Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury
Abstract
:1. Introduction
2. Materials and Methods
2.1. Mice
2.2. Processing of scRNA-Seq Data
2.3. Gene Set Enrichment Analysis
2.4. Partial Sciatic Nerve Ligation (PSL) Model
2.5. Administration of Pexidartinib (PLX3397)
2.6. Immunohistochemistry
2.7. Reverse Transcription-Quantitative Polymerase Chain Reaction (RT-qPCR)
2.8. Drug Administration
2.9. Von Frey Test
2.10. Gene Expression Analysis
2.11. Statistical Analysis
3. Results
3.1. Expression of CCL4 by Male-Dominant Subpopulation of Spinal Microglia After Nerve Injury
3.2. Male Microglia-Dominant Upregulation of CCL4 in the Spinal Dorsal Horn
3.3. Sexually Dimorphic Effect of CCR5 Antagonist in Relieving Neuropathic Pain
3.4. Sex-Independent Allodynic Effects of CCL4 in the Spinal Dorsal Horn
4. Discussion
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
CCL4 | CC-chemokine ligand 4 |
CCR5 | CC-chemokine receptor 5 |
CSF1 | colony-stimulating factor 1 |
GSEA | gene set enrichment analysis |
PSL | partial sciatic nerve ligation |
RT-qPCR | reverse transcription-quantitative polymerase chain reaction |
scRNA-seq | single-cell RNA sequencing |
SDH | spinal dorsal horn |
SNI | spared nerve injury |
UMAP | uniform manifold approximation and projection |
References
- Julius, D.; Basbaum, A.I. Molecular mechanisms of nociception. Nature 2001, 413, 203–210. [Google Scholar] [CrossRef] [PubMed]
- Basbaum, A.I.; Bautista, D.M.; Scherrer, G.; Julius, D. Cellular and molecular mechanisms of pain. Cell 2009, 139, 267–284. [Google Scholar] [CrossRef] [PubMed]
- Cohen, S.P.; Vase, L.; Hooten, W.M. Chronic pain: An update on burden, best practices, and new advances. Lancet 2021, 397, 2082–2097. [Google Scholar] [CrossRef]
- Colloca, L.; Ludman, T.; Bouhassira, D.; Baron, R.; Dickenson, A.H.; Yarnitsky, D.; Freeman, R.; Truini, A.; Attal, N.; Finnerup, N.B.; et al. Neuropathic pain. Nat. Rev. Dis. Primers 2017, 3, 17002. [Google Scholar] [CrossRef] [PubMed]
- Ji, R.R.; Chamessian, A.; Zhang, Y.Q. Pain regulation by non-neuronal cells and inflammation. Science 2016, 354, 572–577. [Google Scholar] [CrossRef]
- Tsuda, M.; Masuda, T.; Kohno, K. Microglial diversity in neuropathic pain. Trends Neurosci. 2023, 46, 597–610. [Google Scholar] [CrossRef]
- Midavaine, E.; Cote, J.; Marchand, S.; Sarret, P. Glial and neuroimmune cell choreography in sexually dimorphic pain signaling. Neurosci. Biobehav. Rev. 2021, 125, 168–192. [Google Scholar] [CrossRef]
- Mapplebeck, J.C.S.; Beggs, S.; Salter, M.W. Sex differences in pain: A tale of two immune cells. Pain 2016, 157 (Suppl. S1), S2–S6. [Google Scholar] [CrossRef]
- Saika, F.; Fukazawa, Y.; Hatano, Y.; Kishioka, S.; Hino, Y.; Hino, S.; Suzuki, K.; Kiguchi, N. Sexually dimorphic effects of pexidartinib on nerve injury-induced neuropathic pain in mice. Glia 2024, 72, 1402–1417. [Google Scholar] [CrossRef]
- Sorge, R.E.; Mapplebeck, J.C.; Rosen, S.; Beggs, S.; Taves, S.; Alexander, J.K.; Martin, L.J.; Austin, J.S.; Sotocinal, S.G.; Chen, D.; et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat. Neurosci. 2015, 18, 1081–1083. [Google Scholar] [CrossRef]
- Taves, S.; Berta, T.; Liu, D.L.; Gan, S.; Chen, G.; Kim, Y.H.; Van de Ven, T.; Laufer, S.; Ji, R.R. 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. 2016, 55, 70–81. [Google Scholar] [CrossRef] [PubMed]
- Butovsky, O.; Weiner, H.L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 2018, 19, 622–635. [Google Scholar] [CrossRef]
- Inoue, K.; Tsuda, M. Microglia in neuropathic pain: Cellular and molecular mechanisms and therapeutic potential. Nat. Rev. Neurosci. 2018, 19, 138–152. [Google Scholar] [CrossRef]
- Kuhn, J.A.; Vainchtein, I.D.; Braz, J.; Hamel, K.; Bernstein, M.; Craik, V.; Dahlgren, M.W.; Ortiz-Carpena, J.; Molofsky, A.B.; Molofsky, A.V.; et al. Regulatory T-cells inhibit microglia-induced pain hypersensitivity in female mice. Elife 2021, 10, e69056. [Google Scholar] [CrossRef]
- Sorge, R.E.; LaCroix-Fralish, M.L.; Tuttle, A.H.; Sotocinal, S.G.; Austin, J.S.; Ritchie, J.; Chanda, M.L.; Graham, A.C.; 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, 15450–15454. [Google Scholar] [CrossRef]
- Lee, J.; Hwang, H.; Lee, S.J. Distinct roles of GT1b and CSF-1 in microglia activation in nerve injury-induced neuropathic pain. Mol. Pain. 2021, 17, 17448069211020918. [Google Scholar] [CrossRef] [PubMed]
- Saika, F.; Matsuzaki, S.; Kishioka, S.; Kiguchi, N. Chemogenetic Activation of CX3CR1-Expressing Spinal Microglia Using Gq-DREADD Elicits Mechanical Allodynia in Male Mice. Cells 2021, 10, 874. [Google Scholar] [CrossRef] [PubMed]
- Distefano-Gagne, F.; Bitarafan, S.; Lacroix, S.; Gosselin, D. Roles and regulation of microglia activity in multiple sclerosis: Insights from animal models. Nat. Rev. Neurosci. 2023, 24, 397–415. [Google Scholar] [CrossRef]
- Paolicelli, R.C.; Sierra, A.; Stevens, B.; Tremblay, M.E.; Aguzzi, A.; Ajami, B.; Amit, I.; Audinat, E.; Bechmann, I.; Bennett, M.; et al. Microglia states and nomenclature: A field at its crossroads. Neuron 2022, 110, 3458–3483. [Google Scholar] [CrossRef]
- Tansley, S.; Uttam, S.; Urena Guzman, A.; Yaqubi, M.; Pacis, A.; Parisien, M.; Deamond, H.; Wong, C.; Rabau, O.; Brown, N.; et al. Single-cell RNA sequencing reveals time- and sex-specific responses of mouse spinal cord microglia to peripheral nerve injury and links ApoE to chronic pain. Nat. Commun. 2022, 13, 843. [Google Scholar] [CrossRef]
- Korsunsky, I.; Millard, N.; Fan, J.; Slowikowski, K.; Zhang, F.; Wei, K.; Baglaenko, Y.; Brenner, M.; Loh, P.R.; Raychaudhuri, S. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 2019, 16, 1289–1296. [Google Scholar] [CrossRef] [PubMed]
- Hao, Y.; Stuart, T.; Kowalski, M.H.; Choudhary, S.; Hoffman, P.; Hartman, A.; Srivastava, A.; Molla, G.; Madad, S.; Fernandez-Granda, C.; et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat. Biotechnol. 2024, 42, 293–304. [Google Scholar] [CrossRef] [PubMed]
- Subramanian, A.; Tamayo, P.; Mootha, V.K.; Mukherjee, S.; Ebert, B.L.; Gillette, M.A.; Paulovich, A.; Pomeroy, S.L.; Golub, T.R.; Lander, E.S.; et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 2005, 102, 15545–15550. [Google Scholar] [CrossRef] [PubMed]
- Liberzon, A.; Birger, C.; Thorvaldsdottir, H.; Ghandi, M.; Mesirov, J.P.; Tamayo, P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015, 1, 417–425. [Google Scholar] [CrossRef] [PubMed]
- Kiguchi, N.; Kobayashi, D.; Saika, F.; Matsuzaki, S.; Kishioka, S. Inhibition of peripheral macrophages by nicotinic acetylcholine receptor agonists suppresses spinal microglial activation and neuropathic pain in mice with peripheral nerve injury. J. Neuroinflamm. 2018, 15, 96. [Google Scholar] [CrossRef]
- Seltzer, Z.; Dubner, R.; Shir, Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990, 43, 205–218. [Google Scholar] [CrossRef]
- Elmore, M.R.; Najafi, A.R.; Koike, M.A.; Dagher, N.N.; Spangenberg, E.E.; Rice, R.A.; Kitazawa, M.; Matusow, B.; Nguyen, H.; West, B.L.; et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014, 82, 380–397. [Google Scholar] [CrossRef]
- Kwiatkowski, K.; Piotrowska, A.; Rojewska, E.; Makuch, W.; Jurga, A.; Slusarczyk, J.; Trojan, E.; Basta-Kaim, A.; Mika, J. Beneficial properties of maraviroc on neuropathic pain development and opioid effectiveness in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 2016, 64, 68–78. [Google Scholar] [CrossRef]
- Kiguchi, N.; Uta, D.; Ding, H.; Uchida, H.; Saika, F.; Matsuzaki, S.; Fukazawa, Y.; Abe, M.; Sakimura, K.; Ko, M.C.; et al. GRP receptor and AMPA receptor cooperatively regulate itch-responsive neurons in the spinal dorsal horn. Neuropharmacology 2020, 170, 108025. [Google Scholar] [CrossRef]
- Chaplan, S.R.; Bach, F.W.; Pogrel, J.W.; Chung, J.M.; Yaksh, T.L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 1994, 53, 55–63. [Google Scholar] [CrossRef]
- Comerford, I.; McColl, S.R. Atypical chemokine receptors in the immune system. Nat. Rev. Immunol. 2024, 24, 753–769. [Google Scholar] [CrossRef]
- Proudfoot, A.E. Chemokine receptors: Multifaceted therapeutic targets. Nat. Rev. Immunol. 2002, 2, 106–115. [Google Scholar] [CrossRef]
- Dorr, P.; Westby, M.; Dobbs, S.; Griffin, P.; Irvine, B.; Macartney, M.; Mori, J.; Rickett, G.; Smith-Burchnell, C.; Napier, C.; et al. Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrob. Agents Chemother. 2005, 49, 4721–4732. [Google Scholar] [CrossRef] [PubMed]
- Tsuda, M.; Shigemoto-Mogami, Y.; Koizumi, S.; Mizokoshi, A.; Kohsaka, S.; Salter, M.W.; Inoue, K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 2003, 424, 778–783. [Google Scholar] [CrossRef] [PubMed]
- Milligan, E.D.; Watkins, L.R. Pathological and protective roles of glia in chronic pain. Nat. Rev. Neurosci. 2009, 10, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Matsushita, K.; Tozaki-Saitoh, H.; Kojima, C.; Masuda, T.; Tsuda, M.; Inoue, K.; Hoka, S. Chemokine (C-C motif) receptor 5 is an important pathological regulator in the development and maintenance of neuropathic pain. Anesthesiology 2014, 120, 1491–1503. [Google Scholar] [CrossRef]
- Ji, R.R.; Xu, Z.Z.; Gao, Y.J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 2014, 13, 533–548. [Google Scholar] [CrossRef]
- Mogil, J.S. Qualitative sex differences in pain processing: Emerging evidence of a biased literature. Nat. Rev. Neurosci. 2020, 21, 353–365. [Google Scholar] [CrossRef]
- Mogil, J.S. Animal models of pain: Progress and challenges. Nat. Rev. Neurosci. 2009, 10, 283–294. [Google Scholar] [CrossRef]
- Calvo, M.; Dawes, J.M.; Bennett, D.L. The role of the immune system in the generation of neuropathic pain. Lancet Neurol. 2012, 11, 629–642. [Google Scholar] [CrossRef]
- 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, 334–340. [Google Scholar] [CrossRef]
- Guan, Z.; Kuhn, J.A.; Wang, X.; Colquitt, B.; Solorzano, C.; Vaman, S.; Guan, A.K.; 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. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Wu, L.; Deng, H.; Chen, Y.; Zhou, H.; Liu, M.; Wang, S.; Zheng, L.; Zhu, L.; Lv, X. Anti-inflammatory protein TSG-6 secreted by bone marrow mesenchymal stem cells attenuates neuropathic pain by inhibiting the TLR2/MyD88/NF-kappaB signaling pathway in spinal microglia. J. Neuroinflamm. 2020, 17, 154. [Google Scholar] [CrossRef] [PubMed]
- Koch, A.E.; Kunkel, S.L.; Shah, M.R.; Fu, R.; Mazarakis, D.D.; Haines, G.K.; Burdick, M.D.; Pope, R.M.; Strieter, R.M. Macrophage inflammatory protein-1 beta: A C-C chemokine in osteoarthritis. Clin. Immunol. Immunopathol. 1995, 77, 307–314. [Google Scholar] [CrossRef]
- Chang, T.T.; Chen, J.W. Emerging role of chemokine CC motif ligand 4 related mechanisms in diabetes mellitus and cardiovascular disease: Friends or foes? Cardiovasc. Diabetol. 2016, 15, 117. [Google Scholar] [CrossRef] [PubMed]
- Mukaida, N.; Sasaki, S.I.; Baba, T. CCL4 Signaling in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1231, 23–32. [Google Scholar]
- Ciechanowska, A.; Mika, J. CC Chemokine Family Members’ Modulation as a Novel Approach for Treating Central Nervous System and Peripheral Nervous System Injury-A Review of Clinical and Experimental Findings. Int. J. Mol. Sci. 2024, 25, 3788. [Google Scholar] [CrossRef]
- Saika, F.; Kiguchi, N.; Kobayashi, Y.; Fukazawa, Y.; Kishioka, S. CC-chemokine ligand 4/macrophage inflammatory protein-1beta participates in the induction of neuropathic pain after peripheral nerve injury. Eur. J. Pain. 2012, 16, 1271–1280. [Google Scholar] [CrossRef]
- Xia, M.Q.; Qin, S.X.; Wu, L.J.; Mackay, C.R.; Hyman, B.T. Immunohistochemical study of the beta-chemokine receptors CCR3 and CCR5 and their ligands in normal and Alzheimer’s disease brains. Am. J. Pathol. 1998, 153, 31–37. [Google Scholar] [CrossRef]
- Ciechanowska, A.; Popiolek-Barczyk, K.; Pawlik, K.; Ciapala, K.; Oggioni, M.; Mercurio, D.; De Simoni, M.G.; Mika, J. Changes in macrophage inflammatory protein-1 (MIP-1) family members expression induced by traumatic brain injury in mice. Immunobiology 2020, 225, 151911. [Google Scholar] [CrossRef]
- Rojewska, E.; Zychowska, M.; Piotrowska, A.; Kreiner, G.; Nalepa, I.; Mika, J. Involvement of Macrophage Inflammatory Protein-1 Family Members in the Development of Diabetic Neuropathy and Their Contribution to Effectiveness of Morphine. Front. Immunol. 2018, 9, 494. [Google Scholar] [CrossRef] [PubMed]
- Sorce, S.; Myburgh, R.; Krause, K.H. The chemokine receptor CCR5 in the central nervous system. Prog. Neurobiol. 2011, 93, 297–311. [Google Scholar] [CrossRef] [PubMed]
- Festa, B.P.; Siddiqi, F.H.; Jimenez-Sanchez, M.; Won, H.; Rob, M.; Djajadikerta, A.; Stamatakou, E.; Rubinsztein, D.C. Microglial-to-neuronal CCR5 signaling regulates autophagy in neurodegeneration. Neuron 2023, 111, 2021–2037.e12. [Google Scholar] [CrossRef]
- Dong, F.L.; Yu, L.; Feng, P.D.; Ren, J.X.; Bai, X.H.; Lin, J.Q.; Cao, D.L.; Deng, Y.T.; Zhang, Y.; Shen, H.H.; et al. An atlas of neuropathic pain-associated molecular pathological characteristics in the mouse spinal cord. Commun. Biol. 2025, 8, 70. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Saika, F.; Sato, T.; Nakabayashi, T.; Fukazawa, Y.; Hino, S.; Suzuki, K.; Kiguchi, N. Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury. Cells 2025, 14, 484. https://doi.org/10.3390/cells14070484
Saika F, Sato T, Nakabayashi T, Fukazawa Y, Hino S, Suzuki K, Kiguchi N. Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury. Cells. 2025; 14(7):484. https://doi.org/10.3390/cells14070484
Chicago/Turabian StyleSaika, Fumihiro, Tetsuya Sato, Takeru Nakabayashi, Yohji Fukazawa, Shinjiro Hino, Kentaro Suzuki, and Norikazu Kiguchi. 2025. "Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury" Cells 14, no. 7: 484. https://doi.org/10.3390/cells14070484
APA StyleSaika, F., Sato, T., Nakabayashi, T., Fukazawa, Y., Hino, S., Suzuki, K., & Kiguchi, N. (2025). Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury. Cells, 14(7), 484. https://doi.org/10.3390/cells14070484