Elsevier

Brain, Behavior, and Immunity

Volume 96, August 2021, Pages 279-289
Brain, Behavior, and Immunity

Review Article
Microglial heterogeneity in chronic pain

https://doi.org/10.1016/j.bbi.2021.06.005Get rights and content

Highlights

  • Microglia heterogeneity applies to both healthy and diseased CNS.

  • Regional microglia heterogeneity is associated to altered nociceptive processing.

  • Characterization of microglia heterogeneity will lead to innovative targets for chronic pain.

Abstract

In this review, we report existing preclinical evidence on how the CNS compartment as well as sex affect microglia functions in health. We highlight that recent advances in transcriptomics analyses have led to thorough characterization of disease-associated microglial states in mice and humans. We then consider the specific scenario of peripheral nerve or tissue injury which induce expression of a specific subset of genes in microglia in the dorsal horn of the spinal cord. We suggest the intriguing possibility that future studies may disclose the existence of a unique microglia transcriptional profile that is associated with chronic pain conditions. We also collect evidence that microglial activation in pain-related areas of the brain can be observed in models of neuropathic pain in agreement with recent neuroimaging studies in chronic pain patients. Based on the evidence discussed here, we predict that future studies on the neuroimmune interactions in chronic pain should complement our current understanding of microglia functions, but also adventure in using novel approaches such as scRNA-seq, spatial transcriptomics, CYTOF and transmission electron microscopy to provide a more complete characterization of the function, transcriptome and structure of microglia in chronic pain.

Introduction

In both brain and spinal cord, microglia are ubiquitous resident immune cells that constantly surveil the parenchyma via highly dynamic processes and interactions with neurons, vasculature and other glial cells (Bernier et al., 2019, Cserép et al., 2019). Unlike other tissue-resident and non-parenchymal brain macrophages, microglia originate from erythromyeloid progenitors in the yolk sac that arise earlier than other mononuclear phagocytes (Ginhoux et al., 2010, Utz et al., 2020). However, similar to tissue-resident macrophages, microglia markedly rely on environmental cues that shape their transcriptional profile and functional properties (Gosselin et al., 2014). Through multiple interactions with surrounding cells, microglia assume diverse biological roles that aim to maintenance of homeostasis. Such roles range from securing functional development of neuronal circuits to responding and regulating neuronal excitability and clearing the CNS parenchyma from potential pathogens or dead cells (Sierra et al., 2010, Stevens et al., 2007, Umpierre and Wu, 2020). Upon CNS injury or inflammatory insult, microglia shift from a homeostatic role to a reactive state that guarantees neuroprotection but can also lead to pronounced neuroinflammation (Stratoulias et al., 2019). This divergence from homeostasis is clearly represented at cellular and subcellular levels by the occurrence of morphological changes -rapid regulation of both motility and direction of branches- and prominent transcriptional changes (Davalos et al., 2005, Nimmerjahn et al., 2005). The advent of genome-wide association studies (GWAS) has marked an important paradigm shift in our understanding of microglia in disease states as they implicate genetic variants, that are either solely expressed by microglia or regulate microglia responses, as risk factors for neurodegenerative diseases (Jansen et al., 2019) and neuropsychiatric disorders (Sekar et al., 2016). Therefore, microglia are involved in disease aetiology and actively support and modulate neuronal function.

Although most neuronal and glial cells are known to be diverse across the CNS (Bayraktar et al., 2020, Marques et al., 2016, Saunders et al., 2018), microglial heterogeneity has only recently become the centre of attention. At first, with the intention to classify microglia functional modalities, microglial heterogeneity was based on the expression of membrane markers and morphological configurations, which resulted in the generation of fixed phenotypic categories. For instance, the M1/M2 framework has been adopted from the macrophage field to describe pro-inflammatory (M1) and anti-inflammatory (M2) functions of microglia following CNS insult. However, the use of this oversimplified nomenclature is widely criticized as misleading (Ransohoff, 2016) whereas there is more consensus towards using single-cell RNA sequencing (RNA-seq) analysis to characterize microglial heterogeneity. In the healthy murine brain, RNA-seq approach has demonstrated that microglia share a conserved expression of core genes that regulate homeostatic functions (Bennett et al., 2016, Butovsky et al., 2014, Li et al., 2019). This homeostatic gene signature is associated with various roles played by microglia in a region- and time-dependent fashion (Butovsky et al., 2014, De Biase et al., 2017, Masuda et al., 2019). Furthermore, microglia are endowed with the ability to sense the environment and readily respond to external stimuli across the CNS by changing the expression of the transcriptional homeostatic signature. For instance, in mouse models of Alzheimer’s disease (AD), multiple sclerosis and neuroinflammation, microglial heterogeneity manifests through the establishment of cell subsets that alter their transcriptional profile through down-regulation of some homeostatic genes (Cx3cr1, Tmem119, P2ry12, Sall1, Hexb, Fcrls) and up-regulation of disease-specific genes (Apoe, Trem2, Ctss, Axl) (Jordão et al., 2019, Keren-Shaul et al., 2017, Krasemann et al., 2017). In support of the concept that diverse microglial profiles can be observed in response to pathology, changes in gene expression are accompanied by alteration at the molecular level (Mrdjen et al., 2018), and for instance, microglia adjacent to amyloid plaques, the pathological hallmark of AD, express specific subsets of protein markers (Kamphuis et al., 2016, Parhizkar et al., 2019).

Several comprehensive reviews discuss microglial heterogeneity in the context of regional diversity, sexual dimorphism, healthy development, ageing and nervous system diseases (Masuda et al., 2020, Stratoulias et al., 2019, Tan et al., 2019). We discuss microglial heterogeneity in the context of chronic pain. In the last two decades, it has become apparent that under neuropathic pain conditions following peripheral damage, dorsal horn microglia in the spinal cord play a causal role in the establishment and maintenance of nociceptive mechanisms via well-defined pathways for communication with neurons (Malcangio, 2019). Typical examples of neuroimmune interactions include 1) neuron-derived ATP activation of microglial P2X4/BDNF pathway which results in a TrkB-mediated reduction of GABAergic inhibition of dorsal horn neurons and an overall increase in excitability (Coull et al., 2005, Masuda et al., 2016) 2) microglial P2X7 receptor activation by neuronal ATP that promotes release of the cysteine protease Cathepsin S which liberates neuronal fractalkine (CX3CL1) chemokine domain that further stimulates microglia to secrete pro-nociceptive cytokines via activation of CX3CR1 receptor (Clark et al., 2007a, Malcangio, 2019) and 3) de novo expression of colony-stimulating factor 1 (CSF1) in injured nociceptors which drives the expression of neuropathic pain-associated genes in dorsal horn microglia through binding to CSF1 receptor/DAP12 (Guan et al., 2016). Despite significant progress in our understanding of microglial pathways that facilitate nociceptive transmission, so far very few studies investigated microglial heterogeneous transcriptional changes in chronic pain states. Hence, this review provides an overview on the contemporary framework under which microglial heterogeneity is being studied in the CNS in the context of disease, sex, and intraregional differences. Then against this background, we discuss microglial heterogeneity in chronic pain. Our prediction is that the advent of novel technologies will facilitate a better characterization of functions, transcriptome and structure of microglia and their dynamic role in the establishment and maintenance of chronic pain. This type of approach may lead to the identification of innovative microglial targets for chronic pain treatments.

Section snippets

Membrane markers and morphology

With the aim to understand microglial functions in healthy murine CNS, early studies examined microglial distribution, morphology and membrane marker expression and suggested that CNS regional density accounts for microglia’s ability to adapt and respond to local cues from neuronal environments. Specifically, immunostaining of the membrane marker F4/80 indicates that microglia density in thalamus (101 cells/mm2), hippocampus (120 cells/mm2) and substantia nigra (134 cells/mm2) is higher than in

Chronic pain

In contrast to acute pain which is an adaptive process and a defensive mechanism from harmful stimuli, chronic pain is maladaptive, outlasts noxious stimuli and poorly controlled by current analgesics. Harmful stimuli are detected in the periphery of the body by specialized sensory neurons, namely the nociceptors, which transmit electrical impulses to the dorsal horn of the spinal cord. From this first relay station, nociceptive signalling continues to the higher centres in the brain where pain

Sensory discriminative

In addition to microglia heterogeneous immune responses across the CNS, there is clinical and preclinical evidence that alteration in microglia properties occurs in pain-related areas that are closely linked with both the sensory-discriminative and the affective-motivational component of chronic pain (Austin and Fiore, 2019). Preclinical data in neuropathic pain models indicates that microglia influence nociceptive mechanisms in a variety of brain regions (Fig. 3), as both Iba-1 gene expression

Conclusions

Microglia can assume diverse states to integrate inward and outward signals within microenvironments and under homeostasis, microglia exert multiple functions. Upon stimulation by a pathogen or in disease states, some microglia diverge from homeostatic profiles and acquire distinct modes of activation: cells alter morphology and membrane marker expression, as well as core transcriptional profiles (Mathys et al., 2019, Sala Frigerio et al., 2019). Thus, the association of a microglia

Acknowledgements

The authors acknowledge funds from the European Union’s Horizon 2020 research and innovation programme “TOBeATPAIN” under the Marie Skłodowska-Curie grant agreement No 764860.

We specially thank Ana Rita Alves da Silva for providing the image showing microgliosis in the dorsal horn of the spinal cord in animals with nerve injury (Fig. 2B).

References (102)

  • D. Gosselin et al.

    Environment drives selection and function of enhancers controlling tissue-specific macrophage identities

    Cell

    (2014)
  • N. Gu et al.

    Spinal microgliosis due to resident microglial proliferation is required for pain hypersensitivity after peripheral nerve injury

    Cell Rep.

    (2016)
  • D. Guneykaya et al.

    Transcriptional and translational differences of microglia from male and female brains

    Cell Rep.

    (2018)
  • R.R. Ji et al.

    Glia and pain: is chronic pain a gliopathy?

    Pain

    (2013)
  • W. Kamphuis et al.

    Transcriptional profiling of CD11c-positive microglia accumulating around amyloid plaques in a mouse model for Alzheimer’s disease

    Biochim. Biophys. Acta – Mol. Basis Dis.

    (2016)
  • H. Keren-Shaul et al.

    A unique microglia type associated with restricting development of Alzheimer’s Disease

    Cell

    (2017)
  • S. Krasemann et al.

    The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases

    Immunity

    (2017)
  • L.J. Lawson et al.

    Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain

    Neuroscience

    (1990)
  • Q. Li et al.

    Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing

    Neuron

    (2019)
  • Z. Li et al.

    Spinal versus brain microglial and macrophage activation traits determine the differential neuroinflammatory responses and analgesic effect of minocycline in chronic neuropathic pain

    Brain. Behav. Immun.

    (2016)
  • M. Malcangio

    Spinal mechanisms of neuropathic pain: Is there a P2X4-BDNF controversy? Neurobiol

    Pain

    (2017)
  • T. Masuda et al.

    Microglia heterogeneity in the single-cell era

    Cell Rep.

    (2020)
  • T. Masuda et al.

    IRF8 is a critical transcription factor for transforming microglia into a reactive phenotype

    Cell Rep.

    (2012)
  • D. Mrdjen et al.

    High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease

    Immunity

    (2018)
  • A. Saunders et al.

    Molecular diversity and specializations among the cells of the adult mouse brain

    Cell

    (2018)
  • A. Sierra et al.

    Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis

    Cell Stem Cell

    (2010)
  • R.E. Sorge et al.

    Sex differences in pain responses

    Curr. Opin. Physiol.

    (2018)
  • B. Stevens et al.

    The classical complement cascade mediates CNS synapse elimination

    Cell

    (2007)
  • P. Süß et al.

    Chronic peripheral inflammation causes a region-specific myeloid response in the central nervous system

    Cell Rep.

    (2020)
  • N. Thrupp et al.

    Single-nucleus RNA-Seq is not suitable for detection of microglial activation genes in humans

    Cell Rep.

    (2020)
  • A. Villa et al.

    Sexual differentiation of microglia

    Front. Neuroendocrinol.

    (2019)
  • A. Villa et al.

    Sex-specific features of microglia from adult mice

    Cell Rep.

    (2018)
  • P. Ayata et al.

    Epigenetic regulation of brain region-specific microglia clearance activity

    Nat. Neurosci.

    (2018)
  • A. Badimon et al.

    Negative feedback control of neuronal activity by microglia

    Nature

    (2020)
  • E.E. Barcelon et al.

    Brain microglial activation in chronic pain-associated affective disorder

    Front. Neurosci.

    (2019)
  • Bayraktar, O.A., Bartels, T., Holmqvist, S., Kleshchevnikov, V., Martirosyan, A., Polioudakis, D., Ben Haim, L., Young,...
  • M.L. Bennett et al.

    New tools for studying microglia in the mouse and human CNS

    Proc. Natl. Acad. Sci. U. S. A.

    (2016)
  • M.C. Bushnell et al.

    Cognitive and emotional control of pain and its disruption in chronic pain

    Nat. Rev. Neurosci.

    (2013)
  • O. Butovsky et al.

    Identification of a unique TGF-β-dependent molecular and functional signature in microglia

    Nat. Neurosci.

    (2014)
  • Chu Sin Chung, P., Panigada, T., Cardis, R., Decosterd, I., Gosselin, R.D., 2017. Peripheral nerve injury induces a...
  • A.K. Clark et al.

    Selective activation of microglia facilitates synaptic strength

    J. Neurosci.

    (2015)
  • A.K. Clark et al.

    Inhibition of spinal microglial cathepsin S for the reversal of neuropathic pain

    Proc. Natl. Acad. Sci.

    (2007)
  • J.A.M. Coull et al.

    BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain

    Nature

    (2005)
  • C. Cserép et al.

    Microglia monitor and protect neuronal function via specialized somatic purinergic junctions

    Science (80-.

    (2019)
  • D. Davalos et al.

    ATP mediates rapid microglial response to local brain injury in vivo

    Nat. Neurosci.

    (2005)
  • A.H. de Haas et al.

    Region-specific expression of immunoregulatory proteins on microglia in the healthy CNS

    Glia

    (2008)
  • T. Fernandez-Zafra et al.

    Exploring the transcriptome of resident spinal microglia after collagen antibody-induced arthritis

    Pain

    (2019)
  • A. Forsberg et al.

    Disease activity in rheumatoid arthritis is inversely related to cerebral TSPO binding assessed by [11C]PBR28 positron emission tomography

    J. Neuroimmunol.

    (2019)
  • E. Furube et al.

    Brain region-dependent heterogeneity and dose-dependent difference in transient microglia population increase during lipopolysaccharide-induced inflammation

    Sci. Rep.

    (2018)
  • T.F. Galatro et al.

    Transcriptomic analysis of purified human cortical microglia reveals age-associated changes

    Nat. Neurosci.

    (2017)
  • Cited by (23)

    View all citing articles on Scopus
    View full text