Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-gtxcr Total loading time: 0 Render date: 2024-04-24T03:10:22.327Z Has data issue: false hasContentIssue false

Chapter 15 - Transdiagnostic Features of the Immune System in Major Depressive Disorder, Bipolar Disorder and Schizophrenia

Published online by Cambridge University Press:  02 September 2021

Golam Khandaker
Affiliation:
University of Cambridge
Neil Harrison
Affiliation:
Cardiff University Brain Research Imaging Centre (CUBRIC)
Edward Bullmore
Affiliation:
University of Cambridge
Robert Dantzer
Affiliation:
University of Texas, MD Anderson Cancer Center
Get access

Summary

Defining current psychopathology and optimum treatment selection in psychiatry relies solely on clinical symptom assessment but does not consider underlying biological correlates of psychiatric disorders. In particular, dysregulation of neurotransmitter metabolism and function (1,2), neuroendocrine pathways (3,4) and brain plasticity (5, 6) have been consistently reported across psychiatric disorders. Growing evidence points to a significant role of chronic low-grade inflammation in the pathophysiology of neuropsychiatric disorders such as major depressive disorder (MDD), schizophrenia (SCZ) and bipolar disorder (BD) (7–10). This features immune system dysfunctions including alterations in immune cell regulation (10), complement system (11–14), cytokine (15–18) and chemokine (7,8) pathways. As a result, current and conventional therapeutic strategies are not optimally positioned, with low remission and high relapse rates amongst individuals and treatment resistant numbers being high. Hence, the immune system is becoming a suitable target in personalizing treatment of psychiatric disorders. Unclear as yet is whether, unique immunological signatures exist for different psychiatric disorders including MDD, SCZ and BD and how these originate. Clearly a better characterization of the underlying biological aetiology will lead towards a more personalized and targeted approach.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Schur, RR, Draisma, LW, Wijnen, JP, et al. Brain GABA levels across psychiatric disorders: a systematic literature review and meta-analysis of (1) H-MRS studies. Hum Brain Mapp. 2016;37(9):3337–52.Google Scholar
Allen, PJ. Creatine metabolism and psychiatric disorders: does creatine supplementation have therapeutic value? Neurosci Biobehav Rev. 2012;36(5):1442–62.CrossRefGoogle ScholarPubMed
Landgraf, D, McCarthy, MJ, Welsh, DK. Circadian clock and stress interactions in the molecular biology of psychiatric disorders. Curr Psychiatry Rep. 2014;16(10):483.Google Scholar
McEwen, BS. Protection and damage from acute and chronic stress: allostasis and allostatic overload and relevance to the pathophysiology of psychiatric disorders. Ann N Y Acad Sci. 2004;1032:17.Google Scholar
Andero, R, Choi, DC, Ressler, KJ. BDNF-TrkB receptor regulation of distributed adult neural plasticity, memory formation, and psychiatric disorders. Prog Mol Biol Transl Sci. 2014;122:169–92.CrossRefGoogle ScholarPubMed
Ding, Y, Chang, LC, Wang, X, et al. Molecular and genetic characterization of depression: overlap with other psychiatric disorders and aging. Mol Neuropsychiatry. 2015;1(1):112.Google Scholar
Eyre, HA, Air, T, Pradhan, A, et al. A meta-analysis of chemokines in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016;68:18.Google Scholar
Stuart, MJ, Baune, BT. Chemokines and chemokine receptors in mood disorders, schizophrenia, and cognitive impairment: a systematic review of biomarker studies. Neurosci Biobehav Rev. 2014;42:93115.CrossRefGoogle ScholarPubMed
Stuart, MJ, Singhal, G, Baune, BT. Systematic review of the neurobiological relevance of chemokines to psychiatric disorders. Front Cell Neurosci. 2015;9:357.Google Scholar
Mazza, MG, Lucchi, S, Tringali, AGM, et al. Neutrophil/lymphocyte ratio and platelet/lymphocyte ratio in mood disorders: a meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2018;84(Pt A):229–36.Google Scholar
Ishii, T, Hattori, K, Miyakawa, T, et al. Increased cerebrospinal fluid complement C5 levels in major depressive disorder and schizophrenia. Biochem Biophys Res Commun. 2018;497(2):683–8.Google Scholar
Mayilyan, KR, Weinberger, DR, Sim, RB. The complement system in schizophrenia. Drug News Perspect. 2008;21(4):200–10.Google Scholar
Rus, H, Cudrici, C, David, S, Niculescu, F. The complement system in central nervous system diseases. Autoimmunity. 2006;39(5):395402.CrossRefGoogle ScholarPubMed
Ratajczak, MZ, Pedziwiatr, D, Cymer, M, et al. Sterile Inflammation of Brain, due to Activation of Innate Immunity, as a Culprit in Psychiatric Disorders. Front Psychiatry. 2018;9:60.Google Scholar
Sayana, P, Colpo, GD, Simoes, LR, et al. A systematic review of evidence for the role of inflammatory biomarkers in bipolar patients. J Psychiatr Res. 2017;92:160–82.CrossRefGoogle ScholarPubMed
Capuron, L, Lasselin, J, Castanon, N. Role of adiposity-driven inflammation in depressive morbidity. Neuropsychopharmacology. 2017;42(1):115–28.Google Scholar
Rodrigues-Amorim, D, Rivera-Baltanas, T, Spuch, C, et al. Cytokines dysregulation in schizophrenia: A systematic review of psychoneuroimmune relationship. Schizophr Res. 2018;197:1933.Google Scholar
Suvisaari, J, Mantere, O. Inflammation theories in psychotic disorders: a critical review. Infect Disord Drug Targets. 2013;13(1):5970.CrossRefGoogle ScholarPubMed
Herkenham, M, Kigar, SL. Contributions of the adaptive immune system to mood regulation: mechanisms and pathways of neuroimmune interactions. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2017;79:4957.Google Scholar
Haapakoski, R, Ebmeier, KP, Alenius, H, Kivimaki, M. Innate and adaptive immunity in the development of depression: an update on current knowledge and technological advances. Prog Neuropsychopharmacol Biol Psychiatry. 2016;66:6372.Google Scholar
Kendler, KS, Karkowski, LM, Prescott, CA. Causal relationship between stressful life events and the onset of major depression. Am J Psychiatry. 1999;156(6):837–41.Google Scholar
Shastri, A, Bonifati, DM, Kishore, U. Innate immunity and neuroinflammation. Mediators Inflamm. 2013;2013:342931.Google Scholar
Merad, M, Sathe, P, Helft, J, Miller, J, Mortha, A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013;31:563604.Google Scholar
Tian, L, Ma, L, Kaarela, T, Li, Z. Neuroimmune crosstalk in the central nervous system and its significance for neurological diseases. J Neuroinflammation. 2012;9:155.Google Scholar
Aspelund, A, Antila, S, Proulx, ST, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. The Journal of Experimental Medicine. 2015;212(7):991–9.Google Scholar
Louveau, A, Smirnov, I, Keyes, TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337–41.Google Scholar
Wohleb, ES, McKim, DB, Shea, DT, et al. Re-establishment of anxiety in stress-sensitized mice is caused by monocyte trafficking from the spleen to the brain. Biol Psychiatry. 2014;75(12):970–81.Google Scholar
Sawicki, CM, McKim, DB, Wohleb, ES, 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. 2015;302:151–64.Google Scholar
Seidel, A, Arolt, V, Hunstiger, M, et al. Major depressive disorder is associated with elevated monocyte counts. Acta Psychiatr Scand. 1996;94(3):198204.Google Scholar
Maes, M, Van der Planken, M, Stevens, WJ, et al. Leukocytosis, monocytosis and neutrophilia: hallmarks of severe depression. J Psychiatr Res. 1992;26(2):125–34.Google Scholar
Rothermundt, M, Arolt, V, Weitzsch, C, Eckhoff, D, Kirchner, H. Immunological dysfunction in schizophrenia: a systematic approach. Neuropsychobiology. 1998;37(4):186–93.CrossRefGoogle ScholarPubMed
Zorrilla, EP, Cannon, TD, Gur, RE, Kessler, J. Leukocytes and organ-nonspecific autoantibodies in schizophrenics and their siblings: markers of vulnerability or disease? Biol Psychiatry. 1996;40(9):825–33.Google Scholar
Theodoropoulou, S, Spanakos, G, Baxevanis, CN, et al. Cytokine serum levels, autologous mixed lymphocyte reaction and surface marker analysis in never medicated and chronically medicated schizophrenic patients. Schizophr Res. 2001;47(1):1325.Google Scholar
Drexhage, RC, Hoogenboezem, TA, Cohen, D, et al. An activated set point of T-cell and monocyte inflammatory networks in recent-onset schizophrenia patients involves both pro- and anti-inflammatory forces. Int J Neuropsychopharmacol. 2011;14(6):746–55.Google Scholar
Nikkila, HV, Muller, K, Ahokas, A, et al. Accumulation of macrophages in the CSF of schizophrenic patients during acute psychotic episodes. Am J Psychiatry. 1999;156(11):1725–9.Google Scholar
Drexhage, RC, Hoogenboezem, TH, Versnel, MA, et al. The activation of monocyte and T cell networks in patients with bipolar disorder. Brain Behav Immun. 2011;25(6):1206–13.Google Scholar
Drexhage, RC, Knijff, EM, Padmos, RC, et al. The mononuclear phagocyte system and its cytokine inflammatory networks in schizophrenia and bipolar disorder. Expert Rev Neurother. 2010;10(1):5976.Google Scholar
Grosse, L, Carvalho, LA, Wijkhuijs, AJ, et al. Clinical characteristics of inflammation-associated depression: monocyte gene expression is age-related in major depressive disorder. Brain Behav Immun. 2015;44:4856.Google Scholar
Padmos, RC, Hillegers, MH, Knijff, EM, et al. A discriminating messenger RNA signature for bipolar disorder formed by an aberrant expression of inflammatory genes in monocytes. Arch Gen Psychiatry. 2008;65(4):395407.Google Scholar
Zorrilla, EP, Luborsky, L, McKay, JR, et al. The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav Immun. 2001;15(3):199226.Google Scholar
Miller, BJ, Gassama, B, Sebastian, D, Buckley, P, Mellor, A. Meta-analysis of lymphocytes in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2013;73(10):993–9.Google Scholar
Torres, KCL, Souza, BR, Miranda, DM, et al. The leukocytes expressing DARPP-32 are reduced in patients with schizophrenia and bipolar disorder. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2009;33(2):214–9.Google Scholar
Sperner-Unterweger, B, Whitworth, A, Kemmler, G, et al. T-cell subsets in schizophrenia: a comparison between drug-naive first episode patients and chronic schizophrenic patients. Schizophr Res. 1999;38(1):6170.Google Scholar
Hunter, JE, Schmidt, FL. Fixed effects vs. random effects meta-analysis models: implications for cumulative research knowledge. International Journal of Selection and Assessment. 2000;8(4):275–92.Google Scholar
Blume, J, Douglas, SD, Evans, DL. Immune suppression and immune activation in depression.. 2011;25(2):221–9.Google Scholar
Seidel, A, Arolt, V, Hunstiger, M, et al. Increased CD56+ natural killer cells and related cytokines in major depression. Clin Immunol Immunopathol. 1996;78(1):83–5.Google Scholar
Ravindran, AV, Griffiths, J, Merali, Z, Anisman, H. Circulating lymphocyte subsets in major depression and dysthymia with typical or atypical features. Psychosom Med. 1998;60(3):283–9.CrossRefGoogle ScholarPubMed
Yovel, G, Sirota, P, Mazeh, D, et al. Higher natural killer cell activity in schizophrenic patients: the impact of serum factors, medication, and smoking. Brain Behav Immun. 2000;14(3):153–69.Google Scholar
Karpinski, P, Frydecka, D, Sasiadek, MM, Misiak, B. Reduced number of peripheral natural killer cells in schizophrenia but not in bipolar disorder. Brain Behav Immun. 2016;54:194200.Google Scholar
Rolle, A, Pollmann, J, Cerwenka, A. Memory of infections: an emerging role for natural killer cells. PLoS Pathog. 2013;9(9):e1003548.Google Scholar
Toben, C, Baune, BT. An act of balance between adaptive and maladaptive immunity in depression: a role for T lymphocytes. J Neuroimmune Pharmacol. 2015;10(4):595609.Google Scholar
Maes, M, Stevens, WJ, Declerck, LS, et al. Significantly increased expression of T-cell activation markers (interleukin-2 and HLA-DR) in depression: further evidence for an inflammatory process during that illness. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 1993;17(2):241–55.CrossRefGoogle ScholarPubMed
Miller, AH. Depression and immunity: a role for T cells? Brain Behav Immun. 2010;24(1):18.Google Scholar
Pavon, L, Sandoval-Lopez, G, Eugenia Hernandez, M, et al. Th2 cytokine response in major depressive disorder patients before treatment. Journal of Neuroimmunology. 2006;172(1–2):156–65.Google Scholar
Darko, DF, Gillin, JC, Risch, SC, et al. Mitogen-stimulated lymphocyte proliferation and pituitary hormones in major depression. Biol Psychiatry. 1989;26(2):145–55.Google Scholar
Rothermundt, M, Arolt, V, Fenker, J, et al. Different immune patterns in melancholic and non-melancholic major depression. Eur Arch Psychiatry Clin Neurosci. 2001;251(2):90–7.Google Scholar
Seidel, A, Arolt, V, Hunstiger, M, et al. Major depressive disorder is associated with elevated monocyte counts. Acta Psychiatrica Scandinavica. 1996;94(3):198204.CrossRefGoogle ScholarPubMed
Maes, M, Stevens, W, DeClerck, L, et al. Immune disorders in depression: higher T helper/T suppressor-cytotoxic cell ratio. Acta Psychiatr Scand. 1992;86(6):423–31.Google Scholar
Nikkila, HV, Muller, K, Ahokas, A, Rimon, R, Andersson, LC. Increased frequency of activated lymphocytes in the cerebrospinal fluid of patients with acute schizophrenia. Schizophr Res. 2001;49(1–2):99105.CrossRefGoogle ScholarPubMed
Barbosa, IG, Rocha, NP, Assis, F, et al. Monocyte and lymphocyte activation in bipolar disorder: a new piece in the puzzle of immune dysfunction in mood disorders. Int J Neuropsychopharmacol. 2014;18(1):pyu021.Google Scholar
Breunis, MN, Kupka, RW, Nolen, WA, et al. High numbers of circulating activated T cells and raised levels of serum IL-2 receptor in bipolar disorder. Biol Psychiatry. 2003;53(2):157–65.Google Scholar
Becking, K, Haarman, BCM, Grosse, L, et al. The circulating levels of CD4+ t helper cells are higher in bipolar disorder as compared to major depressive disorder. Journal of Neuroimmunology. 2018;319:2836.Google Scholar
Wu, W, Zheng, YL, Tian, LP, et al. Circulating T lymphocyte subsets, cytokines, and immune checkpoint inhibitors in patients with bipolar II or major depression: a preliminary study. Sci Rep. 2017;7:40530.CrossRefGoogle ScholarPubMed
Hickie, I, Hickie, C, Bennett, B, et al. Biochemical correlates of in vivo cell-mediated immune dysfunction in patients with depression: a preliminary report. International Journal of Immunopharmacology. 1995;17(8):685–90.Google Scholar
Pietruczuk, K, Lisowska, KA, Grabowski, K, Landowski, J, Witkowski, JM. Proliferation and apoptosis of T lymphocytes in patients with bipolar disorder. Sci Rep. 2018;8(1):3327.Google Scholar
Frydecka, D, Beszlej, A, Karabon, L, et al. The role of genetic variations of immune system regulatory molecules CD28 and CTLA-4 in schizophrenia. Psychiatry Res. 2013;208(2):197–8.Google Scholar
Jun, TY, Pae, CU, Chae, JH, Bahk, WM, Kim, KS. Polymorphism of CTLA-4 gene for major depression in the Korean population. Psychiatry Clin Neurosci. 2001;55(5):533–7.Google Scholar
Grosse, L, Hoogenboezem, T, Ambrée, O, et al. Deficiencies of the T and natural killer cell system in major depressive disorder: T regulatory cell defects are associated with inflammatory monocyte activation. Brain, Behavior, and Immunity. 2016;54:3844.Google Scholar
Mikova, O, Yakimova, R, Bosmans, E, Kenis, G, Maes, M. Increased serum tumor necrosis factor alpha concentrations in major depression and multiple sclerosis. Eur Neuropsychopharmacol. 2001;11(3):203–8.Google Scholar
do Prado, CH, Rizzo, LB, Wieck, A, et al. Reduced regulatory T cells are associated with higher levels of Th1/TH17 cytokines and activated MAPK in type 1 bipolar disorder. Psychoneuroendocrinology. 2013;38(5):667–76.Google Scholar
Najjar, S, Pearlman, DM, Alper, K, Najjar, A, Devinsky, O. Neuroinflammation and psychiatric illness. J Neuroinflammation. 2013;10:43.Google Scholar
Robertson, MJ, Schacterle, RS, Mackin, GA, et al. Lymphocyte subset differences in patients with chronic fatigue syndrome, multiple sclerosis and major depression. Clin Exp Immunol. 2005;141(2):326–32.Google Scholar
Steiner, J, Jacobs, R, Panteli, B, et al. Acute schizophrenia is accompanied by reduced T cell and increased B cell immunity. Eur Arch Psychiatry Clin Neurosci. 2010;260(7):509–18.Google Scholar
Schwarz, MJ, Chiang, S, Muller, N, Ackenheil, M. T-helper-1 and T-helper-2 responses in psychiatric disorders. Brain Behav Immun. 2001;15(4):340–70.CrossRefGoogle ScholarPubMed
Schwarz, MJ, Muller, N, Riedel, M, Ackenheil, M. The Th2-hypothesis of schizophrenia: a strategy to identify a subgroup of schizophrenia caused by immune mechanisms. Med Hypotheses. 2001;56(4):483–6.Google Scholar
Debnath, M, Berk, M. Th17 pathway-mediated immunopathogenesis of schizophrenia: mechanisms and implications. Schizophr Bull. 2014;40(6):1412–21.Google Scholar
Slyepchenko, A, Maes, M, Köhler, C, et al. T helper 17 cells may drive neuroprogression in major depressive disorder: proposal of an integrative model. Neuroscience & Biobehavioral Reviews. 2016;64:83100.CrossRefGoogle ScholarPubMed
Beurel, E, Lowell, JA. Th17 cells in depression. Brain, Behavior, and Immunity. 2018;69:2834.Google Scholar
Poletti, S, de Wit, H, Mazza, E, et al. Th17 cells correlate positively to the structural and functional integrity of the brain in bipolar depression and healthy controls. Brain, Behavior, and Immunity. 2017;61:317–25.Google Scholar
Vogels, RJ, Koenders, MA, van Rossum, EF, Spijker, AT, Drexhage, HA. T cell deficits and overexpression of hepatocyte growth factor in anti-inflammatory circulating monocytes of middle-aged patients with bipolar disorder characterized by a high prevalence of the metabolic syndrome. Front Psychiatry. 2017;8:34.CrossRefGoogle ScholarPubMed
Walport, MJ. Complement. Second of two parts. N Engl J Med. 2001;344(15):1140–4.Google Scholar
Walport, MJ. Complement. First of two parts. N Engl J Med. 2001;344(14):1058–66.Google Scholar
Stephan, AH, Barres, BA, Stevens, B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89.Google Scholar
Crider, A, Feng, T, Pandya, CD, et al. Complement component 3a receptor deficiency attenuates chronic stress-induced monocyte infiltration and depressive-like behavior. Brain Behav Immun. 2018;70:246–56.Google Scholar
Wei, J, Liu, Y, Zhao, L, et al. Plasma complement component 4 increases in patients with major depressive disorder. Neuropsychiatr Dis Treat. 2018;14:3741.Google Scholar
Zhang, C, Zhang, DF, Wu, ZG, et al. Complement factor H and susceptibility to major depressive disorder in Han Chinese. Br J Psychiatry. 2016;208(5):446–52.Google Scholar
Stelzhammer, V, Haenisch, F, Chan, MK, et al. Proteomic changes in serum of first onset, antidepressant drug-naive major depression patients. Int J Neuropsychopharmacol. 2014;17(10):1599–608.CrossRefGoogle ScholarPubMed
Morera, AL, Henry, M, Garcia-Hernandez, A, Fernandez-Lopez, L. Acute phase proteins as biological markers of negative psychopathology in paranoid schizophrenia. Actas Esp Psiquiatr. 2007;35(4):249–52.Google Scholar
Nimgaonkar, VL, Prasad, KM, Chowdari, KV, Severance, EG, Yolken, RH. The complement system: a gateway to gene-environment interactions in schizophrenia pathogenesis. Mol Psychiatry. 2017;22(11):1554–61.Google Scholar
Kucharska-Mazur, J, Jablonski, M, Misiak, B, et al. Adult stem cells in psychiatric disorders – new discoveries in peripheral blood. Prog Neuropsychopharmacol Biol Psychiatry. 2018;80(Pt A):23–7.Google Scholar
Wadee, AA, Kuschke, RH, Wood, LA, et al. Serological observations in patients suffering from acute manic episodes. Hum Psychopharmacol. 2002;17(4):175–9.Google Scholar
Maes, M, Delange, J, Ranjan, R, et al. Acute phase proteins in schizophrenia, mania and major depression: modulation by psychotropic drugs. Psychiatry Res. 1997;66(1):111.CrossRefGoogle ScholarPubMed
Akcan, U, Karabulut, S, Ismail Kucukali, C, Cakir, S, Tuzun, E. Bipolar disorder patients display reduced serum complement levels and elevated peripheral blood complement expression levels. Acta Neuropsychiatr. 2018;30(2):70–8.Google Scholar
Dantzer, R. Cytokine, sickness behavior, and depression. Immunol Allergy Clin North Am. 2009;29(2):247–64.Google Scholar
Capuron, L, Castanon, N. Role of inflammation in the development of neuropsychiatric symptom domains: evidence and mechanisms. Curr Top Behav Neurosci. 2017;31:3144.Google Scholar
Dowlati, Y, Herrmann, N, Swardfager, , et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67(5):446–57.CrossRefGoogle ScholarPubMed
Liu, Y, Ho, RC, Mak, A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. J Affect Disord. 2012;139(3):230–9.Google Scholar
Jiang, M, Qin, P, Yang, X. Comorbidity between depression and asthma via immune-inflammatory pathways: a meta-analysis. J Affect Disord. 2014;166:22–9.Google Scholar
Haapakoski, R, Mathieu, J, Ebmeier, KP, Alenius, H, Kivimaki, M. Cumulative meta-analysis of interleukins 6 and 1beta, tumour necrosis factor alpha and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.Google Scholar
Goldsmith, DR, Rapaport, MH, Miller, BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21(12):1696–709.Google Scholar
Kohler, CA, Freitas, TH, Maes, M, et al. Peripheral cytokine and chemokine alterations in depression: a meta-analysis of 82 studies. Acta Psychiatr Scand. 2017;135(5):373–87.Google Scholar
Wang, AK, Miller, BJ. Meta-analysis of cerebrospinal fluid cytokine and tryptophan catabolite alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder, and depression. Schizophr Bull. 2018;44(1):7583.Google Scholar
Valkanova, V, Ebmeier, KP, Allan, CL. CRP, IL-6 and depression: a systematic review and meta-analysis of longitudinal studies. J Affect Disord. 2013;150(3):736–44.Google Scholar
Ellul, P, Boyer, L, Groc, L, Leboyer, M, Fond, G. Interleukin-1 beta-targeted treatment strategies in inflammatory depression: toward personalized care. Acta Psychiatr Scand. 2016;134(6):469–84.Google Scholar
Potvin, S, Stip, E, Sepehry, AA, et al. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry. 2008;63(8):801–8.Google Scholar
Miller, BJ, Buckley, P, Seabolt, W, Mellor, A, Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70(7):663–71.Google Scholar
Pillinger, T, Osimo, EF, Brugger, S, et al. A meta-analysis of immune parameters, variability, and assessment of modal distribution in psychosis and test of the immune subgroup hypothesis. Schizophr Bull. 2019;45(5):1120–33.Google Scholar
Muller, N, Weidinger, E, Leitner, B, Schwarz, MJ. The role of inflammation in schizophrenia. Front Neurosci. 2015;9:372.Google Scholar
Frydecka, D, Misiak, B, Pawlak-Adamska, E, et al. Interleukin-6: the missing element of the neurocognitive deterioration in schizophrenia? The focus on genetic underpinnings, cognitive impairment and clinical manifestation. Eur Arch Psychiatry Clin Neurosci. 2015;265(6):449–59.Google Scholar
Shi, J, Levinson, DF, Duan, J, et al. Common variants on chromosome 6p22.1 are associated with schizophrenia. Nature. 2009;460(7256):753–7.Google Scholar
Lee, YH, Song, GG. Meta-analysis of associations between tumor necrosis factor-alpha polymorphisms and schizophrenia susceptibility. Psychiatry Res. 2015;226(2–3):521–2.Google Scholar
Qin, H, Zhang, L, Xu, G, Pan, X. Lack of association between TNFalpha rs1800629 polymorphism and schizophrenia risk: a meta-analysis. Psychiatry Res. 2013;209(3):314–9.Google Scholar
Sacchetti, E, Bocchio-Chiavetto, L, Valsecchi, P, et al. -G308A tumor necrosis factor alpha functional polymorphism and schizophrenia risk: meta-analysis plus association study. Brain Behav Immun. 2007;21(4):450–7.Google Scholar
Ripke, S, O’Dushlaine, C, Chambert, K, et al. Genome-wide association analysis identifies 13 new risk loci for schizophrenia. Nat Genet. 2013;45(10):1150–9.Google Scholar
Gao, L, Li, Z, Chang, S, Wang, J. Association of interleukin-10 polymorphisms with schizophrenia: a meta-analysis. PLoS One. 2014;9(3):e90407.Google Scholar
Shibuya, M, Watanabe, Y, Nunokawa, A, et al. Interleukin 1 beta gene and risk of schizophrenia: detailed case-control and family-based studies and an updated meta-analysis. Hum Psychopharmacol. 2014;29(1):31–7.CrossRefGoogle Scholar
Modabbernia, A, Taslimi, S, Brietzke, E, Ashrafi, M. Cytokine alterations in bipolar disorder: a meta-analysis of 30 studies. Biol Psychiatry. 2013;74(1):1525.Google Scholar
Munkholm, K, Brauner, JV, Kessing, LV, Vinberg, M. Cytokines in bipolar disorder vs. healthy control subjects: a systematic review and meta-analysis. J Psychiatr Res. 2013;47(9):1119–33.Google Scholar
Munkholm, K, Vinberg, M, Vedel Kessing, L. Cytokines in bipolar disorder: a systematic review and meta-analysis. J Affect Disord. 2013;144(1–2):1627.Google Scholar
Horn, SR, Long, MM, Nelson, BW, et al. Replication and reproducibility issues in the relationship between C-reactive protein and depression: a systematic review and focused meta-analysis. Brain Behav Immun. 2018;73:85114.Google Scholar
van den Ameele, S, van Diermen, L, Staels, W, et al. The effect of mood-stabilizing drugs on cytokine levels in bipolar disorder: a systematic review. J Affect Disord. 2016;203:364–73.Google Scholar
Capuzzi, E, Bartoli, F, Crocamo, C, Clerici, M, Carra, G. Acute variations of cytokine levels after antipsychotic treatment in drug-naive subjects with a first-episode psychosis: a meta-analysis. Neurosci Biobehav Rev. 2017;77:122–8.Google Scholar
Kohler, CA, Freitas, TH, Stubbs, B, et al. Peripheral alterations in cytokine and chemokine levels after antidepressant drug treatment for major depressive disorder: systematic review and meta-analysis. Mol Neurobiol. 2018;55(5):4195–206.Google Scholar
Ono, SJ, Nakamura, T, Miyazaki, D, et al. Chemokines: roles in leukocyte development, trafficking, and effector function. J Allergy Clin Immunol. 2003 Jun;111(6):1185–99.Google Scholar
Le, Y, Zhou, Y, Iribarren, P, Wang, J. Chemokines and chemokine receptors: their manifold roles in homeostasis and disease. Cell Mol Immunol. 2004;1(2):95104.Google Scholar
Jo, WK, Law, AC, Chung, SK. The neglected co-star in the dementia drama: the putative roles of astrocytes in the pathogeneses of major neurocognitive disorders. Mol Psychiatry. 2014;19(2):159–67.Google Scholar
Rostene, W, Buckingham, JC. Chemokines as modulators of neuroendocrine functions. J Mol Endocrinol. 2007;38(3):351–3.Google Scholar
Rostene, W, Kitabgi, P, Parsadaniantz, SM. Chemokines: a new class of neuromodulator? Nat Rev Neurosci. 2007;8(11):895903.Google Scholar
Bergon, A, Belzeaux, R, Comte, M, et al. CX3CR1 is dysregulated in blood and brain from schizophrenia patients. Schizophr Res. 2015;168(1–2):434–43.Google Scholar
Zakharyan, R, Boyajyan, A. Inflammatory cytokine network in schizophrenia. World J Biol Psychiatry. 2014;15(3):174–87.Google Scholar
Barbosa, IG, Rocha, NP, Bauer, ME, et al. Chemokines in bipolar disorder: trait or state? Eur Arch Psychiatry Clin Neurosci. 2013;263(2):159–65.Google Scholar
Panizzutti, B, Gubert, C, Schuh, AL, et al. Increased serum levels of eotaxin/CCL11 in late-stage patients with bipolar disorder: an accelerated aging biomarker? J Affect Disord. 2015;182:64–9.CrossRefGoogle ScholarPubMed
Tokac, D, Tuzun, E, Gulec, H, et al. Chemokine and chemokine receptor polymorphisms in bipolar disorder. Psychiatry Investig. 2016;13(5):541–8.Google Scholar
Altamura, AC, Mundo, E, Cattaneo, E, et al. The MCP-1 gene (SCYA2) and mood disorders: preliminary results of a case-control association study. Neuroimmunomodulation. 2010;17(2):126–31.Google Scholar
Pae, CU, Kim, JJ, Yu, HS, et al. Monocyte chemoattractant protein-1 promoter -2518 polymorphism may have an influence on clinical heterogeneity of bipolar I disorder in the Korean population. Neuropsychobiology. 2004;49(3):111–4.Google Scholar
Rosenblat, JD, Cha, DS, Mansur, RB, McIntyre, RS. Inflamed moods: a review of the interactions between inflammation and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2014;53:2334.Google Scholar
Robinson, MW, Harmon, C, O’Farrelly, C. Liver immunology and its role in inflammation and homeostasis. Cell Mol Immunol. 2016;13(3):267–76.Google Scholar
Niciu, MJ, Ionescu, DF, Mathews, DC, Richards, EM, Zarate, CA, Jr. Second messenger/signal transduction pathways in major mood disorders: moving from membrane to mechanism of action, part I: major depressive disorder. CNS Spectr. 2013;18(5):231–41.Google Scholar
Niciu, MJ, Ionescu, DF, Mathews, DC, Richards, EM, Zarate, CA, Jr. Second messenger/signal transduction pathways in major mood disorders: moving from membrane to mechanism of action, part II: bipolar disorder. CNS Spectr. 2013;18(5):242–51.Google ScholarPubMed
Miklowitz, DJ, Portnoff, LC, Armstrong, CC, et al. Inflammatory cytokines and nuclear factor-kappa B activation in adolescents with bipolar and major depressive disorders. Psychiatry Res. 2016;241:315–22.Google Scholar
Altamura, AC, Buoli, M, Pozzoli, S. Role of immunological factors in the pathophysiology and diagnosis of bipolar disorder: comparison with schizophrenia. Psychiatry Clin Neurosci. 2014;68(1):2136.Google Scholar
Muller, N, Schwarz, MJ. Immune system and schizophrenia. Curr Immunol Rev. 2010;6(3):213–20.Google Scholar
Kubera, M, Basta-Kaim, A, Wrobel, A, Maes, M, Dudek, D. Increased mitogen-induced lymphocyte proliferation in treatment resistant depression: a preliminary study. Neuro Endocrinol Lett. 2004;25(3):207–10.Google Scholar
Muller, N, Schwarz, MJ. The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Mol Psychiatry. 2007;12(11):9881000.Google Scholar
Al-Amin, MM, Nasir Uddin, MM, Mahmud Reza, H. Effects of antipsychotics on the inflammatory response system of patients with schizophrenia in peripheral blood mononuclear cell cultures. Clin Psychopharmacol Neurosci. 2013;11(3):144–51.Google Scholar
Nazimek, K, Strobel, S, Bryniarski, P, et al. The role of macrophages in anti-inflammatory activity of antidepressant drugs. Immunobiology. 2017;222(6):823–30.Google Scholar
Brietzke, E, Kauer-Sant’Anna, M, Teixeira, AL, Kapczinski, F. Abnormalities in serum chemokine levels in euthymic patients with bipolar disorder. Brain Behav Immun. 2009;23(8):1079–82.Google Scholar
Miller, AH, Raison, CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16(1):2234.Google Scholar
Fillman, SG, Sinclair, D, Fung, SJ, Webster, MJ, Shannon Weickert, C. Markers of inflammation and stress distinguish subsets of individuals with schizophrenia and bipolar disorder. Transl Psychiatry. 2014;4:e365.Google Scholar
Belvederi Murri, M, Prestia, D, Mondelli, V, et al. The HPA axis in bipolar disorder: Systematic review and meta-analysis. Psychoneuroendocrinology. 2016;63:327–42.Google Scholar
Jacobson, L. Hypothalamic-pituitary-adrenocortical axis: neuropsychiatric aspects. Compr Physiol. 2014;4(2):715–38.Google Scholar
Maes, M, Carvalho, AF. The compensatory immune-regulatory reflex system (CIRS) in depression and bipolar disorder. Molecular Neurobiology. 2018;55(12):8885–903.Google Scholar
Jara, LJ, Navarro, C, Medina, G, Vera-Lastra, O, Blanco, F. Immune-neuroendocrine interactions and autoimmune diseases. Clin Dev Immunol. 2006;13(2–4):109–23.Google Scholar
Chen, Y, Jiang, T, Chen, P, et al. Emerging tendency towards autoimmune process in major depressive patients: a novel insight from Th17 cells. Psychiatry Research. 2011;188(2):224–30.Google Scholar
Steiner, J, Bielau, H, Brisch, R, 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(2):151–7.Google Scholar
Setiawan, E, Wilson, AA, Mizrahi, R, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry. 2015;72(3):268–75.Google Scholar
Wohleb, ES, Franklin, T, Iwata, M, Duman, RS. Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci. 2016;17(8):497511.Google Scholar
Szepesi, Z, Manouchehrian, O, Bachiller, S, Deierborg, T. Bidirectional microglia-neuron communication in health and disease. Front Cell Neurosci. 2018;12:323.Google Scholar
Neniskyte, U, Gross, CT. Errant gardeners: glial-cell-dependent synaptic pruning and neurodevelopmental disorders. Nat Rev Neurosci. 2017;18(11):658–70.Google Scholar
Kempton, MJ, Salvador, Z, Munafo, MR, et al. Structural neuroimaging studies in major depressive disorder. Meta-analysis and comparison with bipolar disorder. Arch Gen Psychiatry. 2011;68(7):675–90.Google Scholar
van Erp, TG, Hibar, DP, Rasmussen, JM, et al. Subcortical brain volume abnormalities in 2028 individuals with schizophrenia and 2540 healthy controls via the ENIGMA consortium. Mol Psychiatry. 2016;21(4):585.Google Scholar
Hibar, DP, Westlye, LT, van Erp, TG, et al. Subcortical volumetric abnormalities in bipolar disorder. Mol Psychiatry. 2016;21(12):1710–6.Google Scholar
Schmaal, L, Veltman, DJ, van Erp, TG, et al. Subcortical brain alterations in major depressive disorder: findings from the ENIGMA Major Depressive Disorder working group. Mol Psychiatry. 2016;21(6):806–12.Google Scholar
Harrison, NA. Brain structures implicated in inflammation-associated depression. Curr Top Behav Neurosci. 2017;31:221–48.Google Scholar
Gemechu, JM, Bentivoglio, M. T cell recruitment in the brain during normal aging. Front Cell Neurosci. 2012;6:38.Google Scholar
Jha, MK, Miller, AH, Minhajuddin, A, Trivedi, MH. Association of T and non-T cell cytokines with anhedonia: role of gender differences. Psychoneuroendocrinology. 2018;95:17.Google Scholar
Cuthbert, BN, Insel, TR. Toward new approaches to psychotic disorders: the NIMH Research Domain Criteria project. Schizophr Bull. 2010;36(6):1061–2.Google Scholar
Sanislow, CA, Pine, DS, Quinn, KJ, et al. Developing constructs for psychopathology research: research domain criteria. J Abnorm Psychol. 2010;119(4):631–9.Google Scholar
Dooley, LN, Kuhlman, KR, Robles, TF, et al. The role of inflammation in core features of depression: Insights from paradigms using exogenously induced inflammation. Neuroscience & Biobehavioral Reviews. 2018;94:219–37.Google Scholar
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(1):4656.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×