Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter October 30, 2023

Dominant mechanism in spinal cord injury-induced immunodeficiency syndrome (SCI-IDS): sympathetic hyperreflexia

  • Ping Yang EMAIL logo , Zhi-qun Bian , Zhen-bo Song , Cheng-ying Yang , Li Wang and Zhong-xiang Yao ORCID logo EMAIL logo

Abstract

Clinical studies have shown that individuals with spinal cord injury (SCI) are particularly susceptible to infectious diseases, resulting in a syndrome called SCI-induced immunodeficiency syndrome (SCI-IDS), which is the leading cause of death after SCI. It is believed that SCI-IDS is associated with exaggerated activation of sympathetic preganglionic neurons (SPNs). After SCI, disruption of bulbospinal projections from the medulla oblongata C1 neurons to the SPNs results in the loss of sympathetic inhibitory modulation from the brain and brainstem and the occurrence of abnormally high levels of spinal sympathetic reflexes (SSR), named sympathetic hyperreflexia. As the post-injury survival time lengthens, mass recruitment and anomalous sprouting of excitatory interneurons within the spinal cord result in increased SSR excitability, resulting in an excess sympathetic output that disrupts the immune response. Therefore, we first analyze the structural underpinnings of the spinal cord-sympathetic nervous system-immune system after SCI, then demonstrate the progress in highlighting mechanisms of SCI-IDS focusing on norepinephrine (NE)/Beta 2-adrenergic receptor (β2-AR) signal pathways, and summarize recent preclinical studies examining potential means such as regulating SSR and inhibiting β2-AR signal pathways to improve immune function after SCI. Finally, we present research perspectives such as to promote the effective regeneration of C1 neurons to rebuild the connection of C1 neurons with SPNs, to regulate excitable or inhibitory interneurons, and specifically to target β2-AR signal pathways to re-establish neuroimmune balance. These will help us design effective strategies to reverse post-SCI sympathetic hyperreflexia and improve the overall quality of life for individuals with SCI.


Corresponding authors: Ping Yang, Department of Neurobiology, Army Medical University (Third Military Medical University), Chongqing 400038, China, E-mail: ; and Zhong-xiang Yao, Department of Physiology, Army Medical University (Third Military Medical University), Chongqing 400038, China, E-mail:

Funding source: National Natural Science Foundation of China

Award Identifier / Grant number: 82271423, 82071761

  1. Research ethics: Not applicable.

  2. Author contributions: PY collected the literature, analyzed the data, and wrote the manuscript. ZBS was involved in the schematic drawing. LW, CYY, and ZQB participated in part in the collection and discussion of the document. PY and ZXY conceived, designed, supervised, initialed, reviewed, edited and revised the manuscript. The authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: The authors state no conflict of interest.

  4. Research funding: This work was supported by grants from the National Natural Science Foundation of China (No. 82271423 to PY and No. 82071761 to CYY).

  5. Data availability: Not applicable.

References

Abe, C. and Inoue, T. (2018). Role of C1 neurons in anti-inflammatory reflex: mediation between afferents and efferents. Neurosci. Res. 136: 6–12, https://doi.org/10.1016/j.neures.2018.05.001.Search in Google Scholar PubMed

Abe, C., Inoue, T., Inglis, M.A., Viar, K.E., Huang, L., Ye, H., Rosin, D.L., Stornetta, R.L., Okusa, M.D., and Guyenet, P.G. (2017). C1 neurons mediate a stress-induced anti-inflammatory reflex in mice. Nat. Neurosci. 20: 700–707, https://doi.org/10.1038/nn.4526.Search in Google Scholar PubMed PubMed Central

Adib, Y., Bensussan, A., and Michel, L. (2022). Cutaneous wound healing: a review about innate immune response and current therapeutic applications. Mediat. Inflamm. 2022: 5344085, https://doi.org/10.1155/2022/5344085.Search in Google Scholar PubMed PubMed Central

Bauer, C.S., Nieto-Rostro, M., Rahman, W., Tran-Van-Minh, A., Ferron, L., Douglas, L., Kadurin, I., Sri Ranjan, Y., Fernandez-Alacid, L., Millar, N.S., et al.. (2009). The increased trafficking of the calcium channel subunit alpha2delta-1 to presynaptic terminals in neuropathic pain is inhibited by the alpha2delta ligand pregabalin. J. Neurosci. 29: 4076–4088, https://doi.org/10.1523/jneurosci.0356-09.2009.Search in Google Scholar

Bellinger, D.L. and Lorton, D. (2014). Autonomic regulation of cellular immune function. Auton. Neurosci. 182: 15–41, https://doi.org/10.1016/j.autneu.2014.01.006.Search in Google Scholar PubMed

Bethea, J.R., Nagashima, H., Acosta, M.C., Briceno, C., Gomez, F., Marcillo, A.E., Loor, K., Green, J., and Dietrich, W.D. (1999). Systemically administered interleukin-10 reduces tumor necrosis factor-α production and significantly improves functional recovery following traumatic spinal cord injury in rats. J. Neurotrauma 16: 851–863, https://doi.org/10.1089/neu.1999.16.851.Search in Google Scholar PubMed

Bikbaev, A., Ciuraszkiewicz-Wojciech, A., Heck, J., Klatt, O., Freund, R., Mitlöhner, J., Enrile Lacalle, S., Sun, M., Repetto, D., Frischknecht, R., et al.. (2020). Auxiliary α2δ1 and α2δ3 subunits of calcium channels drive excitatory and inhibitory neuronal network development. J. Neurosci. 40: 4824–4841, https://doi.org/10.1523/jneurosci.1707-19.2020.Search in Google Scholar PubMed PubMed Central

Bracchi-Ricard, V., Zha, J., Smith, A., Lopez-Rodriguez, D.M., Bethea, J.R., and Andreansky, S. (2016). Chronic spinal cord injury attenuates influenza virus-specific antiviral immunity. J. Neuroinflamm. 13: 125–138, https://doi.org/10.1186/s12974-016-0574-y.Search in Google Scholar PubMed PubMed Central

Brennan, F.H., Noble, B.T., Wang, Y., Guan, Z., Davis, H., Mo, X., Harris, C., Eroglu, C., Ferguson, A.R., and Popovich, P.G. (2021). Acute post-injury blockade of α2δ-1 calcium channel subunits prevents pathological autonomic plasticity after spinal cord injury. Cell Rep 34: 108667, https://doi.org/10.1016/j.celrep.2020.108667.Search in Google Scholar PubMed PubMed Central

Brommer, B., Engel, O., Kopp, M.A., Watzlawick, R., Müller, S., Prüss, H., Chen, Y., DeVivo, M.J., Finkenstaedt, F.W., Dirnagl, U., et al.. (2016). Spinal cord injury-induced immune deficiency syndrome enhances infection susceptibility dependent on lesion level. Brain 139: 692–707, https://doi.org/10.1093/brain/awv375.Search in Google Scholar PubMed PubMed Central

Carpenter, R.S., Marbourg, J.M., Brennan, F.H., Mifflin, K.A., Hall, J.C.E., Jiang, R.R., Mo, X.M., Karunasiri, M., Burke, M.H., Dorrance, A.M., et al.. (2020). Spinal cord injury causes chronic bone marrow failure. Nat. Commun. 11: 3702, https://doi.org/10.1038/s41467-020-17564-z.Search in Google Scholar PubMed PubMed Central

Chavan, S.S., Pavlov, V.A., and Tracey, K.J. (2017). Mechanisms and therapeutic relevance of neuro-immune communication. Immun. 46: 927–942, https://doi.org/10.1016/j.immuni.2017.06.008.Search in Google Scholar PubMed PubMed Central

Chen, J., Li, L., Chen, S.R., Chen, H., Xie, J.D., Sirrieh, R.E., MacLean, D.M., Zhang, Y., Zhou, M.-H., Jayaraman, V., et al.. (2018). The α2δ-1-NMDA receptor complex is critically involved in neuropathic pain development and gabapentin therapeutic actions. Cell Rep. 22: 2307–2321, https://doi.org/10.1016/j.celrep.2018.02.021.Search in Google Scholar PubMed PubMed Central

Chhatar, S. and Lal, G. (2021). Role of adrenergic receptor signalling in neuroimmune communication. Curr. Res. Immunol. 2: 202–217, https://doi.org/10.1016/j.crimmu.2021.11.001.Search in Google Scholar PubMed PubMed Central

Christopherson, K.S., Ullian, E.M., Stokes, C.C., Mullowney, C.E., Hell, J.W., Agah, A., Lawler, J., Mosher, D.F., Bornstein, P., and Barres, B.A. (2005). Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120: 421–433, https://doi.org/10.1016/j.cell.2004.12.020.Search in Google Scholar PubMed

Cleypool, C.G.J., Mackaaij, C., Lotgerink Bruinenberg, D., Schurink, B., and Bleys, R.L.A.W. (2021). Sympathetic nerve distribution in human lymph nodes. J. Anat. 239: 282–289, https://doi.org/10.1111/joa.13422.Search in Google Scholar PubMed PubMed Central

Davoodzadeh Gholami, M., Kardar, G.A., Saeedi, Y., Heydari, S., Garssen, J., and Falak, R. (2017). Exhaustion of T lymphocytes in the tumor microenvironment: significance and effective mechanisms. Cell. Immunol. 322: 1–14, https://doi.org/10.1016/j.cellimm.2017.10.002.Search in Google Scholar PubMed

Devi, S., Alexandre, Y.O., Loi, J.K., Gillis, R., Ghazanfari, N., Creed, S.J., Holz, L.E., Shackleford, D., Mackay, L.K., Heath, W.R., et al.. (2021). Adrenergic regulation of the vasculature impairs leukocyte interstitial migration and suppresses immune responses. Immun. 54: 1219–1230, https://doi.org/10.1016/j.immuni.2021.03.025.Search in Google Scholar PubMed

DeVivo, M.J., Kartus, P.L., Stover, S.L., Rutt, R.D., and Fine, P.R. (1989). Cause of death for patients with spinal cord injuries. Arch Intern. Med. 149: 1761–1766, https://doi.org/10.1001/archinte.1989.00390080043011.Search in Google Scholar

Druschel, C., Ossami Saidy, R.R., Grittner, U., Nowak, C.P., Meisel, A., Schaser, K.D., Niedeggen, A., Liebscher, T., Kopp, M.A., and Schwab, J.M. (2000). Clinical decision-making on spinal cord injury-associated pneumonia: a nationwide survey in Germany. Spinal Cord 38: 873–881, https://doi.org/10.1038/s41393-020-0435-5.Search in Google Scholar PubMed PubMed Central

Edsberg, L.E., Wyffels, J.T., Ogrin, R., Craven, B.C., and Houghton, P. (2015). A pilot study evaluating protein abundance in pressure ulcer fluid from people with and without spinal cord injury. J. Spinal Cord Med. 38: 456–467, https://doi.org/10.1179/2045772314y.0000000212.Search in Google Scholar

Eldahan, K.C. and Rabchevsky, A.G. (2018). Autonomic dysreflexia after spinal cord injury: systemic pathophysiology and methods of management. Auton. Neurosci. 209: 59–70, https://doi.org/10.1016/j.autneu.2017.05.002.Search in Google Scholar PubMed PubMed Central

Estrada, L.D., Ağaç Çobanoğlu, D., Wise, A., Maples, R.W., Çobanoğlu, M.C., and Farrar, J.D. (2022). Adrenergic signaling controls early transcriptional programs during CD8+ T cell responses to viral infection. PLoS One 17: e0272017, https://doi.org/10.1371/journal.pone.0272017.Search in Google Scholar PubMed PubMed Central

Falvey, A., Metz, C.N., Tracey, K.J., and Pavlov, V.A. (2022). Peripheral nerve stimulation and immunity: the expanding opportunities for providing mechanistic insight and therapeutic intervention. Int. Immunol. 34: 107–118, https://doi.org/10.1093/intimm/dxab068.Search in Google Scholar PubMed PubMed Central

Finestone, H.M. and Teasell, R.W. (1993). Autonomic dysreflexia after brainstem tumor resection. A case report. Am. J. Phys. Med. Rehabil. 72: 395–397, https://doi.org/10.1097/00002060-199312000-00011.Search in Google Scholar

Frauwirth, K.A., Riley, J.L., Harris, M.H., Parry, R.V., Rathmell, J.C., Plas, D.R., Elstrom, R.L., June, C.H., and Thompson, C.B. (2002). The CD28 signaling pathway regulates glucose metabolism. Immun. 16: 769–777, https://doi.org/10.1016/s1074-7613(02)00323-0.Search in Google Scholar PubMed

Geng, Q., Li, L., Shen, Z., Zheng, Y., Wang, L., Xue, R., Xue, W., Peng, M., and Zhao, J. (2023). Norepinephrine inhibits CD8+ T-cell infiltration and function, inducing anti-PD-1 mAb resistance in lung adenocarcinoma. Br. J. Cancer 128: 1223–1235, https://doi.org/10.1038/s41416-022-02132-7.Search in Google Scholar PubMed PubMed Central

Glaser, R. and Kiecolt-Glaser, J.K. (2005). Stress-induced immune dysfunction: implications for health. Nat. Rev. Immunol. 5: 243–251, https://doi.org/10.1038/nri1571.Search in Google Scholar PubMed

Gong, C., Zheng, X., Guo, F., Wang, Y., Zhang, S., Chen, J., Sun, X., Zulfiqar Ali Shah, S., Zheng, Y., Li, X., et al.. (2021). Human spinal gaba neurons alleviate spasticity and improve locomotion in rats with spinal cord injury. Cell Rep. 34: 108889, https://doi.org/10.1016/j.celrep.2021.108889.Search in Google Scholar PubMed

Held, K.S. and Lane, T.E. (2014). Spinal cord injury, immunodepression, and antigenic challenge. Semin. Immunol. 26: 415–420, https://doi.org/10.1016/j.smim.2014.03.003.Search in Google Scholar PubMed PubMed Central

Hendrich, J., Van Minh, A.T., Heblich, F., Nieto-Rostro, M., Watschinger, K., Striessnig, J., Wratten, J., Davies, A., and Dolphin, A.C. (2008). Pharmacological disruption of calcium channel trafficking by the alpha2delta ligand gabapentin. Proc. Natl. Acad. Sci. U. S. A. 105: 3628–3633, https://doi.org/10.1073/pnas.0708930105.Search in Google Scholar PubMed PubMed Central

Hou, S., Duale, H., Cameron, A.A., Abshire, S.M., Lyttle, T.S., and Rabchevsky, A.G. (2008). Plasticity of lumbosacral propriospinal neurons is associated with the development of autonomic dysreflexia after thoracic spinal cord transection. J. Comp. Neurol. 509: 382–399, https://doi.org/10.1002/cne.21771.Search in Google Scholar PubMed PubMed Central

Jeffries, M.A. and Tom, V.J. (2021). Peripheral immune dysfunction: a problem of central importance after spinal cord injury. Biology 10: 928–947, https://doi.org/10.3390/biology10090928.Search in Google Scholar PubMed PubMed Central

Kohm, A.P. and Sanders, V.M. (2000). Norepinephrine: a messenger from the brain to the immune system. Immunol. Today 21: 539–542, https://doi.org/10.1016/s0167-5699(00)01747-3.Search in Google Scholar PubMed

Krassioukov, A.V. and Weaver, L.C. (1996). Morphological changes in sympathetic preganglionic neurons after spinal cord injury in rats. Neurosci. 70: 211–225, https://doi.org/10.1016/0306-4522(95)00294-s.Search in Google Scholar PubMed

Krishnan, S., Karg, P.E., Boninger, M.L., and Brienza, D.M. (2017). Association between presence of pneumonia and pressure ulcer formation following traumatic spinal cord injury. J. Spinal Cord Med. 40: 415–422, https://doi.org/10.1080/10790268.2016.1180099.Search in Google Scholar PubMed PubMed Central

Larouche, J., Sheoran, S., Maruyama, K., and Martino, M.M. (2018). Immune regulation of skin wound healing: mechanisms and novel therapeutic targets. Adv. Wound Care 7: 209–231, https://doi.org/10.1089/wound.2017.0761.Search in Google Scholar PubMed PubMed Central

Liu, T., Yang, L., Han, X., Ding, X., Li, J., and Yang, J. (2020). Local sympathetic innervations modulate the lung innate immune responses. Sci. Adv. 6: eaay1497, https://doi.org/10.1126/sciadv.aay1497.Search in Google Scholar PubMed PubMed Central

Llewellyn-Smith, I.J. and Weaver, L.C. (2001). Changes in synaptic inputs to sympathetic preganglionic neurons after spinal cord injury. J. Comp. Neurol. 435: 226–240, https://doi.org/10.1002/cne.1204.Search in Google Scholar PubMed

Lucin, K.M., Sanders, V.M., Jones, T.B., Malarkey, W.B., and Popovich, P.G. (2007). Impaired antibody synthesis after spinal cord injury is level dependent and is due to sympathetic nervous system dysregulation. Exp. Neurol. 207: 75–84, https://doi.org/10.1016/j.expneurol.2007.05.019.Search in Google Scholar PubMed PubMed Central

Lucin, K.M., Sanders, V.M., and Popovich, P.G. (2009). Stress hormones collaborate to induce lymphocyte apoptosis after high level spinal cord injury. J. Neurochem. 110: 140914–140921, https://doi.org/10.1111/j.1471-4159.2009.06232.x.Search in Google Scholar PubMed PubMed Central

Marbourg, J.M., Bratasz, A., Mo, X., and Popovich, P.G. (2017). Spinal cord injury suppresses cutaneous inflammation: implications for peripheral wound healing. J. Neurotrauma 34: 1149–1155, https://doi.org/10.1089/neu.2016.4611.Search in Google Scholar PubMed PubMed Central

Mazzone, G.L., Mohammadshirazi, A., Aquino, J.B., Nistri, A., and Taccola, G. (2021). GABAergic mechanisms can redress the tilted balance between excitation and inhibition in damaged spinal networks. Mol. Neurobiol. 58: 3769–3786, https://doi.org/10.1007/s12035-021-02370-5.Search in Google Scholar PubMed PubMed Central

Mifflin, K.A., Brennan, F.H., Guan, Z., Kigerl, K.A., Filous, A.R., Mo, X., Schwab, J.M., and Popovich, P.G. (2022). Spinal cord injury impairs lung immunity in mice. J. Immunol. 209: 157–170, https://doi.org/10.4049/jimmunol.2200192.Search in Google Scholar PubMed PubMed Central

Mironets, E., Fischer, R., Bracchi-Ricard, V., Saltos, T.M., Truglio, T.S., O’Reilly, M.L., Swanson, K.A., Bethea, J.R., and Tom, V.J. (2020). Attenuating neurogenic sympathetic hyperreflexia robustly improves antibacterial immunity after chronic spinal cord injury. J. Neurosci. 40: 478–492, https://doi.org/10.1523/jneurosci.2417-19.2019.Search in Google Scholar PubMed PubMed Central

Mironets, E., Osei-Owusu, P., Bracchi-Ricard, V., Fischer, R., Owens, E.A., Ricard, J., Wu, D., Saltos, T., Collyer, E., Hou, S., et al.. (2018). Soluble TNFα signaling within the spinal cord contributes to the development of autonomic dysreflexia and ensuing vascular and immune dysfunction after spinal cord injury. J. Neurosci. 38: 4146–4162, https://doi.org/10.1523/jneurosci.2376-17.2018.Search in Google Scholar

Mota, C.M.D. and Madden, C.J. (2022). Neural control of the spleen as an effector of immune responses to inflammation: mechanisms and treatments. Am. J. Physiol. Regul. Integr. Comp. Physiol. 323: R375–R384, https://doi.org/10.1152/ajpregu.00151.2022.Search in Google Scholar PubMed PubMed Central

Noble, B.T., Brennan, F.H., and Popovich, P.G. (2018). The spleen as a neuroimmune interface after spinal cord injury. J. Neuroimmunol. 321: 1–11, https://doi.org/10.1016/j.jneuroim.2018.05.007.Search in Google Scholar PubMed

Noble, B.T., Brennan, F.H., Wang, Y., Guan, Z., Mo, X., Schwab, J.M., and Popovich, P.G. (2000). Thoracic VGluT2+ spinal interneurons regulate structural and functional plasticity of sympathetic networks after high-level spinal cord injury. J. Neurosci. 42: 3659–3675, https://doi.org/10.1523/jneurosci.2134-21.2022.Search in Google Scholar PubMed PubMed Central

Norden, D.M., Bethea, J.R., and Jiang, J. (2018). Impaired CD8 T cell antiviral immunity following acute spinal cord injury. J. Neuroinflamm. 15: 149, https://doi.org/10.1186/s12974-018-1191-8.Search in Google Scholar PubMed PubMed Central

Prüss, H., Tedeschi, A., Thiriot, A., Lynch, L., Loughhead, S.M., Stutte, S., Mazo, I.B., Kopp, M.A., Brommer, B., Blex, C., et al.. (2017). Spinal cord injury-induced immunodeficiency is mediated by a sympathetic-neuroendocrine adrenal reflex. Nat. Neurosci. 20: 1549–1559, https://doi.org/10.1038/nn.4643.Search in Google Scholar PubMed

Qiao, G., Bucsek, M.J., Winder, N.M., Chen, M., Giridharan, T., Olejniczak, S.H., Hylander, B.L., and Repasky, E.A. (2019). β-Adrenergic signaling blocks murine CD8+ T-cell metabolic reprogramming during activation: a mechanism for immunosuppression by adrenergic stress. Cancer Immunol. Immunother. 68: 11–22, https://doi.org/10.1007/s00262-018-2243-8.Search in Google Scholar PubMed PubMed Central

Riegger, T., Conrad, S., Schluesener, H.J., Kaps, H.P., Badke, A., Baron, C., Gerstein, J., Dietz, K., Abdizahdeh, M., and Schwab, J.M. (2009). Immune depression syndrome following human spinal cord injury (SCI): a pilot study. Neurosci. 158: 1194–1199, https://doi.org/10.1016/j.neuroscience.2008.08.021.Search in Google Scholar PubMed

Rodgers, K.A., Kigerl, K.A., Schwab, J.M., and Popovich, P.G. (2022). Immune dysfunction after spinal cord injury - a review of autonomic and neuroendocrine mechanisms. Curr. Opin. Pharmacol. 64: 102230, https://doi.org/10.1016/j.coph.2022.102230.Search in Google Scholar PubMed PubMed Central

Salvador, A.F., de Lima, K.A., and Kipnis, J. (2021). Neuromodulation by the immune system: a focus on cytokines. Nat. Rev. Immunol. 21: 526–541, https://doi.org/10.1038/s41577-021-00508-z.Search in Google Scholar PubMed

Sharif, H. and Hou, S. (2017). Autonomic dysreflexia: a cardiovascular disorder following spinal cord injury. Neural Regen. Res. 12: 1390–1400, https://doi.org/10.4103/1673-5374.215241.Search in Google Scholar PubMed PubMed Central

Soriano, J.E., Hudelle, R., Squair, J.W., Mahe, L., Amir, S., Gautier, M., Perez Puchalt, V., Barraud, Q., Phillips, A.A., and Courtine, G. (2023). Longitudinal interrogation of sympathetic neural circuits and hemodynamics in preclinical models. Nat. Protoc. 18: 340–373, https://doi.org/10.1038/s41596-022-00764-w.Search in Google Scholar PubMed

Sribnick, E.A., Popovich, P.G., and Hall, M.W. (2022). Central nervous system injury-induced immune suppression. Neurosurg. Focus 52: E10, https://doi.org/10.3171/2021.11.focus21586.Search in Google Scholar

Stellwagen, D., Beattie, E.C., Seo, J.Y., and Malenka, R.C. (2005). Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-α. J. Neurosci. 25: 3219–3228, https://doi.org/10.1523/jneurosci.4486-04.2005.Search in Google Scholar

Stornetta, R.L. and Guyenet, P.G. (2018). C1 neurons: a nodal point for stress? Exp. Physiol. 103: 332–336, https://doi.org/10.1113/ep086435.Search in Google Scholar PubMed PubMed Central

Toulon, A., Breton, L., Taylor, K.R., Tenenhaus, M., Bhavsar, D., Lanigan, C., Rudolph, R., Jameson, J., and Havran, W.L. (2009). A role for human skin-resident T cells in wound healing. J. Exp. Med. 206: 743–750, https://doi.org/10.1084/jem.20081787.Search in Google Scholar PubMed PubMed Central

Trautmann, L., Janbazian, L., Chomont, N., Said, E.A., Gimmig, S., Bessette, B., Boulassel, M.-R., Delwart, E., Sepulveda, H., Balderas, R.S., et al.. (2006). Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 12: 1198–1202, https://doi.org/10.1038/nm1482.Search in Google Scholar PubMed

Udit, S., Blake, K., and Chiu, I.M. (2022). Somatosensory and autonomic neuronal regulation of the immune response. Nat. Rev. Neurosci. 23: 157–171, https://doi.org/10.1038/s41583-021-00555-4.Search in Google Scholar PubMed PubMed Central

Ueno, M., Ueno-Nakamura, Y., Niehaus, J., Popovich, P.G., and Yoshida, Y. (2016). Silencing spinal interneurons inhibits immune suppressive autonomic reflexes caused by spinal cord injury. Nat. Neurosci. 19: 784–487, https://doi.org/10.1038/nn.4289.Search in Google Scholar PubMed PubMed Central

Ulndreaj, A., Tzekou, A., Siddiqui, A.M., and Fehlings, M.G. (2020). Effects of experimental cervical spinal cord injury on peripheral adaptive immunity. PLoS One 15: e0241285, https://doi.org/10.1371/journal.pone.0241285.Search in Google Scholar PubMed PubMed Central

Wang, C.X., Nuttin, B., Heremans, H., Dom, R., and Gybels, J. (1996). Production of tumor necrosis factor in spinal cord following traumatic injury in rats. J. Neuroimmunol. 69: 151–156, https://doi.org/10.1016/0165-5728(96)00080-x.Search in Google Scholar PubMed

Yang, Y., Ye, Y., Chen, C., Kong, C., Su, X., Zhang, X., Bai, W., and He, X. (2019). Acute traumatic brain injury induces CD4+ and CD8+ T cell functional impairment by upregulating the expression of PD-1 via the activated sympathetic nervous system. Neuroimmunomodulation 26: 43–57, https://doi.org/10.1159/000495465.Search in Google Scholar PubMed

Zha, J., Smith, A., Andreansky, S., Bracchi-Ricard, V., and Bethea, J.R. (2014). Chronic thoracic spinal cord injury impairs CD8+ T-cell function by up-regulating programmed cell death-1 expression. J. Neuroinflamm. 11: 65, https://doi.org/10.1186/1742-2094-11-65.Search in Google Scholar PubMed PubMed Central

Zhang, Y., Guan, Z., Reader, B., Shawler, T., Mandrekar-Colucci, S., Huang, K., Weil, Z., Bratasz, A., Wells, J., Powell, N.D., et al.. (2013). Autonomic dysreflexia causes chronic immune suppression after spinal cord injury. J. Neurosci. 33: 12970–12981, https://doi.org/10.1523/jneurosci.1974-13.2013.Search in Google Scholar

Received: 2023-08-16
Accepted: 2023-10-13
Published Online: 2023-10-30
Published in Print: 2024-04-25

© 2023 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 27.4.2024 from https://www.degruyter.com/document/doi/10.1515/revneuro-2023-0090/html
Scroll to top button