Nervenheilkunde 2020; 39(01/02): 55-66
DOI: 10.1055/a-1037-2032
Schwerpunkt
© Georg Thieme Verlag KG Stuttgart · New York

„Old Friends“, Immunregulation und Stressresilienz

Teil 2: Mechanismen[ 1 ] Old friends, immune regulation and stress resilience
Dominik Langgartner
1   Sektion für Molekulare Psychosomatik, Klinik für Psychosomatische Medizin und Psychotherapie, Universität Ulm
,
Christopher A. Lowry
2   Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, CO, USA
3   Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
4   Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver Veterans Affairs Medical Center (VAMC), Denver, CO, USA
5   Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO, USA
,
Stefan O. Reber
1   Sektion für Molekulare Psychosomatik, Klinik für Psychosomatische Medizin und Psychotherapie, Universität Ulm
› Author Affiliations
Further Information

Publication History

Publication Date:
12 February 2020 (online)

ZUSAMMENFASSUNG

Es gibt zahlreiche Hinweise darauf, dass chronische negative Erfahrungen, insbesondere chronischer psychosozialer Stress, einen wesentlichen Risikofaktor für die Entstehung vieler somatischer und affektiver Störungen darstellen, einschließlich chronischer entzündlicher Darmerkrankungen (CED) und der posttraumatischen Belastungsstörung (PTBS). Die Mechanismen, die der Entstehung chronischer stressbedingter Erkrankungen zugrunde liegen, sind jedoch noch weitgehend unbekannt und die derzeitigen Behandlungs- und Präventionsstrategien sind unzureichend wirksam und zuverlässig. Ein besseres Verständnis der Mechanismen, die an der Entstehung und Aufrechterhaltung chronischer stressbedingter Erkrankungen beteiligt sind, kann zu neuen Ansätzen in der Prävention und Behandlung dieser Erkrankungen führen. In diesem 2-teiligen Review fassen wir die theoretischen Grundlagen (Teil 1) sowie die zugrunde liegenden Mechanismen (Teil 2) zusammen, welche die Hypothese stützen, dass ein Anstieg der Immun(re)aktivität und des Entzündungsgeschehens in der heutigen modernen Gesellschaft, potenziell gefördert durch einen verminderten Kontakt mit immunregulatorischen Mikroorganismen („Old Friends“), ursächlich an der gesteigerten Anfälligkeit für stressbedingte Pathologien sein könnte. Darüber hinaus diskutieren wir den künstlich herbeigeführten Kontakt mit Old Friends als vielversprechende Strategie zur Förderung der Stressresilienz und zur Vorbeugung/Behandlung chronischer stressbedingter Erkrankungen.

ABSTRACT

There is a considerable body of evidence indicating that chronic adverse experience, especially chronic psychosocial stress/trauma,represents a major risk factor for the development of many somatic and affective disorders including inflammatory bowel disease (IBD) and posttraumatic stress disorder (PTSD). However, the mechanisms underlying the development of chronic stress-induced disorders are still in large part unknown, and current treatment and prevention strategies lack efficacy and reliability. A greater understanding of mechanisms involved in the development and persistence of chronic stress-induced disorders may lead to novel approaches to prevention and treatment of these disorders. In this two-part review, we summarize the theoretical background (Part 1) and underlying mechanisms (Part 2) that support the hypothesis that increases in immune (re-) activity and inflammation, potentially promoted by a reduced exposure to immunoregulatory microorganisms (“Old Friends”) in today’s modern society, may be causal factors in mediating the vulnerability to development and persistence of stress-related pathologies. Moreover, we discuss strategies to increase immunoregulatory processes and attenuate inflammation, as for instance contact with immunoregulatory “Old Friends“, which appears to be a promising strategy to promote stress resilience and to prevent/treat chronic stress-related disorder

1 Bei dem vorliegenden Artikel handelt es sich um die deutsche Übersetzung des im Journal Pflügers Archiv – European Journal of Physiology erschienen Artikels Langgartner und Kollegen, 2018 [1].


 
  • Literatur

  • 1 Langgartner D. et al Old Friends, immunoregulation, and stress resilience. Pflügers Archiv – European Journal of Physiology 2018; 1: 33
  • 2 Langgartner D. et al Chronic subordinate colony housing paradigm: a mouse model to characterize the consequences of insufficient glucocorticoid signaling. Front Psychiatry 2015; 6: 18
  • 3 Mathew SJ. et al Ketamine for treatment-resistant unipolar depression: current evidence. CNS Drugs 2012; 26 (03) 189-204
  • 4 Freedman R. Abrupt withdrawal of antidepressant treatment. Am J Psychiatry 2010; 167 (08) 886-8
  • 5 Lin EH. et al The role of the primary care physician in patients’ adherence to antidepressant therapy. Med Care 1995; 33 (01) 67-74
  • 6 Andrews PW. et al Blue again: perturbational effects of antidepressants suggest monoaminergic homeostasis in major depression. Frontiers in Psychology 2011; 2
  • 7 Rush AJet al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: A STAR*D report. American Journal of Psychiatry 2006; 163 (11) 1905-1917
  • 8 Li X. et al Review of pharmacological treatment in mood disorders and future directions for drug development. Neuropsychopharmacology 2012; 37 (01) 77-101
  • 9 Papakostas GI. et al Does the probability of receiving placebo influence clinical trial outcome? A meta-regression of double-blind, randomized clinical trials in MDD. Eur Neuropsychopharmacol 2009; 19 (01) 34-40
  • 10 Inse TR. et al Cure therapeutics and strategic prevention: raising the bar for mental health research. Mol Psychiatry 2006; 11 (01) 11-7
  • 11 Dantzer R. et al Cytokines and sickness behavior. Ann N Y Acad Sci 1998; 840: 586-90
  • 12 Capuron L. et al Cytokines and depression: The need for a new paradigm. Brain, Behavior, and Immunity 2003; 17 1 (Suppl. 01) 119-124
  • 13 Dantzer R. Cytokine-induced sickness behaviour: a neuroimmune response to activation of innate immunity. European Journal of Pharmacology 2004; 500 1–3 399-411
  • 14 Dantzer R. et al From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci 2008; 9 (01) 46-56
  • 15 Hanski I. et al Environmental biodiversity, human microbiota, and allergy are interrelated. Proceedings of the National Academy of Sciences 2012; 109 (21) 8334-8339
  • 16 Blaser MJ. The theory of disappearing microbiota and the epidemics of chronic diseases. Nat Rev Immunol 2017; 17 (08) 461-463
  • 17 Lowry CA. et al The Microbiota, Immunoregulation, and Mental Health: Implications for Public Health. Curr Environ Health Rep 2016; 3 (03) 270-86
  • 18 Rook GA. et al Microbial ‘Old Friends’, immunoregulation and stress resilience. Evol Med Public Health 2013; 2013 (01) 46-64
  • 19 Rohleder N. Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom Med 2014; 76 (03) 181-9
  • 20 Li Y. et al Altered expression of CD4(+)CD25(+) regulatory T cells and its 5-HT(1a) receptor in patients with major depression disorder. J Affect Disord 2010; 124 1–2 68-75
  • 21 Sommershof A. et al Substantial reduction of naive and regulatory T cells following traumatic stress. Brain Behav Immun 2009; 23 (08) 1117-24
  • 22 Dimsdale JE. Psychological stress and cardiovascular disease. J Am Coll Cardiol 2008; 51 (13) 1237-46
  • 23 Mausbach BT. et al A longitudinal analysis of the relations among stress, depressive symptoms, leisure satisfaction, and endothelial function in caregivers. Health Psychol 2012; 31 (04) 433-40
  • 24 Unden AL. et al Cardiovascular effects of social support in the work place: twenty-four-hour ECG monitoring of men and women. Psychosom Med 1991; 53 (01) 50-60
  • 25 Buckley T. et al Prospective study of early bereavement on psychological and behavioural cardiac risk factors. Intern Med J 2009; 39 (06) 370-8
  • 26 Buckley T. et al Effect of early bereavement on heart rate and heart rate variability. Am J Cardiol 2012; 110 (09) 1378-83
  • 27 Kivimaki M. et al Job strain as a risk factor for coronary heart disease: a collaborative meta-analysis of individual participant data. Lancet 2012; 380 9852 1491-7
  • 28 Almansa C. et al Prevalence of functional gastrointestinal disorders in patients with fibromyalgia and the role of psychologic distress. Clin Gastroenterol Hepatol 2009; 7 (04) 438-45
  • 29 Wright RJ. et al Chronic caregiver stress and IgE expression, allergen-induced proliferation, and cytokine profiles in a birth cohort predisposed to atopy. J Allergy Clin Immunol 2004; 113 (06) 1051-7
  • 30 Wright RJ. et al Community violence and asthma morbidity: the Inner-City Asthma Study. Am J Public Health 2004; 94 (04) 625-32
  • 31 Buske-Kirschbaum A. et al Psychobiological aspects of atopic dermatitis: an overview. Psychother Psychosom 2001; 70 (01) 6-16
  • 32 Coker AL. et al Physical health consequences of physical and psychological intimate partner violence. Arch Fam Med 2000; 9 (05) 451-7
  • 33 Herrmann M. et al Stress and rheumatic diseases. Rheum Dis Clin North Am 2000; 26 (04) 737-63 viii
  • 34 Levenstein S. et al Stress and exacerbation in ulcerative colitis: a prospective study of patients enrolled in remission. The American Journal of Gastroenterology 2000; 95 (05) 1213-1220
  • 35 Salem SN. et al Non-specific ulcerative colitis in Bedouin Arabs. Lancet 1967; 1 7488 473-5
  • 36 Duffy LC. et al Relevance of major stress events as an indicator of disease activity prevalence in inflammatory bowel disease. Behav Med 1991; 17 (03) 101-10
  • 37 Robertson DA. et al Personality profile and affective state of patients with inflammatory bowel disease. Gut 1989; 30 (05) 623-6
  • 38 Bernstein CN. et al A prospective population-based study of triggers of symptomatic flares in IBD. Am J Gastroenterol 2010; 105 (09) 1994-2002
  • 39 Bernstein CN. New insights into IBD epidemiology: are there any lessons for treatment?. Dig Dis 2010; 28 (03) 406-10
  • 40 Mawdsley J. et al Psychological stress in IBD: new insights into pathogenic and therapeutic implications. Gut 2005; 54 (10) 1481-91
  • 41 Mawdsley JE. et al The role of psychological stress in inflammatory bowel disease. Neuroimmunomodulation 2006; 13 5–6 327-36
  • 42 Bitton A. et al Psychosocial determinants of relapse in ulcerative colitis: a longitudinal study. Am J Gastroenterol 2003; 98 (10) 2203-2208
  • 43 Campbell J. et al Intimate partner violence and physical health consequences. Arch Intern Med 2002; 162 (10) 1157-63
  • 44 Cohen S. et al Psychological stress and susceptibility to the common cold. N Engl J Med 1991; 325 (09) 606-12
  • 45 Kiecolt-Glaser JK. et al Chronic stress alters the immune response to influenza virus vaccine in older adults. Proc Natl Acad Sci U S A 1996; 93 (07) 3043-7
  • 46 Marucha PT. et al Mucosal wound healing is impaired by examination stress. Psychosom Med 1998; 60 (03) 362-5
  • 47 Kiecolt-Glaser JK. et al Psychological influences on surgical recovery. Perspectives from psychoneuroimmunology. Am Psychol 1998; 53 (11) 1209-18
  • 48 Kiecolt-Glaser JK. et al Slowing of wound healing by psychological stress. Lancet 1995; 346 8984 1194-6
  • 49 Kiecolt-Glaser JK, Glaser R. Psychoneuroimmunology and health consequences: data and shared mechanisms. Psychosom Med 1995; 57 (03) 269-74
  • 50 Levav I. et al Cancer incidence and survival following bereavement. Am J Public Health 2000; 90 (10) 1601-7
  • 51 Reiche EM. et al Stress, depression, the immune system, and cancer. Lancet Oncol 2004; 5 (10) 617-25
  • 52 Risch N. et al Interaction between the serotonin transporter gene (5-httlpr), stressful life events, and risk of depression: A meta-analysis. JAMA 2009; 301 (23) 2462-2471
  • 53 Agid O. et al Environment and vulnerability to major psychiatric illness: a case control study of early parental loss in major depression, bipolar disorder and schizophrenia. Mol Psychiatry 1999; 4 (02) 163-72
  • 54 Heim C, Nemeroff CB. The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biol Psychiatry 2001; 49 (12) 1023-39
  • 55 Holsboer F. Stress, hypercortisolism and corticosteroid receptors in depression: implicatons for therapy. Journal of Affective Disorders 2001; 62 1–2 77-91
  • 56 Virtanen M. et al Overtime work as a predictor of major depressive episode: a 5-year follow-up of the Whitehall II study. PLoS One 2012; 7 (01) e30719
  • 57 Virtanen M. et al Saved by the bell: does working too much increase the likelihood of depression?. Expert Rev Neurother 2012; 12 (05) 497-9
  • 58 Hammen C. Stress and Depression. Annual Review of Clinical Psychology 2005; 1 (01) 293-319
  • 59 de Kloet ER. et al Stress and the brain: from adaptation to disease. Nat Rev Neurosci 2005; 6 (06) 463-475
  • 60 Cryan JF. et al The ascent of mouse: advances in modelling human depression and anxiety. Nat Rev Drug Discov 2005; 4 (09) 775-90
  • 61 Cryan JF. et al Animal models of mood disorders: Recent developments. Curr Opin Psychiatry 2007; 20 (01) 1-7
  • 62 Heim C. et al Childhood trauma and risk for chronic fatigue syndrome: association with neuroendocrine dysfunction. Arch Gen Psychiatry 2009; 66 (01) 72-80
  • 63 Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med 2002; 347 (12) 911-20
  • 64 AJ W. et al Evidence for the increase in asthma worldwide. CIBA Found Symp 1997; 206: 122-34
  • 65 HC W. Is the prevalence of atopic dermatitis increasing?. Clinical and Experimental Dermatology 1992; 17 (06) 385-391
  • 66 Upton MN. et al Intergenerational 20 year trends in the prevalence of asthma and hay fever in adults: the Midspan family study surveys of parents and offspring. BMJ 2000; 321 7253 88-92
  • 67 Ret G al. Incidence of multiple sclerosis in the town of Sassari, Sardinia, 1965 to 1985: evidence for increasing occurrence of the disease. Neurology 1988; 38: 384-8
  • 68 Poser S. et al Increasing Incidence of Multiple Sclerosis in South Lower Saxony, Germany. Neuroepidemiology 1989; 8 (04) 207-213
  • 69 Group EAS. Variation and trends in incidence of childhood diabetes in Europe. The Lancet 2000; 355 9207 873-876
  • 70 F.F A critical review of epidemiological studies in inflammatory bowel disease. Scand J Gastroenterol 2001; 36 (01) 2-15
  • 71 Ng SC. et al Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 2018; 390 10114 2769-2778
  • 72 Xu F. et al Health-Risk Behaviors and Chronic Conditions Among Adults with Inflammatory Bowel Disease — United States, 2015 and 2016. MMWR Morb Mortal Wkly Rep 2018; 67: 190-195
  • 73 Murray CJL. et al Evidence-Based Health Policy – Lessons from the Global Burden of Disease Study. Science 1996; 274 5288 740-743
  • 74 K S. Mental health: A world of depression. Nature 2014; 515: 180-81
  • 75 Alonso J. et al Days out of role due to common physical and mental conditions: results from the WHO World Mental Health surveys. Mol Psychiatry 2010: 1-13
  • 76 Littlejohn G. et al Neurogenic inflammation in fibromyalgia. Seminars in Immunopathology. 2018
  • 77 Pearson TA. et al Markers of Inflammation and Cardiovascular Disease. Application to Clinical and Public Health Practice: A Statement for Healthcare Professionals From the Centers for Disease Control and Prevention and the American Heart Association 2003; 107 (03) 499-511
  • 78 Podolsky DK. Inflammatory Bowel Disease. N Engl J Med 2002; 347 (06) 417-429
  • 79 Covic T. et al Depression and anxiety in patients with rheumatoid arthritis: prevalence rates based on a comparison of the Depression, Anxiety and Stress Scale (DASS) and the hospital, Anxiety and Depression Scale (HADS). BMC Psychiatry 2012; 12: 6
  • 80 Ryanna K. et al Regulatory T cells in bronchial asthma. Allergy 2009; 64 (03) 335-347
  • 81 Ali Z. et al Is atopic dermatitis associated with obesity? A systematic review of observational studies. Journal of the European Academy of Dermatology and Venereology. 0(0)
  • 82 Breser ML. et al Immunological Mechanisms Underlying Chronic Pelvic Pain and Prostate Inflammation in Chronic Pelvic Pain Syndrome. Frontiers in Immunology 2017; 8: 898
  • 83 Gola H. et al Posttraumatic stress disorder is associated with an enhanced spontaneous production of pro-inflammatory cytokines by peripheral blood mononuclear cells. BMC Psychiatry 2013; 13: 40
  • 84 Michopoulos V. et al Inflammation in Fear- and Anxiety-Based Disorders: PTSD, GAD, and Beyond. Neuropsychopharmacology 2017; 42 (01) 254-270
  • 85 Howren MB. et al Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med 2009; 71 (02) 171-86
  • 86 Pace TW. et al Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. Am J Psychiatry 2006; 163 (09) 1630-3
  • 87 Maes M. et al Relationships between interleukin-6 activity, acute phase proteins, and function of the hypothalamic-pituitary-adrenal axis in severe depression. Psychiatry Res 1993; 49 (01) 11-27
  • 88 Grossi G. et al Physiological correlates of burnout among women. J Psychosom Res 2003; 55 (04) 309-16
  • 89 von Kanel R. et al Association between burnout and circulating levels of pro- and anti-inflammatory cytokines in schoolteachers. J Psychosom Res 2008; 65 (01) 51-9
  • 90 Strahler J. et al Dysregulated stress signal sensitivity and inflammatory disinhibition as a pathophysiological mechanism of stress-related chronic fatigue. Neurosci Biobehav Rev 2016; 68: 298-318
  • 91 Rohleder N. et al Role of interleukin-6 in stress, sleep, and fatigue. Ann N Y Acad Sci 2012; 1261: 88-96
  • 92 Bower JE. et al Fatigue and gene expression in human leukocytes: Increased NF-[kappa]B and decreased glucocorticoid signaling in breast cancer survivors with persistent fatigue. Brain, Behavior, and Immunity 2010; 25 (01) 147-150
  • 93 Feigenson KA. et al Inflammation and the Two-Hit Hypothesis of Schizophrenia. Neuroscience and biobehavioral reviews 2014; 38: 72-93
  • 94 Pace TW. et al Increased peripheral NF-kappaB pathway activity in women with childhood abuse-related posttraumatic stress disorder. Brain Behav Immun 2012; 26 (01) 13-7
  • 95 Lindqvist D. et al Proinflammatory milieu in combat-related PTSD is independent of depression and early life stress. Brain Behav Immun 2014; 42: 81-8
  • 96 O’Donovan A. et al Elevated risk for autoimmune disorders in iraq and afghanistan veterans with posttraumatic stress disorder. Biol Psychiatry 2015; 77 (04) 365-74
  • 97 Camara RJ. et al Post-traumatic stress in Crohn’s disease and its association with disease activity. Frontline Gastroenterol 2011; 2 (01) 2-9
  • 98 Spitzer C. et al Association of posttraumatic stress disorder with low-grade elevation of C-reactive protein: Evidence from the general population. Journal of Psychiatric Research 2010; 44 (01) 15-21
  • 99 Lehrner A. et al Cortisol and the Hypothalamic – Pituitary–Adrenal Axis in PTSD, in Posttraumatic Stress Disorder. New York: John Wiley & Sons, Inc; 2016: 265-290
  • 100 Mason JW. et al Urinary free-cortisol levels in posttraumatic stress disorder patients. J Nerv Ment Dis 1986; 174 (03) 145-9
  • 101 Yehuda R. Status of glucocorticoid alterations in post-traumatic stress disorder. Ann N Y Acad Sci 2009; 1179: 56-69
  • 102 Yehuda R. Post-traumatic stress disorder. N Engl J Med 2002; 346 (02) 108-14
  • 103 Yehudy R. et al Mini-Review: Stress-Related Psychiatric Disorders with Low Cortisol Levels: A Metabolic Hypothesis. 2011
  • 104 Yehuda R. et al The cortisol and glucocorticoid receptor response to low dose dexamethasone administration in aging combat veterans and holocaust survivors with and without posttraumatic stress disorder. Biol Psychiatry 2002; 52 (05) 393-403
  • 105 Morris MC. et al Relations among posttraumatic stress disorder, comorbid major depression, and HPA function: a systematic review and meta-analysis. Clin Psychol Rev 2012; 32 (04) 301-15
  • 106 de Kloet CS. et al Assessment of HPA-axis function in posttraumatic stress disorder: Pharmacological and non-pharmacological challenge tests, a review. Journal of Psychiatric Research 2006; 40 (06) 550-567
  • 107 Vogelzangs N. et al Anxiety disorders and inflammation in a large adult cohort. Transl Psychiatry 2013; 3: e249
  • 108 Copeland WE. et al Generalized anxiety and C-reactive protein levels: a prospective, longitudinal analysis. Psychol Med 2012; 42 (12) 2641-50
  • 109 Wagner EY. et al Evidence for chronic low-grade systemic inflammation in individuals with agoraphobia from a population-based prospective study. PLoS One 2015; 10 (04) e0123757
  • 110 Hoge EA. et al Broad spectrum of cytokine abnormalities in panic disorder and posttraumatic stress disorder. Depress Anxiety 2009; 26 (05) 447-55
  • 111 Vieira MM. et al Enhanced Th17 phenotype in individuals with generalized anxiety disorder. J Neuroimmunol 2010; 229 1–2 212-8
  • 112 Miller AH. et al The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol 2016; 16 (01) 22-34
  • 113 Raison CL. et al Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol 2006; 27 (01) 24-31
  • 114 Dantzer R. Role of the Kynurenine Metabolism Pathway in Inflammation-Induced Depression: Preclinical Approaches, in Inflammation-Associated Depression: Evidence, Mechanisms and Implications. Dantzer R, Capuron L. eds 2017. Springer International Publishing:; Cham: 117-138
  • 115 Miller AH. et al Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry 2009; 65 (09) 732-41
  • 116 Maes M. Major depression and activation of the inflammatory response system. Adv Exp Med Biol 1999; 461: 25-46
  • 117 Maes M. et al Evidence for a systemic immune activation during depression: results of leukocyte enumeration by flow cytometry in conjunction with monoclonal antibody staining. Psychol Med 1992; 22 (01) 45-53
  • 118 Maes M. Evidence for an immune response in major depression: A review and hypothesis. Progress in Neuro-Psychopharmacology and Biological Psychiatry 1995; 19 (01) 11-38
  • 119 Dunbar PR. et al Neopterin measurement provides evidence of altered cell-mediated immunity in patients with depression, but not with schizophrenia. Psychol Med 1992; 22 (04) 1051-7
  • 120 Maes M. et al Depression-related disturbances in mitogen-induced lymphocyte responses and interleukin-1 beta and soluble interleukin-2 receptor production. Acta Psychiatr Scand 1991; 84 (04) 379-86
  • 121 Maslach C. et al Job burnout. Annu Rev Psychol 2001; 52: 397-422
  • 122 American-Psychiatric-Association Diagnostic and Statistical Manual of Mental Disorders Fifth Edition ed. Washington, DC: American Psychiatric Association Press; 2013
  • 123 Friberg T. Burnout: from popular culture to psychiatric diagnosis in Sweden. Cult Med Psychiatry 2009; 33 (04) 538-58
  • 124 Melamed S. et al Burnout and risk of cardiovascular disease: evidence, possible causal paths, and promising research directions. Psychol Bull 2006; 132 (03) 327-53
  • 125 Sharpe M. Psychiatric diagnosis and chronic fatigue syndrome: controversies and conflicts. J Mental Health 2005; 14: 269-76
  • 126 Maes M. et al Evidence for inflammation and activation of cell-mediated immunity in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Increased interleukin-1, tumor necrosis factor- & #x3b1; PMN-elastase, lysozyme and neopterin. Journal of Affective Disorders 2012; 136 (03) 933-939
  • 127 Fletcher MA. et al Plasma cytokines in women with chronic fatigue syndrome. Journal of Translational Medicine 2009; 7 (01) 96
  • 128 Raison CL. et al Association of peripheral inflammatory markers with chronic fatigue in a population-based sample. Brain, Behavior, and Immunity 2009; 23 (03) 327-337
  • 129 Brenu EW. et al Immunological abnormalities as potential biomarkers in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis. Journal of Translational Medicine 2011; 9 (01) 81
  • 130 Miller GE. et al Clinical depression and regulation of the inflammatory response during acute stress. Psychosom Med 2005; 67 (05) 679-87
  • 131 Steptoe A. et al The effects of acute psychological stress on circulating inflammatory factors in humans: a review and meta-analysis. Brain Behav Immun 2007; 21 (07) 901-12
  • 132 Bierhaus A. et al A mechanism converting psychosocial stress into mononuclear cell activation. Proc Natl Acad Sci U S A 2003; 100 (04) 1920-5
  • 133 Wolf JM. et al Determinants of the NF-kappaB response to acute psychosocial stress in humans. Brain Behav Immun 2009; 23 (06) 742-9
  • 134 Pace TW. et al A short review on the psychoneuroimmunology of posttraumatic stress disorder: from risk factors to medical comorbidities. Brain Behav Immun 2011; 25 (01) 6-13
  • 135 Kaufman J. Depressive disorders in maltreated children. J Am Acad Child Adolesc Psychiatry 1991; 30 (02) 257-65
  • 136 Carpenter LL. et al Association between plasma IL-6 response to acute stress and early-life adversity in healthy adults. Neuropsychopharmacology 2010; 35 (13) 2617-23
  • 137 Danese A. et al Childhood maltreatment predicts adult inflammation in a life-course study. Proc Natl Acad Sci U S A 2007; 104 (04) 1319-24
  • 138 Surtees P. et al Association of depression with peripheral leukocyte counts in EPIC-Norfolk – role of sex and cigarette smoking. J Psychosom Res 2003; 54 (04) 303-6
  • 139 Baumeister D. et al Childhood trauma and adulthood inflammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-alpha. Mol Psychiatry 2016; 21 (05) 642-9
  • 140 Danese A. et al Biological embedding of stress through inflammation processes in childhood. Mol Psychiatry 2011; 16 (03) 244-6
  • 141 Brenhouse HC. et al Andersen, Nonsteroidal anti-inflammatory treatment prevents delayed effects of early life stress in rats. Biol Psychiatry 2011; 70 (05) 434-40
  • 142 Boeck C. et al Inflammation in adult women with a history of child maltreatment: The involvement of mitochondrial alterations and oxidative stress. Mitochondrion 2016; 30: 197-207
  • 143 Miller GE. et al Harsh Family Climate in Early Life Presages the Emergence of a Proinflammatory Phenotype in Adolescence. Psychological Science 2010; 21 (06) 848-856
  • 144 Ehrlich KB. et al Testing the biological embedding hypothesis: Is early life adversity associated with a later proinflammatory phenotype?. Development and Psychopathology 2016; 28 4pt2 1273-1283
  • 145 Elwenspoek M. et al The effects of early life adversity on the immune system. Psychoneuroendocrinology 2017; 82: 140-154
  • 146 Miller GE. et al Low early-life social class leaves a biological residue manifested by decreased glucocorticoid and increased proinflammatory signaling. Proc Natl Acad Sci U S A 2009; 106 (34) 14716-21
  • 147 Veenema AH. et al Early life stress enhances the vulnerability to chronic psychosocial stress and experimental colitis in adult mice. Endocrinology 2008; 144 (06) 2727-36
  • 148 Varghese AK. et al Antidepressants Attenuate Increased Susceptibility to Colitis in a Murine Model of Depression. Gastroenterology 2006; 130 (06) 1743-1753
  • 149 Barreau F. et al Neonatal maternal deprivation triggers long term alterations in colonic epithelial barrier and mucosal immunity in rats. Gut 2004; 53 (04) 501-6
  • 150 Romeo RD. et al Anxiety and fear behaviors in adult male and female C57BL/6 mice are modulated by maternal separation. Hormones and Behavior 2003; 43 (05) 561-567
  • 151 Veenema AH. et al Opposite effects of maternal separation on intermale and maternal aggression in C57BL/6 mice: Link to hypothalamic vasopressin and oxytocin immunoreactivity. Psychoneuroendocrinology 2007; 32 (05) 437-450
  • 152 Ladd CO. et al Persistent changes in corticotropin-releasing factor neuronal systems induced by maternal deprivation. Endocrinology 1996; 137 (04) 1212-1218
  • 153 Sanchez MM. et al Early adverse experience as a developmental risk factor for later psychopathology: evidence from rodent and primate models. Dev Psychopathol 2001; 13 (03) 419-49
  • 154 Gutman DA. et al Neurobiology of early life stress: rodent studies. Semin Clin Neuropsychiatry 2002; 7 (02) 89-95
  • 155 Huot RL. et al Neonatal maternal separation reduces hippocampal mossy fiber density in adult Long Evans rats. Brain Res 2002; 950 1–2 52-63
  • 156 Kalinichev M. et al Long-lasting changes in stress-induced corticosterone response and anxiety-like behaviors as a consequence of neonatal maternal separation in Long-Evans rats. Pharmacology Biochemistry and Behavior 2002; 73 (01) 131-140
  • 157 Newport DJ. et al Parental depression: animal models of an adverse life event. Am J Psychiatry 2002; 159 (08) 1265-83
  • 158 Veenema AH. Early life stress, the development of aggression and neuroendocrine and neurobiological correlates: What can we learn from animal models?. Frontiers in Neuroendocrinology 2009; 30 (04) 497-518
  • 159 Plotsky PM. et al Early, postnatal experience alters hypothalamic corticotropin-releasing factor (CRF) mRNA, median eminence CRF content and stress-induced release in adult rats. Brain Res Mol Brain Res 1993; 18 (03) 195-200
  • 160 Wigger A. et al Neumann, Periodic maternal deprivation induces gender-dependent alterations in behavioral and neuroendocrine responses to emotional stress in adult rats. Physiol Behav 1999; 66 (02) 293-302
  • 161 Derry HM. et al Lower subjective social status exaggerates interleukin-6 responses to a laboratory stressor. Psychoneuroendocrinology 2013; 38 (11) 2676-2685
  • 162 Singh-Manoux A. et al Subjective social status: its determinants and its association with measures of ill-health in the Whitehall II study. Social Science & Medicine 2003; 56 (06) 1321-1333
  • 163 Hu PF. et al Relationship between subjective social status and measures of health in older Taiwanese persons. Journal of the American Geriatrics Society 2005; 53 (03) 483-488
  • 164 Demakakos P. et al Socioeconomic status and health: the role of subjective social status. Soc Sci Med 2008; 67 (02) 330-40
  • 165 Breines JG. et al Self-compassion as a predictor of interleukin-6 response to acute psychosocial stress. Brain Behav Immun 2014; 37: 109-14
  • 166 Pace T. et al Engagement with Cognitively-Based Compassion Training is associated with reduced salivary C-reactive protein from before to after training in foster care program adolescents. Psychoneuroendocrinology 2013; 38 (02) 294-299
  • 167 McInnis CM. et al Measures of adiposity predict interleukin-6 responses to repeated psychosocial stress. Brain Behav Immun 2014; 42: 33-40
  • 168 Steptoe A. et al The effect of acute psychological stress on circulating inflammatory factors in humans: a review and meta-analysis. Brain, Behavior, & Immunity 2007; 7: 901-912
  • 169 von Kanel R. et al Delayed response and lack of habituation in plasma interleukin-6 to acute mental stress in men. Brain Behav Immun 2006; 20 (01) 40-8
  • 170 Lapidus L. et al Distribution of adipose tissue and risk of cardiovascular disease and death: a 12 year follow up of participants in the population study of women in Gothenburg, Sweden. British Medical Journal (Clinical research ed.) 1984; 289 6454 1257-1261
  • 171 Lavie CJ. et al Obesity and Cardiovascular Disease: Risk Factor, Paradox, and Impact of Weight Loss. Journal of the American College of Cardiology 2009; 53 (21) 1925-1932
  • 172 Kissebah AH. et al Regional adiposity and morbidity. Physiological Reviews 1994; 74 (04) 761-811
  • 173 Shelton RC. et al Eating ourselves to death (and despair): the contribution of adiposity and inflammation to depression. Prog Neurobiol 2010; 91 (04) 275-99
  • 174 Shoelson SE. et al Obesity, inflammation, and insulin resistance. Gastroenterology 2007; 132 (06) 2169-80
  • 175 de Wit L. et al Depression and obesity: a meta-analysis of community-based studies. Psychiatry Res 2010; 178 (02) 230-5
  • 176 Luppino S. et al Overweight, obesity, and depression: a systematic review and meta-analysis of longitudinal studies. Arch Gen Psychiatry 2010; 67 (03) 220-9
  • 177 von Kanel R. et al Effect of Alzheimer caregiving stress and age on frailty markers interleukin-6, C-reactive protein, and D-dimer. J Gerontol A Biol Sci Med Sci 2006; 61 (09) 963-9
  • 178 Gouin JP. et al Chronic stress, daily stressors, and circulating inflammatory markers. Health Psychology 2012; 31 (02) 264-268
  • 179 Lutgendorf SK. et al Life stress, mood disturbance, and elevated interleukin-6 in healthy older women. J Gerontol A Biol Sci Med Sci 1999; 54 (09) M434-9
  • 180 Mausbach BT. et al Self-efficacy buffers the relationship between dementia caregiving stress and circulating concentrations of the proinflammatory cytokine interleukin-6. Am J Geriatr Psychiatry 2011; 19 (01) 64-71
  • 181 Gouin JP. et al Chronic stress, daily stressors, and circulating inflammatory markers. Health Psychol 2012; 31 (02) 264-8
  • 182 Rohleder N. et al Biologic cost of caring for a cancer patient: dysregulation of pro- and anti-inflammatory signaling pathways. J Clin Oncol 2009; 27 (18) 2909-15
  • 183 Miller GE. et al A functional genomic fingerprint of chronic stress in humans: blunted glucocorticoid and increased NF-kappaB signaling. Biol Psychiatry 2008; 64 (04) 266-72
  • 184 Miller GE. et al Greater inflammatory activity and blunted glucocorticoid signaling in monocytes of chronically stressed caregivers. Brain Behav Immun 2014; 41: 191-9
  • 185 Miller G. et al Unfavorable socioeconomic conditions in early life presage expression of proinflammatory phenotype in adolescence. Psychosom Med 2007; 69 (05) 402-9
  • 186 Packard CJ. et al Early life socioeconomic adversity is associated in adult life with chronic inflammation, carotid atherosclerosis, poorer lung function and decreased cognitive performance: a cross-sectional, population-based study. BMC Public Health 2011; 11: 42
  • 187 Lanier P. et al Child Maltreatment and Pediatric Health Outcomes: A Longitudinal Study of Low-income Children. Journal of Pediatric Psychology 2010; 35 (05) 511-522
  • 188 Graham-Bermann SA. et al Violence exposure and traumatic stress symptoms as additional predictors of health problems in high-risk children. The Journal of Pediatrics 2005; 146 (03) 349-354
  • 189 Pace TW. et al Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. American Journal of Psychiatry 2006; 163 (09) 1630-1633
  • 190 Khandaker GM. et al 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 (10) 1121-8
  • 191 Kivimaki M. et al Long-term inflammation increases risk of common mental disorder: a cohort study. Mol Psychiatry 2014; 19 (02) 149-50
  • 192 Duivis HE. et al Depressive symptoms, health behaviors, and subsequent inflammation in patients with coronary heart disease: prospective findings from the heart and soul study. Am J Psychiatry 2011; 168 (09) 913-20
  • 193 Margaretten M. et al Depression in patients with rheumatoid arthritis: description, causes and mechanisms. Int J Clin Rheumtol 2011; 6 (06) 617-623
  • 194 Zhang Y. et al Antidepressant drug, desipramine, alleviates allergic rhinitis by regulating Treg and Th17 cells. Int J Immunopathol Pharmacol 2013; 26 (01) 107-15
  • 195 Dijkstra HP. et al The prevalence and current opinion of treatment of allergic rhinitis in elite athletes. Curr Opin Allergy Clin Immunol 2011; 11 (02) 103-8
  • 196 Reichenberg A. et al Cytokine-associated emotional and cognitive disturbances in humans. Arch Gen Psychiatry 2001; 58 (05) 445-52
  • 197 van den Biggelaar A. et al Inflammation and interleukin-1 signaling network contribute to depressive symptoms but not cognitive decline in old age. Experimental Gerontology 2007; 42 (07) 693-701
  • 198 Moieni M. et al Sex Differences in Depressive and Socioemotional Responses to an Inflammatory Challenge: Implications for Sex Differences in Depression. Neuropsychopharmacology 2015; 40 (07) 1709-1716
  • 199 Kessler RC. et al Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbidity Survey. Arch Gen Psychiatry 1994; 51 (01) 8-19
  • 200 Grigoriadis S. et al Gender issues in depression. Ann Clin Psychiatry 2007; 19 (04) 247-55
  • 201 Engler H. et al Selective increase of cerebrospinal fluid IL-6 during experimental systemic inflammation in humans: association with depressive symptoms. Mol Psychiatry 2017
  • 202 Bull SJ. et al Functional polymorphisms in t he interleukin-6 and serotonin transporter genes, and depression and fatigue induced by interferon-[alpha] and ribavirin treatment. Mol Psychiatry 2009; 14 (12) 1095-1104
  • 203 Eraly SA. et al Assessment of plasma C-reactive protein as a biomarker of posttraumatic stress disorder risk. JAMA Psychiatry 2014; 71 (04) 423-31
  • 204 Vasiliki Michopoulos, et al Association of CRP Genetic Variation and CRP Level With Elevated PTSD Symptoms and Physiological Responses in a Civilian Population With High Levels of Trauma. American Journal of Psychiatry 2015; 172 (04) 353-362
  • 205 Pervanidou P. et al Elevated morning serum interleukin (IL)-6 or evening salivary cortisol concentrations predict posttraumatic stress disorder in children and adolescents six months after a motor vehicle accident. Psychoneuroendocrinology 2007; 32: 991-999
  • 206 Cattaneo A. et al Absolute Measurements of Macrophage Migration Inhibitory Factor and Interleukin-1-β mRNA Levels Accurately Predict Treatment Response in Depressed Patients. International Journal of Neuropsychopharmacology 2016
  • 207 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 (01) 31-41
  • 208 Na KS. et al Efficacy of adjunctive celecoxib treatment for patients with major depressive disorder: a meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48: 79-85
  • 209 Köhler 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
  • 210 Karl S. et al Low-Dose Aspirin for Prevention of Cardiovascular Risk in Bereavement: Results from a Feasibility Study. Psychotherapy and Psychosomatics 2018; 87 (02) 112-113
  • 211 Hiles SA. et al Interleukin-6, C-reactive protein and interleukin-10 after antidepressant treatment in people with depression: a meta-analysis. Psychol Med 2012; 42 (10) 2015-26
  • 212 Yoshimura R. et al Higher plasma interleukin-6 (IL-6) level is associated with SSRI- or SNRI-refractory depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry 2009; 33 (04) 722-726
  • 213 Dantzer R. Role of the Kynurenine Metabolism Pathway in Inflammation-Induced Depression: Preclinical Approaches. Curr Top Behav Neurosci 2017; 31: 117-138
  • 214 Dantze R. Cytokine-Induced Sickness Behavior: Mechanisms and Implications. Ann NY Acad Sci 2001; 933 (01) 222-234
  • 215 Dantzer R. Cytokine-Induced Sickness Behavior: Where Do We Stand?. Brain, Behavior, and Immunity 2001; 15 (01) 7-24
  • 216 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 (45) 16136-41
  • 217 Fleshner M. Stress-evoked sterile inflammation, danger associated molecular patterns (DAMPs), microbial associated molecular patterns (MAMPs) and the inflammasome. Brain Behav Immun 2013; 27 (01) 1
  • 218 Schroder K. et al The inflammasomes. Cell 2010; 140 (06) 821-32
  • 219 Menu P. et al The NLRP3 inflammasome in health and disease: the good, the bad and the ugly. Clin Exp Immunol 2011; 166 (01) 1-15
  • 220 Strowig T. et al Inflammasomes in health and disease. Nature 2012; 481 7381 278-86
  • 221 Menzel CL. et al Caspase-1 Is Hepatoprotective during Trauma and Hemorrhagic Shock by Reducing Liver Injury and Inflammation. Molecular Medicine 2011; 17 9–10 1031-1038
  • 222 Straub RH. et al Key role of the sympathetic microenvironment for the interplay of tumour necrosis factor and interleukin 6 in normal but not in inflamed mouse colon mucosa. Gut 2005; 54 (08) 1098-1106
  • 223 Härle P. et al An opposing time-dependent immune-modulating effect of the sympathetic nervous system conferred by altering the cytokine profile in the local lymph nodes and spleen of mice with type II collagen-induced arthritis. Arthritis & Rheumatism 2005; 52 (04) 1305-1313
  • 224 Härle P, Pongratz G, Straub RH. The sympathetic nervous system stimulates collagen-induced arthritis (CIA) in the induction phase and inhibits CIA in the late effector phase in DBA-1 mice. Arthritis Rheum 2003; 48 (Suppl. 09) 350
  • 225 Straub RH. et al Uncoupling of the sympathetic nervous system and the hypothalamic-pituitary-adrenal axis in inflammatory bowel disease?. J Neuroimmunol 2002; 126 1–2 116-25
  • 226 Miller LE. et al The loss of sympathetic nerve fibers in the synovial tissue of patients with rheumatoid arthritis is accompanied by increased norepinephrine release from synovial macrophages. FASEB J 2000; 14 (13) 2097-2107
  • 227 Bowers SL. et al Stressor-specific alterations in corticosterone and immune responses in mice. Brain, Behavior, and Immunity 2008; 22 (01) 105-113
  • 228 Viswanathan K, Dhabhar FS. Stress-induced enhancement of leukocyte trafficking into sites of surgery or immune activation. PNAS 2005; 102 (16) 5808-5813
  • 229 Dhabhar FS, McEwen BS. Enhancing versus suppressive effects of stress hormones on skin immune function. Proc Natl Acad Sci U S A 1999; 96 (03) 1059-64
  • 230 Dhabhar FS. et al Stress-induced changes in blood leukocyte distribution. Role of adrenal steroid hormones. J Immunol 1996; 157 (04) 1638-44
  • 231 Dhabhar FS. et al Effects of stress on immune cell distribution. Dynamics and hormonal mechanisms. J Immunol 1995; 154 (10) 5511-27
  • 232 Rock KL. et al The sterile inflammatory response. Annu Rev Immunol 2010; 28: 321-42
  • 233 Johnson JD, Fleshner M. Releasing signals, secretory pathways, and immune function of endogenous extracellular heat shock protein 72. J Leukoc Biol 2006; 79 (03) 425-34
  • 234 Maslanik T. et al Commensal Bacteria and MAMPs Are Necessary for Stress-Induced Increases in IL-1β and IL-18 but Not IL-6, IL-10 or MCP-1. PLOS ONE 2012; 7 (12) e50636
  • 235 Maslanik T. et al The inflammasome and danger associated molecular patterns (DAMPs) are implicated in cytokine and chemokine responses following stressor exposure. Brain, Behavior, and Immunity 2013; 28 (00) 54-62
  • 236 Johnson JD. et al Adrenergic receptors mediate stress-induced elevations in extracellular Hsp72. J Appl Physiol 2005; 99 (05) 1789-95
  • 237 Allen RG. et al The intestinal microbiota are necessary for stressor-induced enhancement of splenic macrophage microbicidal activity. Brain Behav Immun 2012; 26 (03) 371-82
  • 238 Bailey MT. The contributing role of the intestinal microbiota in stressor-induced increases in susceptibility to enteric infection and systemic immunomodulation. Horm Behav 2012; 62 (03) 286-94
  • 239 Bailey MT. et al Exposure to a social stressor alters the structure of the intestinal microbiota: Implications for stressor-induced immunomodulation. Brain, Behavior, and Immunity 2011; 25 (03) 397-407
  • 240 Füchsl AM, Neumann ID, Reber SO. Stress Resilience: A Low-Anxiety Genotype Protects Male Mice From the Consequences of Chronic Psychosocial Stress. Endocrinology 2014; 155 (01) 117-126
  • 241 Füchsl AM, Uschold-Schmidt N, Reber SO. Chronic psychosocial stress in male mice causes an up-regulation of scavenger receptor class B type 1 protein in the adrenal glands. Stress 2013; 16 (04) 461-468
  • 242 Czech B. et al Effect of chronic psychosocial stress on nonalcoholic steatohepatitis in mice. Int J Clin Exp Pathol 2013; 6 (08) 1585-93
  • 243 Reber SO. Stress and animal models of inflammatory bowel disease—An update on the role of the hypothalamo–pituitary–adrenal axis. Psychoneuroendocrinology 2012; 37 (01) 1-19
  • 244 Singewald GM. et al Effect of chronic psychosocial stress-induced by subordinate colony (CSC) housing on brain neuronal activity patterns in mice. Stress 2009; 12 (01) 58-69
  • 245 Reber SO. et al Chronic subordinate colony housing paradigm: A mouse model for mechanisms of PTSD vulnerability, targeted prevention, and treatment—2016 Curt Richter Award Paper. Psychoneuroendocrinology 2016; 74: 221-230
  • 246 Foertsch S. et al Splenic glucocorticoid resistance following psychosocial stress requires physical injury. Scientific Reports 2017; 7 (01) 15730
  • 247 Reber SO. et al Adrenal insufficiency and colonic inflammation after a novel chronic psycho-social stress paradigm in mice: implications and mechanisms. Endocrinology 2007; 148 (02) 670-82
  • 248 Langgartner D. et al Effects of Prior Psychosocial Trauma on Subsequent Immune Response After Experimental Thorax Trauma. Shock 2018; 49 (06) 690-697
  • 249 Langgartner D. et al Individual differences in stress vulnerability: The role of gut pathobionts in stress-induced colitis. Brain Behav Immun 2017; 64: 23-32
  • 250 Netea MG. et al IL-1beta processing in host defense: beyond the inflammasomes. PLoS Pathog 2010; 6 (02) e1000661
  • 251 Eisenbarth SC. et al Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature 2008; 453 7198 1122-6
  • 252 Berk M. et al So depression is an inflammatory disease, but where does the inflammation come from?. BMC Med 2013; 11: 200
  • 253 Galea I, Bechmann I, Perry VH. What is immune privilege (not)?. Trends Immunol 2007; 28 (01) 12-8
  • 254 Louveau A, Harris TH, Kipnis J. Revisiting the Mechanisms of CNS Immune Privilege. Trends Immunol 2015; 36 (10) 569-577
  • 255 Dantzer R. et al Neural and humoral pathways of communication from the immune system to the brain: parallel or convergent?. Auton Neurosci 2000; 85 1–3 60-5
  • 256 Lang CH, Cooney R, Vary TC. Central interleukin-1 partially mediates endotoxin-induced changes in glucose metabolism. Am J Physiol 1996; 271 2 Pt 1 E309-16
  • 257 Kakucska I. et al Endotoxin-induced corticotropin-releasing hormone gene expression in the hypothalamic paraventricular nucleus is mediated centrally by interleukin-1. Endocrinology 1993; 133 (02) 815-21
  • 258 Garcia-Lecea M. et al Development of Circumventricular Organs in the Mirror of Zebrafish Enhancer-Trap Transgenics. Front Neuroanat 2017; 11: 114
  • 259 Takahashi Y. et al Circumventricular organs and fever. Am J Physiol 1997; 273 5 Pt 2 R1690-5
  • 260 Lee HY, Whiteside MB, Herkenham M. Area postrema removal abolishes stimulatory effects of intravenous interleukin-1beta on hypothalamic-pituitary-adrenal axis activity and c-fos mRNA in the hypothalamic paraventricular nucleus. Brain Res Bull 1998; 46 (06) 495-503
  • 261 Quan N, Whiteside M, Herkenham M. Time course and localization patterns of interleukin-1beta messenger RNA expression in brain and pituitary after peripheral administration of lipopolysaccharide. Neuroscience 1998; 83 (01) 281-93
  • 262 Laflamme N, Rivest S. Toll-like receptor 4: the missing link of the cerebral innate immune response triggered by circulating gram-negative bacterial cell wall components. FASEB J 2001; 15 (01) 155-163
  • 263 Ericsson A. et al Type 1 interleukin-1 receptor in the rat brain: distribution, regulation, and relationship to sites of IL-1-induced cellular activation. J Comp Neurol 1995; 361 (04) 681-98
  • 264 Cao C. et al Endothelial cells of the rat brain vasculature express cyclooxygenase-2 mRNA in response to systemic interleukin-1 beta: a possible site of prostaglandin synthesis responsible for fever. Brain Res 1996; 733 (02) 263-72
  • 265 Nadeau S, Rivest S. Effects of circulating tumor necrosis factor on the neuronal activity and expression of the genes encoding the tumor necrosis factor receptors (p55 and p75) in the rat brain: a view from the blood-brain barrier. Neuroscience 1999; 93 (04) 1449-64
  • 266 Vallieres L, Rivest S. Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1beta. J Neurochem 1997; 69 (04) 1668-83
  • 267 Lacroix S, Feinstein D, Rivest S. The bacterial endotoxin lipopolysaccharide has the ability to target the brain in upregulating its membrane CD14 receptor within specific cellular populations. Brain Pathol 1998; 8 (04) 625-40
  • 268 Goehler LE, Erisir A, Gaykema RP. Neural-immune interface in the rat area postrema. Neuroscience 2006; 140 (04) 1415-34
  • 269 Nadeau S, Rivest S. Regulation of the gene encoding tumor necrosis factor alpha (TNF-alpha) in the rat brain and pituitary in response in different models of systemic immune challenge. J Neuropathol Exp Neurol 1999; 58 (01) 61-77
  • 270 Wong ML. et al Inducible nitric oxide synthase gene expression in the brain during systemic inflammation. Nat Med 1996; 2 (05) 581-4
  • 271 Stitt JT. Prostaglandin E as the neural mediator of the febrile response. Yale J Biol Med 1986; 59 (02) 137-49
  • 272 Bernheim HA. Is prostaglandin E2 involved in the pathogenesis of fever? Effects of interleukin-1 on the release of prostaglandins. Yale J Biol Med 1986; 59 (02) 151-8
  • 273 Bebo Jr BF, Linthicum DS. Expression of mRNA for 55-kDa and 75-kDa tumor necrosis factor (TNF) receptors in mouse cerebrovascular endothelium: effects of interleukin-1 beta, interferon-gamma and TNF-alpha on cultured cells. J Neuroimmunol 1995; 62 (02) 161-7
  • 274 Quan N. et al Induction of inhibitory factor kappaBalpha mRNA in the central nervous system after peripheral lipopolysaccharide administration: an in situ hybridization histochemistry study in the rat. Proc Natl Acad Sci U S A 1997; 94 (20) 10985-90
  • 275 Stitt JT. Passage of immunomodulators across the blood-brain barrier. Yale J Biol Med 1990; 63 (02) 121-31
  • 276 Konsman JP. et al Rat brain vascular distribution of interleukin-1 type-1 receptor immunoreactivity: relationship to patterns of inducible cyclooxygenase expression by peripheral inflammatory stimuli. J Comp Neurol 2004; 472 (01) 113-29
  • 277 Ek M. et al Inflammatory response: pathway across the blood-brain barrier. Nature 2001; 410 6827 430-1
  • 278 Banks WA. The blood-brain barrier: connecting the gut and the brain. Regul Pept 2008; 149 1–3 11-4
  • 279 Banks WA. Blood-brain barrier transport of cytokines: a mechanism for neuropathology. Curr Pharm Des 2005; 11 (08) 973-84
  • 280 Banks WA, Kastin AJ, Durham DA. Bidirectional transport of interleukin-1 alpha across the blood-brain barrier. Brain Res Bull 1989; 23 (06) 433-7
  • 281 Banks WA. et al Human interleukin (IL) 1 alpha, murine IL-1 alpha and murine IL-1 beta are transported from blood to brain in the mouse by a shared saturable mechanism. J Pharmacol Exp Ther 1991; 259 (03) 988-96
  • 282 Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol 1993; 47 (02) 169-76
  • 283 Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murine blood-brain barrier. Neurosci Lett 1994; 179 1–2 53-6
  • 284 Banks WA, Niehoff ML, Zalcman SS. Permeability of the mouse blood-brain barrier to murin interleukin-2: predominance of a saturable efflux system. Brain Behav Immun 2004; 18 (05) 434-42
  • 285 Kastin AJ, Akerstrom V, Pan W. Interleukin-10 as a CNS therapeutic: the obstacle of the blood-brain/blood-spinal cord barrier. Brain Res Mol Brain Res 2003; 114 (02) 168-71
  • 286 Maier SF. et al The role of the vagus nerve in cytokine-to-brain communication. Ann N Y Acad Sci 1998; 840: 289-300
  • 287 Obreja O. et al IL-1 beta potentiates heat-activated currents in rat sensory neurons: involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J 2002; 16 (12) 1497-503
  • 288 Long NC. et al Roles of interleukin 1 beta and tumor necrosis factor in lipopolysaccharide fever in rats. Am J Physiol 1990; 259 4 Pt 2 R724-8
  • 289 Goehler LE. et al Vagal paraganglia bind biotinylated interleukin-1 receptor antagonist: a possible mechanism for immune-to-brain communication. Brain Res Bull 1997; 43 (03) 357-64
  • 290 Ek M. et al Activation of vagal afferents after intravenous injection of interleukin-1beta: role of endogenous prostaglandins. J Neurosci 1998; 18 (22) 9471-9
  • 291 Bonaz B, Sinniger V, Pellissier S. The Vagus Nerve in the Neuro-Immune Axis: Implications in the Pathology of the Gastrointestinal Tract. Front Immunol 2017; 8: 1452
  • 292 Bonaz B, Sinniger V, Pellissier S. Anti-inflammatory properties of the vagus nerve: potential therapeutic implications of vagus nerve stimulation. J Physiol 2016; 594 (20) 5781-5790
  • 293 Watkins LR. et al Blockade of interleukin-1 induced hyperthermia by subdiaphragmatic vagotomy: evidence for vagal mediation of immune-brain communication. Neurosci Lett 1995; 183 1–2 27-31
  • 294 Laye S. et al Subdiaphragmatic vagotomy blocks induction of IL-1 beta mRNA in mice brain in response to peripheral LPS. Am J Physiol Regul Integr Comp Physiol 1995; 268 (05) R1327-1331
  • 295 Hendry S, Hsiao S. The somatosensory system; in Fundamental Neuroscience. Squire LR, Berg D, Bloom FE. et al. (eds) München: Elsevier; 2013
  • 296 Kayalioglu G. Projections form the spinal cord to the brain; in The Spinal Cord. Watson C, Paxinos G, Kayalioglu G. eds Washington: Academic Press; 2009: 148-167
  • 297 Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci 2011; 12 (08) 453-66
  • 298 Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest 2015; 125 (03) 926-38
  • 299 Kerfoot SM. et al TNF-alpha-secreting monocytes are recruited into the brain of cholestatic mice. Hepatology 2006; 43 (01) 154-62
  • 300 D’Mello C, Le T, Swain MG. Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factoralpha signaling during peripheral organ inflammation. J Neurosci 2009; 29 (07) 2089-102
  • 301 Wohleb ES. et al Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J Neurosci 2013; 33 (34) 13820-33
  • 302 Albert PR, Vahid-Ansari F, Luckhart C. Serotonin-prefrontal cortical circuitry in anxiety and depression phenotypes: pivotal role of pre- and post-synaptic 5-HT1A receptor expression. Front Behav Neurosci 2014; 8: 199
  • 303 Delgado PL. Depression: the case for a monoamine deficiency. J Clin Psychiatry 2000; 61 (Suppl. 06) 7-11
  • 304 Sperner-Unterweger B, Kohl C, Fuchs D. Immune changes and neurotransmitters: possible interactions in depression?. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48: 268-76
  • 305 Neurauter G. et al Chronic immune stimulation correlates with reduced phenylalanine turnover. Curr Drug Metab 2008; 9 (07) 622-7
  • 306 Felger JC. et al Tyrosine metabolism during interferon-alpha administration: association with fatigue and CSF dopamine concentrations. Brain Behav Immun 2013; 31: 153-60
  • 307 Maes M. et al 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 (03) 702-21
  • 308 Heyes MP. et al Different kynurenine pathway enzymes limit quinolinic acid formation by various human cell types. Biochem J 1997; 326 Pt 2 351-6
  • 309 Pemberton LA. et al Quinolinic acid production by macrophages stimulated with IFN-gamma, TNF-alpha, and IFN-alpha. J Interferon Cytokine Res 1997; 17 (10) 589-95
  • 310 Taylor MW, Feng GS. Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism. FASEB J 1991; 5 (11) 2516-22
  • 311 Takikawa O. et al Mechanism of interferon-gamma action. Characterization of indoleamine 2,3-dioxygenase in cultured human cells induced by interferon-gamma and evaluation of the enzyme-mediated tryptophan degradation in its anticellular activity. J Biol Chem 1988; 263 (04) 2041-8
  • 312 Kim H. et al Brain indoleamine 2,3-dioxygenase contributes to the comorbidity of pain and depression. J Clin Invest 2012; 122 (08) 2940-54
  • 313 Van der Does AJ. The effects of tryptophan depletion on mood and psychiatric symptoms. J Affect Disord 2001; 64 2–3 107-19
  • 314 Maes M. et al Relationships between lower plasma L-tryptophan levels and immune-inflammatory variables in depression. Psychiatry Res 1993; 49 (02) 151-65
  • 315 Elovainio M. et al Indoleamine 2,3-dioxygenase activation and depressive symptoms: results from the Young Finns Study. Psychosom Med 2012; 74 (07) 675-81
  • 316 O’Connor JC. et al Induction of IDO by bacille Calmette-Guerin is responsible for development of murine depressive-like behavior. J Immunol 2009; 182 (05) 3202-12
  • 317 Capuron L. et al Dopaminergic mechanisms of reduced basal ganglia responses to hedonic reward during interferon alfa administration. Arch Gen Psychiatry 2012; 69 (10) 1044-53
  • 318 Zhu CB, Blakely RD, Hewlett WA. The proinflammatory cytokines interleukin-1beta and tumor necrosis factor-alpha activate serotonin transporters. Neuropsychopharmacology 2006; 31 (10) 2121-31
  • 319 Zhu CB. et al Interleukin-1 receptor activation by systemic lipopolysaccharide induces behavioral despair linked to MAPK regulation of CNS serotonin transporters. Neuropsychopharmacology 2010; 35 (13) 2510-20
  • 320 Sanacora G, Treccani G, Popoli M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2012; 62 (01) 63-77
  • 321 Kucukibrahimoglu E. et al The change in plasma GABA, glutamine and glutamate levels in fluoxetine- or S-citalopram-treated female patients with major depression. Eur J Clin Pharmacol 2009; 65 (06) 571-7
  • 322 Blakely PK. et al Disrupted glutamate transporter expression in the spinal cord with acute flaccid paralysis caused by West Nile virus infection. J Neuropathol Exp Neurol 2009; 68 (10) 1061-72
  • 323 Vesce S. et al Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation. Int Rev Neurobiol 2007; 82: 57-71
  • 324 Maes M. et al Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine 1997; 9 (11) 853-8
  • 325 de Carvalho LP, Bochet P, Rossier J. The endogenous agonist quinolinic acid and the non endogenous homoquinolinic acid discriminate between NMDAR2 receptor subunits. Neurochem Int 1996; 28 (04) 445-52
  • 326 Sender R, Fuchs S, Milo R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016; 164 (03) 337-40
  • 327 Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour – epigenetic regulation of the gut-brain axis. Genes Brain Behav 2014; 13 (01) 69-86
  • 328 Stilling RM, Dinan TG, Cryan JF. The brain’s Geppetto-microbes as puppeteers of neural function and behaviour?. J Neurovirol 2016; 22 (01) 14-21
  • 329 Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci 2012; 13 (10) 701-12
  • 330 Carabotti M. et al The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol 2015; 28 (02) 203-209
  • 331 Dinan TG, Cryan JF. Melancholic microbes: a link between gut microbiota and depression?. Neurogastroenterol Motil 2013; 25 (09) 713-9
  • 332 Forsythe P, Kunze WA. Voices from within: gut microbes and the CNS. Cell Mol Life Sci 2013; 70 (01) 55-69
  • 333 Forsythe P, Kunze WA, Bienenstock J. On communication between gut microbes and the brain. Curr Opin Gastroenterol 2012; 28 (06) 557-62
  • 334 Tanoue T, Umesaki Y, Honda K. Immune responses to gut microbiota-commensals and pathogens. Gut Microbes 2010; 1 (04) 224-233
  • 335 Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 2009; 9 (05) 313-323
  • 336 Lozupone CA. et al Diversity, stability and resilience of the human gut microbiota. Nature 2012; 489 7415 220-30
  • 337 Stilling RM. et al Microbes & neurodevelopment – Absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain Behav Immun 2015; 50: 209-220
  • 338 Hooper LV, Macpherson AJ. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol 2010; 10 (03) 159-69
  • 339 Wu HJ, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012; 3 (01) 4-14
  • 340 Macpherson AJ, Uhr T. Gut flora – mechanisms of regulation. Eur J Surg Suppl 2002 587 53-7
  • 341 Ohnmacht C. et al MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science 2015; 349 6251 989-93
  • 342 Sefik E. et al MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORgamma(+) regulatory T cells. Science 2015; 349 6251 993-7
  • 343 O’Mahony C. et al Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog 2008; 4 (08) e1000112
  • 344 Konieczna P. et al Bifidobacterium infantis, 35624 administration induces Foxp3 T regulatory cells in human peripheral blood: potential role for myeloid and plasmacytoid dendritic cells. Gut 2012; 61 (03) 354-66
  • 345 Ivanov II. et al Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009; 139 (03) 485-98
  • 346 Mazmanian SK. et al An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005; 122 (01) 107-18
  • 347 Atarashi K. et al Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013; 500 7461 232-6
  • 348 Horwitz DA, Zheng SG, Gray JD. The role of the combination of IL-2 and TGF-beta or IL-10 in the generation and function of CD4 + CD25 + and CD8 + regulatory T cell subsets. J Leukoc Biol 2003; 74 (04) 471-8
  • 349 Chu H, Mazmanian SK. Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat Immunol 2013; 14 (07) 668-75
  • 350 Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008; 453 7195 620-5
  • 351 Jeon SG. et al Probiotic Bifidobacterium breve induces IL-10-producing Tr1 cells in the colon. PLoS Pathog 2012; 8 (05) e1002714
  • 352 Vinolo MA. et al Regulation of inflammation by short chain fatty acids. Nutrients 2011; 3 (10) 858-76
  • 353 Arpaia N. et al Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013; 504 7480 451-5
  • 354 Singh N. et al Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014; 40 (01) 128-39
  • 355 Tedelind S. et al Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J Gastroenterol 2007; 13 (20) 2826-32
  • 356 Inagaki T. et al Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A 2006; 103 (10) 3920-5
  • 357 Hofmann AF, Eckmann L. How bile acids conter gut mucosal protection against bacteria. Proc Natl Acad Sci U S A 2006; 103 (12) 4333-4
  • 358 Rowley TJ. et al Antinociceptive and anti-inflammatory effects of choline in a mouse model of postoperative pain. Br J Anaesth 2010; 105 (02) 201-7
  • 359 Lewis ED. et al The Form of Choline in the Maternal Diet Affects Immune Development in Suckled Rat Offspring. J Nutr 2016; 146 (04) 823-30
  • 360 Baganz NL, Blakely RD. A dialogue between the immune system and brain, spoken in the language of serotonin. ACS Chem Neurosci 2013; 4 (01) 48-63
  • 361 Marsland BJ. Regulating inflammation with microbial metabolites. Nat Med 2016; 22 (06) 581-583
  • 362 Wikoff WR. et al Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A 2009; 106 (10) 3698-703
  • 363 Clarke G. et al The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 2013; 18 (06) 666-73
  • 364 Tsavkelova EA. et al [Hormones and hormone-like substances of microorganisms: a review]. Prikl Biokhim Mikrobiol 2006; 42 (03) 261-8
  • 365 Alexeev EE. et al Microbiota-Derived Indole Metabolites Promote Human and Murine Intestinal Homeostasis through Regulation of Interleukin-10 Receptor. Am J Pathol 2018; 188 (05) 1183-1194
  • 366 Lanis JM. et al Tryptophan metabolite activation of the aryl hydrocarbon receptor regulates IL-10 receptor expression on intestinal epithelia. Mucosal Immunol 2017; 10 (05) 1133-1144
  • 367 Reigstad CS. et al Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J 2015; 29 (04) 1395-403
  • 368 Singh RK. et al Influence of diet on the gut microbiome and implications for human health. J Transl Med 2017; 15 (01) 73
  • 369 Mariat D. et al The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 2009; 9: 123
  • 370 Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med 2016; 8 (01) 39
  • 371 O’Mahony SM. et al Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry 2009; 65 (03) 263-7
  • 372 Reber SO. et al Immunization with a heat-killed preparation of the environmental bacterium Mycobacterium vaccae promotes stress resilience in mice. Proc Natl Acad Sci U S A 2016; 113 (22) E3130-9
  • 373 De Palma G. et al The microbiota-gut-brain axis in gastrointestinal disorders: stressed bugs, stressed brain or both?. J Physiol 2014; 592 (14) 2989-97
  • 374 Hemmings SMJ. et al The Microbiome in Posttraumatic Stress Disorder and Trauma-Exposed Controls: An Exploratory Study. Psychosom Med 2017; 79 (08) 936-946
  • 375 De Palma G. et al Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice. Sci Transl Med 2017; 9: 379
  • 376 Kelly JR. et al Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res 2016; 82: 109-18
  • 377 Bercik P. et al The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 2011; 141 (02) 599-609 609 e1-3
  • 378 Collins SM, Kassam Z, Bercik P. The adoptive transfer of behavioral phenotype via the intestinal microbiota: experimental evidence and clinical implications. Curr Opin Microbiol 2013; 16 (03) 240-5
  • 379 Postolache TT, Komarow H, Tonelli LH. Allergy: a risk factor for suicide?. Curr Treat Options Neurol 2008; 10 (05) 363-76
  • 380 Lin W. et al Allergic dysregulation and hyperimmunoglobulinemia E in Foxp3 mutant mice. J Allergy Clin Immunol 2005; 116 (05) 1106-15
  • 381 Bennett CL. et al The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001; 27 (01) 20-1
  • 382 Kriegel MA. et al Defective suppressor function of human CD4 + CD25 + regulatory T cells in autoimmune polyglandular syndrome type II. J Exp Med 2004; 199 (09) 1285-91
  • 383 Rook GA, Raison CL, Lowry CA. Childhood microbial experience, immunoregulation, inflammation and adult susceptibility to psychosocial stressors and depression in rich and poor countries. Evol Med Public Health 2013; 2013 (01) 14-7
  • 384 Depis F. et al Unstable FoxP3 + T regulatory cells in NZW mice. Proc Natl Acad Sci U S A 2016; 113 (05) 1345-50
  • 385 Lynch SV, Boushey HA. The microbiome and development of allergic disease. Curr Opin Allergy Clin Immunol 2016; 16 (02) 165-71
  • 386 Martínez I. et al The gut microbiota of rural papua new guineans: composition, diversity patterns, and ecological processes. Cell Reports 2015; 11 (04) 527-538
  • 387 Stamper CE. et al The microbiome of the built environment and human behavior: implications for emotional health and well-being in postmodern western societies. Int Rev Neurobiol 2016; 131: 289-323
  • 388 Stein MM. et al Innate immunity and asthma risk in amish and hutterite farm children. New England Journal of Medicine 2016; 375 (05) 411-421
  • 389 Fall T. et al Early exposure to dogs and farm animals and the risk of childhood asthma. JAMA Pediatr 2015; 169 (11) e153219
  • 390 Mubanga M. et al Dog ownership and the risk of cardiovascular disease and death – a nationwide cohort study. Sci Rep 2017; 7 (01) 15821
  • 391 Farez MF. et al Melatonin Contributes to the Seasonality of Multiple Sclerosis Relapses. Cell 2015; 162 (06) 1338-52
  • 392 Zelante T. et al Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013; 39 (02) 372-85
  • 393 Zhang L. et al Persistent Organic Pollutants Modify Gut Microbiota-Host Metabolic Homeostasis in Mice Through Aryl Hydrocarbon Receptor Activation. Environ Health Perspect 2015; 123 (07) 679-88
  • 394 Quintana FJ. et al Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature 2008; 453 7191 65-71
  • 395 Mezrich JD. et al An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 2010; 185 (06) 3190-8
  • 396 Qiu J, Zhou L. Aryl hydrocarbon receptor promotes RORgammat(+) group 3 ILCs and controls intestinal immunity and inflammation. Semin Immunopathol 2013; 35 (06) 657-70
  • 397 Wang YC. et al The suppression effect of dendritic cells maturation by adipose-derived stem cells through TGF-beta1 related pathway. Exp Cell Res 2018; 370 (02) 708-717
  • 398 Harden JL, Egilmez NK. Indoleamine 2,3-dioxygenase and dendritic cell tolerogenicity. Immunol Invest 2012; 41 6–7 738-64
  • 399 Laumet G. et al Resolution of inflammation-induced depression requires T lymphocytes and endogenous brain interleukin-10 signaling. Neuropsychopharmacology 2018
  • 400 Smith PM. et al The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013; 341 6145 569-73
  • 401 Zeng H. et al Metabolic control of regulatory T cell development and function. Trends Immunol 2015; 36 (01) 3-12
  • 402 Vergnu IM. et al Manipulation of the endocytic pathway and phagocyte functions by Mycobacterium tuberculosis lipoarabinomannan. Front Cell Infect Microbiol 2014; 4: 187
  • 403 Konieczna P. et al Human dendritic cell DC-SIGN and TLR-2 mediate complementary immune regulatory activities in response to Lactobacillus rhamnosus JB-1. PLoS One 2015; 10 (03) e0120261
  • 404 United Nations, D. o. E. a. S. A., Population Division, World Urbanization Prospects: The, 2014 Revision Highlights (ST/ESA/SER.A/352) 2014
  • 405 Nicolaou N. et al Allergic disease in urban and rural populations: increasing prevalence with increasing urbanization. Allergy 2005; 60 (11) 1357-1360
  • 406 Braun-Fahrländer C. et al Environmental Exposure to Endotoxin and Its Relation to Asthma in School-Age Children. New England Journal of Medicine 2002; 347 (12) 869-877
  • 407 Riedler J. et al Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. The Lancet 2001; 358 9288 1129-1133
  • 408 Ege MJ. et al Exposure to Environmental Microorganisms and Childhood Asthma. New England Journal of Medicine 2011; 364 (08) 701-709
  • 409 Hakanen E. et al Urban environment predisposes dogs and their owners to allergic symptoms. Sci Rep 2018; 8 (01) 1585
  • 410 Lehtimaki J. et al Skin microbiota and allergic symptoms associate with exposure to environmental microbes. Proc Natl Acad Sci U S A 2018; 115 (19) 4897-4902
  • 411 Peen J. et al The current status of urban-rural differences in psychiatric disorders. Acta Psychiatrica Scandinavica 2010; 121 (02) 84-93
  • 412 Rook GA. Regulation of the immune system by biodiversity from the natural environment: an ecosystem service essential to health. Proc Natl Acad Sci U S A 2013; 110 (46) 18360-7
  • 413 Vassos E. et al Urban-rural differences in incidence rates of psychiatric disorders in Denmark. The British Journal of Psychiatry 2016; 208 (05) 435-440
  • 414 Pedersen CB. et al Evidence of a dose-response relationship between urbanicity during upbringing and schizophrenia risk. Arch Gen Psychiatry 2001; 58 (11) 1039-46
  • 415 Mortensen PB. et al Effects of Family History and Place and Season of Birth on the Risk of Schizophrenia. New England Journal of Medicine 1999; 340 (08) 603-608
  • 416 Pedersen CB. et al Family history, place and season of birth as risk factors for schizophrenia in Denmark: A replication and reanalysis. British Journal of Psychiatry 2001; 179 (01) 46-52
  • 417 Lewis G. et al Schizophrenia and city life. The Lancet 1992; 340 8812 137-140
  • 418 Pedersen C. et al Evidence of a dose-response relationship between urbanicity during upbringing and schizophrenia risk. Archives of General Psychiatry 2001; 58 (11) 1039-1046
  • 419 Kirschbaum C. et al The ‘Trier Social Stress Test’ – a tool for investigating psychobiological stress responses in a laboratory setting. Neuropsychobiology 1993; 28 1–2 76-81
  • 420 Rook GA. et al The Hygiene Hypothesis and Darwinian Medicine. 2009 Birkhäuser
  • 421 Hopfenspirger MT. et al Airway hyperresponsiveness, late allergic response, and eosinophilia are reversed with mycobacterial antigens in ovalbumin-presensitized mice. J Immunol 2002; 168 (05) 2516-22
  • 422 Hunt JR. et al Intragastric administration of Mycobacterium vaccae inhibits severe pulmonary allergic inflammation in a mouse model. Clin Exp Allergy 2005; 35 (05) 685-90
  • 423 Zuany-Amorim C. et al Long-term protective and antigen-specific effect of heat-killed Mycobacterium vaccae in a murine model of allergic pulmonary inflammation. J Immunol 2002; 169 (03) 1492-9
  • 424 Zuany-Amorim C. et al Suppression of airway eosinophilia by killed Mycobacterium vaccae-induced allergen-specific regulatory T-cells. Nat Med 2002; 8 (06) 625-9
  • 425 Akkoc T. et al Mycobacterium vaccae immunization to OVA sensitized pregnant BALB/c mice suppressed placental and postnatal IL-5 and inducing IFN-gamma secretion. Immunopharmacol Immunotoxicol 2008; 30 (01) 1-11
  • 426 Akkoc T. et al Mycobacterium vaccae immunization to pregnant BALB/c mice ameliorated lung histophathology and bone marrow eosinophila in ovalbumin sensitized offsprings. Open Journal of Immunology 2015; 4: 31-41
  • 427 Dwivedi M. et al Induction of regulatory T cells: A role for probiotics and prebiotics to suppress autoimmunity. Autoimmun Rev 2016; 15 (04) 379-92
  • 428 O’Brien ME. et al A randomized phase II study of SRL172 (Mycobacterium vaccae) combined with chemotherapy in patients with advanced inoperable non-small-cell lung cancer and mesothelioma. Br J Cancer 2000; 83 (07) 853-7
  • 429 O’Brien ME. et al SRL172 (killed Mycobacterium vaccae) in addition to standard chemotherapy improves quality of life without affecting survival, in patients with advanced non-small-cell lung cancer: phase III results. Ann Oncol 2004; 15 (06) 906-14
  • 430 Lowry CA. et al Identification of an immune-responsive mesolimbocortical serotonergic system: potential role in regulation of emotional behavior. Neuroscience 2007; 146 (02) 756-72
  • 431 Siebler PH. et al Acute Administration of the Nonpathogenic, Saprophytic Bacterium, Mycobacterium vaccae, Induces Activation of Serotonergic Neurons in the Dorsal Raphe Nucleus and Antidepressant-Like Behavior in Association with Mild Hypothermia. Cellular and Molecular Neurobiology 2017
  • 432 Amat J. Medial prefrontal cortex determines how stressor controllability affects behavior and dorsal raphe nucleus. Nat Neurosci 2005; 8 (03) 365-71
  • 433 Fox JH. et al Preimmunization with a heat-killed preparation of Mycobacterium vaccae enhances fear extinction in the fear-potentiated startle paradigm. Brain, Behavior, and Immunity 2017
  • 434 Russo AS. et al Acoustic startle response in rats predicts inter-individual variation in fear extinction. Neurobiology of Learning and Memory 2017; 139: 157-164
  • 435 Frank MG. et al Immunization with Mycobacterium vaccae induces an anti-inflammatory milieu in the CNS: Attenuation of stress-induced microglial priming, alarmins and anxiety-like behavior. Brain Behav Immun 2018
  • 436 Fonken LK. et al Mycobacterium vaccae immunization protects aged rats from surgery-elicited neuroinflammation and cognitive dysfunction. Neurobiology of Aging 2018 in press
  • 437 Martinez I. et al The gut microbiota of rural papua new guineans: composition, diversity patterns, and ecological processes. Cell Rep 2015; 11 (04) 527-38
  • 438 Arnold IC. et al The immunomodulatory properties of Helicobacter pylori confer protection against allergic and cronic inflammatory disorders. Front Cell Infect Microbiol 2012; 2: 10
  • 439 Pounder RE. et al The prevalence of Helicobacter pylori infection in different countries. Aliment Pharmacol Ther 1995; 9 (Suppl. 02) 33-9
  • 440 American-Psychiatric-Association Diagnostic and statistical manual of mental disorders. Washington, DC: 2011
  • 441 Boscarino JA. Diseases among men 20 years after exposure to severe stress: implications for clinical research and medical care. Psychosom Med 1997; 59 (06) 605-14
  • 442 Daskalakis NP. et al Endocrine aspects of post-traumatic stress disorder and implications for diagnosis and treatment. Endocrinol Metab Clin North Am 2013; 42 (03) 503-13
  • 443 Yehuda R. et al Circadian rhythm of salivary cortisol in Holocaust survivors with and without PTSD. Am J Psychiatry 2005; 162 (05) 998-1000
  • 444 Yehuda R. et al The ACTH response to dexamethasone in PTSD. Am J Psychiatry 2004; 161 (08) 1397-403
  • 445 Wieck A. et al Neuroimmunoendocrine Interactions in Post-Traumatic Stress Disorder: Focus on Long-Term Implications of Childhood Maltreatment. Neuroimmunomodulation 2014; 21 2–3 145-151
  • 446 Zhou J. et al Dysregulation in microRNA expression is associated with alterations in immune functions in combat veterans with post-traumatic stress disorder. PLoS One 2014; 9 (04) e94075
  • 447 Glaesmer H. et al The association of traumatic experiences and posttraumatic stress disorder with physical morbidity in old age: a German population-based study. Psychosom Med 2011; 73 (05) 401-6
  • 448 Glaesmer H. et al Posttraumatic stress disorder and its comorbidity with depression and somatisation in the elderly – a German community-based study. Aging Ment Health 2012; 16 (04) 403-12
  • 449 Foertsch S. et al Chronic psychosocial stress disturbs long-bone growth in adolescent mice. Disease Models & amp; Mechanisms 2017; 10 (12) 1399-1409
  • 450 Haffner-Luntzer M. Chronic psychosocial stress disturbs bone fracture healing via β-adrenoceptor signaling. Unpublished 2018
  • 451 Petrakis IL. et al Noradrenergic vs Serotonergic Antidepressant with or without Naltrexone for Veterans with PTSD and Comorbid Alcohol Dependence. Neuropsychopharmacology 2011; 37: 996
  • 452 Fear NT. et al What are the consequences of deployment to Iraq and Afghanistan on the mental health of the UK armed forces? A cohort study. The Lancet 2010; 375 9728 1783-1797
  • 453 Peters S. et al Differential effects of baclofen and oxytocin on the increased ethanol consumption following chronic psychosocial stress in mice. Addiction Biology 2013; 18 (01) 66-77