Social stress induces neurovascular pathology promoting depression

Caroline Menard, Madeline L Pfau, Georgia E Hodes, Veronika Kana, Victoria X Wang, Sylvain Bouchard, Aki Takahashi, Meghan E Flanigan, Hossein Aleyasin, Katherine B LeClair, William G Janssen, Benoit Labonté, Eric M Parise, Zachary S Lorsch, Sam A Golden, Mitra Heshmati, Carol Tamminga, Gustavo Turecki, Matthew Campbell, Zahi A Fayad, Cheuk Ying Tang, Miriam Merad, Scott J Russo, Caroline Menard, Madeline L Pfau, Georgia E Hodes, Veronika Kana, Victoria X Wang, Sylvain Bouchard, Aki Takahashi, Meghan E Flanigan, Hossein Aleyasin, Katherine B LeClair, William G Janssen, Benoit Labonté, Eric M Parise, Zachary S Lorsch, Sam A Golden, Mitra Heshmati, Carol Tamminga, Gustavo Turecki, Matthew Campbell, Zahi A Fayad, Cheuk Ying Tang, Miriam Merad, Scott J Russo

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.

Figures

Figure 1. Social stress vulnerability and MDD…
Figure 1. Social stress vulnerability and MDD is associated with reduced cldn5 expression
A) Experimental timeline of 10-day social defeat stress (CSDS), social interaction (SI) behavioral screening and tissue collection of nucleus accumbens (NAc). B) Quantitative PCR revealed significant changes in the NAc of stress-susceptible (SS) and resilient (RES) mice when compared to unstressed controls (CTRL) for gene expression related to endothelial cell biology, angiogenesis, tight junctions, blood-brain barrier (BBB) formation and cytokines/chemokines. The range of color indicates individual differences within a group – standard error of the mean (SEM) from the average represented by the dashed line. C) Several patterns of gene expression correlated with SI ratio (D). Representative heat maps of SI test are shown on the right. Cldn5 mRNA (E) and protein (F, G) levels were lower in the NAc of SS mice and correlated with social avoidance. Scale bar is set at 20 µm. H) Chronic treatment with imipramine reversed social avoidance and rescued cldn5 loss in SS mice. I) Transmission electron microscopy revealed discontinuity in tight junctions of SS (red arrows), but not resilient, mice. Scale bar is set at 500 nm (250 nm for insets), 52–66 tight junctions/mouse and three mice/group. J)CLDN5 mRNA level was significantly lower in the NAc of MDD patients from two different cohorts. Data represent mean ± SEM, number of animals or patients (n) is indicated on graphs. Two-way ANOVA followed by Bonferroni’s multiple comparison test for antidepressant treatment, correlations were evaluated with Pearson’s correlation coefficient, and one-way ANOVA followed by Bonferroni’s multiple comparison test for other graphs, *p < 0.05; **p < 0.01; ***p < 0.001.
Figure 2. Conditional knockdown of cldn5 expression…
Figure 2. Conditional knockdown of cldn5 expression in the NAc is sufficient to induce depression-like behaviors
A) Experimental timeline of NAc bilateral injection of AAV-shRNA or AAV-shRNA-cldn5 viruses and behavioral studies. Cldn5 mRNA (B) and protein (C) levels are reduced following AAV-shRNA-cldn5 injection in the NAc of stress-naïve mice when compared to AAV-shRNA-injected animals. Co-staining with CD31 confirmed lower cldn5 protein level in blood vessels following AAV-shRNA-cldn5 virus injection and doxycycline (Dox) treatment (C). Scale bar is set at 2 µm. Downregulation of cldn5 expression prior to subthreshold micro-defeat (stressed) induced depression-like behaviors as assessed by splash (D), sucrose preference (E), forced swim (F) and social interaction (G) tests. The effect was significant when compared to unstressed mice and stressed AAV-shRNA. H) Experimental timeline of cldn5 rescue experiment. I) Stressed AAV-shRNA-cldn5 mice displayed depression-like behaviors when subjected to doxycycline treatment as assessed with forced swim test and sucrose preference. J) No difference was observed between groups for sucrose preference and social interaction when doxycycline was removed from the water, allowing normal cldn5 expression. Data represent mean ± SEM, number of animals (n) is indicated on graphs. Unpaired t-test for virus validation or two-way ANOVA followed by Bonferroni’s multiple comparison test for behaviors, *p < 0.05; **p < 0.01; ***p < 0.001.
Figure 3. Social stress vulnerability is associated…
Figure 3. Social stress vulnerability is associated with increased blood-brain barrier (BBB) permeability
A) Experimental timeline of 10-day CSDS, SI screening, and magnetic resonance imaging (MRI) scans. Pictures display baseline MRI scan compared to 25 minutes after injection of the gadolinium-contrasting agent (Gd-DTPA). B) Higher Gd-DTPA signal was detected in different brain regions, including the NAc in SS mice and negatively correlated with SI ratio (C). The range of color indicates individual differences within a group - SEM from the average represented by the dashed line. Assessment of BBB permeability with cadaverine Alexa Fluor-555 and Evans blue (EB) dyes revealed significant BBB leakiness in the NAc of SS mice (D, E). EB dye is present in all vessels within 10 min of injection and prior to perfusion (E, left) but can only be detected in the perivascular space of NAc blood vessels in SS mice after circulating 16 hours followed by 5-min perfusion (E, right). Scale bar is set at 20 µm. F, G) Downregulation of cldn5 expression increased BBB permeability to cadaverine Alexa Fluor-555 and EB dyes (H) in AAV-shRNA-cldn5 mice. I) Transmission electron microscopy, conducted 2 hours after injection of horseradish peroxidase (HRP) followed by 20-min perfusion, confirmed increased BBB permeability in the NAc of SS mice with HRP detected within the endothelial cell lining (red arrows). Quantification was performed on 34–61 blood vessels/mouse in three mice/group. Scale bar is set at 500 nm. Data represent mean ± SEM, number of animals (n) is indicated on graphs. Correlations were evaluated with Pearson’s correlation coefficient, two-way ANOVA followed by Bonferroni’s multiple comparison test for BBB permeability in virus-injected mice, one-way ANOVA followed by Bonferroni’s multiple comparison test for other graphs, *p < 0.05; **p < 0.01; ***p < 0.001.
Figure 4. Chronic social stress induces peripheral…
Figure 4. Chronic social stress induces peripheral monocyte accumulation and IL-6 passage into the NAc
A) Experimental timeline of 10-day CSDS, SI behavioral screening and tissue collection of ccr2RFP∷ cx3cr1GFP mice. B) Stressed ccr2RFP∷ cx3cr1GFP mice showed decreased SI scores following 10-day CSDS. Representative heat maps are shown on the right. C) Flow cytometric analysis of forebrain ccr2RFP+ and cx3cr1GFP+ cells revealed higher level of ccr2+ peripheral monocyte accumulation into the brain of stressed mice. Cells were gated on live CD11b+F4/80+ (left). Numbers adjacent to gates represent percentages of ccr2+cx3cr1 cells among CD11b+F4/80+ cells. Right panel shows CD45 expression of F4/80+ccr2+ monocytes and F4/80+gfp+ microglia. Bar graph shows percentage of ccr2+cx3cr1 cells among forebrain CD11b+F4/80+ cells. D) IL-6 protein level is increased in the blood and NAc of SS mice after 10-day CSDS. E) Dose response IL-6 i.p. injection did not change IL-6 protein level in the NAc of stress-naïve mice. F) Direct infusion of IL-6 into the NAc (5 min) induces social avoidance when subthreshold micro-defeat was conducted 20 min after the end of the infusion. G) Experimental timeline of peripheral biotinylated-IL6 injection after 10-day CSDS. IL-6-biotin-Neuravidin-Oregon 488 was detectable in the NAc parenchyma of SS mice only after 2h circulation and 5-min perfusion with PBS. H) Biotinylated IL-6 was also detectable in the NAc of AAV-shRNA-cldn5-injected mice. Data represent mean ± SEM, number of animals (n) is indicated on graphs. T-test for CX3CR1-GFP/CCR2-RFP mouse data, one-way ANOVA followed by Bonferroni’s multiple comparison test for IL-6 protein levels and two-way ANOVA followed by Bonferroni’s multiple comparison test for SI ratio after IL-6 or saline infusion, *p < 0.05; **p < 0.01; ***p < 0.001.

References

    1. Menard C, Pfau ML, Hodes GE, Russo SJ. Immune and Neuroendocrine Mechanisms of Stress Vulnerability and Resilience. Neuropsychopharmacology. 2016;42:62–80.
    1. Kessler RC, Chiu WT, Demler O, Merikangas KR, Walters EE. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617–627.
    1. Seligman F, Nemeroff CB. The interface of depression and cardiovascular disease: therapeutic implications. Ann N Y Acad Sci. 2015;1345:25–35.
    1. Carney RM, Freedland KE. Depression and coronary heart disease. Nat Rev Cardiol. 2016;14:145–155.
    1. Wood SK. Individual differences in the neurobiology of social stress: implications for depression-cardiovascular disease comorbidity. Curr Neuropharmacol. 2014;12:205–211.
    1. Huffman JC, Celano CM, Beach SR, Motiwala SR, Januzzi JL. Depression and cardiac disease: epidemiology, mechanisms, and diagnosis. Cardiovasc Psychiatry Neurol. 2013;2013:695925.
    1. Hodes GE, Kana V, Menard C, Merad M, Russo SJ. Neuroimmune mechanisms of depression. Nat Neurosci. 2015;18:1386–1393.
    1. Barnes J, Mondelli V, Pariante CM. Genetic Contributions of Inflammation to Depression. Neuropsychopharmacology. 2016;42:81–98.
    1. Dantzer R. Cytokine, sickness behavior, and depression. Immunol Allergy Clin North Am. 2009;29:247–264.
    1. Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.
    1. Hodes GE, et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci U S A. 2014;111:16136–16141.
    1. Powell ND, et al. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via beta-adrenergic induction of myelopoiesis. Proc Natl Acad Sci U S A. 2013;110:16574–16579.
    1. Heidt T, et al. Chronic variable stress activates hematopoietic stem cells. Nat Med. 2014;20:754–758.
    1. Wohleb ES, Powell ND, Godbout JP, Sheridan JF. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J Neurosci. 2013;33:13820–13833.
    1. Weber MD, Godbout JP, Sheridan JF. Repeated Social Defeat, Neuroinflammation, and Behavior: Monocytes Carry the Signal. Neuropsychopharmacology. 2016;42:46–61.
    1. Esposito P, et al. Acute stress increases permeability of the blood-brain-barrier through activation of brain mast cells. Brain Res. 2001;888:117–127.
    1. Santha P, et al. Restraint Stress-Induced Morphological Changes at the Blood-Brain Barrier in Adult Rats. Front Mol Neurosci. 2015;8:88.
    1. Friedman A, et al. Pyridostigmine brain penetration under stress enhances neuronal excitability and induces early immediate transcriptional response. Nat Med. 1996;2:1382–1385.
    1. Sharma HS, Dey PK. Impairment of blood-brain barrier (BBB) in rat by immobilization stress: role of serotonin (5-HT) Indian J Physiol Pharmacol. 1981;25:111–122.
    1. 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. 1984;19:1183–1206.
    1. Roszkowski M, Bohacek J. Stress does not increase blood-brain barrier permeability in mice. J Cereb Blood Flow Metab. 2016;36:1304–1315.
    1. Gunzel D, Yu AS. Claudins and the modulation of tight junction permeability. Physiol Rev. 2013;93:525–569.
    1. Nitta T, et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol. 2003;161:653–660.
    1. Meltzer H, Vostanis P, Ford T, Bebbington P, Dennis MS. Victims of bullying in childhood and suicide attempts in adulthood. Eur Psychiatry. 2011;26:498–503.
    1. Berton O, Nestler EJ. New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci. 2006;7:137–151.
    1. Menard C, Hodes GE, Russo SJ. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience. 2016;321:138–162.
    1. Golden SA, Covington HE, 3rd, Berton O, Russo SJ. A standardized protocol for repeated social defeat stress in mice. Nat Protoc. 2011;6:1183–1191.
    1. Russo SJ, Nestler EJ. The brain reward circuitry in mood disorders. Nat Rev Neurosci. 2013;14:609–625.
    1. Zhang Y, et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci. 2014;34:11929–11947.
    1. Zhang Y, et al. Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse. Neuron. 2016;89:37–53.
    1. Hodes GE, et al. Sex Differences in Nucleus Accumbens Transcriptome Profiles Associated with Susceptibility versus Resilience to Subchronic Variable Stress. J Neurosci. 2015;35:16362–16376.
    1. Golden SA, et al. Epigenetic regulation of RAC1 induces synaptic remodeling in stress disorders and depression. Nat Med. 2013;19:337–344.
    1. Campbell M, et al. Systemic low-molecular weight drug delivery to pre-selected neuronal regions. EMBO Mol Med. 2011;3:235–245.
    1. McKim DB, et al. Microglial recruitment of IL-1beta-producing monocytes to brain endothelium causes stress-induced anxiety. Mol Psychiatry, in press. 2017
    1. Saederup N, et al. Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS One. 2010;5:e13693.
    1. Mizutani M, et al. The fractalkine receptor but not CCR2 is present on microglia from embryonic development throughout adulthood. J Immunol. 2012;188:29–36.
    1. Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol. 2014;14:392–404.
    1. Maes M, et al. Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine. 1997;9:853–858.
    1. Dowlati Y, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–457.
    1. Coppen AJ. Abnormality of the blood-cerebrospinal fluid barrier of patients suffering from a depressive illness. J Neurol Neurosurg Psychiatry. 1960;23:156–161.
    1. Hambardzumyan D, Gutmann DH, Kettenmann H. The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci. 2016;19:20–27.
    1. Shichita T, et al. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15:946–950.
    1. Golden SA, et al. Basal forebrain projections to the lateral habenula modulate aggression reward. Nature. 2016;534:688–692.
    1. Mishra V, et al. Primary blast causes mild, moderate, severe and lethal TBI with increasing blast overpressures: Experimental rat injury model. Sci Rep. 2016;6:26992.
    1. Keaney J, et al. Autoregulated paracellular clearance of amyloid-beta across the blood-brain barrier. Sci Adv. 2015;1:e1500472.
    1. Campbell M, et al. RNAi-mediated reversible opening of the blood-brain barrier. J Gene Med. 2008;10:930–947.
    1. Doyle SL, et al. IL-18 attenuates experimental choroidal neovascularization as a potential therapy for wet age-related macular degeneration. Sci Transl Med. 2014;6:230ra244.
    1. Walchli T, et al. Quantitative assessment of angiogenesis, perfused blood vessels and endothelial tip cells in the postnatal mouse brain. Nat Protoc. 2015;10:53–74.
    1. Blank T, et al. Brain Endothelial- and Epithelial-Specific Interferon Receptor Chain 1 Drives Virus-Induced Sickness Behavior and Cognitive Impairment. Immunity. 2016;44:901–912.
    1. Janssen WG, 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. 2005;191(Suppl 1):S28–44.
    1. Armulik A, et al. Pericytes regulate the blood-brain barrier. Nature. 2010;468:557–561.

Source: PubMed

3
订阅