Skip to main content
Top
Published in: Molecular Brain 1/2017

Open Access 01-12-2017 | Review

Molecules in pain and sex: a developing story

Authors: Josiane C. S. Mapplebeck, Simon Beggs, Michael W. Salter

Published in: Molecular Brain | Issue 1/2017

Login to get access

Abstract

Microglia are dynamic immune cells with diverse roles in maintaining homeostasis of the central nervous system. Dysregulation of microglia has been critically implicated in the genesis of neuropathic pain. Peripheral nerve injury, a common cause of neuropathic pain, engages microglia-neuronal signalling which causes disinhibition and facilitated excitation of spinal nociceptive pathways. However, recent literature indicates that the role of microglia in neuropathic pain is sexually dimorphic, and that female pain processing appears to be independent of microglia, depending rather on T cells. Despite this sex difference, pain signalling in the spinal cord converges downstream of microglia, as NMDAR-mediated facilitated excitation in pain transmitting neurons is consistent between males and females. Determining whether pain signalling is sexually dimorphic in humans and, further, addressing the sex bias in pain research will increase the translational relevance of preclinical findings and advance our understanding of chronic pain in women.
Literature
1.
go back to reference Pizzo P, Clark N. Relieving pain in America: a blueprint for transforming prevention, care, education, and research (Institute of medicine). Washington, DC: The National Academies Press; 2011. Pizzo P, Clark N. Relieving pain in America: a blueprint for transforming prevention, care, education, and research (Institute of medicine). Washington, DC: The National Academies Press; 2011.
2.
go back to reference Bouhassira D, Lantéri-Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain. 2008;136(3):380–7.CrossRefPubMed Bouhassira D, Lantéri-Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain. 2008;136(3):380–7.CrossRefPubMed
3.
go back to reference Merskey H, Bogduk N. Classification of chronic pain. 2nd ed. Seattle: IASP Press; 1994. Merskey H, Bogduk N. Classification of chronic pain. 2nd ed. Seattle: IASP Press; 1994.
4.
go back to reference Woolf CJ, Mannion RJ. Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet (London, England). 1999;353(9168):1959–64.CrossRef Woolf CJ, Mannion RJ. Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet (London, England). 1999;353(9168):1959–64.CrossRef
5.
go back to reference Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature. 2003;424(6950):778–83.CrossRefPubMed Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature. 2003;424(6950):778–83.CrossRefPubMed
6.
go back to reference Coull JA, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, Gravel C, Salter MW, De Koninck Y. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature. 2005;438(7070):1017–21.CrossRefPubMed Coull JA, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, Gravel C, Salter MW, De Koninck Y. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature. 2005;438(7070):1017–21.CrossRefPubMed
8.
go back to reference Sorge RE, Mapplebeck JCS, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin J-S, Sotocinal SG, Chen D, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci. 2015;18:1081–3.CrossRefPubMedPubMedCentral Sorge RE, Mapplebeck JCS, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin J-S, Sotocinal SG, Chen D, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci. 2015;18:1081–3.CrossRefPubMedPubMedCentral
9.
go back to reference Klein SL, Schiebinger L, Stefanick ML, Cahill L, Danska J, de Vries GJ, Kibbe MR, McCarthy MM, Mogil JS, Woodruff TK, et al. Opinion: sex inclusion in basic research drives discovery. Proc Natl Acad Sci U S A. 2015;112(17):5257–8.CrossRefPubMedPubMedCentral Klein SL, Schiebinger L, Stefanick ML, Cahill L, Danska J, de Vries GJ, Kibbe MR, McCarthy MM, Mogil JS, Woodruff TK, et al. Opinion: sex inclusion in basic research drives discovery. Proc Natl Acad Sci U S A. 2015;112(17):5257–8.CrossRefPubMedPubMedCentral
10.
go back to reference Prendergast BJ, Onishi KG, Zucker I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci Biobehav Rev. 2014;40:1–5.CrossRefPubMed Prendergast BJ, Onishi KG, Zucker I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci Biobehav Rev. 2014;40:1–5.CrossRefPubMed
11.
go back to reference Mogil JS, Chanda ML. The case for the inclusion of female subjects in basic science studies of pain. Pain. 2005;117(1–2):1–5.CrossRefPubMed Mogil JS, Chanda ML. The case for the inclusion of female subjects in basic science studies of pain. Pain. 2005;117(1–2):1–5.CrossRefPubMed
12.
go back to reference Beery AK, Zucker I. Sex bias in neuroscience and biomedical research. Neurosci Biobehav Rev. 2011;35(3):565–72.CrossRefPubMed Beery AK, Zucker I. Sex bias in neuroscience and biomedical research. Neurosci Biobehav Rev. 2011;35(3):565–72.CrossRefPubMed
13.
go back to reference Fillingim RBKC, Ribeiro-Dasilva MC, Rahim-Williams B, Riley 3rd JL. Sex, gender, and pain: a review of recent clinical and experimental findings. J Pain. 2009;10(5):447–85.CrossRefPubMedPubMedCentral Fillingim RBKC, Ribeiro-Dasilva MC, Rahim-Williams B, Riley 3rd JL. Sex, gender, and pain: a review of recent clinical and experimental findings. J Pain. 2009;10(5):447–85.CrossRefPubMedPubMedCentral
14.
go back to reference Torrance NSB, Bennett MI, Lee AJ. The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. J Pain. 2006;7(4):281–9.CrossRefPubMed Torrance NSB, Bennett MI, Lee AJ. The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. J Pain. 2006;7(4):281–9.CrossRefPubMed
15.
go back to reference Mogil JS. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci. 2012;13(12):859–66.CrossRefPubMed Mogil JS. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci. 2012;13(12):859–66.CrossRefPubMed
17.
go back to reference Niesters M, Dahan A, Kest B, Zacny J, Stijnen T, Aarts L, Sarton E. Do sex differences exist in opioid analgesia? A systematic review and meta-analysis of human experimental and clinical studies. Pain. 2010;151(1):61–8.CrossRefPubMed Niesters M, Dahan A, Kest B, Zacny J, Stijnen T, Aarts L, Sarton E. Do sex differences exist in opioid analgesia? A systematic review and meta-analysis of human experimental and clinical studies. Pain. 2010;151(1):61–8.CrossRefPubMed
18.
go back to reference Aguzzi A, Barres BA, Bennett ML. Microglia: scapegoat, saboteur, or something else? Science (New York, NY). 2013;339(6116):156–61.CrossRef Aguzzi A, Barres BA, Bennett ML. Microglia: scapegoat, saboteur, or something else? Science (New York, NY). 2013;339(6116):156–61.CrossRef
20.
go back to reference Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19(8):312–8.CrossRefPubMed Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19(8):312–8.CrossRefPubMed
22.
go back to reference Ginhoux FGM, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science (New York, NY). 2010;330(6005):841–5.CrossRef Ginhoux FGM, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science (New York, NY). 2010;330(6005):841–5.CrossRef
23.
go back to reference Fogg DK, Sibon C, Miled C, Jung S, Aucouturier P, Littman DR, Cumano A, Geissmann F. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science (New York, NY). 2006;311(5757):83–7.CrossRef Fogg DK, Sibon C, Miled C, Jung S, Aucouturier P, Littman DR, Cumano A, Geissmann F. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science (New York, NY). 2006;311(5757):83–7.CrossRef
24.
go back to reference Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FMV. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007;10(12):1538–43.CrossRefPubMed Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FMV. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007;10(12):1538–43.CrossRefPubMed
25.
go back to reference Guan Z, Kuhn JA, Wang X, Colquitt B, Solorzano C, Vaman S, Guan AK, Evans-Reinsch Z, Braz J, Devor M, et al. Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Nat Neurosci. 2016;19:94–101. Guan Z, Kuhn JA, Wang X, Colquitt B, Solorzano C, Vaman S, Guan AK, Evans-Reinsch Z, Braz J, Devor M, et al. Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Nat Neurosci. 2016;19:94–101.
26.
go back to reference Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8(6):752–8.CrossRefPubMed Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8(6):752–8.CrossRefPubMed
27.
go back to reference Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science (New York, NY). 2005;308(5726):1314–8.CrossRef Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science (New York, NY). 2005;308(5726):1314–8.CrossRef
28.
go back to reference Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9(12):1512–9.CrossRefPubMed Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9(12):1512–9.CrossRefPubMed
29.
go back to reference Li Y, Du XF, Liu CS, Wen ZL, Du JL. Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo. Dev Cell. 2012;23(6):1189–202.CrossRefPubMed Li Y, Du XF, Liu CS, Wen ZL, Du JL. Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo. Dev Cell. 2012;23(6):1189–202.CrossRefPubMed
30.
go back to reference Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, et al. Synaptic pruning by microglia is necessary for normal brain development. Science (New York, NY). 2011;333(6048):1456–8.CrossRef Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, et al. Synaptic pruning by microglia is necessary for normal brain development. Science (New York, NY). 2011;333(6048):1456–8.CrossRef
31.
go back to reference Roumier A, Bechade C, Poncer JC, Smalla KH, Tomasello E, Vivier E, Gundelfinger ED, Triller A, Bessis A. Impaired synaptic function in the microglial KARAP/DAP12-deficient mouse. J Neurosci. 2004;24(50):11421–8.CrossRefPubMed Roumier A, Bechade C, Poncer JC, Smalla KH, Tomasello E, Vivier E, Gundelfinger ED, Triller A, Bessis A. Impaired synaptic function in the microglial KARAP/DAP12-deficient mouse. J Neurosci. 2004;24(50):11421–8.CrossRefPubMed
32.
go back to reference Parkhurst CN, Yang G, Ninan I, Savas JN, Yates 3rd JR, Lafaille JJ, Hempstead BL, Littman DR, Gan WB. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell. 2013;155(7):1596–609.CrossRefPubMedPubMedCentral Parkhurst CN, Yang G, Ninan I, Savas JN, Yates 3rd JR, Lafaille JJ, Hempstead BL, Littman DR, Gan WB. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell. 2013;155(7):1596–609.CrossRefPubMedPubMedCentral
33.
go back to reference Neumann H, Kotter MR, Franklin RJ. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain. 2009;132(Pt 2):288–95.PubMed Neumann H, Kotter MR, Franklin RJ. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain. 2009;132(Pt 2):288–95.PubMed
34.
go back to reference Salter M, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014;158(1):15–24.CrossRefPubMed Salter M, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014;158(1):15–24.CrossRefPubMed
35.
go back to reference Tashima R, Mikuriya S, Tomiyama D, Shiratori-Hayashi M, Yamashita T, Kohro Y, Tozaki-Saitoh H, Inoue K, Tsuda M. Bone marrow-derived cells in the population of spinal microglia after peripheral nerve injury. Sci Rep. 2016;6:23701.CrossRefPubMedPubMedCentral Tashima R, Mikuriya S, Tomiyama D, Shiratori-Hayashi M, Yamashita T, Kohro Y, Tozaki-Saitoh H, Inoue K, Tsuda M. Bone marrow-derived cells in the population of spinal microglia after peripheral nerve injury. Sci Rep. 2016;6:23701.CrossRefPubMedPubMedCentral
36.
go back to reference Gu N, Peng J, Murugan M, Wang X, Eyo UB, Sun D, Ren Y, DiCicco-Bloom E, Young W, Dong H, et al. Spinal microgliosis Due to resident microglial proliferation is required for pain hypersensitivity after peripheral nerve injury. Cell Rep. 2016;16(3):605–14.CrossRefPubMedPubMedCentral Gu N, Peng J, Murugan M, Wang X, Eyo UB, Sun D, Ren Y, DiCicco-Bloom E, Young W, Dong H, et al. Spinal microgliosis Due to resident microglial proliferation is required for pain hypersensitivity after peripheral nerve injury. Cell Rep. 2016;16(3):605–14.CrossRefPubMedPubMedCentral
37.
go back to reference Masuda T, Iwamoto S, Yoshinaga R, Tozaki-Saitoh H, Nishiyama A, Mak TW, Tamura T, Tsuda M, Inoue K. Transcription factor IRF5 drives P2X4R + −reactive microglia gating neuropathic pain. Nat Commun. 2014;5:3771.CrossRefPubMedPubMedCentral Masuda T, Iwamoto S, Yoshinaga R, Tozaki-Saitoh H, Nishiyama A, Mak TW, Tamura T, Tsuda M, Inoue K. Transcription factor IRF5 drives P2X4R + −reactive microglia gating neuropathic pain. Nat Commun. 2014;5:3771.CrossRefPubMedPubMedCentral
38.
go back to reference Ulmann L, Hatcher JP, Hughes JP, Chaumont S, Green PJ, Conquet F, Buell GN, Reeve AJ, Chessell IP, Rassendren F. Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain. J Neurosci. 2008;28(44):11263–8.CrossRefPubMed Ulmann L, Hatcher JP, Hughes JP, Chaumont S, Green PJ, Conquet F, Buell GN, Reeve AJ, Chessell IP, Rassendren F. Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain. J Neurosci. 2008;28(44):11263–8.CrossRefPubMed
39.
go back to reference Tozaki-Saitoh H, Tsuda M, Miyata H, Ueda K, Kohsaka S, Inoue K. P2Y12 receptors in spinal microglia are required for neuropathic pain after peripheral nerve injury. J Neurosci. 2008;28(19):4949–56.CrossRefPubMed Tozaki-Saitoh H, Tsuda M, Miyata H, Ueda K, Kohsaka S, Inoue K. P2Y12 receptors in spinal microglia are required for neuropathic pain after peripheral nerve injury. J Neurosci. 2008;28(19):4949–56.CrossRefPubMed
40.
go back to reference Mapplebeck JCS, Beggs S, Salter MW. Sex Differences in Pain: A Tale of Two Immune Cells. Pain. 2015;Publish Ahead of Print. Mapplebeck JCS, Beggs S, Salter MW. Sex Differences in Pain: A Tale of Two Immune Cells. Pain. 2015;Publish Ahead of Print.
41.
go back to reference Beggs S, Salter MW. The known knowns of microglia-neuronal signalling in neuropathic pain. Neurosci Lett. 2013;557 Pt A:37–42.CrossRefPubMed Beggs S, Salter MW. The known knowns of microglia-neuronal signalling in neuropathic pain. Neurosci Lett. 2013;557 Pt A:37–42.CrossRefPubMed
42.
go back to reference Masuda T, Tsuda M, Yoshinaga R, Tozaki-Saitoh H, Ozato K, Tamura T, Inoue K. IRF8 is a critical transcription factor for transforming microglia into a reactive phenotype. Cell Rep. 2012;1(4):334–40.CrossRefPubMedPubMedCentral Masuda T, Tsuda M, Yoshinaga R, Tozaki-Saitoh H, Ozato K, Tamura T, Inoue K. IRF8 is a critical transcription factor for transforming microglia into a reactive phenotype. Cell Rep. 2012;1(4):334–40.CrossRefPubMedPubMedCentral
43.
go back to reference Guo W, Miyoshi K, Dubner R, Gu M, Li M, Liu J, Yang J, Zou S, Ren K, Noguchi K, et al. Spinal 5-HT3 receptors mediate descending facilitation and contribute to behavioral hypersensitivity via a reciprocal neuron-glial signaling cascade. Mol Pain. 2014;10(1):35.PubMedPubMedCentral Guo W, Miyoshi K, Dubner R, Gu M, Li M, Liu J, Yang J, Zou S, Ren K, Noguchi K, et al. Spinal 5-HT3 receptors mediate descending facilitation and contribute to behavioral hypersensitivity via a reciprocal neuron-glial signaling cascade. Mol Pain. 2014;10(1):35.PubMedPubMedCentral
44.
go back to reference Wei F, Dubner R, Zou S, Ren K, Bai G, Wei D, Guo W. Molecular depletion of descending serotonin unmasks its novel facilitatory role in the development of persistent pain. J Neurosci. 2010;30:8624–36.CrossRefPubMedPubMedCentral Wei F, Dubner R, Zou S, Ren K, Bai G, Wei D, Guo W. Molecular depletion of descending serotonin unmasks its novel facilitatory role in the development of persistent pain. J Neurosci. 2010;30:8624–36.CrossRefPubMedPubMedCentral
45.
go back to reference Keller AF, Beggs S, Salter MW, De Koninck Y. Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol Pain. 2007;3:27.CrossRefPubMedPubMedCentral Keller AF, Beggs S, Salter MW, De Koninck Y. Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol Pain. 2007;3:27.CrossRefPubMedPubMedCentral
46.
go back to reference Masuda T, Ozono Y, Mikuriya S, Kohro Y, Tozaki-Saitoh H, Iwatsuki K, Uneyama H, Ichikawa R, Salter MW, Tsuda M, et al. Dorsal horn neurons release extracellular ATP in a VNUT-dependent manner that underlies neuropathic pain. Nat Commun. 2016;7:12529.CrossRefPubMedPubMedCentral Masuda T, Ozono Y, Mikuriya S, Kohro Y, Tozaki-Saitoh H, Iwatsuki K, Uneyama H, Ichikawa R, Salter MW, Tsuda M, et al. Dorsal horn neurons release extracellular ATP in a VNUT-dependent manner that underlies neuropathic pain. Nat Commun. 2016;7:12529.CrossRefPubMedPubMedCentral
47.
go back to reference Trang T, Beggs S, Wan X, Salter MW. P2X4-receptor-mediated synthesis and release of brain-derived neurotrophic factor in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation. J Neurosci. 2009;29(11):3518–28.CrossRefPubMedPubMedCentral Trang T, Beggs S, Wan X, Salter MW. P2X4-receptor-mediated synthesis and release of brain-derived neurotrophic factor in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation. J Neurosci. 2009;29(11):3518–28.CrossRefPubMedPubMedCentral
48.
go back to reference Coull JA, Boudreau D, Bachand K, Prescott SA, Nault F, Sik A, De Koninck P, De Koninck Y. Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain. Nature. 2003;424(6951):938–42.CrossRefPubMed Coull JA, Boudreau D, Bachand K, Prescott SA, Nault F, Sik A, De Koninck P, De Koninck Y. Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain. Nature. 2003;424(6951):938–42.CrossRefPubMed
49.
go back to reference Payne JA. Functional characterization of the neuronal-specific K-Cl cotransporter: implications for [K+]oregulation. Am J Physiol Cell Physiol. 1997;273(5):C1516–25. Payne JA. Functional characterization of the neuronal-specific K-Cl cotransporter: implications for [K+]oregulation. Am J Physiol Cell Physiol. 1997;273(5):C1516–25.
50.
go back to reference Hübner CA, Stein V, Hermans-Borgmeyer I, Meyer T, Ballanyi K, Jentsch TJ. Disruption of KCC2 reveals an essential role of K-Cl cotransport already in early synaptic inhibition. Neuron. 2001;30(2):515–24.CrossRefPubMed Hübner CA, Stein V, Hermans-Borgmeyer I, Meyer T, Ballanyi K, Jentsch TJ. Disruption of KCC2 reveals an essential role of K-Cl cotransport already in early synaptic inhibition. Neuron. 2001;30(2):515–24.CrossRefPubMed
51.
go back to reference Gagnon M, Bergeron MJ, Lavertu G, Castonguay A, Tripathy S, Bonin RP, Perez-Sanchez J, Boudreau D, Wang B, Dumas L, et al. Chloride extrusion enhancers as novel therapeutics for neurological diseases. Nat Med. 2013;19(11):1524–8.CrossRefPubMedPubMedCentral Gagnon M, Bergeron MJ, Lavertu G, Castonguay A, Tripathy S, Bonin RP, Perez-Sanchez J, Boudreau D, Wang B, Dumas L, et al. Chloride extrusion enhancers as novel therapeutics for neurological diseases. Nat Med. 2013;19(11):1524–8.CrossRefPubMedPubMedCentral
52.
go back to reference Asiedu M, Ossipov MH, Kaila K, Price TJ. Acetazolamide and midazolam act synergistically to inhibit neuropathic pain. Pain. 2010;148(2):302–8.CrossRefPubMed Asiedu M, Ossipov MH, Kaila K, Price TJ. Acetazolamide and midazolam act synergistically to inhibit neuropathic pain. Pain. 2010;148(2):302–8.CrossRefPubMed
53.
go back to reference Maren TH. Carbonic anhydrase: chemistry, physiology, and inhibition. Physiol Rev. 1967;47(4):595–781.PubMed Maren TH. Carbonic anhydrase: chemistry, physiology, and inhibition. Physiol Rev. 1967;47(4):595–781.PubMed
54.
go back to reference Salter MW, Kalia LV. Src kinases: a hub for NMDA receptor regulation. Nat Rev Neurosci. 2004;5(4):317–28.CrossRefPubMed Salter MW, Kalia LV. Src kinases: a hub for NMDA receptor regulation. Nat Rev Neurosci. 2004;5(4):317–28.CrossRefPubMed
55.
go back to reference Liu XJ, Gingrich JR, Vargas-Caballero M, Dong YN, Sengar A, Beggs S, Wang SH, Ding HK, Frankland PW, Salter MW. Treatment of inflammatory and neuropathic pain by uncoupling Src from the NMDA receptor complex. Nat Med. 2008;14(12):1325–32.CrossRefPubMedPubMedCentral Liu XJ, Gingrich JR, Vargas-Caballero M, Dong YN, Sengar A, Beggs S, Wang SH, Ding HK, Frankland PW, Salter MW. Treatment of inflammatory and neuropathic pain by uncoupling Src from the NMDA receptor complex. Nat Med. 2008;14(12):1325–32.CrossRefPubMedPubMedCentral
56.
go back to reference Parsons SJ, Parsons JT. Src family kinases, key regulators of signal transduction. Oncogene. 2004;23(48):7906–9.CrossRefPubMed Parsons SJ, Parsons JT. Src family kinases, key regulators of signal transduction. Oncogene. 2004;23(48):7906–9.CrossRefPubMed
57.
go back to reference Gingrich JR, Pelkey KA, Fam SR, Huang Y, Petralia RS, Wenthold RJ, Salter MW. Unique domain anchoring of Src to synaptic NMDA receptors via the mitochondrial protein NADH dehydrogenase subunit 2. Proc Natl Acad Sci U S A. 2004;101(16):6237–42.CrossRefPubMedPubMedCentral Gingrich JR, Pelkey KA, Fam SR, Huang Y, Petralia RS, Wenthold RJ, Salter MW. Unique domain anchoring of Src to synaptic NMDA receptors via the mitochondrial protein NADH dehydrogenase subunit 2. Proc Natl Acad Sci U S A. 2004;101(16):6237–42.CrossRefPubMedPubMedCentral
58.
go back to reference Chen W, Walwyn W, Ennes HS, Kim H, McRoberts JA, Marvizon JC. BDNF released during neuropathic pain potentiates NMDA receptors in primary afferent terminals. Eur J Neurosci. 2014;39(9):1439–54.CrossRefPubMedPubMedCentral Chen W, Walwyn W, Ennes HS, Kim H, McRoberts JA, Marvizon JC. BDNF released during neuropathic pain potentiates NMDA receptors in primary afferent terminals. Eur J Neurosci. 2014;39(9):1439–54.CrossRefPubMedPubMedCentral
59.
go back to reference Hildebrand ME, Xu J, Dedek A, Li Y, Sengar AS, Beggs S, Lombroso PJ, Salter MW. Potentiation of synaptic GluN2B NMDAR currents by Fyn kinase is gated through BDNF-mediated disinhibition in spinal pain processing. Cell Rep. 2016;17(10):2753–65.CrossRefPubMed Hildebrand ME, Xu J, Dedek A, Li Y, Sengar AS, Beggs S, Lombroso PJ, Salter MW. Potentiation of synaptic GluN2B NMDAR currents by Fyn kinase is gated through BDNF-mediated disinhibition in spinal pain processing. Cell Rep. 2016;17(10):2753–65.CrossRefPubMed
60.
go back to reference Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T, Binaglia M, Corsini E, Di Luca M, Galli CL, et al. Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J Neurosci. 2003;23(25):8692–700.PubMed Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T, Binaglia M, Corsini E, Di Luca M, Galli CL, et al. Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J Neurosci. 2003;23(25):8692–700.PubMed
61.
go back to reference Sorge RE, LaCroix-Fralish ML, Tuttle AH, Sotocinal SG, Austin JS, Ritchie J, Chanda ML, Graham AC, Topham L, Beggs S, et al. Spinal cord toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J Neurosci. 2011;31(43):15450–4.CrossRefPubMedPubMedCentral Sorge RE, LaCroix-Fralish ML, Tuttle AH, Sotocinal SG, Austin JS, Ritchie J, Chanda ML, Graham AC, Topham L, Beggs S, et al. Spinal cord toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J Neurosci. 2011;31(43):15450–4.CrossRefPubMedPubMedCentral
62.
go back to reference Miller SI, Ernst RK, Bader MW. LPS, TLR4 and infectious disease diversity. Nat Rev Microbiol. 2005;3(1):36–46.CrossRefPubMed Miller SI, Ernst RK, Bader MW. LPS, TLR4 and infectious disease diversity. Nat Rev Microbiol. 2005;3(1):36–46.CrossRefPubMed
63.
go back to reference Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87(2):149–58.CrossRefPubMed Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87(2):149–58.CrossRefPubMed
64.
go back to reference Taves S, Berta T, Liu DL, Gan S, Chen G, Kim YH, Van de Ven T, Laufer S, Ji RR. Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: Sex-dependent microglial signaling in the spinal cord. Brain Behav Immun. 2015;55:70-81. Taves S, Berta T, Liu DL, Gan S, Chen G, Kim YH, Van de Ven T, Laufer S, Ji RR. Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: Sex-dependent microglial signaling in the spinal cord. Brain Behav Immun. 2015;55:70-81. 
65.
go back to reference Yang Y, Li H, Li T-T, Luo H, Gu X-Y, Lü N, Ji R-R, Zhang Y-Q. Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female wistar rats via P2X7 receptor and IL-18. J Neurosci. 2015;35(20):7950–63.CrossRefPubMed Yang Y, Li H, Li T-T, Luo H, Gu X-Y, Lü N, Ji R-R, Zhang Y-Q. Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female wistar rats via P2X7 receptor and IL-18. J Neurosci. 2015;35(20):7950–63.CrossRefPubMed
66.
go back to reference Rosen S, Ham B, Mogil JS. Sex differences in neuroimmunity and pain. J Neurosci Res. 2017;95(1–2):500–8.CrossRefPubMed Rosen S, Ham B, Mogil JS. Sex differences in neuroimmunity and pain. J Neurosci Res. 2017;95(1–2):500–8.CrossRefPubMed
68.
go back to reference Farza H, Salmon AM, Hadchouel M, Moreau JL, Babinet C, Tiollais P, Pourcel C. Hepatitis B surface antigen gene expression is regulated by sex steroids and glucocorticoids in transgenic mice. Proc Natl Acad Sci U S A. 1987;84(5):1187–91.CrossRefPubMedPubMedCentral Farza H, Salmon AM, Hadchouel M, Moreau JL, Babinet C, Tiollais P, Pourcel C. Hepatitis B surface antigen gene expression is regulated by sex steroids and glucocorticoids in transgenic mice. Proc Natl Acad Sci U S A. 1987;84(5):1187–91.CrossRefPubMedPubMedCentral
69.
go back to reference Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert T, Barrett L, Brenner G, Vardeh D, Woolf C, et al. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci. 2009;29(46):14415–22.CrossRefPubMedPubMedCentral Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert T, Barrett L, Brenner G, Vardeh D, Woolf C, et al. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci. 2009;29(46):14415–22.CrossRefPubMedPubMedCentral
70.
go back to reference Cao L, DeLeo J. CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. Eur J Immunol. 2008;38(2):448–58.CrossRefPubMedPubMedCentral Cao L, DeLeo J. CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. Eur J Immunol. 2008;38(2):448–58.CrossRefPubMedPubMedCentral
71.
go back to reference Sweitzer SM, Hickey WF, Rutkowski MD, Pahl JL, DeLeo JA. Focal peripheral nerve injury induces leukocyte trafficking into the central nervous system: potential relationship to neuropathic pain. Pain. 2002;100(1–2):163–70.CrossRefPubMed Sweitzer SM, Hickey WF, Rutkowski MD, Pahl JL, DeLeo JA. Focal peripheral nerve injury induces leukocyte trafficking into the central nervous system: potential relationship to neuropathic pain. Pain. 2002;100(1–2):163–70.CrossRefPubMed
72.
go back to reference Zhang MA, Rego D, Moshkova M, Kebir H, Chruscinski A, Nguyen H, Akkermann R, Stanczyk FZ, Prat A, Steinman L, et al. Peroxisome proliferator-activated receptor (PPAR)alpha and -gamma regulate IFNgamma and IL-17A production by human T cells in a sex-specific way. Proc Natl Acad Sci U S A. 2012;109(24):9505–10.CrossRefPubMedPubMedCentral Zhang MA, Rego D, Moshkova M, Kebir H, Chruscinski A, Nguyen H, Akkermann R, Stanczyk FZ, Prat A, Steinman L, et al. Peroxisome proliferator-activated receptor (PPAR)alpha and -gamma regulate IFNgamma and IL-17A production by human T cells in a sex-specific way. Proc Natl Acad Sci U S A. 2012;109(24):9505–10.CrossRefPubMedPubMedCentral
73.
go back to reference Draleau K, Maddula S, Slaiby A, Nutile-McMenemy N, De Leo J, Cao L. Phenotypic identification of spinal cord-infiltrating CD4(+) T lymphocytes in a Murine model of neuropathic pain. J Pain Relief. 2014;Suppl 3:003.PubMedPubMedCentral Draleau K, Maddula S, Slaiby A, Nutile-McMenemy N, De Leo J, Cao L. Phenotypic identification of spinal cord-infiltrating CD4(+) T lymphocytes in a Murine model of neuropathic pain. J Pain Relief. 2014;Suppl 3:003.PubMedPubMedCentral
74.
go back to reference Ko JS, Eddinger KA, Angert M, Chernov AV, Dolkas J, Strongin AY, Yaksh TL, Shubayev VI. Spinal activity of interleukin 6 mediates myelin basic protein-induced allodynia. Brain Behav Immun. 2016;56:378–89.CrossRefPubMed Ko JS, Eddinger KA, Angert M, Chernov AV, Dolkas J, Strongin AY, Yaksh TL, Shubayev VI. Spinal activity of interleukin 6 mediates myelin basic protein-induced allodynia. Brain Behav Immun. 2016;56:378–89.CrossRefPubMed
75.
go back to reference Liu H, Shiryaev SA, Chernov AV, Kim Y, Shubayev I, Remacle AG, Baranovskaya S, Golubkov VS, Strongin AY, Shubayev VI. Immunodominant fragments of myelin basic protein initiate T cell-dependent pain. J Neuroinflammation. 2012;9:119.PubMedPubMedCentral Liu H, Shiryaev SA, Chernov AV, Kim Y, Shubayev I, Remacle AG, Baranovskaya S, Golubkov VS, Strongin AY, Shubayev VI. Immunodominant fragments of myelin basic protein initiate T cell-dependent pain. J Neuroinflammation. 2012;9:119.PubMedPubMedCentral
76.
go back to reference Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald-Smith GP, Gao H, Hennessy L, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2013;110(9):3507–12.CrossRefPubMedPubMedCentral Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald-Smith GP, Gao H, Hennessy L, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2013;110(9):3507–12.CrossRefPubMedPubMedCentral
77.
go back to reference Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol (Baltimore, Md : 1950). 2004;172(5):2731–8.CrossRef Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol (Baltimore, Md : 1950). 2004;172(5):2731–8.CrossRef
78.
go back to reference Del Valle L, Schwartzman RJ, Alexander G. Spinal cord histopathological alterations in a patient with longstanding complex regional pain syndrome. Brain Behav Immun. 2009;23(1):85–91.CrossRefPubMed Del Valle L, Schwartzman RJ, Alexander G. Spinal cord histopathological alterations in a patient with longstanding complex regional pain syndrome. Brain Behav Immun. 2009;23(1):85–91.CrossRefPubMed
79.
go back to reference Shi Y, Gelman BB, Lisinicchia JG, Tang SJ. Chronic-pain-associated astrocytic reaction in the spinal cord dorsal horn of human immunodeficiency virus-infected patients. J Neurosci. 2012;32(32):10833–40.CrossRefPubMedPubMedCentral Shi Y, Gelman BB, Lisinicchia JG, Tang SJ. Chronic-pain-associated astrocytic reaction in the spinal cord dorsal horn of human immunodeficiency virus-infected patients. J Neurosci. 2012;32(32):10833–40.CrossRefPubMedPubMedCentral
80.
go back to reference Loggia ML, Chonde DB, Akeju O, Arabasz G, Catana C, Edwards RR, Hill E, Hsu S, Izquierdo-Garcia D, Ji RR, et al. Evidence for brain glial activation in chronic pain patients. Brain. 2015;138(Pt 3):604–15.CrossRefPubMedPubMedCentral Loggia ML, Chonde DB, Akeju O, Arabasz G, Catana C, Edwards RR, Hill E, Hsu S, Izquierdo-Garcia D, Ji RR, et al. Evidence for brain glial activation in chronic pain patients. Brain. 2015;138(Pt 3):604–15.CrossRefPubMedPubMedCentral
81.
go back to reference Banati RB, Cagnin A, Brooks DJ, Gunn RN, Myers R, Jones T, Birch R, Anand P. Long-term trans-synaptic glial responses in the human thalamus after peripheral nerve injury. Neuroreport. 2001;12(16):3439–42.CrossRefPubMed Banati RB, Cagnin A, Brooks DJ, Gunn RN, Myers R, Jones T, Birch R, Anand P. Long-term trans-synaptic glial responses in the human thalamus after peripheral nerve injury. Neuroreport. 2001;12(16):3439–42.CrossRefPubMed
82.
go back to reference Landry RP, Jacobs VL, Romero-Sandoval EA, DeLeo JA. Propentofylline, a CNS glial modulator does not decrease pain in post-herpetic neuralgia patients: in vitro evidence for differential responses in human and rodent microglia and macrophages. Exp Neurol. 2012;234(2):340–50.CrossRefPubMed Landry RP, Jacobs VL, Romero-Sandoval EA, DeLeo JA. Propentofylline, a CNS glial modulator does not decrease pain in post-herpetic neuralgia patients: in vitro evidence for differential responses in human and rodent microglia and macrophages. Exp Neurol. 2012;234(2):340–50.CrossRefPubMed
Metadata
Title
Molecules in pain and sex: a developing story
Authors
Josiane C. S. Mapplebeck
Simon Beggs
Michael W. Salter
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Brain / Issue 1/2017
Electronic ISSN: 1756-6606
DOI
https://doi.org/10.1186/s13041-017-0289-8

Other articles of this Issue 1/2017

Molecular Brain 1/2017 Go to the issue