Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Spinal cord injury-induced immunodeficiency is mediated by a sympathetic-neuroendocrine adrenal reflex

Abstract

Acute spinal cord injury (SCI) causes systemic immunosuppression and life-threatening infections, thought to result from noradrenergic overactivation and excess glucocorticoid release via hypothalamus–pituitary–adrenal axis stimulation. Instead of consecutive hypothalamus–pituitary–adrenal axis activation, we report that acute SCI in mice induced suppression of serum norepinephrine and concomitant increase in cortisol, despite suppressed adrenocorticotropic hormone, indicating primary (adrenal) hypercortisolism. This neurogenic effect was more pronounced after high-thoracic level (Th1) SCI disconnecting adrenal gland innervation, compared with low-thoracic level (Th9) SCI. Prophylactic adrenalectomy completely prevented SCI-induced glucocorticoid excess and lymphocyte depletion but did not prevent pneumonia. When adrenalectomized mice were transplanted with denervated adrenal glands to restore physiologic glucocorticoid levels, the animals were completely protected from pneumonia. These findings identify a maladaptive sympathetic-neuroendocrine adrenal reflex mediating immunosuppression after SCI, implying that therapeutic normalization of the glucocorticoid and catecholamine imbalance in SCI patients could be a strategy to prevent detrimental infections.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: SCI-IDS is lesion-height-dependent and associated with major changes in CA and GC levels in both murine experimental SCI and human SCI.
Figure 2: SCI resulted in lymphoid organ involution, leukocyte depletion and spontaneous pneumonia.
Figure 3: Interventional effects of ADX after acute SCI.
Figure 4: Interventional effects of autologous ATX after acute SCI.
Figure 5: Liver iNKT cells do not interfere with infection susceptibility.
Figure 6: SCI-induced depletion of B and T cell progenitors.
Figure 7: Impaired lymphocyte trafficking behavior after acute SCI.

Similar content being viewed by others

References

  1. Steinman, L. Elaborate interactions between the immune and nervous systems. Nat. Immunol. 5, 575–581 (2004).

    CAS  PubMed  Google Scholar 

  2. Irwin, M.R. & Cole, S.W. Reciprocal regulation of the neural and innate immune systems. Nat. Rev. Immunol. 11, 625–632 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Meisel, C., Schwab, J.M., Prass, K., Meisel, A. & Dirnagl, U. Central nervous system injury-induced immune deficiency syndrome. Nat. Rev. Neurosci. 6, 775–786 (2005).

    CAS  PubMed  Google Scholar 

  4. Lucin, K.M., Sanders, V.M. & Popovich, P.G. Stress hormones collaborate to induce lymphocyte apoptosis after high level spinal cord injury. J. Neurochem. 110, 1409–1421 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Riegger, T. et al. Spinal cord injury-induced immune depression syndrome (SCI-IDS). Eur. J. Neurosci. 25, 1743–1747 (2007).

    PubMed  Google Scholar 

  6. Oropallo, M.A. et al. Chronic spinal cord injury impairs primary antibody responses but spares existing humoral immunity in mice. J. Immunol. 188, 5257–5266 (2012).

    CAS  PubMed  Google Scholar 

  7. Riegger, T. et al. Immune depression syndrome following human spinal cord injury (SCI): a pilot study. Neuroscience 158, 1194–1199 (2009).

    CAS  PubMed  Google Scholar 

  8. Furlan, J.C., Krassioukov, A.V. & Fehlings, M.G. Hematologic abnormalities within the first week after acute isolated traumatic cervical spinal cord injury: a case-control cohort study. Spine 31, 2674–2683 (2006).

    PubMed  Google Scholar 

  9. Brommer, B. et al. Spinal cord injury-induced immune deficiency syndrome enhances infection susceptibility dependent on lesion level. Brain 139, 692–707 (2016).

    PubMed  PubMed Central  Google Scholar 

  10. DeVivo, M.J., Kartus, P.L., Stover, S.L., Rutt, R.D. & Fine, P.R. Cause of death for patients with spinal cord injuries. Arch. Intern. Med. 149, 1761–1766 (1989).

    CAS  PubMed  Google Scholar 

  11. Jackson, A.B. & Groomes, T.E. Incidence of respiratory complications following spinal cord injury. Arch. Phys. Med. Rehabil. 75, 270–275 (1994).

    CAS  PubMed  Google Scholar 

  12. Failli, V. et al. Functional neurological recovery after spinal cord injury is impaired in patients with infections. Brain 135, 3238–3250 (2012).

    PubMed  Google Scholar 

  13. Kopp, M.A. et al. Long-term functional outcome in patients with acquired infections after acute spinal cord injury. Neurology 88, 892–900 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Borovikova, L.V. et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 405, 458–462 (2000).

    CAS  PubMed  Google Scholar 

  15. Martelli, D., Yao, S.T., McKinley, M.J. & McAllen, R.M. Reflex control of inflammation by sympathetic nerves, not the vagus. J. Physiol. (Lond.) 592, 1677–1686 (2014).

    CAS  Google Scholar 

  16. Zhang, Y. et al. Autonomic dysreflexia causes chronic immune suppression after spinal cord injury. J. Neurosci. 33, 12970–12981 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Ueno, M., Ueno-Nakamura, Y., Niehaus, J., Popovich, P.G. & Yoshida, Y. Silencing spinal interneurons inhibits immune suppressive autonomic reflexes caused by spinal cord injury. Nat. Neurosci. 19, 784–787 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Meador, K.J. et al. Role of cerebral lateralization in control of immune processes in humans. Ann. Neurol. 55, 840–844 (2004).

    PubMed  Google Scholar 

  19. Walter, U. et al. Insular stroke is associated with acute sympathetic hyperactivation and immunodepression. Eur. J. Neurol. 20, 153–159 (2013).

    CAS  PubMed  Google Scholar 

  20. Williams, J.M. et al. Sympathetic innervation of murine thymus and spleen: evidence for a functional link between the nervous and immune systems. Brain Res. Bull. 6, 83–94 (1981).

    CAS  PubMed  Google Scholar 

  21. Felten, D.L., Ackerman, K.D., Wiegand, S.J. & Felten, S.Y. Noradrenergic sympathetic innervation of the spleen: I. Nerve fibers associate with lymphocytes and macrophages in specific compartments of the splenic white pulp. J. Neurosci. Res. 18, 28–36, 118–121 (1987).

    CAS  PubMed  Google Scholar 

  22. Previnaire, J.G., Soler, J.M., El Masri, W. & Denys, P. Assessment of the sympathetic level of lesion in patients with spinal cord injury. Spinal Cord 47, 122–127 (2009).

    CAS  PubMed  Google Scholar 

  23. Wong, C.H., Jenne, C.N., Lee, W.Y., Léger, C. & Kubes, P. Functional innervation of hepatic iNKT cells is immunosuppressive following stroke. Science 334, 101–105 (2011).

    CAS  PubMed  Google Scholar 

  24. Massberg, S. et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell 131, 994–1008 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Dembowsky, K., Czachurski, J., Amendt, K. & Seller, H. Tonic descending inhibition of the spinal somato-sympathetic reflex from the lower brain stem. J. Auton. Nerv. Syst. 2, 157–182 (1980).

    CAS  PubMed  Google Scholar 

  26. Tibbs, P.A., Young, B., McAllister, R.G. Jr. & Todd, E.P. Studies of experimental cervical spinal cord transection. Part III: Effects of acute cervical spinal cord transection on cerebral blood flow. J. Neurosurg. 50, 633–638 (1979).

    CAS  PubMed  Google Scholar 

  27. Rawe, S.E. & Perot, P.L. Jr. Pressor response resulting from experimental contusion injury to the spinal cord. J. Neurosurg. 50, 58–63 (1979).

    CAS  PubMed  Google Scholar 

  28. Young, W., DeCrescito, V., Tomasula, J.J. & Ho, V. The role of the sympathetic nervous system in pressor responses induced by spinal injury. J. Neurosurg. 52, 473–481 (1980).

    CAS  PubMed  Google Scholar 

  29. Edwards, A.V. & Jones, C.T. Autonomic control of adrenal function. J. Anat. 183, 291–307 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Parker, T.L., Kesse, W.K., Mohamed, A.A. & Afework, M. The innervation of the mammalian adrenal gland. J. Anat. 183, 265–276 (1993).

    PubMed  PubMed Central  Google Scholar 

  31. Holzwarth, M.A., Cunningham, L.A. & Kleitman, N. The role of adrenal nerves in the regulation of adrenocortical functions. Ann. NY Acad. Sci. 512, 449–464 (1987).

    CAS  PubMed  Google Scholar 

  32. Dhabhar, F.S. & McEwen, B.S. Enhancing versus suppressive effects of stress hormones on skin immune function. Proc. Natl. Acad. Sci. USA 96, 1059–1064 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Andersson, U. & Tracey, K.J. Neural reflexes in inflammation and immunity. J. Exp. Med. 209, 1057–1068 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Dimitrov, S. et al. Cortisol and epinephrine control opposing circadian rhythms in T cell subsets. Blood 113, 5134–5143 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Scheiermann, C. et al. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 37, 290–301 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Schedlowski, M. et al. Catecholamines modulate human NK cell circulation and function via spleen-independent beta 2-adrenergic mechanisms. J. Immunol. 156, 93–99 (1996).

    CAS  PubMed  Google Scholar 

  37. Prass, K. et al. Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation. J. Exp. Med. 198, 725–736 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Lucin, K.M., Sanders, V.M., Jones, T.B., Malarkey, W.B. & Popovich, P.G. Impaired antibody synthesis after spinal cord injury is level dependent and is due to sympathetic nervous system dysregulation. Exp. Neurol. 207, 75–84 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Rouleau, P., Ung, R.V., Lapointe, N.P. & Guertin, P.A. Hormonal and immunological changes in mice after spinal cord injury. J. Neurotrauma 24, 367–378 (2007).

    PubMed  Google Scholar 

  40. von Andrian, U.H. Intravital microscopy of the peripheral lymph node microcirculation in mice. Microcirculation 3, 287–300 (1996).

    CAS  PubMed  Google Scholar 

  41. Kopp, M.A. et al. The SCIentinel study--prospective multicenter study to define the spinal cord injury-induced immune depression syndrome (SCI-IDS)--study protocol and interim feasibility data. BMC Neurol. 13, 168 (2013).

    PubMed  PubMed Central  Google Scholar 

  42. Fatima, G., Sharma, V.P. & Verma, N.S. Circadian variations in melatonin and cortisol in patients with cervical spinal cord injury. Spinal Cord 54, 364–367 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank P. Popovich for critically reading the manuscript and insightful suggestions. We are grateful for the excellent technical help of D. Brandl, L. Mosch, C. Josties and I. Przesdzing. This work has been supported by grants from the German Academic Exchange Service (DAAD, D/10/43923) and German Research Foundation (DFG, PR 1274/2-1 to H.P.; STU 528/1-1, CRC-914 to S.S. and Cluster of Excellence NeuroCure to U.D.), by the Wings for Life Spinal Cord Research Foundation (WfL-DE-006/12), Else Kröner Fresenius Stiftung, German legal accident insurance (DGUV), the Era-Net-NEURON Program of the European Union, NIDILRR (#90SI5020), the Ohio State University Discovery Theme and the W.E. Hunt & C.M. Curtis Endowment to J.M.S. The National Spinal Cord Injury Database (NSCID) is funded by the National Institute on Disability, Independent Living, and Rehabilitation Research (NIDILRR, Grant number 90DP0083), US Department of Health and Human Services. This work was supported by the HMS Center for Immune Imaging and NIH grants AI112521 and AR068383 (to U.H.v.A.).

Author information

Authors and Affiliations

Authors

Contributions

H.P., U.H.v.A. and J.M.S. designed the research study; H.P., A. Tedeschi, L.L., A. Thiriot, S.M.L., S.S., I.B.M., M.A.K., B.B., C.B., L.-C.G., T.L., A.N., F.B., M.S.V., M.J.D. and Y.C. conducted experiments and acquired data; all authors analyzed data; H.P., A.Thiriot, U.D., U.H.v.A. and J.M.S. wrote the manuscript; all authors contributed discussion to the manuscript.

Corresponding author

Correspondence to Ulrich H von Andrian.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Progressive reduction of organ cellularity after SCI.

The SCI-induced reduction in organ size was detectable already after 24 hours (a) and progresses continuously to 48 hours (b). Already at these early time points, cell loss is related to the level of SCI. Data are mean ± SEM, n=3 animals per group. * p<0.05; ** p<0.01; 1-way ANOVA with Tukey’s multiple comparison test.

Supplementary Figure 2 SCI-induced cell depletion affects all analyzed types of immune cells.

FACS gating strategy exemplarily shown for CD19+ B cells (a, leukocytes; b, single cells; c, CD19+ cells). The profound cell loss affected all major immune cell populations including CD19+ B cells (d), CD4+ T cells (e), CD8+ T cells (f), CD11c+ dendritic cells (g), CD11b+ monocytes (h), and NK1.1+ NK cells (i). Data are mean ± SEM, n=3-5 animals per group. * p<0.05; ** p<0.01; *** p<0.001, 1-way ANOVA with Tukey’s multiple comparison test.

Supplementary Figure 3 Sciatic nerve lesion has no effect on immune cell composition of the innervated bone marrow.

(a) Deafferentiation of the tibial bone marrow by right sciatic nerve injury (compared to sham) did not change the total number of bone marrow cells. (b-d) Right sciatic nerve lesion did not change the number and frequency of CD19+ (b), CD4+ (c), CD8+ (d), Gr-1+, NK1.1+, CD11c+, and CD11b+ cells (not shown) in the ipsi- and contralateral bone marrow. (e-f) Homing of donor splenocytes for 2 hours (e) as well as redistribution for 15 hours (f) to the bone marrow was equal in mice with right sciatic nerve lesion and sham-operated animals. Data are mean ± SEM, n=6-9 animals per group. * p<0.05; 1-way ANOVA with Tukey’s multiple comparison test.

Supplementary Figure 4 Adrenalectomy reversed the SCI-induced cell depletion in all analyzed types of immune cells.

Adrenalectomy prevented the loss of all examined cell populations 72 hours after SCI. In particular, the number of CD19+ (a), CD4+ (b), CD8+ (c), CD11c+ and CD11b+ cells (not shown) were unchanged after SCI. Data are mean ± SEM, n=3 animals per group. * p<0.05; ** p<0.01; *** p<0.001, 1-way ANOVA with Tukey’s multiple comparison test.

Supplementary Figure 5 Splenectomy did not reverse SCI-induced immune cell depletion.

In contrast to surgical removal of the adrenals, removal of the spleen which is similarly innervated by sympathetic fibers via the splanchnic nerve, could not re-establish the SCI-induced cell death in thymus and lymph nodes (a) and did not change the characteristic SCI-induced pattern of B cell populations in the bone marrow (b-e) with massive enrichment of mature B cells (e).

Supplementary Figure 6 Adrenotransplantation reduced effects of lesion height and protected from pneumonia after SCI.

Disconnection of the adrenal glands from spinal innervation (adrenotransplantation) resulted in only subtle differences in organ shrinkage between high versus low thoracic level SCI (Th1/Th9). This held true for all examined cell populations, including CD4+ (a), CD19+ (b), CD8+ (c), CD11c+, and CD11b+ cells (not shown). Data are mean ± SEM, n=3-7 animals per group. * p<0.05; ** p<0.01; *** p<0.001, 1-way ANOVA with Tukey’s multiple comparison test.

Supplementary Figure 7 The phenotype of liver iNKT cells did not change after SCI.

Numbers and percentages of liver iNKT cells were not altered after experimental SCI in wild-type mice after adrenalectomy (a) and adrenotransplantation (b). Data are mean ± SEM, n=3-4 animals per group. * p<0.05; 1-way ANOVA with Tukey’s multiple comparison test.

Supplementary Figure 8 Phenotypic characterization of the lymphocyte homing defect.

Homing of injected donor splenocytes (composed of ~52% CD19+, ~23% CD4+, ~17% CD8+ cells) was not equally impaired for all donor cell populations or target organs. (a) The homing deficit was most pronounced for B cells with reduced percentage of CD19+ cells (only 30% B cells after SCI compared to almost 60% after sham), while the percentage of T cells in the donor population was increased (despite total reduction). (b) In contrast, homing of donor cells to the cervical LNs was equally impaired for all cell populations after SCI. (c) The 2-hour homing deficit of donor cells to most organs was detectable already 1 d post SCI, most pronounced to peripheral LNs. (d) Not only homing, but also redistribution of donor cells (15 hours cell migration) was severely disturbed after SCI. Data are mean ± SEM, n=3-5 (a-b) or 3 (c-d) animals per group. * p<0.05; ** p<0.01; *** p<0.001, unpaired Student’s t test (a-b) or 1-way ANOVA with Tukey’s multiple comparison test (c-d).

Supplementary Figure 9 Homing behavior of T cells after SCI.

(a) In vivo imaging of the inguinal LN showed that rolling of T lymphocytes after SCI was not impaired. (b) In the shrunken brachial LN after SCI, the expression level of PNAd (MECA-79 antibody) appeared increased. (c) In contrast to spleen and LNs, homing to the bone marrow is only slightly changed with a subtle increase of CD4+ and CD8+ T cells. (d) BM mRNA expression of CXCL12 was increased. (e) BM of injured mice (3d post SCI) recruits ~5 times more mature B cells from the circulation 24 hours after injection. Homing to BM is completely abolished if B cells were pre-treated with pertussis toxin in vitro. Data are mean ± SEM, n=3-5 animals per group. * p<0.05; ** p<0.01; *** p<0.001, unpaired Student’s t test. Bar represents 1 mm in b.

Supplementary Figure 10 Cascade of events showing how SCI ultimately results in neuroendocrine dysfunction involving the adrenal glands and leading to infection.

(a) High-level (Th1) SCI interrupts neural vegetative innervation of the adrenal glands from the spinal cord via splanchnic and adrenal nerves. (b) Adrenal denervation results in a drop of CA release and in disinhibition of GC release. Increased GCs then suppress ACTH production (primary hypercortisolism). Dysfunctional neuro-endocrine signaling leads to high GC and low NE levels which promote infections via pathways including reduced cardiac output, disturbed lymphocyte trafficking, or increased immune cell apoptosis. (c) The susceptibility to pneumonia was reversed if GC levels remained balanced in adrenotransplanted animals. The figure additionally shows how further pathways of immune dysfunction after SCI interfere with the here presented cascade. For example, vagus nerve-mediated parasympathetic innervation of the cardiovascular system (top) or spleen and liver (bottom) intersect with the sympathetic route. This might be of particular relevance to lesions of the central nervous system, which are located above the originating vagus fibers in the brainstem, such as in stroke.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Prüss, H., Tedeschi, A., Thiriot, A. et al. Spinal cord injury-induced immunodeficiency is mediated by a sympathetic-neuroendocrine adrenal reflex. Nat Neurosci 20, 1549–1559 (2017). https://doi.org/10.1038/nn.4643

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4643

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing