., Dan, Y. 2012; 488 (7411): 379-?

    Abstract

    Inhibitory interneurons are essential components of the neural circuits underlying various brain functions. In the neocortex, a large diversity of GABA (γ-aminobutyric acid) interneurons has been identified on the basis of their morphology, molecular markers, biophysical properties and innervation pattern. However, how the activity of each subtype of interneurons contributes to sensory processing remains unclear. Here we show that optogenetic activation of parvalbumin-positive (PV+) interneurons in the mouse primary visual cortex (V1) sharpens neuronal feature selectivity and improves perceptual discrimination. Using multichannel recording with silicon probes and channelrhodopsin-2 (ChR2)-mediated optical activation, we found that increased spiking of PV+ interneurons markedly sharpened orientation tuning and enhanced direction selectivity of nearby neurons. These effects were caused by the activation of inhibitory neurons rather than a decreased spiking of excitatory neurons, as archaerhodopsin-3 (Arch)-mediated optical silencing of calcium/calmodulin-dependent protein kinase IIα (CAMKIIα)-positive excitatory neurons caused no significant change in V1 stimulus selectivity. Moreover, the improved selectivity specifically required PV+ neuron activation, as activating somatostatin or vasointestinal peptide interneurons had no significant effect. Notably, PV+ neuron activation in awake mice caused a significant improvement in their orientation discrimination, mirroring the sharpened V1 orientation tuning. Together, these results provide the first demonstration that visual coding and perception can be improved by increased spiking of a specific subtype of cortical inhibitory interneurons.

    View details for DOI 10.1038/nature11312

    View details for Web of Science ID 000307501000042

    View details for PubMedID 22878719

    View details for PubMedCentralID PMC3422431

  • When the electricity (and the lights) go out: transient changes in excitability NATURE NEUROSCIENCE Ferenczi, E., Deisseroth, K. 2012; 15 (8): 1058-1060

    Abstract

    Natural or artificially induced electrical activity changes can alter ion balance so as to briefly influence firing. An optogenetics study delineates one mechanism: Cl- shifts causing seconds-long excitability changes after silencing.

    View details for DOI 10.1038/nn.3172

    View details for Web of Science ID 000306844500003

    View details for PubMedID 22837032

  • Expanding the Repertoire of Optogenetically Targeted Cells with an Enhanced Gene Expression System CELL REPORTS Tanaka, K. F., Matsui, K., Sasaki, T., Sano, H., Sugio, S., Fan, K., Hen, R., Nakai, J., Yanagawa, Y., Hasuwa, H., Okabe, M., Deisseroth, K., Ikenaka, K., Yamanaka, A. 2012; 2 (2): 397-406

    Abstract

    Optogenetics has been enthusiastically pursued in recent neuroscience research, and the causal relationship between neural activity and behavior is becoming ever more accessible. Here, we established knockin-mediated enhanced gene expression by improved tetracycline-controlled gene induction (KENGE-tet) and succeeded in generating transgenic mice expressing a highly light-sensitive channelrhodopsin-2 mutant at levels sufficient to drive the activities of multiple cell types. This method requires two lines of mice: one that controls the pattern of expression and another that determines the protein to be produced. The generation of new lines of either type readily expands the repertoire to choose from. In addition to neurons, we were able to manipulate the activity of nonexcitable glial cells in vivo. This shows that our system is applicable not only to neuroscience but also to any biomedical study that requires understanding of how the activity of a selected population of cells propagates through the intricate organic systems.

    View details for DOI 10.1016/j.celrep.2012.06.011

    View details for Web of Science ID 000309715100018

    View details for PubMedID 22854021

  • Striatal Dopamine Release Is Triggered by Synchronized Activity in Cholinergic Interneurons NEURON Threlfell, S., Lalic, T., Platt, N. J., Jennings, K. A., Deisseroth, K., Cragg, S. J. 2012; 75 (1): 58-64

    Abstract

    Striatal dopamine plays key roles in our normal and pathological goal-directed actions. To understand dopamine function, much attention has focused on how midbrain dopamine neurons modulate their firing patterns. However, we identify a presynaptic mechanism that triggers dopamine release directly, bypassing activity in dopamine neurons. We paired electrophysiological recordings of striatal channelrhodopsin2-expressing cholinergic interneurons with simultaneous detection of dopamine release at carbon-fiber microelectrodes in striatal slices. We reveal that activation of cholinergic interneurons by light flashes that cause only single action potentials in neurons from a small population triggers dopamine release via activation of nicotinic receptors on dopamine axons. This event overrides ascending activity from dopamine neurons and, furthermore, is reproduced by activating ChR2-expressing thalamostriatal inputs, which synchronize cholinergic interneurons in vivo. These findings indicate that synchronized activity in cholinergic interneurons directly generates striatal dopamine signals whose functions will extend beyond those encoded by dopamine neuron activity.

    View details for DOI 10.1016/j.neuron.2012.04.038

    View details for Web of Science ID 000306539600008

    View details for PubMedID 22794260

  • Altered profile of basket cell afferent synapses in hyper-excitable dentate gyrus revealed by optogenetic and two-pathway stimulations EUROPEAN JOURNAL OF NEUROSCIENCE Ledri, M., Nikitidou, L., Erdelyi, F., Szabo, G., Kirik, D., Deisseroth, K., Kokaia, M. 2012; 36 (1): 1971-1983

    Abstract

    Cholecystokinin (CCK-) positive basket cells form a distinct class of inhibitory GABAergic interneurons, proposed to act as fine-tuning devices of hippocampal gamma-frequency (30-90 Hz) oscillations, which can convert into higher frequency seizure activity. Therefore, CCK-basket cells may play an important role in regulation of hyper-excitability and seizures in the hippocampus. In normal conditions, the endogenous excitability regulator neuropeptide Y (NPY) has been shown to modulate afferent inputs onto dentate gyrus CCK-basket cells, providing a possible novel mechanism for excitability control in the hippocampus. Using GAD65-GFP mice for CCK-basket cell identification, and whole-cell patch-clamp recordings, we explored whether the effect of NPY on afferent synapses to CCK-basket cells is modified in the hyper-excitable dentate gyrus. To induce a hyper-excitable state, recurrent seizures were evoked by electrical stimulation of the hippocampus using the well-characterized rapid kindling protocol. The frequency of spontaneous and miniature excitatory and inhibitory post-synaptic currents recorded in CCK-basket cells was decreased by NPY. The excitatory post-synaptic currents evoked in CCK-basket cells by optogenetic activation of principal neurons were also decreased in amplitude. Interestingly, we observed an increased proportion of spontaneous inhibitory post-synaptic currents with slower rise times, indicating that NPY may inhibit gamma aminobutyric acid release preferentially in peri-somatic synapses. These findings indicate that increased levels and release of NPY observed after seizures can modulate afferent inputs to CCK-basket cells, and therefore alter their impact on the oscillatory network activity and excitability in the hippocampus.

    View details for DOI 10.1111/j.1460-9568.2012.08080.x

    View details for Web of Science ID 000305903000003

    View details for PubMedID 22512307

  • Optogenetics and Psychiatry: Applications, Challenges, and Opportunities BIOLOGICAL PSYCHIATRY Deisseroth, K. 2012; 71 (12): 1030-1032
  • The best of times, the worst of times for psychiatric disease NATURE NEUROSCIENCE Karayiorgou, M., Flint, J., Gogos, J. A., Malenka, R. C. 2012; 15 (6): 811-812

    View details for DOI 10.1038/nn.3115

    View details for Web of Science ID 000304546700005

    View details for PubMedID 22627793

    View details for PubMedCentralID PMC4416402

  • A critical role for NMDA receptors in parvalbumin interneurons for gamma rhythm induction and behavior MOLECULAR PSYCHIATRY Carlen, M., Meletis, K., Siegle, J. H., Cardin, J. A., Futai, K., Vierling-Claassen, D., Ruehlmann, C., Jones, S. R., Deisseroth, K., Sheng, M., Moore, C. I., Tsai, L. 2012; 17 (5): 537-548

    Abstract

    Synchronous recruitment of fast-spiking (FS) parvalbumin (PV) interneurons generates gamma oscillations, rhythms that emerge during performance of cognitive tasks. Administration of N-methyl-D-aspartate (NMDA) receptor antagonists alters gamma rhythms, and can induce cognitive as well as psychosis-like symptoms in humans. The disruption of NMDA receptor (NMDAR) signaling specifically in FS PV interneurons is therefore hypothesized to give rise to neural network dysfunction that could underlie these symptoms. To address the connection between NMDAR activity, FS PV interneurons, gamma oscillations and behavior, we generated mice lacking NMDAR neurotransmission only in PV cells (PV-Cre/NR1f/f mice). Here, we show that mutant mice exhibit enhanced baseline cortical gamma rhythms, impaired gamma rhythm induction after optogenetic drive of PV interneurons and reduced sensitivity to the effects of NMDAR antagonists on gamma oscillations and stereotypies. Mutant mice show largely normal behaviors except for selective cognitive impairments, including deficits in habituation, working memory and associative learning. Our results provide evidence for the critical role of NMDAR in PV interneurons for expression of normal gamma rhythms and specific cognitive behaviors.

    View details for DOI 10.1038/mp.2011.31

    View details for Web of Science ID 000303110800009

    View details for PubMedID 21468034

  • Optogenetic stimulation of a hippocampal engram activates fear memory recall NATURE Liu, X., Ramirez, S., Pang, P. T., Puryear, C. B., Govindarajan, A., Deisseroth, K., Tonegawa, S. 2012; 484 (7394): 381-U415

    Abstract

    A specific memory is thought to be encoded by a sparse population of neurons. These neurons can be tagged during learning for subsequent identification and manipulation. Moreover, their ablation or inactivation results in reduced memory expression, suggesting their necessity in mnemonic processes. However, the question of sufficiency remains: it is unclear whether it is possible to elicit the behavioural output of a specific memory by directly activating a population of neurons that was active during learning. Here we show in mice that optogenetic reactivation of hippocampal neurons activated during fear conditioning is sufficient to induce freezing behaviour. We labelled a population of hippocampal dentate gyrus neurons activated during fear learning with channelrhodopsin-2 (ChR2) and later optically reactivated these neurons in a different context. The mice showed increased freezing only upon light stimulation, indicating light-induced fear memory recall. This freezing was not detected in non-fear-conditioned mice expressing ChR2 in a similar proportion of cells, nor in fear-conditioned mice with cells labelled by enhanced yellow fluorescent protein instead of ChR2. Finally, activation of cells labelled in a context not associated with fear did not evoke freezing in mice that were previously fear conditioned in a different context, suggesting that light-induced fear memory recall is context specific. Together, our findings indicate that activating a sparse but specific ensemble of hippocampal neurons that contribute to a memory engram is sufficient for the recall of that memory. Moreover, our experimental approach offers a general method of mapping cellular populations bearing memory engrams.

    View details for DOI 10.1038/nature11028

    View details for Web of Science ID 000302946500034

    View details for PubMedID 22441246

    View details for PubMedCentralID PMC3331914

  • Synaptic Activity Unmasks Dopamine D2 Receptor Modulation of a Specific Class of Layer V Pyramidal Neurons in Prefrontal Cortex JOURNAL OF NEUROSCIENCE Gee, S., Ellwood, I., Patel, T., Luongo, F., Deisseroth, K., Sohal, V. S. 2012; 32 (14): 4959-4971

    Abstract

    Dopamine D2 receptors (D2Rs) play a major role in the function of the prefrontal cortex (PFC), and may contribute to prefrontal dysfunction in conditions such as schizophrenia. Here we report that in mouse PFC, D2Rs are selectively expressed by a subtype of layer V pyramidal neurons that have thick apical tufts, prominent h-current, and subcortical projections. Within this subpopulation, the D2R agonist quinpirole elicits a novel afterdepolarization that generates voltage fluctuations and spiking for hundreds of milliseconds. Surprisingly, this afterdepolarization is masked in quiescent brain slices, but is readily unmasked by physiologic levels of synaptic input which activate NMDA receptors, possibly explaining why this phenomenon has not been reported previously. Notably, we could still elicit this afterdepolarization for some time after the cessation of synaptic stimulation. In addition to NMDA receptors, the quinpirole-induced afterdepolarization also depended on L-type Ca(2+) channels and was blocked by the selective L-type antagonist nimodipine. To confirm that D2Rs can elicit this afterdepolarization by enhancing Ca(2+) (and Ca(2+)-dependent) currents, we measured whole-cell Ca(2+) potentials that occur after blocking Na(+) and K(+) channels, and found quinpirole enhanced these potentials, while the selective D2R antagonist sulpiride had the opposite effect. Thus, D2Rs can elicit a Ca(2+)-channel-dependent afterdepolarization that powerfully modulates activity in specific prefrontal neurons. Through this mechanism, D2Rs might enhance outputs to subcortical structures, contribute to reward-related persistent firing, or increase the level of noise in prefrontal circuits.

    View details for DOI 10.1523/JNEUROSCI.5835-11.2012

    View details for Web of Science ID 000302783500026

    View details for PubMedID 22492051

    View details for PubMedCentralID PMC3352768

  • Optogenetic investigation of neural circuits underlying brain disease in animal models NATURE REVIEWS NEUROSCIENCE Tye, K. M., Deisseroth, K. 2012; 13 (4): 251-266

    Abstract

    Optogenetic tools have provided a new way to establish causal relationships between brain activity and behaviour in health and disease. Although no animal model captures human disease precisely, behaviours that recapitulate disease symptoms may be elicited and modulated by optogenetic methods, including behaviours that are relevant to anxiety, fear, depression, addiction, autism and parkinsonism. The rapid proliferation of optogenetic reagents together with the swift advancement of strategies for implementation has created new opportunities for causal and precise dissection of the circuits underlying brain diseases in animal models.

    View details for DOI 10.1038/nrn3171

    View details for Web of Science ID 000301942500009

    View details for PubMedID 22430017

  • GABA Neurons of the VTA Drive Conditioned Place Aversion NEURON Tan, K. R., Yvon, C., Turiault, M., Mirzabekov, J. J., Doehner, J., Labouebe, G., Deisseroth, K., Tye, K. M., Luescher, C. 2012; 73 (6): 1173-1183

    Abstract

    Salient but aversive stimuli inhibit the majority of dopamine (DA) neurons in the ventral tegmental area (VTA) and cause conditioned place aversion (CPA). The cellular mechanism underlying DA neuron inhibition has not been investigated and the causal link to behavior remains elusive. Here, we show that GABA neurons of the VTA inhibit DA neurons through neurotransmission at GABA(A) receptors. We also observe that GABA neurons increase their firing in response to a footshock and provide evidence that driving GABA neurons with optogenetic effectors is sufficient to affect behavior. Taken together, our data demonstrate that synaptic inhibition of DA neurons drives place aversion.

    View details for DOI 10.1016/j.neuron.2012.02.015

    View details for Web of Science ID 000301998700014

    View details for PubMedID 22445344

  • Structural Model of Channelrhodopsin JOURNAL OF BIOLOGICAL CHEMISTRY Watanabe, H. C., Welke, K., Schneider, F., Tsunoda, S., Zhang, F., Deisseroth, K., Hegemann, P., Elstner, M. 2012; 287 (10): 7456-7466

    Abstract

    Channelrhodopsins (ChRs) are light-gated cation channels that mediate ion transport across membranes in microalgae (vectorial catalysis). ChRs are now widely used for the analysis of neural networks in tissues and living animals with light (optogenetics). For elucidation of functional mechanisms at the atomic level, as well as for further engineering and application, a detailed structure is urgently needed. In the absence of an experimental structure, here we develop a structural ChR model based on several molecular computational approaches, capitalizing on characteristic patterns in amino acid sequences of ChR1, ChR2, Volvox ChRs, Mesostigma ChR, and the recently identified ChR of the halophilic alga Dunaliella salina. In the present model, we identify remarkable structural motifs that may explain fundamental electrophysiological properties of ChR2, ChR1, and their mutants, and in a crucial validation of the model, we successfully reproduce the excitation energy predicted by absorption spectra.

    View details for DOI 10.1074/jbc.M111.320309

    View details for Web of Science ID 000301060200046

    View details for PubMedID 22241469

    View details for PubMedCentralID PMC3293557

  • Integrated device for combined optical neuromodulation and electrical recording for chronic in vivo applications JOURNAL OF NEURAL ENGINEERING Wang, J., Wagner, F., Borton, D. A., Zhang, J., Ozden, I., Burwell, R. D., Nurmikko, A. V., Van Wagenen, R., Diester, I., Deisseroth, K. 2012; 9 (1)

    Abstract

    Studying brain function and its local circuit dynamics requires neural interfaces that can record and stimulate the brain with high spatiotemporal resolution. Optogenetics, a technique that genetically targets specific neurons to express light-sensitive channel proteins, provides the capability to control central nervous system neuronal activity in mammals with millisecond time precision. This technique enables precise optical stimulation of neurons and simultaneous monitoring of neural response by electrophysiological means, both in the vicinity of and distant to the stimulation site. We previously demonstrated, in vitro, the dual capability (optical delivery and electrical recording) while testing a novel hybrid device (optrode-MEA), which incorporates a tapered coaxial optical electrode (optrode) and a 100 element microelectrode array (MEA). Here we report a fully chronic implant of a new version of this device in ChR2-expressing rats, and demonstrate its use in freely moving animals over periods up to 8 months. In its present configuration, we show the device delivering optical excitation to a single cortical site while mapping the neural response from the surrounding 30 channels of the 6 × 6 element MEA, thereby enabling recording of optically modulated single-unit and local field potential activity across several millimeters of the neocortical landscape.

    View details for DOI 10.1088/1741-2560/9/1/016001

    View details for Web of Science ID 000300618700003

    View details for PubMedID 22156042

  • Principles for applying optogenetic tools derived from direct comparative analysis of microbial opsins NATURE METHODS Mattis, J., Tye, K. M., Ferenczi, E. A., Ramakrishnan, C., O'Shea, D. J., Prakash, R., Gunaydin, L. A., Hyun, M., Fenno, L. E., Gradinaru, V., Yizhar, O., Deisseroth, K. 2012; 9 (2): 159-172

    Abstract

    Diverse optogenetic tools have allowed versatile control over neural activity. Many depolarizing and hyperpolarizing tools have now been developed in multiple laboratories and tested across different preparations, presenting opportunities but also making it difficult to draw direct comparisons. This challenge has been compounded by the dependence of performance on parameters such as vector, promoter, expression time, illumination, cell type and many other variables. As a result, it has become increasingly complicated for end users to select the optimal reagents for their experimental needs. For a rapidly growing field, critical figures of merit should be formalized both to establish a framework for further development and so that end users can readily understand how these standardized parameters translate into performance. Here we systematically compared microbial opsins under matched experimental conditions to extract essential principles and identify key parameters for the conduct, design and interpretation of experiments involving optogenetic techniques.

    View details for DOI 10.1038/NMETH.1808

    View details for Web of Science ID 000300029600026

  • Principles for applying optogenetic tools derived from direct comparative analysis of microbial opsins. Nature methods Mattis, J., Tye, K. M., Ferenczi, E. A., Ramakrishnan, C., O'Shea, D. J., Prakash, R., Gunaydin, L. A., Hyun, M., Fenno, L. E., Gradinaru, V., Yizhar, O., Deisseroth, K. 2012; 9 (2): 159-172

    Abstract

    Diverse optogenetic tools have allowed versatile control over neural activity. Many depolarizing and hyperpolarizing tools have now been developed in multiple laboratories and tested across different preparations, presenting opportunities but also making it difficult to draw direct comparisons. This challenge has been compounded by the dependence of performance on parameters such as vector, promoter, expression time, illumination, cell type and many other variables. As a result, it has become increasingly complicated for end users to select the optimal reagents for their experimental needs. For a rapidly growing field, critical figures of merit should be formalized both to establish a framework for further development and so that end users can readily understand how these standardized parameters translate into performance. Here we systematically compared microbial opsins under matched experimental conditions to extract essential principles and identify key parameters for the conduct, design and interpretation of experiments involving optogenetic techniques.

    View details for DOI 10.1038/nmeth.1808

    View details for PubMedID 22179551

  • Yellow Optogenetics with Volvox Channelrhodopsin Variants Schneider, F., Prigge, M., Tsunoda, S. P., Yizhar, O., Deisseroth, K., Hegemann, P. CELL PRESS. 2012: 681A-682A
  • Optetrode: a multichannel readout for optogenetic control in freely moving mice NATURE NEUROSCIENCE Anikeeva, P., Andalman, A. S., Witten, I., Warden, M., Goshen, I., Grosenick, L., Gunaydin, L. A., Frank, L. M., Deisseroth, K. 2012; 15 (1): 163-U204

    Abstract

    Recent advances in optogenetics have improved the precision with which defined circuit elements can be controlled optically in freely moving mammals; in particular, recombinase-dependent opsin viruses, used with a growing pool of transgenic mice expressing recombinases, allow manipulation of specific cell types. However, although optogenetic control has allowed neural circuits to be manipulated in increasingly powerful ways, combining optogenetic stimulation with simultaneous multichannel electrophysiological readout of isolated units in freely moving mice remains a challenge. We designed and validated the optetrode, a device that allows for colocalized multi-tetrode electrophysiological recording and optical stimulation in freely moving mice. Optetrode manufacture employs a unique optical fiber-centric coaxial design approach that yields a lightweight (2 g), compact and robust device that is suitable for behaving mice. This low-cost device is easy to construct (2.5 h to build without specialized equipment). We found that the drive design produced stable high-quality recordings and continued to do so for at least 6 weeks following implantation. We validated the optetrode by quantifying, for the first time, the response of cells in the medial prefrontal cortex to local optical excitation and inhibition, probing multiple different genetically defined classes of cells in the mouse during open field exploration.

    View details for DOI 10.1038/nn.2992

    View details for Web of Science ID 000298414400027

  • A critical role for NMDA receptors in parvalbumin interneurons for gamma rhythm induction and behavior. Mol Psychiatry. Carlén, M., Meletis, K., Siegle, J. H., Cardin, J. A., Futai, K., Vierling-Claassen, D., Deisseroth, K. 2012; 5 (17): 537-48
  • Optetrode: a multichannel readout for optogenetic control in freely moving mice. Nature neuroscience Anikeeva, P., Andalman, A. S., Witten, I., Warden, M., Goshen, I., Grosenick, L., Gunaydin, L. A., Frank, L. M., Deisseroth, K. 2012; 15 (1): 163-170

    Abstract

    Recent advances in optogenetics have improved the precision with which defined circuit elements can be controlled optically in freely moving mammals; in particular, recombinase-dependent opsin viruses, used with a growing pool of transgenic mice expressing recombinases, allow manipulation of specific cell types. However, although optogenetic control has allowed neural circuits to be manipulated in increasingly powerful ways, combining optogenetic stimulation with simultaneous multichannel electrophysiological readout of isolated units in freely moving mice remains a challenge. We designed and validated the optetrode, a device that allows for colocalized multi-tetrode electrophysiological recording and optical stimulation in freely moving mice. Optetrode manufacture employs a unique optical fiber-centric coaxial design approach that yields a lightweight (2 g), compact and robust device that is suitable for behaving mice. This low-cost device is easy to construct (2.5 h to build without specialized equipment). We found that the drive design produced stable high-quality recordings and continued to do so for at least 6 weeks following implantation. We validated the optetrode by quantifying, for the first time, the response of cells in the medial prefrontal cortex to local optical excitation and inhibition, probing multiple different genetically defined classes of cells in the mouse during open field exploration.

    View details for DOI 10.1038/nn.2992

    View details for PubMedID 22138641

  • GABAergic circuits mediate the reinforcement-related signals of striatal cholinergic interneurons. Nature neuroscience English, D. F., Ibanez-Sandoval, O., Stark, E., Tecuapetla, F., Buzsáki, G., Deisseroth, K., Tepper, J. M., Koos, T. 2012; 15 (1): 123-130

    Abstract

    Neostriatal cholinergic interneurons are believed to be important for reinforcement-mediated learning and response selection by signaling the occurrence and motivational value of behaviorally relevant stimuli through precisely timed multiphasic population responses. An important problem is to understand how these signals regulate the functioning of the neostriatum. Here we describe the synaptic organization of a previously unknown circuit that involves direct nicotinic excitation of several classes of GABAergic interneurons, including neuroptide Y-expressing neurogilaform neurons, and enables cholinergic interneurons to exert rapid inhibitory control of the activity of projection neurons. We also found that, in vivo, the dominant effect of an optogenetically reproduced pause-excitation population response of cholinergic interneurons was powerful and rapid inhibition of the firing of projection neurons that is coincident with synchronous cholinergic activation. These results reveal a previously unknown circuit mechanism that transmits reinforcement-related information of ChAT interneurons in the mouse neostriatal network.

    View details for DOI 10.1038/nn.2984

    View details for PubMedID 22158514

  • GABAergic circuits mediate the reinforcement-related signals of striatal cholinergic interneurons NATURE NEUROSCIENCE English, D. F., Ibanez-Sandoval, O., Stark, E., Tecuapetla, F., Buzsaki, G., Deisseroth, K., Tepper, J. M., Koos, T. 2012; 15 (1): 123-U155

    Abstract

    Neostriatal cholinergic interneurons are believed to be important for reinforcement-mediated learning and response selection by signaling the occurrence and motivational value of behaviorally relevant stimuli through precisely timed multiphasic population responses. An important problem is to understand how these signals regulate the functioning of the neostriatum. Here we describe the synaptic organization of a previously unknown circuit that involves direct nicotinic excitation of several classes of GABAergic interneurons, including neuroptide Y-expressing neurogilaform neurons, and enables cholinergic interneurons to exert rapid inhibitory control of the activity of projection neurons. We also found that, in vivo, the dominant effect of an optogenetically reproduced pause-excitation population response of cholinergic interneurons was powerful and rapid inhibition of the firing of projection neurons that is coincident with synchronous cholinergic activation. These results reveal a previously unknown circuit mechanism that transmits reinforcement-related information of ChAT interneurons in the mouse neostriatal network.

    View details for DOI 10.1038/nn.2984

    View details for Web of Science ID 000298414400022

    View details for PubMedCentralID PMC3245803

  • Recombinase-Driver Rat Lines: Tools, Techniques, and Optogenetic Application to Dopamine-Mediated Reinforcement NEURON Witten, I. B., Steinberg, E. E., Lee, S. Y., Davidson, T. J., Zalocusky, K. A., Brodsky, M., Yizhar, O., Cho, S. L., Gong, S., Ramakrishnan, C., Stuber, G. D., Tye, K. M., Janak, P. H., Deisseroth, K. 2011; 72 (5): 721-733

    Abstract

    Currently there is no general approach for achieving specific optogenetic control of genetically defined cell types in rats, which provide a powerful experimental system for numerous established neurophysiological and behavioral paradigms. To overcome this challenge we have generated genetically restricted recombinase-driver rat lines suitable for driving gene expression in specific cell types, expressing Cre recombinase under the control of large genomic regulatory regions (200-300 kb). Multiple tyrosine hydroxylase (Th)::Cre and choline acetyltransferase (Chat)::Cre lines were produced that exhibited specific opsin expression in targeted cell types. We additionally developed methods for utilizing optogenetic tools in freely moving rats and leveraged these technologies to clarify the causal relationship between dopamine (DA) neuron firing and positive reinforcement, observing that optical stimulation of DA neurons in the ventral tegmental area (VTA) of Th::Cre rats is sufficient to support vigorous intracranial self-stimulation (ICSS). These studies complement existing targeting approaches by extending the generalizability of optogenetics to traditionally non-genetically-tractable but vital animal models.

    View details for DOI 10.1016/j.neuron.2011.10.028

    View details for Web of Science ID 000297971100008

    View details for PubMedID 22153370

    View details for PubMedCentralID PMC3282061

  • Leptin regulates the reward value of nutrient NATURE NEUROSCIENCE Domingos, A. I., Vaynshteyn, J., Voss, H. U., Ren, X., Gradinaru, V., Zang, F., Deisseroth, K., de Araujo, I. E., Friedman, J. 2011; 14 (12): 1562-U92

    Abstract

    We developed an assay for quantifying the reward value of nutrient and used it to analyze the effects of metabolic state and leptin. In this assay, mice chose between two sippers, one of which dispensed water and was coupled to optogenetic activation of dopaminergic (DA) neurons and the other of which dispensed natural or artificial sweeteners. This assay measured the reward value of sweeteners relative to lick-induced optogenetic activation of DA neurons. Mice preferred optogenetic stimulation of DA neurons to sucralose, but not to sucrose. However, the mice preferred sucralose plus optogenetic stimulation versus sucrose. We found that food restriction increased the value of sucrose relative to sucralose plus optogenetic stimulation, and that leptin decreased it. Our data suggest that leptin suppresses the ability of sucrose to drive taste-independent DA neuronal activation and provide new insights into the mechanism of leptin's effects on food intake.

    View details for DOI 10.1038/nn.2977

    View details for Web of Science ID 000297546300016

    View details for PubMedID 22081158

  • Neuronal filtering of multiplexed odour representations NATURE Blumhagen, F., Zhu, P., Shum, J., Schaerer, Y. Z., Yaksi, E., Deisseroth, K., Friedrich, R. W. 2011; 479 (7374): 493-U215

    Abstract

    Neuronal activity patterns contain information in their temporal structure, indicating that information transfer between neurons may be optimized by temporal filtering. In the zebrafish olfactory bulb, subsets of output neurons (mitral cells) engage in synchronized oscillations during odour responses, but information about odour identity is contained mostly in non-oscillatory firing rate patterns. Using optogenetic manipulations and odour stimulation, we found that firing rate responses of neurons in the posterior zone of the dorsal telencephalon (Dp), a target area homologous to olfactory cortex, were largely insensitive to oscillatory synchrony of mitral cells because passive membrane properties and synaptic currents act as low-pass filters. Nevertheless, synchrony influenced spike timing. Moreover, Dp neurons responded primarily during the decorrelated steady state of mitral cell activity patterns. Temporal filtering therefore tunes Dp neurons to components of mitral cell activity patterns that are particularly informative about precise odour identity. These results demonstrate how temporal filtering can extract specific information from multiplexed neuronal codes.

    View details for DOI 10.1038/nature10633

    View details for Web of Science ID 000297285600042

    View details for PubMedID 22080956

  • SNCA Triplication Parkinson's Patient's iPSC-derived DA Neurons Accumulate alpha-Synuclein and Are Susceptible to Oxidative Stress PLOS ONE Byers, B., Cord, B., Ha Nam Nguyen, H. N., Schuele, B., Fenno, L., Lee, P. C., Deisseroth, K., Langston, J. W., Pera, R. R., Palmer, T. D. 2011; 6 (11)

    Abstract

    Parkinson's disease (PD) is an incurable age-related neurodegenerative disorder affecting both the central and peripheral nervous systems. Although common, the etiology of PD remains poorly understood. Genetic studies infer that the disease results from a complex interaction between genetics and environment and there is growing evidence that PD may represent a constellation of diseases with overlapping yet distinct underlying mechanisms. Novel clinical approaches will require a better understanding of the mechanisms at work within an individual as well as methods to identify the specific array of mechanisms that have contributed to the disease. Induced pluripotent stem cell (iPSC) strategies provide an opportunity to directly study the affected neuronal subtypes in a given patient. Here we report the generation of iPSC-derived midbrain dopaminergic neurons from a patient with a triplication in the α-synuclein gene (SNCA). We observed that the iPSCs readily differentiated into functional neurons. Importantly, the PD-affected line exhibited disease-related phenotypes in culture: accumulation of α-synuclein, inherent overexpression of markers of oxidative stress, and sensitivity to peroxide induced oxidative stress. These findings show that the dominantly-acting PD mutation is intrinsically capable of perturbing normal cell function in culture and confirm that these features reflect, at least in part, a cell autonomous disease process that is independent of exposure to the entire complexity of the diseased brain.

    View details for DOI 10.1371/journal.pone.0026159

    View details for Web of Science ID 000297555400007

    View details for PubMedID 22110584

    View details for PubMedCentralID PMC3217921

  • Hemisphere-specific optogenetic stimulation reveals left-right asymmetry of hippocampal plasticity NATURE NEUROSCIENCE Kohl, M. M., Shipton, O. A., Deacon, R. M., Rawlins, J. N., Deisseroth, K., Paulsen, O. 2011; 14 (11): 1413-1415

    Abstract

    Postsynaptic spines at CA3-CA1 synapses differ in glutamate receptor composition according to the hemispheric origin of CA3 afferents. To study the functional consequences of this asymmetry, we used optogenetic tools to selectively stimulate axons of CA3 pyramidal cells originating in either left or right mouse hippocampus. We found that left CA3 input produced more long-term potentiation at CA1 synapses than right CA3 input as a result of differential expression of GluN2B subunit-containing NMDA receptors.

    View details for DOI 10.1038/nn.2915

    View details for Web of Science ID 000296518600015

    View details for PubMedID 21946328

  • Dynamics of Retrieval Strategies for Remote Memories CELL Goshen, I., Brodsky, M., Prakash, R., Wallace, J., Gradinaru, V., Ramakrishnan, C., Deisseroth, K. 2011; 147 (3): 678-689

    Abstract

    Prevailing theory suggests that long-term memories are encoded via a two-phase process requiring early involvement of the hippocampus followed by the neocortex. Contextual fear memories in rodents rely on the hippocampus immediately following training but are unaffected by hippocampal lesions or pharmacological inhibition weeks later. With fast optogenetic methods, we examine the real-time contribution of hippocampal CA1 excitatory neurons to remote memory and find that contextual fear memory recall, even weeks after training, can be reversibly abolished by temporally precise optogenetic inhibition of CA1. When this inhibition is extended to match the typical time course of pharmacological inhibition, remote hippocampus dependence converts to hippocampus independence, suggesting that long-term memory retrieval normally depends on the hippocampus but can adaptively shift to alternate structures. Further revealing the plasticity of mechanisms required for memory recall, we confirm the remote-timescale importance of the anterior cingulate cortex (ACC) and implicate CA1 in ACC recruitment for remote recall.

    View details for DOI 10.1016/j.cell.2011.09.033

    View details for Web of Science ID 000296573700021

    View details for PubMedID 22019004

  • Differential Modulation of Excitatory and Inhibitory Striatal Synaptic Transmission by Histamine JOURNAL OF NEUROSCIENCE Ellender, T. J., Huerta-Ocampo, I., Deisseroth, K., Capogna, M., Bolam, J. P. 2011; 31 (43): 15340-15351

    Abstract

    Information processing in the striatum is critical for basal ganglia function and strongly influenced by neuromodulators (e.g., dopamine). The striatum also receives modulatory afferents from the histaminergic neurons in the hypothalamus which exhibit a distinct diurnal rhythm with high activity during wakefulness, and little or no activity during sleep. In view of the fact that the striatum also expresses a high density of histamine receptors, we hypothesized that released histamine will affect striatal function. We studied the role of histamine on striatal microcircuit function by performing whole-cell patch-clamp recordings of neurochemically identified striatal neurons combined with electrical and optogenetic stimulation of striatal afferents in mouse brain slices. Bath applied histamine had many effects on striatal microcircuits. Histamine, acting at H(2) receptors, depolarized both the direct and indirect pathway medium spiny projection neurons (MSNs). Excitatory, glutamatergic input to both classes of MSNs from both the cortex and thalamus was negatively modulated by histamine acting at presynaptic H(3) receptors. The dynamics of thalamostriatal, but not corticostriatal, synapses were modulated by histamine leading to a facilitation of thalamic input. Furthermore, local inhibitory input to both classes of MSNs was negatively modulated by histamine. Subsequent dual whole-cell patch-clamp recordings of connected pairs of striatal neurons revealed that only lateral inhibition between MSNs is negatively modulated, whereas feedforward inhibition from fast-spiking GABAergic interneurons onto MSNs is unaffected by histamine. These findings suggest that the diurnal rhythm of histamine release entrains striatal function which, during wakefulness, is dominated by feedforward inhibition and a suppression of excitatory drive.

    View details for DOI 10.1523/JNEUROSCI.3144-11.2011

    View details for Web of Science ID 000296446200013

    View details for PubMedID 22031880

  • In Vivo Optogenetic Stimulation of Neocortical Excitatory Neurons Drives Brain-State-Dependent Inhibition CURRENT BIOLOGY Mateo, C., Avermann, M., Gentet, L. J., Zhang, F., Deisseroth, K., Petersen, C. C. 2011; 21 (19): 1593-1602

    Abstract

    Synaptic interactions between excitatory and inhibitory neocortical neurons are important for mammalian sensory perception. Synaptic transmission between identified neurons within neocortical microcircuits has mainly been studied in brain slice preparations in vitro. Here, we investigate brain-state-dependent neocortical synaptic interactions in vivo by combining the specificity of optogenetic stimulation with the precision of whole-cell recordings from postsynaptic excitatory glutamatergic neurons and GFP-labeled inhibitory GABAergic neurons targeted through two-photon microscopy.Channelrhodopsin-2 (ChR2) stimulation of excitatory layer 2/3 barrel cortex neurons evoked larger and faster depolarizing postsynaptic potentials and more synaptically driven action potentials in fast-spiking (FS) GABAergic neurons compared to both non-fast-spiking (NFS) GABAergic neurons and postsynaptic excitatory pyramidal neurons located within the same neocortical microcircuit. The number of action potentials evoked in ChR2-expressing neurons showed low trial-to-trial variability, but postsynaptic responses varied strongly with near-linear dependence upon spontaneously driven changes in prestimulus membrane potential. Postsynaptic responses in excitatory neurons had reversal potentials, which were hyperpolarized relative to action potential threshold and were therefore inhibitory. Reversal potentials measured in postsynaptic GABAergic neurons were close to action potential threshold. Postsynaptic inhibitory neurons preferentially fired synaptically driven action potentials from spontaneously depolarized network states, with stronger state-dependent modulation in NFS GABAergic neurons compared to FS GABAergic neurons.Inhibitory neurons appear to dominate neocortical microcircuit function, receiving stronger local excitatory synaptic input and firing more action potentials compared to excitatory neurons. In mouse layer 2/3 barrel cortex, we propose that strong state-dependent recruitment of inhibitory neurons drives competition among excitatory neurons enforcing sparse coding.

    View details for DOI 10.1016/j.cub.2011.08.028

    View details for Web of Science ID 000295899600016

    View details for PubMedID 21945274

  • Multiscale Computational Models for Optogenetic Control of Cardiac Function BIOPHYSICAL JOURNAL Abilez, O. J., Wong, J., Prakash, R., Deisseroth, K., Zarins, C. K., Kuhl, E. 2011; 101 (6): 1326-1334

    Abstract

    The ability to stimulate mammalian cells with light has significantly changed our understanding of electrically excitable tissues in health and disease, paving the way toward various novel therapeutic applications. Here, we demonstrate the potential of optogenetic control in cardiac cells using a hybrid experimental/computational technique. Experimentally, we introduced channelrhodopsin-2 into undifferentiated human embryonic stem cells via a lentiviral vector, and sorted and expanded the genetically engineered cells. Via directed differentiation, we created channelrhodopsin-expressing cardiomyocytes, which we subjected to optical stimulation. To quantify the impact of photostimulation, we assessed electrical, biochemical, and mechanical signals using patch-clamping, multielectrode array recordings, and video microscopy. Computationally, we introduced channelrhodopsin-2 into a classic autorhythmic cardiac cell model via an additional photocurrent governed by a light-sensitive gating variable. Upon optical stimulation, the channel opens and allows sodium ions to enter the cell, inducing a fast upstroke of the transmembrane potential. We calibrated the channelrhodopsin-expressing cell model using single action potential readings for different photostimulation amplitudes, pulse widths, and frequencies. To illustrate the potential of the proposed approach, we virtually injected channelrhodopsin-expressing cells into different locations of a human heart, and explored its activation sequences upon optical stimulation. Our experimentally calibrated computational toolbox allows us to virtually probe landscapes of process parameters, and identify optimal photostimulation sequences toward pacing hearts with light.

    View details for DOI 10.1016/j.bpj.2011.08.004

    View details for Web of Science ID 000295197300006

    View details for PubMedID 21943413

    View details for PubMedCentralID PMC3177076

  • Cell type-specific channelrhodopsin-2 transgenic mice for optogenetic dissection of neural circuitry function NATURE METHODS Zhao, S., Ting, J. T., Atallah, H. E., Qiu, L., Tan, J., Gloss, B., Augustine, G. J., Deisseroth, K., Luo, M., Graybiel, A. M., Feng, G. 2011; 8 (9): 745-U91

    Abstract

    Optogenetic methods have emerged as powerful tools for dissecting neural circuit connectivity, function and dysfunction. We used a bacterial artificial chromosome (BAC) transgenic strategy to express the H134R variant of channelrhodopsin-2, ChR2(H134R), under the control of cell type–specific promoter elements. We performed an extensive functional characterization of the newly established VGAT-ChR2(H134R)-EYFP, ChAT-ChR2(H134R)-EYFP, Tph2-ChR2(H134R)-EYFP and Pvalb(H134R)-ChR2-EYFP BAC transgenic mouse lines and demonstrate the utility of these lines for precisely controlling action-potential firing of GABAergic, cholinergic, serotonergic and parvalbumin-expressing neuron subsets using blue light. This resource of cell type–specific ChR2(H134R) mouse lines will facilitate the precise mapping of neuronal connectivity and the dissection of the neural basis of behavior.

    View details for DOI 10.1038/nmeth.1668

    View details for Web of Science ID 000294439100011

    View details for PubMedID 21985008

    View details for PubMedCentralID PMC3191888

  • A new mode of corticothalamic transmission revealed in the Gria4(-/-) model of absence epilepsy NATURE NEUROSCIENCE Paz, J. T., Bryant, A. S., Peng, K., Fenno, L., Yizhar, O., Frankel, W. N., Deisseroth, K., Huguenard, J. R. 2011; 14 (9): 1167-U225

    Abstract

    Cortico-thalamo-cortical circuits mediate sensation and generate neural network oscillations associated with slow-wave sleep and various epilepsies. Cortical input to sensory thalamus is thought to mainly evoke feed-forward synaptic inhibition of thalamocortical (TC) cells via reticular thalamic nucleus (nRT) neurons, especially during oscillations. This relies on a stronger synaptic strength in the cortico-nRT pathway than in the cortico-TC pathway, allowing the feed-forward inhibition of TC cells to overcome direct cortico-TC excitation. We found a systemic and specific reduction in strength in GluA4-deficient (Gria4(-/-)) mice of one excitatory synapse of the rhythmogenic cortico-thalamo-cortical system, the cortico-nRT projection, and observed that the oscillations could still be initiated by cortical inputs via the cortico-TC-nRT-TC pathway. These results reveal a previously unknown mode of cortico-thalamo-cortical transmission, bypassing direct cortico-nRT excitation, and describe a mechanism for pathological oscillation generation. This mode could be active under other circumstances, representing a previously unknown channel of cortico-thalamo-cortical information processing.

    View details for DOI 10.1038/nn.2896

    View details for Web of Science ID 000294284900017

    View details for PubMedID 21857658

    View details for PubMedCentralID PMC3308017

  • Optogenetic Interrogation of Dopaminergic Modulation of the Multiple Phases of Reward-Seeking Behavior JOURNAL OF NEUROSCIENCE Adamantidis, A. R., Tsai, H., Boutrel, B., Zhang, F., Stuber, G. D., Budygin, E. A., Tourino, C., Bonci, A., Deisseroth, K., de Lecea, L. 2011; 31 (30): 10829-10835

    Abstract

    Phasic activation of dopaminergic neurons is associated with reward-predicting cues and supports learning during behavioral adaptation. While noncontingent activation of dopaminergic neurons in the ventral tegmental are (VTA) is sufficient for passive behavioral conditioning, it remains unknown whether the phasic dopaminergic signal is truly reinforcing. In this study, we first targeted the expression of channelrhodopsin-2 to dopaminergic neurons of the VTA and optimized optogenetically evoked dopamine transients. Second, we showed that phasic activation of dopaminergic neurons in freely moving mice causally enhances positive reinforcing actions in a food-seeking operant task. Interestingly, such effect was not found in the absence of food reward. We further found that phasic activation of dopaminergic neurons is sufficient to reactivate previously extinguished food-seeking behavior in the absence of external cues. This was also confirmed using a single-session reversal paradigm. Collectively, these data suggest that activation of dopaminergic neurons facilitates the development of positive reinforcement during reward-seeking and behavioral flexibility.

    View details for DOI 10.1523/JNEUROSCI.2246-11.2011

    View details for Web of Science ID 000293171900010

    View details for PubMedID 21795535

    View details for PubMedCentralID PMC3171183

  • Excitatory transmission from the amygdala to nucleus accumbens facilitates reward seeking NATURE Stuber, G. D., Sparta, D. R., Stamatakis, A. M., van Leeuwen, W. A., Hardjoprajitno, J. E., Cho, S., Tye, K. M., Kempadoo, K. A., Zhang, F., Deisseroth, K., Bonci, A. 2011; 475 (7356): 377-U129

    Abstract

    The basolateral amygdala (BLA) has a crucial role in emotional learning irrespective of valence. The BLA projection to the nucleus accumbens (NAc) is thought to modulate cue-triggered motivated behaviours, but our understanding of the interaction between these two brain regions has been limited by the inability to manipulate neural-circuit elements of this pathway selectively during behaviour. To circumvent this limitation, we used in vivo optogenetic stimulation or inhibition of glutamatergic fibres from the BLA to the NAc, coupled with intracranial pharmacology and ex vivo electrophysiology. Here we show that optical stimulation of the pathway from the BLA to the NAc in mice reinforces behavioural responding to earn additional optical stimulation of these synaptic inputs. Optical stimulation of these glutamatergic fibres required intra-NAc dopamine D1-type receptor signalling, but not D2-type receptor signalling. Brief optical inhibition of fibres from the BLA to the NAc reduced cue-evoked intake of sucrose, demonstrating an important role of this specific pathway in controlling naturally occurring reward-related behaviour. Moreover, although optical stimulation of glutamatergic fibres from the medial prefrontal cortex to the NAc also elicited reliable excitatory synaptic responses, optical self-stimulation behaviour was not observed by activation of this pathway. These data indicate that whereas the BLA is important for processing both positive and negative affect, the glutamatergic pathway from the BLA to the NAc, in conjunction with dopamine signalling in the NAc, promotes motivated behavioural responding. Thus, optogenetic manipulation of anatomically distinct synaptic inputs to the NAc reveals functionally distinct properties of these inputs in controlling reward-seeking behaviours.

    View details for DOI 10.1038/nature10194

    View details for Web of Science ID 000292911200042

    View details for PubMedID 21716290

  • OPTOGENETICS: BACKGROUND AND CONCEPTS FOR NEUROSURGERY NEUROSURGERY Lin, S., Deisseroth, K., Henderson, J. M. 2011; 69 (1): 1-3

    View details for DOI 10.1227/NEU.0b013e318224688e

    View details for PubMedID 21792118

  • Challenges and Opportunities for Next-Generation Intracortically Based Neural Prostheses IEEE TRANSACTIONS ON BIOMEDICAL ENGINEERING Gilja, V., Chestek, C. A., Diester, I., Henderson, J. M., Deisseroth, K., Shenoy, K. V. 2011; 58 (7): 1891-1899

    Abstract

    Neural prosthetic systems aim to help disabled patients by translating neural signals from the brain into control signals for guiding computer cursors, prosthetic arms, and other assistive devices. Intracortical electrode arrays measure action potentials and local field potentials from individual neurons, or small populations of neurons, in the motor cortices and can provide considerable information for controlling prostheses. Despite several compelling proof-of-concept laboratory animal experiments and an initial human clinical trial, at least three key challenges remain which, if left unaddressed, may hamper the translation of these systems into widespread clinical use. We review these challenges: achieving able-bodied levels of performance across tasks and across environments, achieving robustness across multiple decades, and restoring able-bodied quality proprioception and somatosensation. We also describe some emerging opportunities for meeting these challenges. If these challenges can be largely or fully met, intracortically based neural prostheses may achieve true clinical viability and help increasing numbers of disabled patients.

    View details for DOI 10.1109/TBME.2011.2107553

    View details for PubMedID 21257365

  • High-efficiency channelrhodopsins for fast neuronal stimulation at low light levels PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Berndt, A., Schoenenberger, P., Mattis, J., Tye, K. M., Deisseroth, K., Hegemann, P., Oertner, T. G. 2011; 108 (18): 7595-7600

    Abstract

    Channelrhodopsin-2 (ChR2) has become an indispensable tool in neuroscience, allowing precise induction of action potentials with short light pulses. A limiting factor for many optophysiological experiments is the relatively small photocurrent induced by ChR2. We screened a large number of ChR2 point mutants and discovered a dramatic increase in photocurrent amplitude after threonine-to-cysteine substitution at position 159. When we tested the T159C mutant in hippocampal pyramidal neurons, action potentials could be induced at very low light intensities, where currently available channelrhodopsins were unable to drive spiking. Biophysical characterization revealed that the kinetics of most ChR2 variants slows down considerably at depolarized membrane potentials. We show that the recently published E123T substitution abolishes this voltage sensitivity and speeds up channel kinetics. When we combined T159C with E123T, the resulting double mutant delivered fast photocurrents with large amplitudes and increased the precision of single action potential induction over a broad range of frequencies, suggesting it may become the standard for light-controlled activation of neurons.

    View details for DOI 10.1073/pnas.1017210108

    View details for Web of Science ID 000290203100063

    View details for PubMedID 21504945

    View details for PubMedCentralID PMC3088623

  • Functional Integration of Grafted Neural Stem Cell-Derived Dopaminergic Neurons Monitored by Optogenetics in an In Vitro Parkinson Model PLOS ONE Tonnesen, J., Parish, C. L., Sorensen, A. T., Andersson, A., Lundberg, C., Deisseroth, K., Arenas, E., Lindvall, O., Kokaia, M. 2011; 6 (3)

    Abstract

    Intrastriatal grafts of stem cell-derived dopamine (DA) neurons induce behavioral recovery in animal models of Parkinson's disease (PD), but how they functionally integrate in host neural circuitries is poorly understood. Here, Wnt5a-overexpressing neural stem cells derived from embryonic ventral mesencephalon of tyrosine hydroxylase-GFP transgenic mice were expanded as neurospheres and transplanted into organotypic cultures of wild type mouse striatum. Differentiated GFP-labeled DA neurons in the grafts exhibited mature neuronal properties, including spontaneous firing of action potentials, presence of post-synaptic currents, and functional expression of DA D₂ autoreceptors. These properties resembled those recorded from identical cells in acute slices of intrastriatal grafts in the 6-hydroxy-DA-induced mouse PD model and from DA neurons in intact substantia nigra. Optogenetic activation or inhibition of grafted cells and host neurons using channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), respectively, revealed complex, bi-directional synaptic interactions between grafted cells and host neurons and extensive synaptic connectivity within the graft. Our data demonstrate for the first time using optogenetics that ectopically grafted stem cell-derived DA neurons become functionally integrated in the DA-denervated striatum. Further optogenetic dissection of the synaptic wiring between grafted and host neurons will be crucial to clarify the cellular and synaptic mechanisms underlying behavioral recovery as well as adverse effects following stem cell-based DA cell replacement strategies in PD.

    View details for DOI 10.1371/journal.pone.0017560

    View details for Web of Science ID 000288025500016

    View details for PubMedID 21394212

    View details for PubMedCentralID PMC3048875

  • An optogenetic toolbox designed for primates NATURE NEUROSCIENCE Diester, I., Kaufman, M. T., Mogri, M., Pashaie, R., Goo, W., Yizhar, O., Ramakrishnan, C., Deisseroth, K., Shenoy, K. V. 2011; 14 (3): 387-397

    Abstract

    Optogenetics is a technique for controlling subpopulations of neurons in the intact brain using light. This technique has the potential to enhance basic systems neuroscience research and to inform the mechanisms and treatment of brain injury and disease. Before launching large-scale primate studies, the method needs to be further characterized and adapted for use in the primate brain. We assessed the safety and efficiency of two viral vector systems (lentivirus and adeno-associated virus), two human promoters (human synapsin (hSyn) and human thymocyte-1 (hThy-1)) and three excitatory and inhibitory mammalian codon-optimized opsins (channelrhodopsin-2, enhanced Natronomonas pharaonis halorhodopsin and the step-function opsin), which we characterized electrophysiologically, histologically and behaviorally in rhesus monkeys (Macaca mulatta). We also introduced a new device for measuring in vivo fluorescence over time, allowing minimally invasive assessment of construct expression in the intact brain. We present a set of optogenetic tools designed for optogenetic experiments in the non-human primate brain.

    View details for DOI 10.1038/nn.2749

    View details for Web of Science ID 000287650100021

    View details for PubMedID 21278729

    View details for PubMedCentralID PMC3150193

  • Active Expiration Induced by Excitation of Ventral Medulla in Adult Anesthetized Rats JOURNAL OF NEUROSCIENCE Pagliardini, S., Janczewski, W. A., Tan, W., Dickson, C. T., Deisseroth, K., Feldman, J. L. 2011; 31 (8): 2895-2905

    Abstract

    Data from perinatal and juvenile rodents support our hypothesis that the preBötzinger complex generates inspiratory rhythm and the retrotrapezoid nucleus-parafacial respiratory group (RTN/pFRG) generates active expiration (AE). Although the role of the RTN/pFRG in adulthood is disputed, we hypothesized that its rhythmogenicity persists but is typically silenced by synaptic inhibition. We show in adult anesthetized rats that local pharmacological disinhibition or optogenetic excitation of the RTN/pFRG can generate AE and transforms previously silent RTN/pFRG neurons into rhythmically active cells whose firing is correlated with late-phase active expiration. Brief excitatory stimuli also reset the respiratory rhythm, indicating strong coupling of AE to inspiration. The AE network location in adult rats overlaps with the perinatal pFRG and appears lateral to the chemosensitive region of adult RTN. We suggest that (1) the RTN/pFRG contains a conditional oscillator that generates AE, and (2) at rest and in anesthesia, synaptic inhibition of RTN/pFRG suppresses AE.

    View details for DOI 10.1523/JNEUROSCI.5338-10.2011

    View details for Web of Science ID 000287670100018

    View details for PubMedID 21414911

    View details for PubMedCentralID PMC3142740

  • In vitro and In silico Optogenetic Control of Differentiated Human Pluripotent Stem Cells Abilez, O., Prakash, R., Wong, J., Kuhl, E., Deisseroth, K., Zarins, C. CELL PRESS. 2011: 368
  • An Implantable Optical Stimulation Delivery System for Actuating an Excitable Biosubstrate IEEE JOURNAL OF SOLID-STATE CIRCUITS Paralikar, K., Cong, P., Yizhar, O., Fenno, L. E., Santa, W., Nielsen, C., Dinsmoor, D., Hocken, B., Munns, G. O., Giftakis, J., Deisseroth, K., Denison, T. 2011; 46 (1): 321-332
  • Approaches to Optical Neuromodulation from Rodents to Non-Human Primates by Integrated Optoelectronic Devices 33rd Annual International Conference of the IEEE Engineering-in-Medicine-and-Biology-Society (EMBS) Wang, J., Ozden, I., Diagne, M., Wagner, F., Borton, D., Brush, B., Agha, N., Burwell, R., Sheinberg, D., Diester, I., Deisseroth, K., Nurmikko, A. IEEE. 2011: 7525–7528

    Abstract

    Methods on rendering neurons in the central nervous system to be light responsive has led to a boom in using optical neuromodulation as a new approach for controlling brain states and understanding neural circuits. In addition to the developing versatility to "optogenetically" labeling of neural cells and their subtypes by microbiological methods, parallel efforts are under way to design and implement optoelectronic devices to achieve simultaneous optical neuromodulation and electrophysiological recording with high spatial and temporal resolution. Such new device-based technologies need to be developed for full exploitation of the promise of optogenetics. In this paper we present single- and multi-element optoelectronic devices developed in our laboratories. The single-unit element, namely the coaxial optrode, was utilized to characterize the neural responses in optogenetically modified rodent and primate models. Furthermore, the multi-element device, integrating the optrode with a 6×6 microelectrode array, was used to characterize the spatiotemporal spread of neural activity in response to single-site optical stimulation in freely moving rats. We suggest that the particular approaches we employed can lead to the emergence of methods where spatio-temporal optical modulation is integrated with real-time read out from neural populations.

    View details for Web of Science ID 000298810005260

    View details for PubMedID 22256079

  • Optogenetics. Nat Methods. Deisseroth, K. 2011; 1 (8): 26-9
  • Amygdala circuitry mediating reversible and bidirectional control of anxiety. Nature. Tye, K. M., Prakash, R., Kim, S. Y., Fenno, L. E., Grosenick, L., Zarabi, H., Deisseroth, K. 2011; 7338 (471): 358-62
  • High-efficiency channelrhodopsins for fast neuronal stimulation at low light levels. A, B., P, S., J, M., KM, T., K, D., P, H. 2011
  • Tracking Stem Cell Differentiation in the Setting of Automated Optogenetic Stimulation STEM CELLS Stroh, A., Tsai, H., Wang, L., Zhang, F., Kressel, J., Aravanis, A., Santhanam, N., Deisseroth, K., Konnerth, A., Schneider, M. B. 2011; 29 (1): 78-88

    Abstract

    Membrane depolarization has been shown to play an important role in the neural differentiation of stem cells and in the survival and function of mature neurons. Here, we introduce a microbial opsin into ESCs and develop optogenetic technology for stem cell engineering applications, with an automated system for noninvasive modulation of ESC differentiation employing fast optogenetic control of ion flux. Mouse ESCs were stably transduced with channelrhodopsin-2 (ChR2)-yellow fluorescent protein and purified by fluorescence activated cell sorting (FACS). Illumination of resulting ChR2-ESCs with pulses of blue light triggered inward currents. These labeled ESCs retained the capability to differentiate into functional mature neurons, assessed by the presence of voltage-gated sodium currents, action potentials, fast excitatory synaptic transmission, and expression of mature neuronal proteins and neuronal morphology. We designed and tested an apparatus for optically stimulating ChR2-ESCs during chronic neuronal differentiation, with high-speed optical switching on a custom robotic stage with environmental chamber for automated stimulation and imaging over days, with tracking for increased expression of neural and neuronal markers. These data point to potential uses of ChR2 technology for chronic and temporally precise noninvasive optical control of ESCs both in vitro and in vivo, ranging from noninvasive control of stem cell differentiation to causal assessment of the specific contribution of transplanted cells to tissue and network function.

    View details for DOI 10.1002/stem.558

    View details for Web of Science ID 000286659300010

    View details for PubMedID 21280159

  • Drug-Driven AMPA Receptor Redistribution Mimicked by Selective Dopamine Neuron Stimulation PLOS ONE Brown, M. T., Bellone, C., Mameli, M., Labouebe, G., Bocklisch, C., Balland, B., Dahan, L., Lujan, R., Deisseroth, K., Luescher, C. 2010; 5 (12)

    Abstract

    Addictive drugs have in common that they cause surges in dopamine (DA) concentration in the mesolimbic reward system and elicit synaptic plasticity in DA neurons of the ventral tegmental area (VTA). Cocaine for example drives insertion of GluA2-lacking AMPA receptors (AMPARs) at glutamatergic synapes in DA neurons. However it remains elusive which molecular target of cocaine drives such AMPAR redistribution and whether other addictive drugs (morphine and nicotine) cause similar changes through their effects on the mesolimbic DA system.We used in vitro electrophysiological techniques in wild-type and transgenic mice to observe the modulation of excitatory inputs onto DA neurons by addictive drugs. To observe AMPAR redistribution, post-embedding immunohistochemistry for GluA2 AMPAR subunit was combined with electron microscopy. We also used a double-floxed AAV virus expressing channelrhodopsin together with a DAT Cre mouse line to selectively express ChR2 in VTA DA neurons. We find that in mice where the effect of cocaine on the dopamine transporter (DAT) is specifically blocked, AMPAR redistribution was absent following administration of the drug. Furthermore, addictive drugs known to increase dopamine levels cause a similar AMPAR redistribution. Finally, activating DA VTA neurons optogenetically is sufficient to drive insertion of GluA2-lacking AMPARs, mimicking the changes observed after a single injection of morphine, nicotine or cocaine.We propose the mesolimbic dopamine system as a point of convergence at which addictive drugs can alter neural circuits. We also show that direct activation of DA neurons is sufficient to drive AMPAR redistribution, which may be a mechanism associated with early steps of non-substance related addictions.

    View details for DOI 10.1371/journal.pone.0015870

    View details for Web of Science ID 000285838900044

    View details for PubMedID 21209835

    View details for PubMedCentralID PMC3013137

  • Tuning arousal with optogenetic modulation of locus coeruleus neurons NATURE NEUROSCIENCE Carter, M. E., Yizhar, O., Chikahisa, S., Nguyen, H., Adamantidis, A., Nishino, S., Deisseroth, K., de Lecea, L. 2010; 13 (12): 1526-U117

    Abstract

    Neural activity in the noradrenergic locus coeruleus correlates with periods of wakefulness and arousal. However, it is unclear whether tonic or phasic activity in these neurons is necessary or sufficient to induce transitions between behavioral states and to promote long-term arousal. Using optogenetic tools in mice, we found that there is a frequency-dependent, causal relationship among locus coeruleus firing, cortical activity, sleep-to-wake transitions and general locomotor arousal. We also found that sustained, high-frequency stimulation of the locus coeruleus at frequencies of 5 Hz and above caused reversible behavioral arrests. These results suggest that the locus coeruleus is finely tuned to regulate organismal arousal and that bursts of noradrenergic overexcitation cause behavioral attacks that resemble those seen in people with neuropsychiatric disorders.

    View details for DOI 10.1038/nn.2682

    View details for Web of Science ID 000284525800018

    View details for PubMedID 21037585

    View details for PubMedCentralID PMC3174240

  • Antidepressant Effect of Optogenetic Stimulation of the Medial Prefrontal Cortex JOURNAL OF NEUROSCIENCE Covington, H. E., Lobo, M. K., Maze, I., Vialou, V., Hyman, J. M., Zaman, S., LaPlant, Q., Mouzon, E., Ghose, S., Tamminga, C. A., Neve, R. L., Deisseroth, K., Nestler, E. J. 2010; 30 (48): 16082-16090

    Abstract

    Brain stimulation and imaging studies in humans have highlighted a key role for the prefrontal cortex in clinical depression; however, it remains unknown whether excitation or inhibition of prefrontal cortical neuronal activity is associated with antidepressant responses. Here, we examined cellular indicators of functional activity, including the immediate early genes (IEGs) zif268 (egr1), c-fos, and arc, in the prefrontal cortex of clinically depressed humans obtained postmortem. We also examined these genes in the ventral portion of the medial prefrontal cortex (mPFC) of mice after chronic social defeat stress, a mouse model of depression. In addition, we used viral vectors to overexpress channel rhodopsin 2 (a light-activated cation channel) in mouse mPFC to optogenetically drive "burst" patterns of cortical firing in vivo and examine the behavioral consequences. Prefrontal cortical tissue derived from clinically depressed humans displayed significant reductions in IEG expression, consistent with a deficit in neuronal activity within this brain region. Mice subjected to chronic social defeat stress exhibited similar reductions in levels of IEG expression in mPFC. Interestingly, some of these changes were not observed in defeated mice that escape the deleterious consequences of the stress, i.e., resilient animals. In those mice that expressed a strong depressive-like phenotype, i.e., susceptible animals, optogenetic stimulation of mPFC exerted potent antidepressant-like effects, without affecting general locomotor activity, anxiety-like behaviors, or social memory. These results indicate that the activity of the mPFC is a key determinant of depression-like behavior, as well as antidepressant responses.

    View details for DOI 10.1523/JNEUROSCI.1731-10.2010

    View details for Web of Science ID 000284999900003

    View details for PubMedID 21123555

    View details for PubMedCentralID PMC3004756

  • Encoding of conditioned fear in central amygdala inhibitory circuits NATURE Ciocchi, S., Herry, C., Grenier, F., Wolff, S. B., Letzkus, J. J., Vlachos, I., Ehrlich, I., Sprengel, R., Deisseroth, K., Stadler, M. B., Mueller, C., Luethi, A. 2010; 468 (7321): 277-U239

    Abstract

    The central amygdala (CEA), a nucleus predominantly composed of GABAergic inhibitory neurons, is essential for fear conditioning. How the acquisition and expression of conditioned fear are encoded within CEA inhibitory circuits is not understood. Using in vivo electrophysiological, optogenetic and pharmacological approaches in mice, we show that neuronal activity in the lateral subdivision of the central amygdala (CEl) is required for fear acquisition, whereas conditioned fear responses are driven by output neurons in the medial subdivision (CEm). Functional circuit analysis revealed that inhibitory CEA microcircuits are highly organized and that cell-type-specific plasticity of phasic and tonic activity in the CEl to CEm pathway may gate fear expression and regulate fear generalization. Our results define the functional architecture of CEA microcircuits and their role in the acquisition and regulation of conditioned fear behaviour.

    View details for DOI 10.1038/nature09559

    View details for Web of Science ID 000284051000045

    View details for PubMedID 21068837

  • Genetic dissection of an amygdala microcircuit that gates conditioned fear NATURE Haubensak, W., Kunwar, P. S., Cai, H., Ciocchi, S., Wall, N. R., Ponnusamy, R., Biag, J., Dong, H., Deisseroth, K., Callaway, E. M., Fanselow, M. S., Luethi, A., Anderson, D. J. 2010; 468 (7321): 270-U230

    Abstract

    The role of different amygdala nuclei (neuroanatomical subdivisions) in processing Pavlovian conditioned fear has been studied extensively, but the function of the heterogeneous neuronal subtypes within these nuclei remains poorly understood. Here we use molecular genetic approaches to map the functional connectivity of a subpopulation of GABA-containing neurons, located in the lateral subdivision of the central amygdala (CEl), which express protein kinase C-δ (PKC-δ). Channelrhodopsin-2-assisted circuit mapping in amygdala slices and cell-specific viral tracing indicate that PKC-δ(+) neurons inhibit output neurons in the medial central amygdala (CEm), and also make reciprocal inhibitory synapses with PKC-δ(-) neurons in CEl. Electrical silencing of PKC-δ(+) neurons in vivo suggests that they correspond to physiologically identified units that are inhibited by the conditioned stimulus, called CEl(off) units. This correspondence, together with behavioural data, defines an inhibitory microcircuit in CEl that gates CEm output to control the level of conditioned freezing.

    View details for DOI 10.1038/nature09553

    View details for Web of Science ID 000284051000044

    View details for PubMedID 21068836

    View details for PubMedCentralID PMC3597095

  • Functional Control of Transplantable Human ESC-Derived Neurons via Optogenetic Targeting STEM CELLS Weick, J. P., Johnson, M. A., Skroch, S. P., Williams, J. C., Deisseroth, K., Zhang, S. 2010; 28 (11): 2008-2016

    Abstract

    Current methods to examine and regulate the functional integration and plasticity of human ESC (hESC)-derived neurons are cumbersome and technically challenging. Here, we engineered hESCs and their derivatives to express the light-gated channelrhodopsin-2 (ChR2) protein to overcome these deficiencies. Optogenetic targeting of hESC-derived neurons with ChR2 linked to the mCherry fluorophore allowed reliable cell tracking as well as light-induced spiking at physiological frequencies. Optically induced excitatory and inhibitory postsynaptic currents could be elicited in either ChR2(+) or ChR2(-) neurons in vitro and in acute brain slices taken from transplanted severe combined immunodeficient (SCID) mice. Furthermore, we created a clonal hESC line that expresses ChR2-mCherry under the control of the synapsin-1 promoter. On neuronal differentiation, ChR2-mCherry expression was restricted to neurons and was stably expressed for at least 6 months, providing more predictable light-induced currents than transient infections. This pluripotent cell line will allow both in vitro and in vivo analysis of functional development as well as the integration capacity of neuronal populations for cell-replacement strategies.

    View details for DOI 10.1002/stem.514

    View details for Web of Science ID 000284395900011

    View details for PubMedID 20827747

    View details for PubMedCentralID PMC2988875

  • Controlling the brain with light. Scientific American Deisseroth, K. 2010; 303 (5): 48-55

    View details for PubMedID 21033283

  • Cell Type-Specific Loss of BDNF Signaling Mimics Optogenetic Control of Cocaine Reward SCIENCE Lobo, M. K., Covington, H. E., Chaudhury, D., Friedman, A. K., Sun, H., Damez-Werno, D., Dietz, D. M., Zaman, S., Koo, J. W., Kennedy, P. J., Mouzon, E., Mogri, M., Neve, R. L., Deisseroth, K., Han, M., Nestler, E. J. 2010; 330 (6002): 385-390

    Abstract

    The nucleus accumbens is a key mediator of cocaine reward, but the distinct roles of the two subpopulations of nucleus accumbens projection neurons, those expressing dopamine D1 versus D2 receptors, are poorly understood. We show that deletion of TrkB, the brain-derived neurotrophic factor (BDNF) receptor, selectively from D1+ or D2+ neurons oppositely affects cocaine reward. Because loss of TrkB in D2+ neurons increases their neuronal excitability, we next used optogenetic tools to control selectively the firing rate of D1+ and D2+ nucleus accumbens neurons and studied consequent effects on cocaine reward. Activation of D2+ neurons, mimicking the loss of TrkB, suppresses cocaine reward, with opposite effects induced by activation of D1+ neurons. These results provide insight into the molecular control of D1+ and D2+ neuronal activity as well as the circuit-level contribution of these cell types to cocaine reward.

    View details for DOI 10.1126/science.1188472

    View details for Web of Science ID 000282986700045

    View details for PubMedID 20947769

    View details for PubMedCentralID PMC3011229

  • Orderly recruitment of motor units under optical control in vivo NATURE MEDICINE Llewellyn, M. E., Thompson, K. R., Deisseroth, K., Delp, S. L. 2010; 16 (10): 1161-U144

    Abstract

    A drawback of electrical stimulation for muscle control is that large, fatigable motor units are preferentially recruited before smaller motor units by the lowest-intensity electrical cuff stimulation. This phenomenon limits therapeutic applications because it is precisely the opposite of the normal physiological (orderly) recruitment pattern; therefore, a mechanism to achieve orderly recruitment has been a long-sought goal in physiology, medicine and engineering. Here we demonstrate a technology for reliable orderly recruitment in vivo. We find that under optical control with microbial opsins, recruitment of motor units proceeds in the physiological recruitment sequence, as indicated by multiple independent measures of motor unit recruitment including conduction latency, contraction and relaxation times, stimulation threshold and fatigue. As a result, we observed enhanced performance and reduced fatigue in vivo. These findings point to an unanticipated new modality of neural control with broad implications for nervous system and neuromuscular physiology, disease research and therapeutic innovation.

    View details for DOI 10.1038/nm.2228

    View details for Web of Science ID 000282644800049

    View details for PubMedID 20871612

  • Astrocytes Control Breathing Through pH-Dependent Release of ATP SCIENCE Gourine, A. V., Kasymov, V., Marina, N., Tang, F., Figueiredo, M. F., Lane, S., Teschemacher, A. G., Spyer, K. M., Deisseroth, K., Kasparov, S. 2010; 329 (5991): 571-575

    Abstract

    Astrocytes provide structural and metabolic support for neuronal networks, but direct evidence demonstrating their active role in complex behaviors is limited. Central respiratory chemosensitivity is an essential mechanism that, via regulation of breathing, maintains constant levels of blood and brain pH and partial pressure of CO2. We found that astrocytes of the brainstem chemoreceptor areas are highly chemosensitive. They responded to physiological decreases in pH with vigorous elevations in intracellular Ca2+ and release of adenosine triphosphate (ATP). ATP propagated astrocytic Ca2+ excitation, activated chemoreceptor neurons, and induced adaptive increases in breathing. Mimicking pH-evoked Ca2+ responses by means of optogenetic stimulation of astrocytes expressing channelrhodopsin-2 activated chemoreceptor neurons via an ATP-dependent mechanism and triggered robust respiratory responses in vivo. This demonstrates a potentially crucial role for brain glial cells in mediating a fundamental physiological reflex.

    View details for DOI 10.1126/science.1190721

    View details for Web of Science ID 000280483500039

    View details for PubMedID 20647426

    View details for PubMedCentralID PMC3160742

  • Regulation of parkinsonian motor behaviours by optogenetic control of basal ganglia circuitry NATURE Kravitz, A. V., Freeze, B. S., Parker, P. R., Kay, K., Thwin, M. T., Deisseroth, K., Kreitzer, A. C. 2010; 466 (7306): 622-U7

    Abstract

    Neural circuits of the basal ganglia are critical for motor planning and action selection. Two parallel basal ganglia pathways have been described, and have been proposed to exert opposing influences on motor function. According to this classical model, activation of the 'direct' pathway facilitates movement and activation of the 'indirect' pathway inhibits movement. However, more recent anatomical and functional evidence has called into question the validity of this hypothesis. Because this model has never been empirically tested, the specific function of these circuits in behaving animals remains unknown. Here we report direct activation of basal ganglia circuitry in vivo, using optogenetic control of direct- and indirect-pathway medium spiny projection neurons (MSNs), achieved through Cre-dependent viral expression of channelrhodopsin-2 in the striatum of bacterial artificial chromosome transgenic mice expressing Cre recombinase under control of regulatory elements for the dopamine D1 or D2 receptor. Bilateral excitation of indirect-pathway MSNs elicited a parkinsonian state, distinguished by increased freezing, bradykinesia and decreased locomotor initiations. In contrast, activation of direct-pathway MSNs reduced freezing and increased locomotion. In a mouse model of Parkinson's disease, direct-pathway activation completely rescued deficits in freezing, bradykinesia and locomotor initiation. Taken together, our findings establish a critical role for basal ganglia circuitry in the bidirectional regulation of motor behaviour and indicate that modulation of direct-pathway circuitry may represent an effective therapeutic strategy for ameliorating parkinsonian motor deficits.

    View details for DOI 10.1038/nature09159

    View details for Web of Science ID 000280412100053

    View details for PubMedID 20613723

    View details for PubMedCentralID PMC3552484

  • Genetic Reactivation of Cone Photoreceptors Restores Visual Responses in Retinitis Pigmentosa SCIENCE Busskamp, V., Duebel, J., Balya, D., Fradot, M., Viney, T. J., Siegert, S., Groner, A. C., Cabuy, E., Forster, V., Seeliger, M., Biel, M., Humphries, P., Paques, M., Mohand-Said, S., Trono, D., Deisseroth, K., Sahel, J. A., Picaud, S., Roska, B. 2010; 329 (5990): 413-417

    Abstract

    Retinitis pigmentosa refers to a diverse group of hereditary diseases that lead to incurable blindness, affecting two million people worldwide. As a common pathology, rod photoreceptors die early, whereas light-insensitive, morphologically altered cone photoreceptors persist longer. It is unknown if these cones are accessible for therapeutic intervention. Here, we show that expression of archaebacterial halorhodopsin in light-insensitive cones can substitute for the native phototransduction cascade and restore light sensitivity in mouse models of retinitis pigmentosa. Resensitized photoreceptors activate all retinal cone pathways, drive sophisticated retinal circuit functions (including directional selectivity), activate cortical circuits, and mediate visually guided behaviors. Using human ex vivo retinas, we show that halorhodopsin can reactivate light-insensitive human photoreceptors. Finally, we identified blind patients with persisting, light-insensitive cones for potential halorhodopsin-based therapy.

    View details for DOI 10.1126/science.1190897

    View details for Web of Science ID 000280196500030

    View details for PubMedID 20576849

  • Optical activation of lateral amygdala pyramidal cells instructs associative fear learning PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Johansen, J. P., Hamanaka, H., Monfils, M. H., Behnia, R., Deisseroth, K., Blair, H. T., LeDoux, J. E. 2010; 107 (28): 12692-12697

    Abstract

    Humans and animals can learn that specific sensory cues in the environment predict aversive events through a form of associative learning termed fear conditioning. This learning occurs when the sensory cues are paired with an aversive event occurring in close temporal proximity. Activation of lateral amygdala (LA) pyramidal neurons by aversive stimuli is thought to drive the formation of these associative fear memories; yet, there have been no direct tests of this hypothesis. Here we demonstrate that viral-targeted, tissue-specific expression of the light-activated channelrhodopsin (ChR2) in LA pyramidal cells permitted optical control of LA neuronal activity. Using this approach we then paired an auditory sensory cue with optical stimulation of LA pyramidal neurons instead of an aversive stimulus. Subsequently presentation of the tone alone produced behavioral fear responses. These results demonstrate in vivo optogenetic control of LA neurons and provide compelling support for the idea that fear learning is instructed by aversive stimulus-induced activation of LA pyramidal cells.

    View details for DOI 10.1073/pnas.1002418107

    View details for Web of Science ID 000279843200054

    View details for PubMedID 20615999

    View details for PubMedCentralID PMC2906568

  • Global and local fMRI signals driven by neurons defined optogenetically by type and wiring NATURE Lee, J. H., Durand, R., Gradinaru, V., Zhang, F., Goshen, I., Kim, D., Fenno, L. E., Ramakrishnan, C., Deisseroth, K. 2010; 465 (7299): 788-792

    Abstract

    Despite a rapidly-growing scientific and clinical brain imaging literature based on functional magnetic resonance imaging (fMRI) using blood oxygenation level-dependent (BOLD) signals, it remains controversial whether BOLD signals in a particular region can be caused by activation of local excitatory neurons. This difficult question is central to the interpretation and utility of BOLD, with major significance for fMRI studies in basic research and clinical applications. Using a novel integrated technology unifying optogenetic control of inputs with high-field fMRI signal readouts, we show here that specific stimulation of local CaMKIIalpha-expressing excitatory neurons, either in the neocortex or thalamus, elicits positive BOLD signals at the stimulus location with classical kinetics. We also show that optogenetic fMRI (of MRI) allows visualization of the causal effects of specific cell types defined not only by genetic identity and cell body location, but also by axonal projection target. Finally, we show that of MRI within the living and intact mammalian brain reveals BOLD signals in downstream targets distant from the stimulus, indicating that this approach can be used to map the global effects of controlling a local cell population. In this respect, unlike both conventional fMRI studies based on correlations and fMRI with electrical stimulation that will also directly drive afferent and nearby axons, this of MRI approach provides causal information about the global circuits recruited by defined local neuronal activity patterns. Together these findings provide an empirical foundation for the widely-used fMRI BOLD signal, and the features of of MRI define a potent tool that may be suitable for functional circuit analysis as well as global phenotyping of dysfunctional circuitry.

    View details for DOI 10.1038/nature09108

    View details for Web of Science ID 000278551800047

    View details for PubMedID 20473285

    View details for PubMedCentralID PMC3177305

  • Glutamatergic Signaling by Mesolimbic Dopamine Neurons in the Nucleus Accumbens JOURNAL OF NEUROSCIENCE Tecuapetla, F., Patel, J. C., Xenias, H., English, D., Tadros, I., Shah, F., Berlin, J., Deisseroth, K., Rice, M. E., Tepper, J. M., Koos, T. 2010; 30 (20): 7105-7110

    Abstract

    Recent evidence suggests the intriguing possibility that midbrain dopaminergic (DAergic) neurons may use fast glutamatergic transmission to communicate with their postsynaptic targets. Because of technical limitations, direct demonstration of the existence of this signaling mechanism has been limited to experiments using cell culture preparations that often alter neuronal function including neurotransmitter phenotype. Consequently, it remains uncertain whether glutamatergic signaling between DAergic neurons and their postsynaptic targets exists under physiological conditions. Here, using an optogenetic approach, we provide the first conclusive demonstration that mesolimbic DAergic neurons in mice release glutamate and elicit excitatory postsynaptic responses in projection neurons of the nucleus accumbens. In addition, we describe the properties of the postsynaptic glutamatergic responses of these neurons during experimentally evoked burst firing of DAergic axons that reproduce the reward-related phasic population activity of the mesolimbic projection. These observations indicate that, in addition to DAergic mechanisms, mesolimbic reward signaling may involve glutamatergic transmission.

    View details for DOI 10.1523/JNEUROSCI.0265-10.2010

    View details for Web of Science ID 000277844700032

    View details for PubMedID 20484653

  • Dlx5 and Dlx6 Regulate the Development of Parvalbumin-Expressing Cortical Interneurons JOURNAL OF NEUROSCIENCE Wang, Y., Dye, C. A., Sohal, V., Long, J. E., Estrada, R. C., Roztocil, T., Lufkin, T., Deisseroth, K., Baraban, S. C., Rubenstein, J. L. 2010; 30 (15): 5334-5345

    Abstract

    Dlx5 and Dlx6 homeobox genes are expressed in developing and mature cortical interneurons. Simultaneous deletion of Dlx5 and 6 results in exencephaly of the anterior brain; despite this defect, prenatal basal ganglia differentiation appeared largely intact, while tangential migration of Lhx6(+) and Mafb(+) interneurons to the cortex was reduced and disordered. The migration deficits were associated with reduced CXCR4 expression. Transplantation of mutant immature interneurons into a wild-type brain demonstrated that loss of either Dlx5 or Dlx5&6 preferentially reduced the number of mature parvalbumin(+) interneurons; those parvalbumin(+) interneurons that were present had increased dendritic branching. Dlx5/6(+/-) mice, which appear normal histologically, show spontaneous electrographic seizures and reduced power of gamma oscillations. Thus, Dlx5&6 appeared to be required for development and function of somal innervating (parvalbumin(+)) neocortical interneurons. This contrasts with Dlx1, whose function is required for dendrite innervating (calretinin(+), somatostatin(+), and neuropeptide Y(+)) interneurons (Cobos et al., 2005).

    View details for DOI 10.1523/JNEUROSCI.5963-09.2010

    View details for Web of Science ID 000276685100021

    View details for PubMedID 20392955

    View details for PubMedCentralID PMC2919857

  • Molecular and Cellular Approaches for Diversifying and Extending Optogenetics CELL Gradinaru, V., Zhang, F., Ramakrishnan, C., Mattis, J., Prakash, R., Diester, I., Goshen, I., Thompson, K. R., Deisseroth, K. 2010; 141 (1): 154-165

    Abstract

    Optogenetic technologies employ light to control biological processes within targeted cells in vivo with high temporal precision. Here, we show that application of molecular trafficking principles can expand the optogenetic repertoire along several long-sought dimensions. Subcellular and transcellular trafficking strategies now permit (1) optical regulation at the far-red/infrared border and extension of optogenetic control across the entire visible spectrum, (2) increased potency of optical inhibition without increased light power requirement (nanoampere-scale chloride-mediated photocurrents that maintain the light sensitivity and reversible, step-like kinetic stability of earlier tools), and (3) generalizable strategies for targeting cells based not only on genetic identity, but also on morphology and tissue topology, to allow versatile targeting when promoters are not known or in genetically intractable organisms. Together, these results illustrate use of cell-biological principles to enable expansion of the versatile fast optogenetic technologies suitable for intact-systems biology and behavior.

    View details for DOI 10.1016/j.cell.2010.02.037

    View details for Web of Science ID 000276211100020

    View details for PubMedID 20303157

  • Ultrafast optogenetic control NATURE NEUROSCIENCE Gunaydin, L. A., Yizhar, O., Berndt, A., Sohal, V. S., Deisseroth, K., Hegemann, P. 2010; 13 (3): 387-U27

    Abstract

    Channelrhodopsins such as channelrhodopsin-2 (ChR2) can drive spiking with millisecond precision in a wide variety of cells, tissues and animal species. However, several properties of this protein have limited the precision of optogenetic control. First, when ChR2 is expressed at high levels, extra spikes (for example, doublets) can occur in response to a single light pulse, with potential implications as doublets may be important for neural coding. Second, many cells cannot follow ChR2-driven spiking above the gamma (approximately 40 Hz) range in sustained trains, preventing temporally stationary optogenetic access to a broad and important neural signaling band. Finally, rapid optically driven spike trains can result in plateau potentials of 10 mV or more, causing incidental upstates with information-processing implications. We designed and validated an engineered opsin gene (ChETA) that addresses all of these limitations (profoundly reducing extra spikes, eliminating plateau potentials and allowing temporally stationary, sustained spike trains up to at least 200 Hz).

    View details for DOI 10.1038/nn.2495

    View details for Web of Science ID 000274860100022

    View details for PubMedID 20081849

  • Optogenetic interrogation of neural circuits: technology for probing mammalian brain structures NATURE PROTOCOLS Zhang, F., Gradinaru, V., Adamantidis, A. R., Durand, R., Airan, R. D., de Lecea, L., Deisseroth, K. 2010; 5 (3): 439-456

    Abstract

    Elucidation of the neural substrates underlying complex animal behaviors depends on precise activity control tools, as well as compatible readout methods. Recent developments in optogenetics have addressed this need, opening up new possibilities for systems neuroscience. Interrogation of even deep neural circuits can be conducted by directly probing the necessity and sufficiency of defined circuit elements with millisecond-scale, cell type-specific optical perturbations, coupled with suitable readouts such as electrophysiology, optical circuit dynamics measures and freely moving behavior in mammals. Here we collect in detail our strategies for delivering microbial opsin genes to deep mammalian brain structures in vivo, along with protocols for integrating the resulting optical control with compatible readouts (electrophysiological, optical and behavioral). The procedures described here, from initial virus preparation to systems-level functional readout, can be completed within 4-5 weeks. Together, these methods may help in providing circuit-level insight into the dynamics underlying complex mammalian behaviors in health and disease.

    View details for DOI 10.1038/nprot.2009.226

    View details for Web of Science ID 000275234900006

    View details for PubMedID 20203662

  • Controlling the brain with light Sci Am. Deisseroth, K. 2010; 5 (303): 48-55
  • Optical activation of lateral amygdala pyramidal cells instructs associative fear leaJohansen JP, Hamanaka H, Monfils MH, Behnia R, Deisseroth K, Blair HT, LeDoux JE. rning. JP, J., H, H., MH, M., R, B., K, D., HT, B. 2010
  • Special issue on optical neural engineering: advances in optical stimulation technology. J Neural Eng. Shoham, S., Deisseroth, K. 2010; 4 (7): 040201
  • Orderly recruitment of motor units under optical control in vivo. Nat Med. Llewellyn, M. E., Thompson, K. R., Deisseroth, K., Delp, S. L. 2010; 10 (16): 1161-5
  • Targeted optogenetic stimulation and recording of neurons in vivo using cell-type-specific expression of Channelrhodopsin-2 NATURE PROTOCOLS Cardin, J. A., Carlen, M., Meletis, K., Knoblich, U., Zhang, F., Deisseroth, K., Tsai, L., Moore, C. I. 2010; 5 (2): 247-254

    Abstract

    A major long-term goal of systems neuroscience is to identify the different roles of neural subtypes in brain circuit function. The ability to causally manipulate selective cell types is critical to meeting this goal. This protocol describes techniques for optically stimulating specific populations of excitatory neurons and inhibitory interneurons in vivo in combination with electrophysiology. Cell type selectivity is obtained using Cre-dependent expression of the light-activated channel Channelrhodopsin-2. We also describe approaches for minimizing optical interference with simultaneous extracellular and intracellular recording. These optogenetic techniques provide a spatially and temporally precise means of studying neural activity in the intact brain and allow a detailed examination of the effect of evoked activity on the surrounding local neural network. Injection of viral vectors requires 30-45 min, and in vivo electrophysiology with optogenetic stimulation requires 1-4 h.

    View details for DOI 10.1038/nprot.2009.228

    View details for Web of Science ID 000275204300007

    View details for PubMedID 20134425

    View details for PubMedCentralID PMC3655719

  • Optogenetic dissection of neuronal circuits in zebrafish using viral gene transfer and the Tet system FRONTIERS IN NEURAL CIRCUITS Zhu, P., Narita, Y., Bundschuh, S. T., Fajardo, O., Schaerer, Y. Z., Chattopadhyaya, B., Bouldoires, E. A., Stepien, A. E., Deisseroth, K., Arber, S., Sprengel, R., Rijli, F. M., Friedrich, R. W. 2009; 3

    Abstract

    The conditional expression of transgenes at high levels in sparse and specific populations of neurons is important for high-resolution optogenetic analyses of neuronal circuits. We explored two complementary methods, viral gene delivery and the iTet-Off system, to express transgenes in the brain of zebrafish. High-level gene expression in neurons was achieved by Sindbis and Rabies viruses. The Tet system produced strong and specific gene expression that could be modulated conveniently by doxycycline. Moreover, transgenic lines showed expression in distinct, sparse and stable populations of neurons that appeared to be subsets of the neurons targeted by the promoter driving the Tet-activator. The Tet system therefore provides the opportunity to generate libraries of diverse expression patterns similar to gene trap approaches or the thy-1 promoter in mice, but with the additional possibility to pre-select cell types of interest. In transgenic lines expressing channelrhodopsin-2, action potential firing could be precisely controlled by two-photon stimulation at low laser power, presumably because the expression levels of the Tet-controlled genes were high even in adults. In channelrhodopsin-2-expressing larvae, optical stimulation with a single blue LED evoked distinct swimming behaviors including backward swimming. These approaches provide new opportunities for the optogenetic dissection of neuronal circuit structure and function.

    View details for DOI 10.3389/neuro.04.021.2009

    View details for Web of Science ID 000207896400001

    View details for PubMedID 20126518

    View details for PubMedCentralID PMC2805431

  • Integrated device for optical stimulation and spatiotemporal electrical recording of neural activity in light-sensitized brain tissue JOURNAL OF NEURAL ENGINEERING Zhang, J., Laiwalla, F., Kim, J. A., Urabe, H., Van Wagenen, R., Song, Y., Connors, B. W., Zhang, F., Deisseroth, K., Nurmikko, A. V. 2009; 6 (5)

    Abstract

    Neural stimulation with high spatial and temporal precision is desirable both for studying the real-time dynamics of neural networks and for prospective clinical treatment of neurological diseases. Optical stimulation of genetically targeted neurons expressing the light sensitive channel protein Channelrhodopsin (ChR2) has recently been reported as a means for millisecond temporal control of neuronal spiking activities with cell-type selectivity. This offers the prospect of enabling local delivery of optical stimulation and the simultaneous monitoring of the neural activity by electrophysiological means, both in the vicinity of and distant to the stimulation site. We report here a novel dual-modality hybrid device, which consists of a tapered coaxial optical waveguide ('optrode') integrated into a 100 element intra-cortical multi-electrode recording array. We first demonstrate the dual optical delivery and electrical recording capability of the single optrode in in vitro preparations of mouse retina, photo-stimulating the native retinal photoreceptors while recording light-responsive activities from ganglion cells. The dual-modality array device was then used in ChR2 transfected mouse brain slices. Specifically, epileptiform events were reliably optically triggered by the optrode and their spatiotemporal patterns were simultaneously recorded by the multi-electrode array.

    View details for DOI 10.1088/1741-2560/6/5/055007

    View details for Web of Science ID 000270670400009

    View details for PubMedID 19721185

    View details for PubMedCentralID PMC2921864

  • Sleep Homeostasis Modulates Hypocretin-Mediated Sleep-to-Wake Transitions JOURNAL OF NEUROSCIENCE Carter, M. E., Adamantidis, A., Ohtsu, H., Deisseroth, K., de Lecea, L. 2009; 29 (35): 10939-10949

    Abstract

    The hypocretins (Hcrts) (also called orexins) are two neuropeptides expressed in the lateral hypothalamus that play a crucial role in the stability of wakefulness. Previously, our laboratory demonstrated that in vivo photostimulation of Hcrt neurons genetically targeted with ChR2, a light-activated cation channel, was sufficient to increase the probability of an awakening event during both slow-wave sleep and rapid eye movement sleep. In the current study, we ask whether Hcrt-mediated sleep-to-wake transitions are affected by light/dark period and sleep pressure. We found that stimulation of Hcrt neurons increased the probability of an awakening event throughout the entire light/dark period but that this effect was diminished with sleep pressure induced by 2 or 4 h of sleep deprivation. Interestingly, photostimulation of Hcrt neurons was still sufficient to increase activity assessed by c-Fos expression in Hcrt neurons after sleep deprivation, although this stimulation did not cause an increase in transitions to wakefulness. In addition, we found that photostimulation of Hcrt neurons increases neural activity assessed by c-Fos expression in the downstream arousal-promoting locus ceruleus and tuberomammilary nucleus but not after 2 h of sleep deprivation. Finally, stimulation of Hcrt neurons was still sufficient to increase the probability of an awakening event in histidine decarboxylase-deficient knock-out animals. Collectively, these results suggest that the Hcrt system promotes wakefulness throughout the light/dark period by activating multiple downstream targets, which themselves are inhibited with increased sleep pressure.

    View details for DOI 10.1523/JNEUROSCI.1205-09.2009

    View details for Web of Science ID 000269518500018

    View details for PubMedID 19726652

  • Optogenetic control of epileptiform activity PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Tonnesen, J., Sorensen, A. T., Deisseroth, K., Lundberg, C., Kokaia, M. 2009; 106 (29): 12162-12167

    Abstract

    The optogenetic approach to gain control over neuronal excitability both in vitro and in vivo has emerged as a fascinating scientific tool to explore neuronal networks, but it also opens possibilities for developing novel treatment strategies for neurologic conditions. We have explored whether such an optogenetic approach using the light-driven halorhodopsin chloride pump from Natronomonas pharaonis (NpHR), modified for mammalian CNS expression to hyperpolarize central neurons, may inhibit excessive hyperexcitability and epileptiform activity. We show that a lentiviral vector containing the NpHR gene under the calcium/calmodulin-dependent protein kinase IIalpha promoter transduces principal cells of the hippocampus and cortex and hyperpolarizes these cells, preventing generation of action potentials and epileptiform activity during optical stimulation. This study proves a principle, that selective hyperpolarization of principal cortical neurons by NpHR is sufficient to curtail paroxysmal activity in transduced neurons and can inhibit stimulation train-induced bursting in hippocampal organotypic slice cultures, which represents a model tissue of pharmacoresistant epilepsy. This study demonstrates that the optogenetic approach may prove useful for controlling epileptiform activity and opens a future perspective to develop it into a strategy to treat epilepsy.

    View details for DOI 10.1073/pnas.0901915106

    View details for Web of Science ID 000268178400062

    View details for PubMedID 19581573

    View details for PubMedCentralID PMC2715517

  • Optical Control of Neurons and Behaviors Deisseroth, K. ELSEVIER SCIENCE INC. 2009: 1S
  • Induced chromosome deletions cause hypersociability and other features of Williams-Beuren syndrome in mice EMBO MOLECULAR MEDICINE Li, H. H., Roy, M., Kuscuoglu, U., Spencer, C. M., Halm, B., Harrison, K. C., Bayle, J. H., Splendore, A., Ding, F., Meltzer, L. A., Wright, E., Paylor, R., Deisseroth, K., Francke, U. 2009; 1 (1): 50-65

    Abstract

    The neurodevelopmental disorder Williams-Beuren syndrome is caused by spontaneous approximately 1.5 Mb deletions comprising 25 genes on human chromosome 7q11.23. To functionally dissect the deletion and identify dosage-sensitive genes, we created two half-deletions of the conserved syntenic region on mouse chromosome 5G2. Proximal deletion (PD) mice lack Gtf2i to Limk1, distal deletion (DD) mice lack Limk1 to Fkbp6, and the double heterozygotes (D/P) model the complete human deletion. Gene transcript levels in brain are generally consistent with gene dosage. Increased sociability and acoustic startle response are associated with PD, and cognitive defects with DD. Both PD and D/P males are growth-retarded, while skulls are shortened and brains are smaller in DD and D/P. Lateral ventricle (LV) volumes are reduced, and neuronal cell density in the somatosensory cortex is increased, in PD and D/P. Motor skills are most impaired in D/P. Together, these partial deletion mice replicate crucial aspects of the human disorder and serve to identify genes and gene networks contributing to the neural substrates of complex behaviours and behavioural disorders.

    View details for DOI 10.1002/emmm.200900003

    View details for Web of Science ID 000273437400010

    View details for PubMedID 20049703

    View details for PubMedCentralID PMC3378107

  • Bi-stable neural state switches NATURE NEUROSCIENCE Berndt, A., Yizhar, O., Gunaydin, L. A., Hegemann, P., Deisseroth, K. 2009; 12 (2): 229-234

    Abstract

    Here we describe bi-stable channelrhodopsins that convert a brief pulse of light into a stable step in membrane potential. These molecularly engineered probes nevertheless retain millisecond-scale temporal precision. Photocurrents can be precisely initiated and terminated with different colors of light, but operate at vastly longer time scales than conventional channelrhodopsins as a result of modification at the C128 position that extends the lifetime of the open state. Because of their enhanced kinetic stability, these step-function tools are also effectively responsive to light at orders of magnitude lower intensity than wild-type channelrhodopsins. These molecules therefore offer important new capabilities for a broad range of in vivo applications.

    View details for DOI 10.1038/nn.2247

    View details for Web of Science ID 000263182000024

    View details for PubMedID 19079251

  • Optogenetics: Development And Application Karl Deisseroth Deisseroth, K. CELL PRESS. 2009: 215A
  • Escape behavior elicited by single, Channelrhodopsin-2-evoked spikes in zebrafish somatosensory neurons CURRENT BIOLOGY Douglass, A. D., Kraves, S., Deisseroth, K., Schier, A. F., Engert, F. 2008; 18 (15): 1133-1137

    Abstract

    Somatosensory neurons in teleosts and amphibians are sensitive to thermal, mechanical, or nociceptive stimuli [1, 2]. The two main types of such cells in zebrafish--Rohon-Beard and trigeminal neurons--have served as models for neural development [3-6], but little is known about how they encode tactile stimuli. The hindbrain networks that transduce somatosensory stimuli into a motor output encode information by using very few spikes in a small number of cells [7], but it is unclear whether activity in the primary receptor neurons is similarly efficient. To address this question, we manipulated the activity of zebrafish neurons with the light-activated cation channel, Channelrhodopsin-2 (ChR2) [8, 9]. We found that photoactivation of ChR2 in genetically defined populations of somatosensory neurons triggered escape behaviors in 24-hr-old zebrafish. Electrophysiological recordings from ChR2-positive trigeminal neurons in intact fish revealed that these cells have extremely low rates of spontaneous activity and can be induced to fire by brief pulses of blue light. Using this technique, we find that even a single action potential in a single sensory neuron was at times sufficient to evoke an escape behavior. These results establish ChR2 as a powerful tool for the manipulation of neural activity in zebrafish and reveal a degree of efficiency in coding that has not been found in primary sensory neurons.

    View details for DOI 10.1016/j.cub.2008.06.077

    View details for Web of Science ID 000258262800026

    View details for PubMedID 18682213

    View details for PubMedCentralID PMC2891506

  • eNpHR: a Natronomonas halorhodopsin enhanced for optogenetic applications BRAIN CELL BIOLOGY Gradinaru, V., Thompson, K. R., Deisseroth, K. 2008; 36 (1-4): 129-139

    Abstract

    Temporally precise inhibition of distinct cell types in the intact nervous system has been enabled by the microbial halorhodopsin NpHR, a fast light-activated electrogenic Cl(-) pump. While neurons can be optically hyperpolarized and inhibited from firing action potentials at moderate NpHR expression levels, we have encountered challenges with pushing expression to extremely high levels, including apparent intracellular accumulations. We therefore sought to molecularly engineer NpHR to achieve strong expression without these cellular side effects. We found that high expression correlated with endoplasmic reticulum (ER) accumulation, and that under these conditions NpHR colocalized with ER proteins containing the KDEL ER retention sequence. We screened a number of different putative modulators of membrane trafficking and identified a combination of two motifs, an N-terminal signal peptide and a C-terminal ER export sequence, that markedly promoted membrane localization and ER export defined by confocal microscopy and whole-cell patch clamp. The modified NpHR displayed increased peak photocurrent in the absence of aggregations or toxicity, and potent optical inhibition was observed not only in vitro but also in vivo with thalamic single-unit recording. The new enhanced NpHR (eNpHR) allows safe, high-level expression in mammalian neurons, without toxicity and with augmented inhibitory function, in vitro and in vivo.

    View details for DOI 10.1007/s11068-008-9027-6

    View details for Web of Science ID 000261177500011

    View details for PubMedID 18677566

    View details for PubMedCentralID PMC2588488

  • Improved expression of halorhodopsin for light-induced silencing of neuronal activity BRAIN CELL BIOLOGY Zhao, S., Cunha, C., Zhang, F., Liu, Q., Gloss, B., Deisseroth, K., Augustine, G. J., Feng, G. 2008; 36 (1-4): 141-154

    Abstract

    The ability to control and manipulate neuronal activity within an intact mammalian brain is of key importance for mapping functional connectivity and for dissecting the neural circuitry underlying behaviors. We have previously generated transgenic mice that express channelrhodopsin-2 for light-induced activation of neurons and mapping of neural circuits. Here we describe transgenic mice that express halorhodopsin (NpHR), a light-driven chloride pump that can be used to silence neuronal activity via light. Using the Thy-1 promoter to target NpHR expression to neurons, we found that neurons in these mice expressed high levels of NpHR-YFP and that illumination of cortical pyramidal neurons expressing NpHR-YFP led to rapid, reversible photoinhibition of action potential firing in these cells. However, NpHR-YFP expression led to the formation of numerous intracellular blebs, which may disrupt neuronal function. Labeling of various subcellular markers indicated that the blebs arise from retention of NpHR-YFP in the endoplasmic reticulum. By improving the signal peptide sequence and adding an ER export signal to NpHR-YFP, we eliminated the formation of blebs and dramatically increased the membrane expression of NpHR-YFP. Thus, the improved version of NpHR should serve as an excellent tool for neuronal silencing in vitro and in vivo.

    View details for DOI 10.1007/s11068-008-9034-7

    View details for Web of Science ID 000261177500012

    View details for PubMedID 18931914

    View details for PubMedCentralID PMC3057022

  • Brain circuit dynamics AMERICAN JOURNAL OF PSYCHIATRY Hu, E. S., Airan, R. D., Vijaykumar, R., Deisseroth, K. 2008; 165 (7): 800-800
  • Red-shifted optogenetic excitation: a tool for fast neural control derived from Volvox carteri NATURE NEUROSCIENCE Zhang, F., Prigge, M., Beyriere, F., Tsunoda, S. P., Mattis, J., Yizhar, O., Hegemann, P., Deisseroth, K. 2008; 11 (6): 631-633

    Abstract

    The introduction of two microbial opsin-based tools, channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), to neuroscience has generated interest in fast, multimodal, cell type-specific neural circuit control. Here we describe a cation-conducting channelrhodopsin (VChR1) from Volvox carteri that can drive spiking at 589 nm, with excitation maximum red-shifted approximately 70 nm compared with ChR2. These results demonstrate fast photostimulation with yellow light, thereby defining a functionally distinct third category of microbial rhodopsin proteins.

    View details for DOI 10.1038/nn.2120

    View details for Web of Science ID 000256133700007

    View details for PubMedID 18432196

    View details for PubMedCentralID PMC2692303

  • Controlling neuronal activity AMERICAN JOURNAL OF PSYCHIATRY Tamminga, C. A., Schneider, M. B., Gradinaru, V., Zhang, F., Deisseroth, K. 2008; 165 (5): 562-562
  • Red-shifted optogenetic excitation: a tool for fast neural control derived from Volvox carteri. Nat Neurosci. Zhang, F., Prigge, M., Beyriere, F., Tsunoda, S. P., Mattis, J., Yizhar, O., Deisseroth, K. 2008; 6 (11): 631-3
  • Brain circuit dynamics Am J Psychiatry. Hu, E. S., Airan, R. D., Vijaykumar, R., Deisseroth, K. 2008; 7 (165): 800
  • Targeting and readout strategies for fast optical neural control in vitro and in vivo JOURNAL OF NEUROSCIENCE Gradinaru, V., Thompson, K. R., Zhang, F., Mogri, M., Kay, K., Schneider, M. B., Deisseroth, K. 2007; 27 (52): 14231-14238
  • Nociceptive neurons protect Drosophila larvae from parasitoid wasps CURRENT BIOLOGY Hwang, R. Y., Zhong, L., Xu, Y., Johnson, T., Zhang, F., Deisseroth, K., Tracey, W. D. 2007; 17 (24): 2105-2116

    Abstract

    Natural selection has resulted in a complex and fascinating repertoire of innate behaviors that are produced by insects. One puzzling example occurs in fruit fly larvae that have been subjected to a noxious mechanical or thermal sensory input. In response, the larvae "roll" with a motor pattern that is completely distinct from the style of locomotion that is used for foraging.We have precisely mapped the sensory neurons that are used by the Drosophila larvae to detect nociceptive stimuli. By using complementary optogenetic activation and targeted silencing of sensory neurons, we have demonstrated that a single class of neuron (class IV multidendritic neuron) is sufficient and necessary for triggering the unusual rolling behavior. In addition, we find that larvae have an innately encoded preference in the directionality of rolling. Surprisingly, the initial direction of rolling locomotion is toward the side of the body that has been stimulated. We propose that directional rolling might provide a selective advantage in escape from parasitoid wasps that are ubiquitously present in the natural environment of Drosophila. Consistent with this hypothesis, we have documented that larvae can escape the attack of Leptopilina boulardi parasitoid wasps by rolling, occasionally flipping the attacker onto its back.The class IV multidendritic neurons of Drosophila larvae are nociceptive. The nociception behavior of Drosophila melanagaster larvae includes an innately encoded directional preference. Nociception behavior is elicited by the ecologically relevant sensory stimulus of parasitoid wasp attack.

    View details for DOI 10.1016/j.cub.2007.11.029

    View details for Web of Science ID 000251852200022

    View details for PubMedID 18060782

    View details for PubMedCentralID PMC2225350

  • Neural substrates of awakening probed with optogenetic control of hypocretin neurons NATURE Adamantidis, A. R., Zhang, F., Aravanis, A. M., Deisseroth, K., de Lecea, L. 2007; 450 (7168): 420-U9

    Abstract

    The neural underpinnings of sleep involve interactions between sleep-promoting areas such as the anterior hypothalamus, and arousal systems located in the posterior hypothalamus, the basal forebrain and the brainstem. Hypocretin (Hcrt, also known as orexin)-producing neurons in the lateral hypothalamus are important for arousal stability, and loss of Hcrt function has been linked to narcolepsy. However, it is unknown whether electrical activity arising from Hcrt neurons is sufficient to drive awakening from sleep states or is simply correlated with it. Here we directly probed the impact of Hcrt neuron activity on sleep state transitions with in vivo neural photostimulation, genetically targeting channelrhodopsin-2 to Hcrt cells and using an optical fibre to deliver light deep in the brain, directly into the lateral hypothalamus, of freely moving mice. We found that direct, selective, optogenetic photostimulation of Hcrt neurons increased the probability of transition to wakefulness from either slow wave sleep or rapid eye movement sleep. Notably, photostimulation using 5-30 Hz light pulse trains reduced latency to wakefulness, whereas 1 Hz trains did not. This study establishes a causal relationship between frequency-dependent activity of a genetically defined neural cell type and a specific mammalian behaviour central to clinical conditions and neurobehavioural physiology.

    View details for DOI 10.1038/nature06310

    View details for Web of Science ID 000250918600055

    View details for PubMedID 17943086

  • Integration of light-controlled neuronal firing and fast circuit imaging CURRENT OPINION IN NEUROBIOLOGY Airan, R. D., Hu, E. S., Vijaykumar, R., Roy, M., Meltzer, L. A., Deisseroth, K. 2007; 17 (5): 587-592

    Abstract

    For understanding normal and pathological circuit function, capitalizing on the full potential of recent advances in fast optical neural circuit control will depend crucially on fast, intact-circuit readout technology. First, millisecond-scale optical control will be best leveraged with simultaneous millisecond-scale optical imaging. Second, both fast circuit control and imaging should be adaptable to intact-circuit preparations from normal and diseased subjects. Here we illustrate integration of fast optical circuit control and fast circuit imaging, review recent work demonstrating utility of applying fast imaging to quantifying activity flow in disease models, and discuss integration of diverse optogenetic and chemical genetic tools that have been developed to precisely control the activity of genetically specified neural populations. Together these neuroengineering advances raise the exciting prospect of determining the role-specific cell types play in modulating neural activity flow in neuropsychiatric disease.

    View details for DOI 10.1016/j.conb.2007.11.003

    View details for Web of Science ID 000252835100013

    View details for PubMedID 18093822

  • An optical neural interface: in vivo control of rodent motor cortex with integrated fiberoptic and optogenetic technology JOURNAL OF NEURAL ENGINEERING Aravanis, A. M., Wang, L., Zhang, F., Meltzer, L. A., Mogri, M. Z., Schneider, M. B., Deisseroth, K. 2007; 4 (3): S143-S156

    Abstract

    Neural interface technology has made enormous strides in recent years but stimulating electrodes remain incapable of reliably targeting specific cell types (e.g. excitatory or inhibitory neurons) within neural tissue. This obstacle has major scientific and clinical implications. For example, there is intense debate among physicians, neuroengineers and neuroscientists regarding the relevant cell types recruited during deep brain stimulation (DBS); moreover, many debilitating side effects of DBS likely result from lack of cell-type specificity. We describe here a novel optical neural interface technology that will allow neuroengineers to optically address specific cell types in vivo with millisecond temporal precision. Channelrhodopsin-2 (ChR2), an algal light-activated ion channel we developed for use in mammals, can give rise to safe, light-driven stimulation of CNS neurons on a timescale of milliseconds. Because ChR2 is genetically targetable, specific populations of neurons even sparsely embedded within intact circuitry can be stimulated with high temporal precision. Here we report the first in vivo behavioral demonstration of a functional optical neural interface (ONI) in intact animals, involving integrated fiberoptic and optogenetic technology. We developed a solid-state laser diode system that can be pulsed with millisecond precision, outputs 20 mW of power at 473 nm, and is coupled to a lightweight, flexible multimode optical fiber, approximately 200 microm in diameter. To capitalize on the unique advantages of this system, we specifically targeted ChR2 to excitatory cells in vivo with the CaMKIIalpha promoter. Under these conditions, the intensity of light exiting the fiber ( approximately 380 mW mm(-2)) was sufficient to drive excitatory neurons in vivo and control motor cortex function with behavioral output in intact rodents. No exogenous chemical cofactor was needed at any point, a crucial finding for in vivo work in large mammals. Achieving modulation of behavior with optical control of neuronal subtypes may give rise to fundamental network-level insights complementary to what electrode methodologies have taught us, and the emerging optogenetic toolkit may find application across a broad range of neuroscience, neuroengineering and clinical questions.

    View details for DOI 10.1088/1741-2560/4/3/S02

    View details for Web of Science ID 000250181600003

    View details for PubMedID 17873414

  • High-speed Imaging reveals neurophysiological links to behavior in an animal model of depression SCIENCE Airan, R. D., Meltzer, L. A., Roy, M., Gong, Y., Chen, H., Deisseroth, K. 2007; 317 (5839): 819-823

    Abstract

    The hippocampus is one of several brain areas thought to play a central role in affective behaviors, but the underlying local network dynamics are not understood. We used quantitative voltage-sensitive dye imaging to probe hippocampal dynamics with millisecond resolution in brain slices after bidirectional modulation of affective state in rat models of depression. We found that a simple measure of real-time activity-stimulus-evoked percolation of activity through the dentate gyrus relative to the hippocampal output subfield-accounted for induced changes in animal behavior independent of the underlying mechanism of action of the treatments. Our results define a circuit-level neurophysiological endophenotype for affective behavior and suggest an approach to understanding circuit-level substrates underlying psychiatric disease symptoms.

    View details for DOI 10.1126/science.1144400

    View details for Web of Science ID 000248624500045

    View details for PubMedID 17615305

  • Circuit-breakers: optical technologies for probing neural signals and systems NATURE REVIEWS NEUROSCIENCE Zhang, F., Aravanis, A. M., Adamantidis, A., de Lecea, L., Deisseroth, K. 2007; 8 (8): 577-581

    Abstract

    Neuropsychiatric disorders, which arise from a combination of genetic, epigenetic and environmental influences, epitomize the challenges faced in understanding the mammalian brain. Elucidation and treatment of these diseases will benefit from understanding how specific brain cell types are interconnected and signal in neural circuits. Newly developed neuroengineering tools based on two microbial opsins, channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), enable the investigation of neural circuit function with cell-type-specific, temporally accurate and reversible neuromodulation. These tools could lead to the development of precise neuromodulation technologies for animal models of disease and clinical neuropsychiatry.

    View details for DOI 10.1038/nrn2192

    View details for Web of Science ID 000248211800012

    View details for PubMedID 17643087

  • High-speed mapping of synaptic connectivity using photostimulation in Channel rhodopsin-2 transgenic mice PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Wang, H., Peca, J., Matsuzaki, M., Matsuzaki, K., Noguchi, J., Qiu, L., Wang, D., Zhangn, F., Boyden, E., Deisserothl, K., Kasai, H., Hall, W. C., Feng, G., Augustine, G. J. 2007; 104 (19): 8143-8148

    Abstract

    To permit rapid optical control of brain activity, we have engineered multiple lines of transgenic mice that express the light-activated cation channel Channelrhodopsin-2 (ChR2) in subsets of neurons. Illumination of ChR2-positive neurons in brain slices produced photocurrents that generated action potentials within milliseconds and with precisely timed latencies. The number of light-evoked action potentials could be controlled by varying either the amplitude or duration of illumination. Furthermore, the frequency of light-evoked action potentials could be precisely controlled up to 30 Hz. Photostimulation also could evoke synaptic transmission between neurons, and, by scanning with a small laser light spot, we were able to map the spatial distribution of synaptic circuits connecting neurons within living cerebral cortex. We conclude that ChR2 is a genetically based photostimulation technology that permits analysis of neural circuits with high spatial and temporal resolution in transgenic mammals.

    View details for DOI 10.1073/pnas.0700384104

    View details for Web of Science ID 000246461500073

    View details for PubMedID 17483470

    View details for PubMedCentralID PMC1876585

  • In vivo light-induced activation of neural circuitry in transgenic mice expressing channelrhodopsin-2 NEURON Arenkiel, B. R., Peca, J., Davison, I. G., Feliciano, C., Deisseroth, K., Augustine, G. J., Ehlers, M. D., Feng, G. 2007; 54 (2): 205-218

    Abstract

    Channelrhodopsin-2 (ChR2) is a light-gated, cation-selective ion channel isolated from the green algae Chlamydomonas reinhardtii. Here, we report the generation of transgenic mice that express a ChR2-YFP fusion protein in the CNS for in vivo activation and mapping of neural circuits. Using focal illumination of the cerebral cortex and olfactory bulb, we demonstrate a highly reproducible, light-dependent activation of neurons and precise control of firing frequency in vivo. To test the feasibility of mapping neural circuits, we exploited the circuitry formed between the olfactory bulb and the piriform cortex in anesthetized mice. In the olfactory bulb, individual mitral cells fired action potentials in response to light, and their firing rate was not influenced by costimulated glomeruli. However, in piriform cortex, the activity of target neurons increased as larger areas of the bulb were illuminated to recruit additional glomeruli. These results support a model of olfactory processing that is dependent upon mitral cell convergence and integration onto cortical cells. More broadly, these findings demonstrate a system for precise manipulation of neural activity in the intact mammalian brain with light and illustrate the use of ChR2 mice in exploring functional connectivity of complex neural circuits in vivo.

    View details for DOI 10.1016/j.neuron.2007.03.005

    View details for Web of Science ID 000246190600007

    View details for PubMedID 17442243

    View details for PubMedCentralID PMC3634585

  • Multimodal fast optical interrogation of neural circuitry NATURE Zhang, F., Wang, L., Brauner, M., Liewald, J. F., Kay, K., Watzke, N., Wood, P. G., Bamberg, E., Nagel, G., Gottschalk, A., Deisseroth, K. 2007; 446 (7136): 633-U4

    Abstract

    Our understanding of the cellular implementation of systems-level neural processes like action, thought and emotion has been limited by the availability of tools to interrogate specific classes of neural cells within intact, living brain tissue. Here we identify and develop an archaeal light-driven chloride pump (NpHR) from Natronomonas pharaonis for temporally precise optical inhibition of neural activity. NpHR allows either knockout of single action potentials, or sustained blockade of spiking. NpHR is compatible with ChR2, the previous optical excitation technology we have described, in that the two opposing probes operate at similar light powers but with well-separated action spectra. NpHR, like ChR2, functions in mammals without exogenous cofactors, and the two probes can be integrated with calcium imaging in mammalian brain tissue for bidirectional optical modulation and readout of neural activity. Likewise, NpHR and ChR2 can be targeted together to Caenorhabditis elegans muscle and cholinergic motor neurons to control locomotion bidirectionally. NpHR and ChR2 form a complete system for multimodal, high-speed, genetically targeted, all-optical interrogation of living neural circuits.

    View details for DOI 10.1038/nature05744

    View details for Web of Science ID 000245438300032

    View details for PubMedID 17410168

  • High-speed mapping of synaptic connectivity using photostimulation in Channelrhodopsin-2 transgenic mice. H, W., J, P., M, M., K, M., J, N., L, Q., Deisseroth, K. 2007
  • Next-generation optical technologies for illuminating genetically targeted brain circuits JOURNAL OF NEUROSCIENCE Deisseroth, K., Feng, G., Majewska, A. K., Miesenbock, G., Ting, A., Schnitzer, M. J. 2006; 26 (41): 10380-10386

    Abstract

    Emerging technologies from optics, genetics, and bioengineering are being combined for studies of intact neural circuits. The rapid progression of such interdisciplinary "optogenetic" approaches has expanded capabilities for optical imaging and genetic targeting of specific cell types. Here we explore key recent advances that unite optical and genetic approaches, focusing on promising techniques that either allow novel studies of neural dynamics and behavior or provide fresh perspectives on classic model systems.

    View details for DOI 10.1523/JNEUROSCI.3863-06.2006

    View details for Web of Science ID 000241192800010

    View details for PubMedID 17035522

    View details for PubMedCentralID PMC2820367

  • Channelrhodopsin-2 and optical control of excitable cells NATURE METHODS Zhang, F., Wang, L., Boyden, E. S., Deisseroth, K. 2006; 3 (10): 785-792

    Abstract

    Electrically excitable cells are important in the normal functioning and in the pathophysiology of many biological processes. These cells are typically embedded in dense, heterogeneous tissues, rendering them difficult to target selectively with conventional electrical stimulation methods. The algal protein Channelrhodopsin-2 offers a new and promising solution by permitting minimally invasive, genetically targeted and temporally precise photostimulation. Here we explore technological issues relevant to the temporal precision, spatial targeting and physiological implementation of ChR2, in the context of other photostimulation approaches to optical control of excitable cells.

    View details for DOI 10.1038/nmeth936

    View details for Web of Science ID 000240942600011

    View details for PubMedID 16990810

  • A role for circuit homeostasis in adult neurogenesis TRENDS IN NEUROSCIENCES Meltzer, L. A., Yabaluri, R., Deisseroth, K. 2005; 28 (12): 653-660

    Abstract

    Insertion of new neurons into adult neural circuits could either promote or impair circuit function, depending on whether homeostatic mechanisms are in place to regulate the resulting changes in neural activity. In the hippocampus (a mammalian forebrain structure important in aspects of memory and mood) several lines of behavioral evidence suggest important adaptive roles for adult-generated neurons, indicating that there could be mechanisms to control the potentially adverse increase in excitation associated with new cells. Here, we delineate behavioral and computational models for the role of circuit homeostasis in enabling neuron insertion to modulate hippocampal function adaptively, and we describe molecular and cellular mechanisms for implementing this circuit-level adaptive regulation of hippocampal activity.

    View details for DOI 10.1016/j.tins.2005.09.007

    View details for Web of Science ID 000234151400004

    View details for PubMedID 16271403

  • GABA excitation in the adult brain: A mechanism for excitation-neurogenesis coupling NEURON Deisseroth, K., Malenka, R. C. 2005; 47 (6): 775-777

    Abstract

    The production of new neurons in the adult hippocampus is exquisitely regulated, and alterations in this process may underlie both normal and pathological hippocampal function. In this issue of Neuron, Tozuka et al. describe electrophysiological recordings that target proliferating progenitor cells in adult mouse hippocampal slices. They report that GABAergic synaptic inputs directly depolarize the proliferating progenitors, thereby activating molecular players that favor neuronal differentiation and providing a mechanism for direct excitation-neurogenesis coupling in vivo.

    View details for DOI 10.1016/j.neuron.2005.08.029

    View details for Web of Science ID 000232085000003

    View details for PubMedID 16157270

  • Millisecond-timescale, genetically targeted optical control of neural activity NATURE NEUROSCIENCE Boyden, E. S., Zhang, F., Bamberg, E., Nagel, G., Deisseroth, K. 2005; 8 (9): 1263-1268

    Abstract

    Temporally precise, noninvasive control of activity in well-defined neuronal populations is a long-sought goal of systems neuroscience. We adapted for this purpose the naturally occurring algal protein Channelrhodopsin-2, a rapidly gated light-sensitive cation channel, by using lentiviral gene delivery in combination with high-speed optical switching to photostimulate mammalian neurons. We demonstrate reliable, millisecond-timescale control of neuronal spiking, as well as control of excitatory and inhibitory synaptic transmission. This technology allows the use of light to alter neural processing at the level of single spikes and synaptic events, yielding a widely applicable tool for neuroscientists and biomedical engineers.

    View details for DOI 10.1038/nn1525

    View details for Web of Science ID 000231483800028

    View details for PubMedID 16116447

  • Excitation-neurogenesis coupling in adult neural stem/progenitor cells NEURON Deisseroth, K., Singla, S., Toda, H., Monje, M., Palmer, T. D., Malenka, R. C. 2004; 42 (4): 535-552

    Abstract

    A wide variety of in vivo manipulations influence neurogenesis in the adult hippocampus. It is not known, however, if adult neural stem/progenitor cells (NPCs) can intrinsically sense excitatory neural activity and thereby implement a direct coupling between excitation and neurogenesis. Moreover, the theoretical significance of activity-dependent neurogenesis in hippocampal-type memory processing networks has not been explored. Here we demonstrate that excitatory stimuli act directly on adult hippocampal NPCs to favor neuron production. The excitation is sensed via Ca(v)1.2/1.3 (L-type) Ca(2+) channels and NMDA receptors on the proliferating precursors. Excitation through this pathway acts to inhibit expression of the glial fate genes Hes1 and Id2 and increase expression of NeuroD, a positive regulator of neuronal differentiation. These activity-sensing properties of the adult NPCs, when applied as an "excitation-neurogenesis coupling rule" within a Hebbian neural network, predict significant advantages for both the temporary storage and the clearance of memories.

    View details for Web of Science ID 000221708300006

    View details for PubMedID 15157417

  • Signaling from synapse to nucleus: the logic behind the mechanisms CURRENT OPINION IN NEUROBIOLOGY Deisseroth, K., Mermelstein, P. G., Xia, H. H., Tsien, R. W. 2003; 13 (3): 354-365

    Abstract

    Signaling from synapse to nucleus is vital for activity-dependent control of neuronal gene expression and represents a sophisticated form of neural computation. The nature of specific signal initiators, nuclear translocators and effectors has become increasingly clear, and supports the idea that the nucleus is able to make sense of a surprising amount of fast synaptic information through intricate biochemical mechanisms. Information transfer to the nucleus can be conveyed by physical translocation of messengers at various stages within the multiple signal transduction cascades that are set in motion by a Ca(2+) rise near the surface membrane. The key role of synapse-to-nucleus signaling in circadian rhythms, long-term memory, and neuronal survival sheds light on the logical underpinning of these signaling mechanisms.

    View details for DOI 10.1016/S0959-4388(03)00076-X

    View details for Web of Science ID 000184245400014

    View details for PubMedID 12850221

  • Dynamic multiphosphorylation passwords for activity-dependent gene expression NEURON Deisseroth, K., Tsien, R. W. 2002; 34 (2): 179-182

    Abstract

    Synapse-to-nucleus signaling leading to CREB-mediated transcription is important for neuronal plasticity. Nuclear CREB phosphorylation at Ser133 allows convergence of multiple kinase pathways driven by neuronal activity and links them to transcriptional activation. But, can various pathways share a common effector mechanism (phosphorylating Ser133) while generating distinct patterns of gene expression? We review three Neuron articles that highlight novel ways Ca(2+) signals can trigger multiple phosphorylation events working in combination to control CREB and its interaction with coactivator molecules.

    View details for Web of Science ID 000174976200004

    View details for PubMedID 11970860

  • Calmodulin priming: Nuclear translocation of a calmodulin complex and the memory of prior neuronal activity PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Mermelstein, P. G., Deisseroth, K., Dasgupta, N., Isaksen, A. L., Tsien, R. W. 2001; 98 (26): 15342-15347

    Abstract

    The neuronal nucleus plays a vital role in information processing, but whether it supports computational functions such as paired-pulse facilitation, comparable to synapses, is unclear. Ca(2+)-dependent movement of calmodulin (CaM) to the nucleus is highly responsive to Ca(2+) entry through L-type channels and promotes activation of the transcription factor CREB (cAMP-responsive element binding protein) through phosphorylation by CaM-sensitive kinases. We characterized key features of this CaM translocation and its possible role in facilitation of nuclear signaling. Nuclear CaM was elevated within 15 s of stimulus onset, preceding the first signs of CREB phosphorylation in hippocampal pyramidal neurons. Depolarization-induced elevation of nuclear CaM also was observed in cerebellar granule cells, neocortical neurons, and dentate gyrus granule cells. Nuclear translocation of CaM was not blocked by disruption of actin filaments or microtubules, or by emptying endoplasmic reticulum Ca(2+) stores with thapsigargin. Translocation of fluorescently tagged CaM was prevented by fusing it with the Ca(2+)/CaM binding peptide M13, suggesting that nuclear CaM accumulation depends on association with endogenous Ca(2+)/CaM binding proteins. To determine whether increased nuclear [CaM] might influence subsequent nuclear signal processing, we compared responses to two consecutive depolarizing stimuli. After a weak "priming" stimulus that caused CaM translocation, CREB phosphorylation caused by a subsequent stimulus was significantly faster, more sensitive to Ca(2+) elevation, and less specifically dependent on Ca(2+) influx through L-type channels. CaM translocation not only supports rapid signaling to the nucleus, but also could provide a "memory" for facilitatory effects of repeated neural activity, seen in altered phosphorylated CREB dynamics and Ca(2+) channel dependence.

    View details for Web of Science ID 000172848800107

    View details for PubMedID 11742070

    View details for PubMedCentralID PMC65031

  • Activity-dependent CREB phosphorylation: Convergence of a fast, sensitive calmodulin kinase pathway and a slow, less sensitive mitogen-activated protein kinase pathway PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Wu, G. Y., Deisseroth, K., Tsien, R. W. 2001; 98 (5): 2808-2813

    Abstract

    The cAMP-responsive element binding protein (CREB), a key regulator of gene expression, is activated by phosphorylation on Ser-133. Several different protein kinases possess the capability of driving this phosphorylation, making it a point of potential convergence for multiple intracellular signaling cascades. Previous work in neurons has indicated that physiologic synaptic stimulation recruits a fast calmodulin kinase IV (CaMKIV)-dependent pathway that dominates early signaling to CREB. Here we show in hippocampal neurons that the fast, CaMK-dependent pathway can be followed by a slower pathway that depends on Ras/mitogen-activated protein kinase (MAPK), along with CaMK. This pathway was blocked by dominant-negative Ras and was specifically recruited by depolarizations that produced strong intracellular Ca(2+) transients. When both pathways were recruited, phosphorylated CREB (pCREB) formation was overwhelmingly dominated by the CaMK pathway between 0 and 10 min, and by the MAPK pathway at 60 min, whereas the two pathways acted in concert at 30 min. The Ca(2+) signals that produced only rapid CaMK signaling to pCREB or both rapid CaMK and slow MAPK signaling deviated significantly for only approximately 1 min, yet their differential impact on pCREB extended over a much longer period, between 20 and 60 min and beyond, which is of likely significance for gene expression. The CaMK-dependent MAPK pathway may inform the nucleus about stimulus amplitude. In contrast, the CaMKIV pathway may be well suited to conveying information on the precise timing of localized synaptic stimuli, befitting its greater speed and sensitivity, whereas the previously described calcineurin pathway may carry information about stimulus duration.

    View details for Web of Science ID 000167258900126

    View details for PubMedID 11226322

    View details for PubMedCentralID PMC30221

  • Spaced stimuli stabilize MAPK pathway activation and its effects on dendritic morphology NATURE NEUROSCIENCE Wu, G. Y., Deisseroth, K., Tsien, R. W. 2001; 4 (2): 151-158

    Abstract

    Memory storage in mammalian neurons probably depends on both biochemical events and morphological alterations in dendrites. Here we report an activity-dependent stabilization of the MAP kinase (MAPK) pathway, prominent in hippocampal dendrites. The longevity of the signal in these dendrites was increased to hours when multiple spaced stimuli were used. Likewise, spaced stimuli and MAPK activation were critical for protrusion of new dendritic filopodia that also remained stable for hours. Our experiments define a new role for stimulus-specific responses of MAPK signaling in activity-dependent neuronal plasticity. The local biochemical signaling in dendrites complements MAPK signaling in gene expression. Together, these processes may support long-lasting behavioral changes.

    View details for Web of Science ID 000167178100013

    View details for PubMedID 11175875

  • Critical dependence of cAMP response element-binding protein phosphorylation on L-type calcium channels supports a selective response to EPSPs in preference to action potentials JOURNAL OF NEUROSCIENCE Mermelstein, P. G., Bito, H., Deisseroth, K., Tsien, R. W. 2000; 20 (1): 266-273

    Abstract

    Activity-dependent gene expression in neurons shows a remarkable ability to differentiate between different types of stimulation: orthodromic inputs that engage synaptic transmission are much more effective than antidromic stimuli that do not. We have studied the basis of such selectivity in cultured hippocampal neurons in which nuclear cAMP response element-binding protein (CREB) phosphorylation is induced by synaptic activity but not by action potential (AP) stimulation in the absence of EPSPs, although spikes by themselves generate large elevations in intracellular Ca(2+). Previous work has shown that Ca(2+) entry through L-type Ca(2+) channels plays a dominant role in triggering calmodulin mobilization and activation of calmodulin-dependent kinases that phosphorylate CREB, raising the possibility that L-type channels contribute to the selective response to EPSPs rather than APs. Accordingly, we performed voltage-clamp experiments to compare the currents carried by L-type channels during depolarizing waveforms that approximated APs or dendritic EPSPs. The integrated current generated by L-type channels was significantly less after mock APs than with EPSP-like depolarizations. The difference was traced to two distinct factors. Compared with other channels, L-type channels activated at relatively negative potentials, favoring their opening with EPSP stimulation; they also exhibited relatively slow activation kinetics, weighing against their contribution during an AP. The relative ineffectiveness of APs as a stimulus for CREB phosphorylation could be overcome by exposure to the agonist Bay K8644, which potentiated the AP-induced influx through L-type channels by approximately 10-fold. Under normal conditions, the unique biophysical properties of L-type channels allow them to act as a kinetic filter to support spike-EPSP discrimination.

    View details for Web of Science ID 000084581800035

    View details for PubMedID 10627604

  • Activity-dependent regulation of communication from synapse to nucleus 22nd International Symposium on Brain Sciences on Challenges for Neuroscience in the 21st Century Bito, H., Deisseroth, K., Tsien, R. W. JAPAN SCIENTIFIC SOC PRESS. 2000: 107–20
  • L-type calcium channels and GSK-3 regulate the activity of NF-ATc4 in hippocampal neurons NATURE Graef, I. A., Mermelstein, P. G., Stankunas, K., Neilson, J. R., Deisseroth, K., Tsien, R. W., Crabtree, G. R. 1999; 401 (6754): 703-708

    Abstract

    The molecular basis of learning and memory has been the object of several recent advances, which have focused attention on calcium-regulated pathways controlling transcription. One of the molecules implicated by pharmacological, biochemical and genetic approaches is the calcium/calmodulin-regulated phosphatase, calcineurin. In lymphocytes, calcineurin responds to specific calcium signals and regulates expression of several immediate early genes by controlling the nuclear import of the NF-ATc family of transcription factors. Here we show that NF-ATc4/NF-AT3 in hippocampal neurons can rapidly translocate from cytoplasm to nucleus and activate NF-AT-dependent transcription in response to electrical activity or potassium depolarization. The calcineurin-mediated translocation is critically dependent on calcium entry through L-type voltage-gated calcium channels. GSK-3 can phosphorylate NF-ATc4, promoting its export from the nucleus and antagonizing NF-ATc4-dependent transcription. Furthermore, we show that induction of the inositol 1,4,5-trisphosphate receptor type 1 is controlled by the calcium/calcineurin/NF-ATc pathway. This provides a new perspective on the function of calcineurin in the central nervous system and indicates that NF-AT-mediated gene expression may be involved in the induction of hippocampal synaptic plasticity and memory formation.

    View details for Web of Science ID 000083207400058

    View details for PubMedID 10537109

  • Calmodulin supports both inactivation and facilitation of L-type calcium channels NATURE Zuhlke, R. D., Pitt, G. S., Deisseroth, K., Tsien, R. W., Reuter, H. 1999; 399 (6732): 159-162

    Abstract

    L-type Ca2+ channels support Ca2+ entry into cells, which triggers cardiac contraction, controls hormone secretion from endocrine cells and initiates transcriptional events that support learning and memory. These channels are examples of molecular signal-transduction units that regulate themselves through their own activity. Among the many types of voltage-gated Ca2+ channel, L-type Ca2+ channels particularly display inactivation and facilitation, both of which are closely linked to the earlier entry of Ca2+ ions. Both forms of autoregulation have a significant impact on the amount of Ca2+ that enters the cell during repetitive activity, with major consequences downstream. Despite extensive biophysical analysis, the molecular basis of autoregulation remains unclear, although a putative Ca2+-binding EF-hand motif and a nearby consensus calmodulin-binding isoleucine-glutamine ('IQ') motif in the carboxy terminus of the alpha1C channel subunit have been implicated. Here we show that calmodulin is a critical Ca2+ sensor for both inactivation and facilitation, and that the nature of the modulatory effect depends on residues within the IQ motif important for calmodulin binding. Replacement of the native isoleucine by alanine removed Ca2+-dependent inactivation and unmasked a strong facilitation; conversion of the same residue to glutamate eliminated both forms of autoregulation. These results indicate that the same calmodulin molecule may act as a Ca2+ sensor for both positive and negative modulation.

    View details for Web of Science ID 000080335700052

    View details for PubMedID 10335846

  • Translocation of calmodulin to the nucleus supports CREB phosphorylation in hippocampal neurons NATURE Deisseroth, K., Heist, E. K., Tsien, R. W. 1998; 392 (6672): 198-202

    Abstract

    Activation of the transcription factor CREB is thought to be important in the formation of long-term memory in several animal species. The phosphorylation of a serine residue at position 133 of CREB is critical for activation of CREB. This phosphorylation is rapid when driven by brief synaptic activity in hippocampal neurons. It is initiated by a highly local, rise in calcium ion concentrations near the cell membrane, but culminates in the activation of a specific calmodulin-dependent kinase known as CaMK IV, which is constitutively present in the neuronal nucleus. It is unclear how the signal is conveyed from the synapse to the nucleus. We show here that brief bursts of activity cause a swift (approximately 1 min) translocation of calmodulin from the cytoplasm to the nucleus, and that this translocation is important for the rapid phosphorylation of CREB. Certain Ca2+ entry systems (L-type Ca2+ channels and NMDA receptors) are able to cause mobilization of calmodulin, whereas others (N- and P/Q-type Ca2+ channels) are not. This translocation of calmodulin provides a form of cellular communication that combines the specificity of local Ca2+ signalling with the ability to produce action at a distance.

    View details for Web of Science ID 000072462700067

    View details for PubMedID 9515967

  • Ca2+-dependent regulation in neuronal gene expression CURRENT OPINION IN NEUROBIOLOGY Bito, H., Deisseroth, K., Tsien, R. W. 1997; 7 (3): 419-429

    Abstract

    Ca2+ is an important signal-transduction molecule that plays a role in many intracellular signaling pathways. Recent advances have indicated that in neurons, Ca2+-controlled signaling mechanisms cooperate in order to discriminate amongst incoming cellular inputs. Ca2+-dependent transcriptional events can thereby be made selectively responsive to bursts of synaptic activity of specific intensity or duration.

    View details for Web of Science ID A1997XW34800018

    View details for PubMedID 9232807

  • CREB phosphorylation and dephosphorylation: A Ca2(+)- and stimulus duration-dependent switch for hippocampal gene expression CELL Bito, H., Deisseroth, K., Tsien, R. W. 1996; 87 (7): 1203-1214

    Abstract

    While changes in gene expression are critical for many brain functions, including long-term memory, little is known about the cellular processes that mediate stimulus-transcription coupling at central synapses. In studying the signaling pathways by which synaptic inputs control the phosphorylation state of cyclic AMP-responsive element binding protein (CREB) and determine expression of CRE-regulated genes, we found two important Ca2+/calmodulin (CaM)-regulated mechanisms in hippocampal neurons: a CaM kinase cascade involving nuclear CaMKIV and a calcineurin-dependent regulation of nuclear protein phosphatase 1 activity. Prolongation of the synaptic input on the time scale of minutes, in part by an activity-induced inactivation of calcineurin, greatly extends the period over which phospho-CREB levels are elevated, thus affecting induction of downstream genes.

    View details for Web of Science ID A1996WA54100009

    View details for PubMedID 8980227

  • Signaling from synapse to nucleus: Postsynaptic CREB phosphorylation during multiple forms of hippocampal synaptic plasticity NEURON Deisseroth, K., Bito, H., Tsien, R. W. 1996; 16 (1): 89-101

    Abstract

    Phosphorylation of the transcription factor CREB is thought to be important in processes underlying long-term memory. It is unclear whether CREB phosphorylation can carry information about the sign of changes in synaptic strength, whether CREB pathways are equally activated in neurons receiving or providing synaptic input, or how synapse-to-nucleus communication is mediated. We found that Ca(2+)-dependent nuclear CREB phosphorylation was rapidly evoked by synaptic stimuli including, but not limited to, those that induced potentiation and depression of synaptic strength. In striking contrast, high frequency action potential firing alone failed to trigger CREB phosphorylation. Activation of a submembranous Ca2+ sensor, just beneath sites of Ca2+ entry, appears critical for triggering nuclear CREB phosphorylation via calmodulin and a Ca2+/calmodulin-dependent protein kinase.

    View details for Web of Science ID A1996TT30700012

    View details for PubMedID 8562094

  • Synaptic plasticity: A molecular mechanism for metaplasticity CURRENT BIOLOGY Deisseroth, K., Bito, H., Schulman, H., Tsien, R. W. 1995; 5 (12): 1334-1338

    View details for Web of Science ID A1995TL10200002

    View details for PubMedID 8749377

  • IDENTIFICATION OF A POINT MUTATION IN THE TOPOISOMERASE-II GENE FROM A HUMAN LEUKEMIA-CELL LINE CONTAINING AN AMSACRINE-RESISTANT FORM OF TOPOISOMERASE-II CANCER RESEARCH Hinds, M., Deisseroth, K., Mayes, J., Altschuler, E., Jansen, R., Ledley, F. D., Zwelling, L. A. 1991; 51 (17): 4729-4731

    Abstract

    HL-60/AMSA is a human leukemia cell line that is 50- to 100-fold more resistant to the cytotoxic actions of the topoisomerase II-reactive intercalator amsacrine than is its drug-sensitive HL-60 parent line. Previously, we have shown that the topoisomerase II from HL-60/AMSA is also resistant to inhibition by amsacrine and other intercalating agents. We therefore sought the molecular basis for the resistance of the topoisomerase II of HL-60/AMSA and, by inference, of the HL-60/AMSA line itself. We report the cloning and sequencing of the topoisomerase II genes from both the sensitive and resistant leukemia cell lines using polymerase chain reaction technology. We have identified a single base change associated with the drug-resistant form of topoisomerase II. This mutation is present in both cloned HL-60/AMSA complementary DNA and extracted HL-60/AMSA genomic DNA. A rapid assay for this mutation in clinical samples has been developed and applied to the DNA of cells from both normal volunteers and leukemia patients. Thus far, the HL-60/AMSA genotype has not been identified in the cells from any individual, suggesting that this genotype is indeed a mutation and not an allelic form of topoisomerase II. The novel assay developed will allow a rapid search for the prevalence of this mutation in clinical samples from patients with leukemia who have relapsed following intercalator therapy.

    View details for Web of Science ID A1991GC74300039

    View details for PubMedID 1651812

  • Cross-resistance of an amsacrine-resistant human leukemia line to topoisomerase II reactive DNA intercalating agents. Evidence for two topoisomerase II directed drug actions. Biochemistry Zwelling, L. A., Mayes, J., Hinds, M., Chan, D., Altschuler, E., Carroll, B., Parker, E., Deisseroth, K., Radcliffe, A., Seligman, M. 1991; 30 (16): 4048-4055

    Abstract

    HL-60/AMSA is a human leukemia cell line that is 50-100-fold more resistant than its drug-sensitive HL-60 parent line to the cytotoxic actions of the DNA intercalator amsacrine (m-AMSA). HL-60/AMSA topoisomerase II is also resistant to the inhibitory actions of m-AMSA. HL-60/AMSA cells and topoisomerase II are cross-resistant to anthracycline and ellipticine intercalators but relatively sensitive to the nonintercalating topoisomerase II reactive epipodophyllotoxin etoposide. We now demonstrate that HL-60/AMSA and its topoisomerase II are cross-resistant to the DNA intercalators mitoxantrone and amonafide, thus strongly indicating that HL-60/AMSA and its topoisomerase II are resistant to topoisomerase II reactive intercalators but not to nonintercalators. At high concentrations, mitoxantrone and amonafide were also found to inhibit their own, m-AMSA's, and etoposide's abilities to stabilize topoisomerase II-DNA complexes. This appears to be due to the ability of these concentrations of mitoxantrone and amonafide to inhibit topoisomerase II mediated DNA strand passage at a point in the topoisomerization cycle prior to the acquisition of the enzyme-DNA configuration that yields DNA cleavage and topoisomerase II-DNA cross-links. In addition, amonafide can inhibit the cytotoxic actions of m-AMSA and etoposide. Taken together, these results suggest that the cytotoxicity of m-AMSA and etoposide is initiated primarily by the stabilization of the topoisomerase II-DNA complex. Other topoisomerase II reactive drugs may inhibit the enzyme at other steps in the topoisomerization cycle, particularly at elevated concentrations.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for PubMedID 1850298

  • CROSS-RESISTANCE OF AN AMSACRINE-RESISTANT HUMAN LEUKEMIA LINE TO TOPOISOMERASE-II REACTIVE DNA INTERCALATING AGENTS - EVIDENCE FOR 2 TOPOISOMERASE-II DIRECTED DRUG ACTIONS BIOCHEMISTRY Zwelling, L. A., Mayes, J., Hinds, M., Chan, D., Altschuler, E., Carroll, B., Parker, E., Deisseroth, K., Radcliffe, A., Seligman, M., Li, L., Farquhar, D. 1991; 30 (16): 4048-4055
  • A RESTRICTION-FRAGMENT-LENGTH-POLYMORPHISM FOR HUMAN TOPOISOMERASE .2. POSSIBLE RELATIONSHIP TO DRUG-RESISTANCE CANCER COMMUNICATIONS Zwelling, L. A., Mayes, J., Deisseroth, K., Hinds, M., GRANT, G., Pathak, S., Ledley, F. D., Vyas, R., Hittelman, W. 1990; 2 (11): 357-361

    Abstract

    In previous studies we used Southern blotting to examine the topoisomerase II locus (on chromosome 17) in human leukemia cell lines and noted a difference in the XmnI restriction endonuclease digestion pattern between an m-AMSA-resistant line and its m-AMSA-sensitive parent line (Zwelling, L. A.; Hinds, M,; Chan, D.; Mayes, J.; Sie, K. L.; Parker, E.; Silberman, L.; Radcliffe, A.; Beran, M.; Blick, M. Characterization of an amsacrine-resistant line of human leukemia cells. Evidence for a drug-resistant form of topoisomerase II. Journal of Biological Chemistry 264:16411-16420; 1989). We now demonstrate that the variable XmnI digestion pattern represents a normal restriction fragment length polymorphism (RFLP) which is observed in subjects without malignant disease and exhibits an autosomal pattern of inheritance. These data suggest that the previously described deviation in the genomic structure of topoisomerase II in the m-AMSA-resistant cell line did not reflect a new mutation, but rather a reduction to homozygosity at the topoisomerase II locus. This reduction to homozygosity is not due to chromosomal loss, as chromosome 17-specific gene probes clearly identify two chromosome 17's in the sensitive line and four in the resistant line, using chromosome painting with a chromosome 17-specific library. Some other genetic change must be the cause of the resistance of HL-60/AMSA and its topoisomerase II to the inhibiting actions of m-AMSA.

    View details for Web of Science ID A1990EJ28800001

    View details for PubMedID 1978687