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:

Social stress induces neurovascular pathology promoting depression

Abstract

Studies suggest that heightened peripheral inflammation contributes to the pathogenesis of major depressive disorder. We investigated the effect of chronic social defeat stress, a mouse model of depression, on blood–brain barrier (BBB) permeability and infiltration of peripheral immune signals. We found reduced expression of the endothelial cell tight junction protein claudin-5 (Cldn5) and abnormal blood vessel morphology in nucleus accumbens (NAc) of stress-susceptible but not resilient mice. CLDN5 expression was also decreased in NAc of depressed patients. Cldn5 downregulation was sufficient to induce depression-like behaviors following subthreshold social stress whereas chronic antidepressant treatment rescued Cldn5 loss and promoted resilience. Reduced BBB integrity in NAc of stress-susceptible or mice injected with adeno-associated virus expressing shRNA against Cldn5 caused infiltration of the peripheral cytokine interleukin-6 (IL-6) into brain parenchyma and subsequent expression of depression-like behaviors. These findings suggest that chronic social stress alters BBB integrity through loss of tight junction protein Cldn5, promoting peripheral IL-6 passage across the BBB and depression.

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

Fig. 1: Social stress vulnerability and MDD is associated with reduced Cldn5 expression.
Fig. 2: Conditional knockdown of Cldn5 expression in the NAc is sufficient to induce depression-like behaviors.
Fig. 3: Social stress vulnerability is associated with increased BBB permeability.
Fig. 4: Chronic social stress induces peripheral monocyte accumulation and IL-6 passage into the NAc.

Similar content being viewed by others

References

  1. Ménard, C., Pfau, M. L., Hodes, G. E. & Russo, S. J. Immune and neuroendocrine mechanisms of stress vulnerability and resilience. Neuropsychopharmacology 42, 62–80 (2017).

    Article  PubMed  Google Scholar 

  2. Kessler, R. C., Chiu, W. T., Demler, O., Merikangas, K. R. & Walters, E. E. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey replication. Arch. Gen. Psychiatry 62, 617–627 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Seligman, F. & Nemeroff, C. B. The interface of depression and cardiovascular disease: therapeutic implications. Ann. NY Acad. Sci. 1345, 25–35 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Carney, R. M. & Freedland, K. E. Depression and coronary heart disease. Nat. Rev. Cardiol. 14, 145–155 (2017).

    Article  PubMed  Google Scholar 

  5. Wood, S. K. Individual differences in the neurobiology of social stress: implications for depression-cardiovascular disease comorbidity. Curr. Neuropharmacol. 12, 205–211 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Huffman, J. C., Celano, C. M., Beach, S. R., Motiwala, S. R. & Januzzi, J. L. Depression and cardiac disease: epidemiology, mechanisms, and diagnosis. Cardiovasc. Psychiatry Neurol. 2013, 695925 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Hodes, G. E., Kana, V., Menard, C., Merad, M. & Russo, S. J. Neuroimmune mechanisms of depression. Nat. Neurosci. 18, 1386–1393 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Barnes, J., Mondelli, V. & Pariante, C. M. Genetic contributions of inflammation to depression. Neuropsychopharmacology 42, 81–98 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Dantzer, R. Cytokine, sickness behavior, and depression. Immunol. Allergy Clin. North Am. 29, 247–264 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Miller, A. H. & Raison, C. L. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 16, 22–34 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hodes, G. E. et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc. Natl. Acad. Sci. USA 111, 16136–16141 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Powell, N. D. et al. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via β-adrenergic induction of myelopoiesis. Proc. Natl. Acad. Sci. USA 110, 16574–16579 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Heidt, T. et al. Chronic variable stress activates hematopoietic stem cells. Nat. Med. 20, 754–758 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wohleb, E. S., Powell, N. D., Godbout, J. P. & Sheridan, J. F. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J. Neurosci. 33, 13820–13833 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Weber, M. D., Godbout, J. P. & Sheridan, J. F. Repeated social defeat, neuroinflammation, and behavior: monocytes carry the signal. Neuropsychopharmacology 42, 46–61 (2017).

    Article  PubMed  Google Scholar 

  16. Esposito, P. et al. Acute stress increases permeability of the blood-brain-barrier through activation of brain mast cells. Brain Res. 888, 117–127 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Sántha, P. et al. Restraint stress-induced morphological changes at the blood-brain barrier in adult rats. Front. Mol. Neurosci. 8, 88 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Friedman, A. et al. Pyridostigmine brain penetration under stress enhances neuronal excitability and induces early immediate transcriptional response. Nat. Med. 2, 1382–1385 (1996).

    Article  CAS  PubMed  Google Scholar 

  19. Sharma, H. S. & Dey, P. K. Impairment of blood-brain barrier (BBB) in rat by immobilization stress: role of serotonin (5-HT). Indian J. Physiol. Pharmacol. 25, 111–122 (1981).

    CAS  PubMed  Google Scholar 

  20. Niklasson, F. & Agren, H. Brain energy metabolism and blood-brain barrier permeability in depressive patients: analyses of creatine, creatinine, urate, and albumin in CSF and blood. Biol. Psychiatry 19, 1183–1206 (1984).

    CAS  PubMed  Google Scholar 

  21. Roszkowski, M. & Bohacek, J. Stress does not increase blood-brain barrier permeability in mice. J. Cereb. Blood Flow Metab. 36, 1304–1315 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Günzel, D. & Yu, A. S. Claudins and the modulation of tight junction permeability. Physiol. Rev. 93, 525–569 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Nitta, T. et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J. Cell Biol. 161, 653–660 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Meltzer, H., Vostanis, P., Ford, T., Bebbington, P. & Dennis, M. S. Victims of bullying in childhood and suicide attempts in adulthood. Eur. Psychiatry 26, 498–503 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Berton, O. & Nestler, E. J. New approaches to antidepressant drug discovery: beyond monoamines. Nat. Rev. Neurosci. 7, 137–151 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Ménard, C., Hodes, G. E. & Russo, S. J. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 321, 138–162 (2016).

    Article  PubMed  Google Scholar 

  27. Golden, S. A., Covington, H. E. III, Berton, O. & Russo, S. J. A standardized protocol for repeated social defeat stress in mice. Nat. Protoc. 6, 1183–1191 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Russo, S. J. & Nestler, E. J. The brain reward circuitry in mood disorders. Nat. Rev. Neurosci. 14, 609–625 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Hodes, G. E. et al. Sex differences in nucleus accumbens transcriptome profiles associated with susceptibility versus resilience to subchronic variable stress. J. Neurosci. 35, 16362–16376 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Golden, S. A. et al. Epigenetic regulation of RAC1 induces synaptic remodeling in stress disorders and depression. Nat. Med. 19, 337–344 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Campbell, M. et al. Systemic low-molecular weight drug delivery to pre-selected neuronal regions. EMBO Mol. Med. 3, 235–245 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. McKim, D.B. et al. Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Mol. Psychiatry https://doi.org/10.1038/mp.2017.64 (2017).

  35. Saederup, N. et al. Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS One 5, e13693 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Mizutani, M. et al. The fractalkine receptor but not CCR2 is present on microglia from embryonic development throughout adulthood. J. Immunol. 188, 29–36 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Maes, M. et al. Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine 9, 853–858 (1997).

    Article  CAS  PubMed  Google Scholar 

  39. Dowlati, Y. et al. A meta-analysis of cytokines in major depression. Biol. Psychiatry 67, 446–457 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Kiraly, D. D. et al. Altered peripheral immune profiles in treatment-resistant depression: response to ketamine and prediction of treatment outcome. Transl. Psychiatry 7, e1065 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Coppen, A. J. Abnormality of the blood-cerebrospinal fluid barrier of patients suffering from a depressive illness. J. Neurol. Neurosurg. Psychiatry 23, 156–161 (1960).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hambardzumyan, D., Gutmann, D. H. & Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 19, 20–27 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Shichita, T. et al. Pivotal role of cerebral interleukin-17-producing γδT cells in the delayed phase of ischemic brain injury. Nat. Med. 15, 946–950 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Golden, S. A. et al. Basal forebrain projections to the lateral habenula modulate aggression reward. Nature 534, 688–692 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mishra, V. et al. Primary blast causes mild, moderate, severe and lethal TBI with increasing blast overpressures: experimental rat injury model. Sci. Rep. 6, 26992 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Keaney, J. et al. Autoregulated paracellular clearance of amyloid-β across the blood-brain barrier. Sci. Adv. 1, e1500472 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Campbell, M. et al. RNAi-mediated reversible opening of the blood-brain barrier. J. Gene Med. 10, 930–947 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Doyle, S. L. et al. IL-18 attenuates experimental choroidal neovascularization as a potential therapy for wet age-related macular degeneration. Sci. Transl. Med. 6, 230ra44 (2014).

    Article  PubMed  Google Scholar 

  49. Wälchli, T. et al. Quantitative assessment of angiogenesis, perfused blood vessels and endothelial tip cells in the postnatal mouse brain. Nat. Protoc. 10, 53–74 (2015).

    Article  PubMed  Google Scholar 

  50. Blank, T. et al. Brain endothelial- and epithelial-specific interferon receptor chain 1 drives virus-induced sickness behavior and cognitive impairment. Immunity 44, 901–912 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Janssen, W. G. et al. Cellular and synaptic distribution of NR2A and NR2B in macaque monkey and rat hippocampus as visualized with subunit-specific monoclonal antibodies. Exp. Neurol. 191 (Suppl. 1), S28–S44 (2005).

    Article  Google Scholar 

  52. Armulik, A. et al. Pericytes regulate the blood-brain barrier. Nature 468, 557–561 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank A. Keller for advice on BBB-related studies and the Center for Comparative Medicine and Surgery housing facilities staff for their work and support. This research was supported by Mental Health grants RO1 MH090264 (S.J.R.), P50 MH096890 (S.J.R.), P50 AT008661-01 (S.J.R.), RO1 MH114882 (S.J.R.), RO1 MH104559 (S.J.R. and M.M.), NIH/NHLBI P01 HL131478 (Z.A.F.), T32 MH087004 (M.L.P., M.H. and M.F.), T32 MH096678 (M.L.P.), F30 MH100835 (M.H.) and F31 MH105217 (M.L.P.), a Janssen/IMHRO Rising Star Translational Research Award (S.J.R.), a Swiss National Science Foundation Advanced Postdoc Mobility Fellowship (V.K.) and a Brain and Behavior Research Foundation NARSAD Young Investigator Award (G.E.H.). C.M. is supported by a Brain and Behavior Research Foundation NARSAD Young Investigator Grant sponsored by the P&S Fund.

Author information

Authors and Affiliations

Authors

Contributions

C.M. and S.J.R. designed the study and wrote the manuscript. C.M., M.L.P., G.E.H., A.T., M.E.F., H.A., K.B.L., Z.S.L., S.A.G. and M.H. performed stereotaxic surgeries, tissue collection and behavioral manipulations and analyzed data. V.K. performed and analyzed flow experiments and Ccr2 RFP::Cx3cr1 GFP mouse immunostaining. V.K. and M.M. provided Ccr2 RFP::Cx3cr1 GFP mice and provided advice on BBB- and immune-related studies. V.X.W., Z.A.F. and C.Y.T. designed, performed and analyzed magnetic resonance imaging scans. S.B. advised on analysis approaches and analyzed data. W.G.J. prepared and imaged transmission electron microscopy samples. B.L., E.M.P., C.T. and G.T. provided post-mortem human tissue samples. M.C. provided viral vectors and advised on viral studies. All the authors read and commented on the manuscript.

Corresponding author

Correspondence to Scott J. Russo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated Supplementary Information

Supplementary Figure 1 Behavioral phenotype of quantitative PCR (qPCR) experiments and Cldn5 expression in other brain regions or animal model of stress

Stress-susceptible (SS) mice spent less time in the interaction zone (***p < 0.0001) (A) and more time in the corners (***p < 0.0001) (B) when the social target (aggressor, AGG) was present compared to unstressed control (CTRL) and resilient (RES). However no significant difference was observed when the social target was absent. C) Stressed mice traveled less distance than CTRL when the social target was either present (***p = 0.0004) or absent (***p = 0.0004) but no difference was observed between SS and RES groups. D) Heat maps of gene expression related to endothelial cells and tight junctions normalized to blood vessel marker pecam1. E) Normalization to pecam1 confirmed specific reduction of cldn5 expression in the nucleus accumbens (NAc) of SS mice (***p = 0.0009). Cldn5 mRNA expression was reduced in the hippocampus (G) of both SS and RES mice after 10-day chronic social defeat stress (CSDS) while no change was measured in the prefrontal cortex (PFC) (F) or hypothalamus (H). Gapdh was used as housekeeping gene for hippocampus (HIPP), prefrontal cortex (PFC) and hypothalamus studies. I) Six days of chronic variable stress (CVS) did not modify cldn5 mRNA expression however, after 28 days of CVS, cldn5 mRNA level was significantly reduced in the NAc of male C57Bl/6 mice (J). Gapdh was used as housekeeping gene for CVS studies. Data represent mean ± SEM, number of animal (n) is indicated on graphs. Unpaired t-test for CUS-related qPCR experiments and one-way ANOVA for other graphs followed by Bonferroni’s multiple comparison test, *p < 0.05; **p < 0.01; ***p < 0.001

Supplementary Figure 2 Behavioral phenotype of immunohistochemistry (IHC) experiments and assessment of Cldn5 endothelial cell specificity and stress-induced loss

SS mice spent less time in the interaction zone (**p = 0.0058) (A) and more time in the corners (**p = 0.0090) (B) when the social target (AGG) was present compared to unstressed CTRL and RES mice. SS mice spent more time in the interaction zone when the social target was absent (*p = 0.0124) (A) but no significant difference was observed for time spent in corners. C) Stressed mice traveled less distance than CTRL when the social target was either present (**p = 0.0024) or absent (*p = 0.0133) but no difference was observed between SS and RES groups. D) Cldn5 mRNA expression is selectively expressed in endothelial cells in the adult mouse and human brain according to RNA-sequencing transcriptome databases29,30. E) Cldn5 protein level was enriched five-fold in half-brain capillary extraction when compared to whole cell homogenates (***p < 0.0001). Purity of the extraction was validated with astrocyte (glial fibrillary acidic protein, gfap) and neuronal (neun) markers. Representative blots in triplicate are shown. F) Following 10-day CSDS, no difference was observed in CD31, occluding or ZO-1 protein levels in the NAc of SS or RES mice. Data represent mean ± SEM, number of animals (n) is indicated on graphs. Unpaired t-test for Western Blot analyses, Pearson’s correlation for tight junction protein level vs social interaction (SI) ratio and one-way ANOVA for other graphs followed by Bonferroni’s multiple comparison test, *p < 0.05; **p < 0.01; ***p < 0.001

Supplementary Figure 3 Tight junction protein levels in the hippocampus (HIPP) and prefrontal cortex (PFC) following 10-d CSDS

A) Cldn5 (*p = 0.0236) and occludin (*p = 0.0327) protein levels were significantly higher in RES mice when compared to SS, but not unstressed control mice. Occludin protein level was positively correlated with SI ratio (*p = 0.0486). No difference was observed for ZO-1 or the endothelial cell marker CD31. B) 10-day CSDS had no effect on tight junction protein levels or CD31 in the PFC. Scale bar at 200 µm. Images corresponded to flattened 1-µm-thick z-stacks at 40x magnification. Data represent mean ± SEM, number of animals (n) is indicated on graphs. One-way ANOVA followed by Bonferroni’s multiple comparison test and Pearson’s correlation for tight junction protein level vs SI ratio, *p < 0.05

Supplementary Figure 4 Blood vessel and capillary morphology in the NAc and PFC of CTRL, SS and RES mice

A) Detailed behavioral phenotyping of the mice imaged with transmission electron microscopy 48 hours after the last defeat. SS mice SI ratio was lower than CTRL and RES mice (***p = 0.0001) as they spent less time in the interaction zone (***p < 0.0001) (A) and more time in the corners (***p = 0.0001) (B) when the social target (AGG) was present. C) No difference was observed between groups for distance traveled. D) Representative pictures of large blood vessels and capillaries in the NAc of CTRL, SS and RES mice. Mice were injected with horseradish peroxidase (HRP) that was allowed to circulate for 2 hours before 20-min perfusion and fixation. Scale bar is set at 2 µm or 500 nm. E) No significant difference was measured for tight junction discontinuity in the PFC of SS or RES mice when compared to CTRL mice. Scale bar is set at 2 µm or 500 nm, 15-24 tight junctions/mouse for three mice/group. Data represent mean ± SEM, number of animals (n) is indicated on graphs. One-way ANOVA followed by Bonferroni’s multiple comparison test, **p < 0.01, ***p < 0.001

Supplementary Figure 5 Behavioral phenotype of SS mice treated chronically with vehicle or the antidepressant imipramine and CLDN5 expression in MDD patients and cocaine users

A) Timeline showing mice were exposed to 10-day CSDS then screened for behavioral phenotype 24 hours later. Vehicle-treated SS mice spent less time in the interaction zone (B) and more time in the corners (C) when the social target (AGG) was present when compared to the other groups, including imipramine-treated SS mice. No difference was observed when the social target was absent. D) Locomotion was similar between all groups despite the presence or absence of the social target. E) Acute treatment with the antidepressant imipramine is not sufficient to rescue cldn5 loss after 10-day CSDS. F) No significant difference was measured for GAPDH in NAc postmortem tissue from healthy controls (CTRL) or MDD patients with (MDD, AD + ) or without (MDD, AD-) antidepressant treatment at the time of death. G) CLDN5 expression in the HIPP and PFC (normalized to GAPDH housekeeping gene) was not significantly different in MDD patients treated or untreated with antidepressants when compared to healthy controls. H) CLDN5 expression is unchanged in cocaine users when compared to healthy controls (normalized to GAPDH housekeeping gene). Data represent mean ± SEM, number of animals or subjects (n) is indicated on graphs. Unpaired t-test for acute treatment and cocaine users, one-way ANOVA for MDD cohorts and two-way ANOVA for other graphs followed by Bonferroni’s multiple comparison test, *p < 0.05

Supplementary Figure 6 Gene expression in NAc of AAV-shRNA-Cldn5-injected mice, supplementary SI behaviors and anxiety test results

A) No significant change was observed at mRNA level in the NAc of AAV-shRNA versus AAV-shRNA-cldn5 for other claudins (cdn1, cldn3, cdn12), cytoskeletal regulators (rac1) or markers of endothelial cells (pecam1), pericytes (pdgfrβ), astrocytes (gfap, s100β) and microglia (iba1) when normalized to the housekeeping gene gapdh. B) CD31 protein level was similar in the NAc of AAV-shRNA-cldn5 and AAV-shRNA-injected mice. C) Stressed AAV-shRNA-injected mice spent less time in the interaction zone and more time in the corners when the social target (AGG) was present. No difference was observed between mice groups for time spent in the interaction zone or corners when the social target was absent or for overall locomotion. D) No significant difference was measured between groups for latency to eat in the novelty-suppressed feeding test (left) or home cage (right). Virus injection in the NAc had no effect on anxiety as measured with the elevated plus maze (E) and open field (F) tests. Data represent mean ± SEM, number of animals (n) is indicated on graphs. Unpaired t-test for virus validation and two-way ANOVA followed by Bonferroni’s multiple comparison test for behavioral experiments, *p < 0.05, ***p < 0.001

Supplementary Figure 7 Experimental timeline for virally mediated HIPP injections and rescue experiment, virus validation and behaviors

Experimental timeline (A) and virus validation (qPCRs, **p = 0.0039, Western Blots, ***p = 0.0006) (B) for AAV-shRNA and AAV-shRNA-cldn5 injections in the HIPP. Full-length blots are included in Supplementary Fig. 12. Downregulation of cldn5 expression had no effect in the splash (C) and sucrose preference tests (D). However mice injected with AAV-shRNA-cldn5 spent more time immobile in the forced swim test (E) and stressed AAV-shRNA-cldn5 mice displayed social avoidance (F). Representative heat maps are shown on the right. G) Experimental timeline of the control cohort for rescue experiment. H) Removal of Dox from the water allowed recovery of cldn5 expression at both mRNA (p = 0.9473) and protein (p = 0.2077) levels in the rescue cohort. Full-length blots are included in Supplementary Fig. 12. I) Conversely, downregulation of cldn5 mRNA (**p = 0.0020) and protein (*p = 0.0116) levels was confirmed in the cohort remaining on Dox. Full-length blots are included in Supplementary Fig. 12. G) Stressed AAV-shRNA-cldn5 mice that remained on dox treatment displayed depression-like behaviors in forced swim (**p = 0.0015) and sucrose preference (**p = 0.0028) tests before (J) and after 5-day rest as measured with sucrose preference (***p = 0.0002) and social interaction tests (**p = 0.0028) (K). Representative heat maps are shown on the left. Data represent mean ± SEM, number of animals (n) is indicated on graphs. Two-way ANOVA followed by Bonferroni’s multiple comparison test, *p < 0.05, **p < 0.01, ***p < 0.001

Supplementary Figure 8 Gd-DTPA and cadaverine Alexa Fluor-555 experiments

A cohort of mice was first behaviorally characterized for MRI studies. SS mice spent less time in the interaction zone (A) and more time in the corners (B) when the social target (AGG) was present when compared to CTRL and RES mice. No difference was observed when the social target was absent. Locomotion was similar in SS and RES mice (C). D) Higher Gd-DTPA signal was detected in different brain regions, including the NAc, in SS mice and negatively correlated with SI ratio. E) Gd-DTPA level was higher in the hippocampus of SS mice versus RES mice and significantly correlated with social avoidance. F) Conversely, no difference was measured between groups for the PFC. A second cohort of mice was behaviorally characterized to assess BBB permeability to cadaverine Alexa Fluor-555. After 10-day CSDS, SS mice spent less time in the interaction zone (G) and more time in the corners (H) versus CTRL and RES groups when the social target was present (AGG). No difference was observed when the social target was absent. I) Stressed mice traveled less distance compared to CTRL when the social target was either present or absent but no significant difference was measured between SS and RES groups. Cadaverine level is significantly correlated with social avoidance in the nucleus accumbens (NAc) (J) and hippocampus (K). L) No difference was measured between groups for the PFC. Data are presented as mean ± SEM, number of animals (n) is indicated on graphs. Correlations were evaluated with Pearson’s correlation coefficient, one-way ANOVA followed by Bonferroni’s multiple comparison test for other graphs, *p < 0.05; **p < 0.01; ***p < 0.001

Supplementary Figure 9 Supplementary behavioral data for Evans blue (EB) extravasation and Ccr2 RFP::Cx3cr1 GFP mice after CSDS

A cohort of mice was behaviorally characterized to assess BBB permeability to circulating EB. SS mice spent less time in the interaction zone (A) and more time in the corners (B) when the social target (AGG) was present when compared to CTRL and RES mice. SS mice spent more time in the interaction zone when the social target was absent (A) but no difference was observed for the corners (B). C) Stressed mice displayed less locomotion versus unstressed CTRL when the social target was either present or absent. However no significant difference was measured between SS and RES groups. D) EB level in the NAc was significantly correlated with social avoidance. E) EB could be detected in hippocampus blood vessels 10 min after the retro-orbital injection was performed (left). No EB extravasation was observed after 16-h circulation followed by 5-min perfusion in the hippocampus (right). F) Similarly, EB was detectable in PFC blood vessels 10-min after the injection but not after 16-h circulation and 5-min perfusion. No difference was measured for the hippocampus (G) or PFC (H) after EB extraction. I) C-C chemokine receptor 2 (ccr2) mRNA expression is specifically elevated in the NAc of SS mice after 10-day CSDS and correlated with social avoidance. J) Stressed ccr2 RFP:: cx3cr1 GFP mice spent less time in the interaction zone when the social target (AGG) was present. Lower overall locomotion was also observed in stressed ccr2 RFP:: cx3cr1 GFP mice when compared to unstressed controls when the AGG was absent. K) Flow cytometry gating strategy for ccr2 RFP monocytes and cx3cr1 GFP microglia. L) No difference was measured between groups for percentage (%) of ccr2-/cx3cr1+ cells (microglia). M) Immunohistochemical analysis of ccr2RFP+ monocytes shows that they accumulate within blood vessels of the NAc (left) and lateral ventricle (right), but not in the parenchyma. Scale bar at 100 µm (50 µm for the insets). Data represent mean ± SEM, number of animals (n) is indicated on graphs. Correlations were evaluated with Pearson’s correlation coefficient, unpaired t-test for ccr2 RFP :: cx3cr1 GFP mice and one-way ANOVA followed by Bonferroni’s multiple comparison test for other graphs, *p < 0.05; **p < 0.01, ***p < 0.001

Supplementary Figure 10 Behavioral data for IL-6 ELISA, IL-6–biotin passage into the parenchyma and NAc IL-6 versus saline infusion

A) Following 10-day CSDS, blood serum and NAc, HIPP and PFC punches were collected to assess IL-6 protein level 48 h after the last defeat. SS mice spent less time in the interaction zone (***p < 0.0001) and more time in the corners (***p < 0.0001) when the aggressor (AGG) was present, No difference was observed when the AGG was absent or between SS and RES mice for overall locomotion. B) IL-6 protein was barely detectable in the HIPP and PFC of CTRL, SS and RES mice. C) Circulating IL-6 level is increased 20 min after recombinant IL-6 i.p. injections. D) SI ratio of mice retro-orbitally injected with biotinylated IL-6 (IL6-biotin) 24h after the SI test (48h after the last defeat or 10-day CSDS). E) SS mice spent less time in the interaction zone, and more time in the corners when the social target was present compared to CTRL and RES animals. No difference was observed for locomotion. F) Minute amount of IL6-biotin-Neutravidin-Oregon488 was detectable in the HIPP of SS mice. None was detectable in the PFC (G). H) Mice administered direct infusion of IL-6 into the NAc spent less time in the interaction zone when the social target AGG was present, but not absent, when compared to the saline group. No difference was observed for the time spent in corners (I) or locomotion (J). Data represent mean ± SEM, number of animals (n) is indicated on graphs. One-way ANOVA followed by Bonferroni’s multiple comparison test for (A-D) and two-way ANOVA followed by Bonferroni’s multiple comparison test for other graphs, *p < 0.05; **p < 0.01

Supplementary Figure 11 Social stress induces neurovascular pathology and BBB leakiness promoting depression-like behaviors

10-day chronic social defeat stress (CSDS) induces loss of Cldn5 expression at mRNA and protein levels, leading to abnormalities in blood vessel morphology and increased BBB permeability in stress-susceptible (SS) mice. These molecular and cellular changes are associated with depression-like behavior such as social avoidance, anhedonia, despair and lack of self-care. Conversely, mice resilient (RES) to CSDS display normal social and stress coping behaviors and neurovascular features similar to unstressed controls (CTRL)

Supplementary Figure 12 Full-length blots of cropped blots in Supplementary Fig. 7

A) Full-length Western blot for cldn5 (top) and actin (bottom) after AAV-shRNA-cldn5 viral injection in the hippocampus (HIPP). B) Full-length Western blot for cldn5 (top) and actin (bottom) after doxycycline (Dox) was removed from the drinking water (Rescue – no Dox) in mice injected with AAV-shRNA-cldn5 in the nucleus accumbens (NAc). C) Full-length Western Blot for cldn5 (top) and actin (bottom) in the NAc of AAV-shRNA-cldn5-injected mice remaining on Dox for the entire experiment

Supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Menard, C., Pfau, M.L., Hodes, G.E. et al. Social stress induces neurovascular pathology promoting depression. Nat Neurosci 20, 1752–1760 (2017). https://doi.org/10.1038/s41593-017-0010-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-017-0010-3

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