Neuroimmune mechanisms of depression

Georgia E Hodes, Veronika Kana, Caroline Menard, Miriam Merad, Scott J Russo, Georgia E Hodes, Veronika Kana, Caroline Menard, Miriam Merad, Scott J Russo

Abstract

Current diagnosis of depression is based solely on behavioral symptomatology. The available US Food and Drug Administration-approved treatments for depression have come from serendipitous discovery and are ineffective in nearly 30-50% of patients, which is thought to reflect a lack of specificity in targeting underlying pathophysiological mechanisms. Recent evidence has identified depression-related disruptions in a neuroimmune axis that interfaces the immune system and CNS to control behavior. This Review examines the evidence in patients and in animal models of depression that demonstrates how the peripheral immune system acts on the brain to alter an individual's response to stress, ultimately contributing to their vulnerability to mood disorders.

Figures

Figure 1. Circuits of the neuro-immune axis
Figure 1. Circuits of the neuro-immune axis
The autonomic nervous system connects peripheral immune organs with the central nervous system and regulates homeostatic and inflammatory functions. A) Sympathetic nerves innervate Nestin+ bone marrow niche cells. Circadian release of NE activates β3 adrenergic receptors (b3AR) on perivascular Nestin+ niche cells, which leads to rhythmic downregulation of CXCL12 and the subsequent release of hematopoietic stem cells (HSC) into the blood stream during the resting period. B) The inflammatory reflex: nerve endings of the vagus nerve sense inflammatory mediators and transport the signals to the brain stem. From there, the signal travels via efferent cholinergic vagal nerves to the celiac ganglion and through adrenergic fibers of the splenic nerve and delivers NE to β2 adrenergic receptors (b2AR) on a subset of splenic T cells, which in turn secrete acetylcholine (ACh) that binds to α7 nicotinic acetylcholine receptors (a7nAChR) on marginal zone macrophages and suppresses the production of inflammatory cytokines, such as TNF-α and IL-1β, . C) Activation of the hypothalamic-pituitary-adrenocortical (HPA) axis leads to glucocorticoid (GC) release from the adrenal glands that exerts anti-inflammatory effects through binding of GC to cytosolic glucocorticoid receptor (GR) and inhibition of IL-6 release by monocytes.
Figure 2. Inflammation and the brain
Figure 2. Inflammation and the brain
In response to chronic stress, there is an increase in circulating monocyte levels, notably LY6Chi. These monocytes are then lured by chemotactic cytokines in brain regions associated with anxiety and depression. A) Once in the brain, monocytes and cytokines affect neuronal synaptic plasticity by modifying cell signaling and gene expression through activation of cytokines receptors, corticotropin-releasing hormone receptor 1 (CRF1) or toll-like receptor 4 (TLR4), by either cytokines themselves, corticotropin-releasing factor (CRF), pathogen-associated molecular patterns (PAMPs) and/or damage-associated molecular pattern molecules (DAMPs). B) Synaptic changes such as down-regulation of CX3CL1 are detected by microglia inducing cytokine release and monocytes recruitment. A proportion of recruited monocytes will remain in the brain and adopt microglia-like properties. C) Cytokines can penetrate into the brain via passive or active mechanisms. Alteration of tight junction protein expression can result in the formation of transient holes allowing small circulating molecules such as cytokines to passively diffuse between endothelial cells. Binding of cytokines to specific receptors on endothelial cell can also induce production and subsequent release of inflammatory mediators into the brain. Finally, if the BBB is weakened and become more permeable monocytes can burrow through affecting neighboring astrocytes, which engulf blood vessels with their astrocytic end-feet. Circulating cytokines into the brain activate microglia stress response and may induce persistent synaptic changes.

References

    1. Maes M, et al. Increased plasma concentrations of interleukin-6, soluble interleukin-6, soluble interleukin-2 and transferrin receptor in major depression. J Affect Disord. 1995;34:301–309.
    1. Dowlati Y, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–457.
    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
    1. Maes M, et al. Leukocytosis, monocytosis and neutrophilia: hallmarks of severe depression. J Psychiatr Res. 1992;26:125–134.
    1. Fenton WS, Stover ES. Mood disorders: cardiovascular and diabetes comorbidity. Curr Opin Psychiatry. 2006;19:421–427.
    1. Maes M, Kubera M, Obuchowiczwa E, Goehler L, Brzeszcz J. Depression's multiple comorbidities explained by (neuro)inflammatory and oxidative & nitrosative stress pathways. Neuro Endocrinol Lett. 2011;32:7–24.
    1. Association, A.P. Diagnostic and Statistical Manual of Mental Disorders. 5th. Arlington, VA: American Psychiatric Association; 2013.
    1. Krishnan V, Nestler EJ. The molecular neurobiology of depression. Nature. 2008;455:894–902.
    1. Renault PF, et al. Psychiatric complications of long-term interferon alfa therapy. Arch Intern Med. 1987;147:1577–1580.
    1. Conversano C, et al. Interferon alpha Therapy in Patients with Chronic Hepatitis C Infection: Quality of Life and Depression. Hematol Rep. 2015;7:5632.
    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. Miller GE, et al. Greater inflammatory activity and blunted glucocorticoid signaling in monocytes of chronically stressed caregivers. Brain Behav Immun. 2014;41:191–199.
    1. Khandaker GM, Pearson RM, Zammit S, Lewis G, Jones PB. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry. 2014;71:1121–1128.
    1. Raison CL, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry. 2013;70:31–41.
    1. Venkiteshwaran A. Tocilizumab. MAbs. 2009;1:432–438.
    1. Brietzke E, Scheinberg M, Lafer B. Therapeutic potential of interleukin-6 antagonism in bipolar disorder. Med Hypotheses. 2011;76:21–23.
    1. Langley RG, et al. Ustekinumab significantly improves symptoms of anxiety, depression, and skin-related quality of life in patients with moderate-to-severe psoriasis: Results from a randomized, double-blind, placebo-controlled phase III trial. J Am Acad Dermatol. 2010;63:457–465.
    1. Iyengar RL, et al. NSAIDs are associated with lower depression scores in patients with osteoarthritis. Am J Med. 2013;126:1017, e1011–e1018.
    1. Kohler O, et al. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry. 2014;71:1381–1391.
    1. Eyre HA, Air T, Proctor S, Rositano S, Baune BT. A critical review of the efficacy of non-steroidal anti-inflammatory drugs in depression. Prog Neuropsychopharmacol Biol Psychiatry. 2015;57:11–16.
    1. Warner-Schmidt JL, Vanover KE, Chen EY, Marshall JJ, Greengard P. Antidepressant effects of selective serotonin reuptake inhibitors (SSRIs) are attenuated by antiinflammatory drugs in mice and humans. Proc Natl Acad Sci U S A. 2011;108:9262–9267.
    1. Sluzewska A, et al. Interleukin-6 serum levels in depressed patients before and after treatment with fluoxetine. Ann N Y Acad Sci. 1995;762:474–476.
    1. Jazayeri S, et al. Effects of eicosapentaenoic acid and fluoxetine on plasma cortisol, serum interleukin-1beta and interleukin-6 concentrations in patients with major depressive disorder. Psychiatry Res. 2010;178:112–115.
    1. Kubera M, et al. Stimulatory effect of antidepressants on the production of IL-6. International immunopharmacology. 2004;4:185–192.
    1. Murrough JW, et al. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry. 2013;170:1134–1142.
    1. De Kock M, Loix S, Lavand'homme P. Ketamine and peripheral inflammation. CNS Neurosci Ther. 2013;19:403–410.
    1. Scheiermann C, Frenette PS, Hidalgo A. Regulation of leucocyte homeostasis in the circulation. Cardiovasc Res. 2015
    1. Hashimoto D, Miller J, Merad M. Dendritic cell and macrophage heterogeneity in vivo. Immunity. 2011;35:323–335.
    1. Spits H, et al. Innate lymphoid cells--a proposal for uniform nomenclature. Nature reviews. Immunology. 2013;13:145–149.
    1. Frank MG, Weber MD, Watkins LR, Maier SF. Stress sounds the alarm in: The role of the danger-associated molecular pattern HMGB1 in stress-induced neuroinflammatory priming. Brain Behav Immun. 2015
    1. Portou MJ, Baker D, Abraham D, Tsui J. The innate immune system, toll-like receptors and dermal wound healing: A review. Vascul Pharmacol. 2015
    1. Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045.
    1. Raz E. Organ-specific regulation of innate immunity. Nat Immunol. 2007;8:3–4.
    1. Marino F, Cosentino M. Adrenergic modulation of immune cells: an update. Amino acids. 2013;45:55–71.
    1. Amsterdam A, Tajima K, Sasson R. Cell-specific regulation of apoptosis by glucocorticoids: implication to their anti-inflammatory action. Biochem Pharmacol. 2002;64:843–850.
    1. Rosas-Ballina M, et al. Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science. 2011;334:98–101.
    1. Tracey KJ. Reflex control of immunity. Nature reviews. Immunology. 2009;9:418–428.
    1. Avitsur R, Powell N, Padgett DA, Sheridan JF. Social interactions, stress, and immunity. Immunol Allergy Clin North Am. 2009;29:285–293.
    1. Engler H, et al. Interleukin-1 receptor type 1-deficient mice fail to develop social stress-associated glucocorticoid resistance in the spleen. Psychoneuroendocrinology. 2008;33:108–117.
    1. Frank MG, Watkins LR, Maier SF. Stress-induced glucocorticoids as a neuroendocrine alarm signal of danger. Brain Behav Immun. 2013;33:1–6.
    1. Manz MG, Boettcher S. Emergency granulopoiesis. Nature reviews. Immunology. 2014;14:302–314.
    1. Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nature reviews. Immunology. 2014;14:392–404.
    1. Nakai A, Hayano Y, Furuta F, Noda M, Suzuki K. Control of lymphocyte egress from lymph nodes through beta2-adrenergic receptors. The Journal of experimental medicine. 2014;211:2583–2598.
    1. Ransohoff RM, Brown MA. Innate immunity in the central nervous system. The Journal of clinical investigation. 2012;122:1164–1171.
    1. Ginhoux F, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330:841–845.
    1. Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nature neuroscience. 2007;10:1538–1543.
    1. Greter M, et al. Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia. Immunity. 2012;37:1050–1060.
    1. Nandi S, et al. The CSF-1 receptor ligands IL-34 and CSF-1 exhibit distinct developmental brain expression patterns and regulate neural progenitor cell maintenance and maturation. Developmental biology. 2012;367:100–113.
    1. Shechter R, London A, Schwartz M. Orchestrated leukocyte recruitment to immune-privileged sites: absolute barriers versus educational gates. Nat Rev Immunol. 2013;13:206–218.
    1. Bulloch K, et al. CD11c/EYFP transgene illuminates a discrete network of dendritic cells within the embryonic, neonatal, adult, and injured mouse brain. J Comp Neurol. 2008;508:687–710.
    1. Baruch K, Schwartz M. CNS-specific T cells shape brain function via the choroid plexus. Brain Behav Immun. 2013;34:11–16.
    1. Louveau A, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015
    1. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308:1314–1318.
    1. Stevens B, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164–1178.
    1. Paolicelli RC, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333:1456–1458.
    1. Limatola C, Ransohoff RM. Modulating neurotoxicity through CX3CL1/CX3CR1 signaling. Frontiers in cellular neuroscience. 2014;8:229.
    1. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci. 2011;14:1142–1149.
    1. Yamasaki R, et al. Differential roles of microglia and monocytes in the inflamed central nervous system. The Journal of experimental medicine. 2014;211:1533–1549.
    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. Sawicki CM, et al. Social defeat promotes a reactive endothelium in a brain region-dependent manner with increased expression of key adhesion molecules, selectins and chemokines associated with the recruitment of myeloid cells to the brain. Neuroscience. 2014
    1. Hickman SE, et al. The microglial sensome revealed by direct RNA sequencing. Nat Neurosci. 2013;16:1896–1905.
    1. Maes M. Evidence for an immune response in major depression: a review and hypothesis. Prog Neuropsychopharmacol Biol Psychiatry. 1995;19:11–38.
    1. Smith RS. The macrophage theory of depression. Med Hypotheses. 1991;35:298–306.
    1. Heidt T, et al. Chronic variable stress activates hematopoietic stem cells. Nat Med. 2014;20:754–758.
    1. Brachman RA, Lehmann ML, Maric D, Herkenham M. Lymphocytes from chronically stressed mice confer antidepressant-like effects to naive mice. J Neurosci. 2015;35:1530–1538.
    1. Pariante CM. The glucocorticoid receptor: part of the solution or part of the problem? J Psychopharmacol. 2006;20:79–84.
    1. Quan N, et al. Molecular mechanisms of glucocorticoid resistance in splenocytes of socially stressed male mice. J Neuroimmunol. 2003;137:51–58.
    1. Kollet O, et al. Physiologic corticosterone oscillations regulate murine hematopoietic stem/progenitor cell proliferation and CXCL12 expression by bone marrow stromal progenitors. Leukemia. 2013;27:2006–2015.
    1. Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murine blood-brain barrier. Neurosci Lett. 1994;179:53–56.
    1. Banks WA, Kastin AJ, Broadwell RD. Passage of cytokines across the blood- brain barrier. Neuroimmunomodulation. 1995;2:241–248.
    1. Gadient RA, Otten UH. Interleukin-6 (IL-6)--a molecule with both beneficial and destructive potentials. Prog Neurobiol. 1997;52:379–390.
    1. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–964.
    1. Reader BF, et al. Peripheral and central effects of repeated social defeat stress: monocyte trafficking, microglial activation, and anxiety. Neuroscience. 2015;289:429–442.
    1. Gudmundsson P, et al. The relationship between cerebrospinal fluid biomarkers and depression in elderly women. Am J Geriatr Psychiatry. 2007;15:832–838.
    1. Torres-Platas SG, Cruceanu C, Chen GG, Turecki G, Mechawar N. Evidence for increased microglial priming and macrophage recruitment in the dorsal anterior cingulate white matter of depressed suicides. Brain Behav Immun. 2014;42:50–59.
    1. Varvel NH, et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc Natl Acad Sci U S A. 2012;109:18150–18155.
    1. Diniz BS, Butters MA, Albert SM, Dew MA, Reynolds CF., 3rd Late-life depression and risk of vascular dementia and Alzheimer's disease: systematic review and meta-analysis of community-based cohort studies. Br J Psychiatry. 2013;202:329–335.
    1. El Khoury J, et al. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med. 2007;13:432–438.
    1. Wang DD, Bordey A. The astrocyte odyssey. Prog Neurobiol. 2008;86:342–367.
    1. Meeuwsen S, Persoon-Deen C, Bsibsi M, Ravid R, van Noort JM. Cytokine, chemokine and growth factor gene profiling of cultured human astrocytes after exposure to proinflammatory stimuli. Glia. 2003;43:243–253.
    1. Pang Y, Cai Z, Rhodes PG. Analysis of genes differentially expressed in astrocytes stimulated with lipopolysaccharide using cDNA arrays. Brain Res. 2001;914:15–22.
    1. Nagy C, et al. Astrocytic abnormalities and global DNA methylation patterns in depression and suicide. Mol Psychiatry. 2014
    1. Leventopoulos M, et al. Long-term effects of early life deprivation on brain glia in Fischer rats. Brain Res. 2007;1142:119–126.
    1. Tynan RJ, et al. Chronic stress-induced disruption of the astrocyte network is driven by structural atrophy and not loss of astrocytes. Acta Neuropathol. 2013;126:75–91.
    1. Steiner J, et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission? J Neuroinflammation. 2011;8:94.
    1. Steiner J, et al. Immunological aspects in the neurobiology of suicide: elevated microglial density in schizophrenia and depression is associated with suicide. J Psychiatr Res. 2008;42:151–157.
    1. Setiawan E, et al. Role of Translocator Protein Density, a Marker of Neuroinflammation, in the Brain During Major Depressive Episodes. JAMA Psychiatry. 2015
    1. Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav Immun. 2007;21:47–59.
    1. Wohleb ES, et al. beta-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J Neurosci. 2011;31:6277–6288.
    1. Frank MG, Miguel ZD, Watkins LR, Maier SF. Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide. Brain Behav Immun. 2010;24:19–30.
    1. Meagher MW, et al. Interleukin-6 as a mechanism for the adverse effects of social stress on acute Theiler's virus infection. Brain Behav Immun. 2007;21:1083–1095.
    1. Koo JW, Russo SJ, Ferguson D, Nestler EJ, Duman RS. Nuclear factor-kappaB is a critical mediator of stress-impaired neurogenesis and depressive behavior. Proc Natl Acad Sci U S A. 2010;107:2669–2674.
    1. Koo JW, Duman RS. IL-1beta is an essential mediator of the antineurogenic and anhedonic effects of stress. Proc Natl Acad Sci U S A. 2008;105:751–756.
    1. Monje FJ, et al. Constant Darkness Induces IL-6-Dependent Depression-Like Behavior through the NF-{kappa}B Signaling Pathway. J Neurosci. 2011;31:9075–9083.
    1. Dantzer R, O'Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:46–56.
    1. Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new '5-HT' hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3- dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:702–721.
    1. Garcia-Oscos F, et al. The stress-induced cytokine interleukin-6 decreases the inhibition/excitation ratio in the rat temporal cortex via trans-signaling. Biol Psychiatry. 2012;71:574–582.
    1. Beattie EC, et al. Control of synaptic strength by glial TNF alpha. Science. 2002;295:2282–2285.
    1. Rolls A, et al. Toll-like receptors modulate adult hippocampal neurogenesis. Nat Cell Biol. 2007;9:1081–1088.
    1. Santarelli L, et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003;301:805–809.
    1. Kettenmann H, Kirchhoff F, Verkhratsky A. Microglia: new roles for the synaptic stripper. Neuron. 2013;77:10–18.
    1. Hawkes CA, McLaurin J. Selective targeting of perivascular macrophages for clearance of beta-amyloid in cerebral amyloid angiopathy. Proc Natl Acad Sci U S A. 2009;106:1261–1266.
    1. Leinenga G, Gotz J. Scanning ultrasound removes amyloid-beta and restores memory in an Alzheimer's disease mouse model. Sci Transl Med. 2015;7:278ra233.
    1. Chen SK, et al. Hematopoietic origin of pathological grooming in Hoxb8 mutant mice. Cell. 2010;141:775–785.
    1. Lin S, et al. Comparison of the transcriptional landscapes between human and mouse tissues. Proc Natl Acad Sci U S A. 2014;111:17224–17229.
    1. Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172:2731–2738.
    1. Maes M, et al. The effects of psychological stress on humans: increased production of pro-inflammatory cytokines and a Th1-like response in stress-induced anxiety. Cytokine. 1998;10:313–318.
    1. Bluthe RM, Dantzer R, Kelley KW. Effects of interleukin-1 receptor antagonist on the behavioral effects of lipopolysaccharide in rat. Brain Res. 1992;573:318–320.
    1. Willner P. Chronic mild stress (CMS) revisited: consistency and behavioural-neurobiological concordance in the effects of CMS. Neuropsychobiology. 2005;52:90–110.
    1. Dalla C, Pitychoutis PM, Kokras N, Papadopoulou-Daifoti Z. Sex differences in animal models of depression and antidepressant response. Basic Clin Pharmacol Toxicol. 2010;106:226–233.
    1. Krishnan V, Berton O, Nestler E. The use of animal models in psychiatric research and treatment. Am J Psychiatry. 2008;165:1109.
    1. Yang C, Shirayama Y, Zhang JC, Ren Q, Hashimoto K. Peripheral interleukin-6 promotes resilience versus susceptibility to inescapable electric stress. Acta Neuropsychiatr. 2015:1–5.
    1. Slattery DA, Cryan JF. The ups and downs of modelling mood disorders in rodents. ILAR J. 2014;55:297–309.
    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. Berton O, et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science. 2006;311:864–868.

Source: PubMed

3
Suscribir