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

Substantial evidence indicates involvement of microglia/macrophages in chronic neuropathic pain. However, the temporal-spatial features of microglial/macrophage activation and their pain-bound roles remain elusive. Here, we evaluated microglia/macrophages and the subtypes in the lumbar spinal cord (SC) and prefrontal cortex (PFC), and analgesic-anxiolytic effect of minocycline at different stages following spared nerve injury (SNI) in rats. While SNI enhanced the number of spinal microglia/macrophages since post-operative day (POD)3, pro-inflammatory MHCII+ spinal microglia/macrophages were unexpectedly less abundant in SNI rats than shams on POD21. By contrast, less abundant anti-inflammatory CD172a (SIRPα)+ microglia/macrophages were found in the PFC of SNI rats. Interestingly in naïve rats, microglial/macrophage expression of CD11b/c, MHCII and MHCII+/CD172a+ ratio were higher in the SC than the cortex. Consistently, multiple immune genes involved in anti-inflammation, phagocytosis, complement activation and M2 microglial/macrophage polarization were upregulated in the spinal dorsal horn and dorsal root ganglia but downregulated in the PFC of SNI rats. Furthermore, daily intrathecal minocycline treatment starting from POD0 for two weeks alleviated mechanical allodynia most robustly before POD3 and attenuated anxiety on POD9. Although minocycline dampened spinal MHCII+ microglia/macrophages until POD13, it failed to do so on cortical microglia/macrophages, indicating that dampening only spinal inflammation may not be enough to alleviate centralized pain at the chronic stage. Taken together, our data provide the first evidence that basal microglial/macrophage traits underlie differential region-specific responses to SNI and minocycline treatment, and suggest that drug treatment efficiently targeting not only spinal but also brain inflammation may be more effective in treating chronic neuropathic pain.

[1]  H. Tozaki-Saitoh,et al.  Microglial regulation of neuropathic pain. , 2013, Journal of pharmacological sciences.

[2]  M. R. Costa,et al.  Astrocyte heterogeneity in the brain: from development to disease , 2015, Front. Cell. Neurosci..

[3]  Ji Zhang,et al.  Characterization of cell proliferation in rat spinal cord following peripheral nerve injury and the relationship with neuropathic pain , 2008, PAIN®.

[4]  I. Nikonenko,et al.  Distribution of microglia and astrocytes in different regions of the normal adult rat brain , 1997, Neurophysiology.

[5]  L. Watkins,et al.  Pathological and protective roles of glia in chronic pain , 2009, Nature Reviews Neuroscience.

[6]  S. Maier,et al.  Pathological pain and the neuroimmune interface , 2014, Nature Reviews Immunology.

[7]  R. Leak,et al.  Neurobiology of microglial action in CNS injuries: Receptor-mediated signaling mechanisms and functional roles , 2014, Progress in Neurobiology.

[8]  R. Leak,et al.  Microglial and macrophage polarization—new prospects for brain repair , 2015, Nature Reviews Neurology.

[9]  Hilla Peretz,et al.  Ju n 20 03 Schrödinger ’ s Cat : The rules of engagement , 2003 .

[10]  S. Verma,et al.  Comorbidities in chronic neuropathic pain. , 2004, Pain medicine.

[11]  A. Piotrowska,et al.  Parthenolide Relieves Pain and Promotes M2 Microglia/Macrophage Polarization in Rat Model of Neuropathy , 2015, Neural plasticity.

[12]  Jason G. Jin,et al.  Complement activation in the peripheral nervous system following the spinal nerve ligation model of neuropathic pain , 2008, PAIN®.

[13]  Clifford J. Woolf,et al.  Complement Induction in Spinal Cord Microglia Results in Anaphylatoxin C5a-Mediated Pain Hypersensitivity , 2007, The Journal of Neuroscience.

[14]  C. Glass,et al.  Microglial cell origin and phenotypes in health and disease , 2011, Nature Reviews Immunology.

[15]  J. Olson Immune response by microglia in the spinal cord , 2010, Annals of the New York Academy of Sciences.

[16]  M. Olmstead,et al.  Neurobiology of Disease Microglia Disrupt Mesolimbic Reward Circuitry in Chronic Pain , 2015 .

[17]  R. Ransohoff,et al.  The myeloid cells of the central nervous system parenchyma , 2010, Nature.

[18]  B. Melchior,et al.  A rose by any other name? the potential consequences of microglial heterogeneity during CNS health and disease , 2007, Neurotherapeutics.

[19]  A. Gourine,et al.  Differential Sensitivity of Brainstem versus Cortical Astrocytes to Changes in pH Reveals Functional Regional Specialization of Astroglia , 2013, The Journal of Neuroscience.

[20]  V. Võikar,et al.  Microglia are polarized to M1 type in high-anxiety inbred mice in response to lipopolysaccharide challenge , 2014, Brain, Behavior, and Immunity.

[21]  H. Neumann,et al.  Microglial activatory (immunoreceptor tyrosine‐based activation motif)‐ and inhibitory (immunoreceptor tyrosine‐based inhibition motif)‐signaling receptors for recognition of the neuronal glycocalyx , 2013, Glia.

[22]  V. Perry,et al.  Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain , 1990, Neuroscience.

[23]  S. McMahon,et al.  Crosstalk between the nociceptive and immune systems in host defence and disease , 2015, Nature Reviews Neuroscience.

[24]  R. Franco,et al.  Alternatively activated microglia and macrophages in the central nervous system , 2015, Progress in Neurobiology.

[25]  J. Deleo,et al.  Inhibition of Microglial Activation Attenuates the Development but Not Existing Hypersensitivity in a Rat Model of Neuropathy , 2003, Journal of Pharmacology and Experimental Therapeutics.

[26]  M. Opp,et al.  Reduced sleep, stress responsivity, and female sex contribute to persistent inflammation-induced mechanical hypersensitivity in rats , 2014, Brain, Behavior, and Immunity.

[27]  T. Sugimoto,et al.  Activated Microglia Contribute to Convergent Nociceptive Inputs to Spinal Dorsal Horn Neurons and the Development of Neuropathic Pain , 2015, Neurochemical Research.

[28]  Maria Fitzgerald,et al.  T-Cell Infiltration and Signaling in the Adult Dorsal Spinal Cord Is a Major Contributor to Neuropathic Pain-Like Hypersensitivity , 2009, The Journal of Neuroscience.

[29]  M. O’Banion,et al.  Are “Resting” Microglia More “M2”? , 2014, Front. Immunol..

[30]  T. Michoel,et al.  Microglial brain regiondependent diversity and selective regional sensitivities to aging , 2015 .

[31]  B. Przewłocka,et al.  Glial inhibitors influence the mRNA and protein levels of mGlu2/3, 5 and 7 receptors and potentiate the analgesic effects of their ligands in a mouse model of neuropathic pain , 2009, PAIN®.

[32]  Zongbin Song,et al.  Microglial polarization dynamics in dorsal spinal cord in the early stages following chronic sciatic nerve damage , 2016, Neuroscience Letters.

[33]  D. Finn,et al.  Minocycline modulates neuropathic pain behaviour and cortical M1–M2 microglial gene expression in a rat model of depression , 2014, Brain, Behavior, and Immunity.

[34]  R. Herrera-Molina,et al.  Transforming growth factor-β1 produced by hippocampal cells modulates microglial reactivity in culture , 2005, Neurobiology of Disease.

[35]  S. Maier,et al.  Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation , 2005, Pain.

[36]  C. Woolf,et al.  Spared nerve injury: an animal model of persistent peripheral neuropathic pain , 2000, Pain.

[37]  A. Rice,et al.  Quantification of the rat spinal microglial response to peripheral nerve injury as revealed by immunohistochemical image analysis and flow cytometry , 2007, Journal of Neuroscience Methods.

[38]  H. Boddeke,et al.  Brain region-specific gene expression profiles in freshly isolated rat microglia , 2015, Front. Cell. Neurosci..

[39]  S. Waxman,et al.  Minocycline attenuates mechanical allodynia and central sensitization following peripheral second-degree burn injury. , 2010, The journal of pain : official journal of the American Pain Society.

[40]  J. Deleo,et al.  Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain , 2004, Neurochemistry International.

[41]  R. Dantzer,et al.  Microglial/macrophage GRK2 determines duration of peripheral IL-1β-induced hyperalgesia: Contribution of spinal cord CX3CR1, p38 and IL-1 signaling , 2010, PAIN.

[42]  H. Aldskogius,et al.  Microglia and neuropathic pain. , 2013, CNS & neurological disorders drug targets.

[43]  V. Perry,et al.  Acute inflammatory responses to mechanical lesions in the CNS: differences between brain and spinal cord , 1999, The European journal of neuroscience.

[44]  R. Meyer,et al.  Mechanisms of Neuropathic Pain , 2006, Neuron.

[45]  E. Benarroch Central neuron-glia interactions and neuropathic pain , 2010, Neurology.

[46]  T. Joensuu,et al.  Abnormal microglial activation in the Cstb−/− mouse, a model for progressive myoclonus epilepsy, EPM1 , 2015, Glia.

[47]  J. Ravetch,et al.  A Novel Role for the IgG Fc Glycan: The Anti-inflammatory Activity of Sialylated IgG Fcs , 2010, Journal of Clinical Immunology.

[48]  Suzana Herculano-Houzel,et al.  The glia/neuron ratio: How it varies uniformly across brain structures and species and what that means for brain physiology and evolution , 2014, Glia.

[49]  E. Senba,et al.  Site-specific subtypes of macrophages recruited after peripheral nerve injury , 2011, Neuroreport.

[50]  A Suzumura,et al.  Minocycline selectively inhibits M1 polarization of microglia , 2013, Cell Death and Disease.

[51]  Shi-Ying Huang,et al.  Minocycline and fluorocitrate suppress spinal nociceptive signaling in intrathecal IL‐1β–induced thermal hyperalgesic rats , 2012, Glia.

[52]  M. Graeber,et al.  Multiple mechanisms of microglia: A gatekeeper's contribution to pain states , 2012, Experimental Neurology.