Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-pjpqr Total loading time: 0 Render date: 2024-07-07T10:42:31.868Z Has data issue: false hasContentIssue false

Chapter 1 - Basic Concepts in Immunobiology

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

Once thought to be an immune-privileged site, we now know that there is a complex and essential bidirectional interplay between the central nervous system (CNS) and the immune system (1). Technological advances in imaging, genomic medicine and immunology have resulted in major revisions to some of the most fundamental and long-held assumptions in neuroscience, and we now understand that the immune system is critically involved not only in brain pathology, but also in the normal processes of brain development and homeostasis.

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

Savitz, J, Harrison, NA. Interoception and Inflammation in Psychiatric Disorders. Biol Psychiatry Cogn Neurosci Neuroimaging. 2018 Jun;3(6):514–24.Google Scholar
Dowlati, Y, Herrmann, N, Swardfager, W, et al. A meta-analysis of cytokines in major depression. Biological Psychiatry. 2010 Mar 1;67(5):446–57.CrossRefGoogle ScholarPubMed
Miller, BJ, Buckley, P, Seabolt, W, Mellor, A, Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biological Psychiatry. 2011 Oct 1;70(7):663–71.Google Scholar
Cunningham, C, Hennessy, E. Co-morbidity and systemic inflammation as drivers of cognitive decline: new experimental models adopting a broader paradigm in dementia research. Alzheimers Res Ther. BioMed Central Ltd. 2015;7(1):33.Google Scholar
Banks, WA. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nature Publishing Group. 2016 Apr;15(4):275–92.Google Scholar
Engelhardt, B, Sorokin, L. The blood-brain and the blood-cerebrospinal fluid barriers: function and dysfunction. Semin Immunopathol. 2009 Nov;31(4):497511.Google Scholar
Liddelow, SA. Development of the choroid plexus and blood-CSF barrier. Front Neurosci. Frontiers. 2015 Mar 3;9(154):319–13.Google Scholar
Sevigny, J, Chiao, P, Bussière, T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016 Sep 1;537(7618):50–6.Google Scholar
Ransohoff, RM, Cardona, AE. The myeloid cells of the central nervous system parenchyma. Nature. 2010 Nov 11;468(7321):253–62.Google Scholar
Li, Q, Barres, BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2017 Nov 20.Google Scholar
Ransohoff, RM, Engelhardt, B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol. 2012 Sep;12(9):623–35.Google Scholar
Esposito, P, Gheorghe, D, Kandere, K, et al. Acute stress increases permeability of the blood-brain-barrier through activation of brain mast cells. Brain Res. 2001 Jan 5;888(1):117–27.Google Scholar
Davies, LC, Jenkins, SJ, Allen, JE, Taylor, PR. Tissue-resident macrophages. Nat Immunol. 2013 Oct;14(10):986–95.CrossRefGoogle ScholarPubMed
Ginhoux, F, Greter, M, Leboeuf, M, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010 Nov 5;330(6005):841–5.Google Scholar
Ajami, B, Bennett, JL, Krieger, C, Tetzlaff, W, Rossi, FMV. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007 Dec;10(12):1538–43.Google Scholar
Mildner, A, Schmidt, H, Nitsche, M, et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci. 2007 Dec;10(12):1544–53.Google Scholar
Nikkilä, HV, Müller, K, Ahokas, A, et al. Accumulation of macrophages in the CSF of schizophrenic patients during acute psychotic episodes. Am J Psychiatry. 1999 Nov;156(11):1725–9.CrossRefGoogle ScholarPubMed
Jackson, AJ, Miller, BJ. Meta-analysis of total and differential white blood cell counts in schizophrenia. Acta Psychiatr Scand. 2020 Jul;142(1):1826.Google Scholar
Padmos, RC, Hillegers, MHJ, 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. American Medical Association; 2008 Apr;65(4):395407.Google Scholar
Nimmerjahn, A, Kirchhoff, F, Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005 May 27;308(5726):1314–8.CrossRefGoogle ScholarPubMed
Davalos, D, Grutzendler, J, Yang, G, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005 Jun;8(6):752–8.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 Mar;72(3):268–75.Google Scholar
Tay, TL, Béchade, C, D’Andrea, I, et al. Microglia gone rogue: impacts on psychiatric disorders across the lifespan. Front Mol Neurosci. 2017;10:421.Google Scholar
Williamson, LL, Sholar, PW, Mistry, RS, Smith, SH, Bilbo, SD. Microglia and memory: modulation by early-life infection. J Neurosci. 2011 Oct 26;31(43):15511–21.Google Scholar
Marin, I, Kipnis, J. Learning and memory … and the immune system. Learn Mem. 2013 Sep 19;20(10):601–6.Google Scholar
Holmes, C, Cunningham, C, Zotova, E, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009 Sep 8;73(10):768–74.Google Scholar
Erblich, B, Zhu, L, Etgen, AM, Dobrenis, K, Pollard, JW. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS ONE. 2011;6(10):e26317.CrossRefGoogle ScholarPubMed
Elmore, MRP, Najafi, AR, Koike, MA, et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron. 2014 Apr 16;82(2):380–97.Google Scholar
Olmos-Alonso, A, Schetters, STT, Sri, S, et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain. 2016 Mar;139(Pt 3):891907.Google Scholar
Acharya, MM, Green, KN, Allen, BD, et al. Elimination of microglia improves cognitive function following cranial irradiation. Sci Rep. 2016 Aug 12;6(1):31545.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 May;54:3844.Google Scholar
Suzuki, H, Savitz, J, Teague, TK, et al. Altered populations of natural killer cells, cytotoxic T lymphocytes, and regulatory T cells in major depressive disorder: Association with sleep disturbance. Brain Behavior and Immunity. 2017 Nov 1;66:193200.Google Scholar
Grosse, L, Carvalho, LA, Birkenhager, TK, et al. Circulating cytotoxic T cells and natural killer cells as potential predictors for antidepressant response in melancholic depression. Restoration of T regulatory cell populations after antidepressant therapy.Psychopharmacology (Berl). 2016 May;233(9):1679–88.Google Scholar
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 Behavior and Immunity. 2000 Sep;14(3):153–69.Google Scholar
Schedlowski, M, Jacobs, R, Stratmann, G, et al. Changes of natural killer cells during acute psychological stress. J Clin Immunol. 1993 Mar;13(2):119–26.Google Scholar
Segerstrom, SC, Miller, GE. Psychological stress and the human immune system: a meta-analytic study of 30 years of inquiry. Psychol Bull. 2004 Jul;130(4):601–30.Google Scholar
Duggal, NA, Upton, J, Phillips, AC, Hampson, P, Lord, JM. NK cell immunesenescence is increased by psychological but not physical stress in older adults associated with raised cortisol and reduced perforin expression. Age (Dordr). 2015 Feb;37(1):9748.Google Scholar
Okun, E, Griffioen, KJ, Mattson, MP. Toll-like receptor signaling in neural plasticity and disease. Trends Neurosci. 2011 May;34(5):269–81.Google Scholar
Tanga, FY, Nutile-McMenemy, N, DeLeo, JA. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proceedings of the National Academy of Sciences. National Academy of Sciences. 2005 Apr 19;102(16):5856–61.Google Scholar
Hutchinson, MR, Zhang, Y, Brown, K, et al. Non-stereoselective reversal of neuropathic pain by naloxone and naltrexone: involvement of toll-like receptor 4 (TLR4). Eur J Neurosci. 2008 Jul;28(1):20–9.CrossRefGoogle ScholarPubMed
Hutchinson, MR, Zhang, Y, Shridhar, M, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behavior and Immunity. 2010 Jan;24(1):8395.Google Scholar
Qu, J, Tao, X-Y, Teng, P, et al. Blocking ATP-sensitive potassium channel alleviates morphine tolerance by inhibiting HSP70-TLR4-NLRP3-mediated neuroinflammation. J Neuroinflammation. BioMed Central. 2017 Nov 25;14(1):228.Google Scholar
Hutchinson, MR, Northcutt, AL, Hiranita, T, et al. Opioid Activation of Toll-Like Receptor 4 Contributes to Drug Reinforcement. Journal of Neuroscience. 2012 Aug 15;32(33):11187–200.Google Scholar
Pandey, GN, Rizavi, HS, Ren, X, Bhaumik, R, Dwivedi, Y. Toll-like receptors in the depressed and suicide brain. Journal of Psychiatric Research. 2014 Jun 1;53(C):62–8.Google Scholar
Hung, Y-Y, Kang, H-Y, Huang, K-W, Huang, T-L. Association between toll-like receptors expression and major depressive disorder. Psychiatry Research. 2014 Dec 15;220(1–2):283–6.Google Scholar
Latz, E, Xiao, TS, Stutz, A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013 Jun;13(6):397411.Google Scholar
Alcocer-Gómez, E, de Miguel, M, Casas-Barquero, N, et al. NLRP3 inflammasome is activated in mononuclear blood cells from patients with major depressive disorder. Brain Behavior and Immunity. 2014 Feb;36:111–7.Google Scholar
Zhang, Y, Liu, L, Liu, YZ, et al. NLRP3 Inflammasome Mediates Chronic Mild Stress-Induced Depression in Mice via Neuroinflammation. Int J Neuropsychopharm. 2015 May 29;18(8):pyv0066.Google Scholar
Kim, HK, Andreazza, AC, Elmi, N, Chen, W, Young, LT. Nod-like receptor pyrin containing 3 (NLRP3) in the post-mortem frontal cortex from patients with bipolar disorder: A potential mediator between mitochondria and immune-activation. Journal of Psychiatric Research. 2016 Jan 1;72(C):4350.Google Scholar
Heneka, MT, Kummer, MP, Stutz, A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013 Jan 23;493(7434):674–8.Google Scholar
Iwata, M, Ota, KT, Li, X-Y, et al. Psychological Stress Activates the Inflammasome via Release of Adenosine Triphosphate and Stimulation of the Purinergic Type 2X7 Receptor. Biological Psychiatry. 2016 Jul 1;80(1):1222.Google Scholar
Smith, KJ, Au, B, Ollis, L, Schmitz, N. The association between C-reactive protein, Interleukin-6 and depression among older adults in the community: A systematic review and meta-analysis. Exp Gerontol. 2018 Feb 1;102:109–32.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 Sep 25;150(3):736–44.Google Scholar
Wium-Andersen, MK, Ørsted, DD, Nielsen, SF, Nordestgaard, BG. Elevated C-reactive protein levels, psychological distress, and depression in 73, 131 individuals. JAMA Psychiatry. 2013 Feb;70(2):176–84.Google Scholar
Khandaker, GM, Pearson, RM, Zammit, S, Lewis, G, Jones, PB. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry. 2014 Oct;71(10):1121–8.CrossRefGoogle ScholarPubMed
Uher, R, Tansey, KE, Dew, T, et al. An inflammatory biomarker as a differential predictor of outcome of depression treatment with escitalopram and nortriptyline. Am J Psychiatry. 2014 Dec 1;171(12):1278–86.Google Scholar
Inoshita, M, Numata, S, Tajima, A, et al. A significant causal association between C-reactive protein levels and schizophrenia. Sci Rep. 2016 May 19;6(1):26105.Google Scholar
Fernandes, BS, Steiner, J, Bernstein, H-G, et al. C-reactive protein is increased in schizophrenia but is not altered by antipsychotics: meta-analysis and implications. Mol Psychiatry. 2016 Apr;21(4):554–64.Google Scholar
Fernandes, BS, Steiner, J, Molendijk, ML, et al. C-reactive protein concentrations across the mood spectrum in bipolar disorder: a systematic review and meta-analysis. 2016 Dec;3(12):1147–56.Google Scholar
Khakzad, MR, Javanbakht, M, Shayegan, MR, et al. The complementary role of high sensitivity C-reactive protein in the diagnosis and severity assessment of autism. Research in Autism Spectrum Disorders. 2012 May 5;6(3):1032–7.Google Scholar
Orsini, F, De Blasio, D, Zangari, R, Zanier, ER, De Simoni, M-G. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci. 2014;8:380.Google Scholar
Sekar, A, Bialas, AR, de Rivera, H, et al. Schizophrenia risk from complex variation of complement component 4. Nature. 2016 Feb 11;530(7589):177–83.Google Scholar
Haijma, SV, Van Haren, N, Cahn, W, et al. Brain volumes in schizophrenia: a meta-analysis in over 18 000 subjects. Schizophr Bull. 2013 Sep;39(5):1129–38.Google Scholar
Osimo, EF, Beck, K, Reis Marques, T, Howes, OD. Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry. 2019 Apr;24(4):549–61.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 Jan;9(1):4656.Google Scholar
Banks, WA. Blood-brain barrier transport of cytokines: a mechanism for neuropathology. Curr Pharm Des. 2005;11(8):973–84.Google Scholar
Krueger, JM, Rector, DM, Roy, S, et al. Sleep as a fundamental property of neuronal assemblies. 2008 Nov 5;9(12):910–9.CrossRefGoogle Scholar
Rachal Pugh, C, Fleshner, M, Watkins, LR, Maier, SF, Rudy, JW. The immune system and memory consolidation: a role for the cytokine IL-1beta. Neurosci Biobehav Rev. 2001 Jan;25(1):2941.Google Scholar
Allan, SM, Tyrrell, PJ, Rothwell, NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005 Aug;5(8):629–40.Google Scholar
Conductier, G, Blondeau, N, Guyon, A, Nahon, J-L, Rovère, C. The role of monocyte chemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. Journal of Neuroimmunology. 2010 Jul 27;224(1–2):93100.Google Scholar
Eyre, HA, Air, T, Pradhan, A, et al. A meta-analysis of chemokines in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2016 Jul 4;68:18.Google Scholar
Leighton, SP, Nerurkar, L, Krishnadas, R, et al. Chemokines in depression in health and in inflammatory illness: a systematic review and meta-analysis. Mol Psychiatry. 2017 Nov 14;7(1):72–58.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 May;42:93115.Google Scholar
Haapakoski, R, Mathieu, J, Ebmeier, KP, Alenius, H, Kivimäki, M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behavior and Immunity. 2015 Oct;49:206–15.Google Scholar
Rodrigues-Amorim, D, Rivera-Baltanás, T, Spuch, C, et al. Cytokines dysregulation in schizophrenia: A systematic review of psychoneuroimmune relationship. Schizophrenia Research. 2018 Jul;197:1933.Google Scholar
Miller, GE, Cole, SW. Clustering of depression and inflammation in adolescents previously exposed to childhood adversity. Biological Psychiatry. 2012 Jul 1;72(1):3440.Google Scholar
Baumeister, D, Akhtar, R, Ciufolini, S, Pariante, CM, Mondelli, V. Childhood trauma and adulthood inflammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α. Mol Psychiatry. Nature Publishing Group; 2016 May;21(5):642–9.Google Scholar
Musselman, DL, Lawson, DH, Gumnick, JF, et al. Paroxetine for the prevention of depression induced by high-dose interferon alfa. N Engl J Med. 2001 Mar 29;344(13):961–6.Google Scholar
Corvin, A, Morris, DW. Genome-wide Association Studies: Findings at the Major Histocompatibility Complex Locus in Psychosis. Biological Psychiatry. 2014 Feb 15;75(4):276–83.Google Scholar
Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014 Jul 24;511(7510):421–7.Google Scholar
Kebir, H, Kreymborg, K, Ifergan, I, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med. 2007 Oct;13(10):1173–5.Google Scholar
Reboldi, A, Coisne, C, Baumjohann, D, et al. C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol. 2009 May;10(5):514–23.Google Scholar
Stromnes, IM, Cerretti, LM, Liggitt, D, Harris, RA, Goverman, JM. Differential regulation of central nervous system autoimmunity by T(H)1 and T(H)17 cells. Nat Med. 2008 Mar;14(3):337–42.Google Scholar
Avgustin, B, Wraber, B, Tavcar, R. Increased Th1 and Th2 immune reactivity with relative Th2 dominance in patients with acute exacerbation of schizophrenia. Croatian Medical Journal. 2005 Apr;46(2):268–74.Google Scholar
Brambilla, P, Bellani, M, Isola, M, et al. Increased M1/decreased M2 signature and signs of Th1/Th2 shift in chronic patients with bipolar disorder, but not in those with schizophrenia. Transl Psychiatry. 2014 Jul 1;4(7):e406–6.Google Scholar
Debnath, M, Berk, M. Th17 pathway-mediated immunopathogenesis of schizophrenia: mechanisms and implications. Schizophrenia Bulletin. Oxford University Press; 2014 Nov;40(6):1412–21.Google Scholar
Ding, M, Song, X, Zhao, J, et al. Activation of Th17 cells in drug naïve, first episode schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2014 Jun 3;51:7882.Google Scholar
Fernandez-Egea, E, Vértes, PE, Flint, SM, et al. Peripheral Immune Cell Populations Associated with Cognitive Deficits and Negative Symptoms of Treatment-Resistant Schizophrenia. PLoS ONE. 2016;11(5):e0155631.Google Scholar
Røge, R, Møller, BK, Andersen, CR, Correll, CU, Nielsen, J. Immunomodulatory effects of clozapine and their clinical implications: what have we learned so far? Schizophrenia Research. Elsevier B.V. 2012 Sep 1;140(1–3):204–13.Google Scholar
Chen, M-L, Tsai, T-C, Lin, Y-Y, et al. Antipsychotic drugs suppress the AKT/NF-kB pathway and regulate the differentiation of T-cell subsets. Immunology Letters. Elsevier B.V. 2011 Oct 30;140(1–2):8191.Google Scholar
Chen, M-L, Tsai, T-C, Wang, L-K, et al. Clozapine inhibits Th1 cell differentiation and causes the suppression of IFN-γ production in peripheral blood mononuclear cells. Immunopharmacol Immunotoxicol. 2012 Aug;34(4):686–94.Google Scholar
O’Sullivan, D, Green, L, Stone, S, et al. Treatment with the antipsychotic agent, risperidone, reduces disease severity in experimental autoimmune encephalomyelitis. PLoS ONE. 2014;9(8):e104430.Google Scholar
Shatz, CJ. MHC class I: an unexpected role in neuronal plasticity. Neuron. 2009 Oct 15;64(1):40–5.Google Scholar
de Graaf, MT, Smitt, PAES, Luitwieler, RL, et al. Central memory CD4+ T cells dominate the normal cerebrospinal fluid. Cytometry B Clin Cytom. 2011 Jan;80(1):4350.Google Scholar
Nikkilä, H, Müller, K, Ahokas, A, et al. Abnormal distributions of T-lymphocyte subsets in the cerebrospinal fluid of patients with acute schizophrenia. Schizophrenia Research. 1995 Feb;14(3):215–21.Google Scholar
Moalem, G, Leibowitz-Amit, R, Yoles, E, et al. Autoimmune T cells protect neurons from secondary degeneration after central nervous system axotomy. Nat Med. 1999 Jan;5(1):4955.Google Scholar
Kipnis, J, Mizrahi, T, Hauben, E, et al. Neuroprotective autoimmunity: naturally occurring CD4+CD25+ regulatory T cells suppress the ability to withstand injury to the central nervous system. Proc Natl Acad Sci USA. National Acad Sciences. 2002 Nov 26;99(24):15620–5.Google Scholar
Kipnis, J, Cardon, M, Avidan, H, et al. Dopamine, through the extracellular signal-regulated kinase pathway, downregulates CD4+CD25+ regulatory T-cell activity: implications for neurodegeneration. J Neurosci. Society for Neuroscience. 2004 Jul 7;24(27):6133–43.Google ScholarPubMed
Walsh, JT, Zheng, J, Smirnov, I, Lorenz, U, Tung, K, Kipnis, J. Regulatory T cells in central nervous system injury: a double-edged sword. J Immunol. 2014 Nov 15;193(10):5013–22.Google Scholar
Kipnis, J, Gadani, S, Derecki, NC. Pro-cognitive properties of T cells. Nat Rev Immunol. 2012 Sep;12(9):663–9.Google Scholar
Dalmau, J, Tüzün, E, Wu, H-Y, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol. 2007 Jan;61(1):2536.Google Scholar
Lennox, BR, Palmer-Cooper, EC, Pollak, T, et al. Prevalence and clinical characteristics of serum neuronal cell surface antibodies in first-episode psychosis: a case-control study. Lancet Psychiatry. 2017 Jan;4(1):42–8.Google Scholar
Brimberg, L, Mader, S, Jeganathan, V, et al. Caspr2-reactive antibody cloned from a mother of an ASD child mediates an ASD-like phenotype in mice. Mol Psychiatry. 2016 Dec;21(12):1663–71.Google Scholar
Brudek, T, Winge, K, Folke, J, et al. Autoimmune antibody decline in Parkinson’s disease and Multiple System Atrophy; a step towards immunotherapeutic strategies. Mol Neurodegener. 2017 Jun 7;12(1):44.Google Scholar
Iliff, JJ, Wang, M, Liao, Y, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Science Translational Medicine. American Association for the Advancement of Science. 2012 Aug 15;4(147):147ra1111.Google Scholar
Louveau, A, Smirnov, I, Keyes, TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015 Jun 1;523(7560):337–41.Google Scholar
Aspelund, A, Antila, S, Proulx, ST, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med. 2015 Jun 29;212(7):991–9.Google Scholar
Da Mesquita, S, Fu, Z, Kipnis, J. The Meningeal Lymphatic System: A New Player in Neurophysiology. Neuron. 2018 Oct 24;100(2):375–88.Google Scholar
Louveau, A, Da Mesquita, S, Kipnis, J. Lymphatics in neurological disorders: a neuro-lympho-vascular component of multiple sclerosis and Alzheimer’s disease? Neuron. Elsevier Inc. 2016 Sep 7;91(5):957–73.Google Scholar
Harrison, NA, Brydon, L, Walker, C, et al. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biological Psychiatry. 2009 Sep 1;66(5):407–14.Google Scholar
Harrison, NA, Doeller, CF, Voon, V, Burgess, N, Critchley, HD. Peripheral Inflammation Acutely Impairs Human Spatial Memory via Actions on Medial Temporal Lobe Glucose Metabolism. Biological Psychiatry. 2014 Oct 1;76(7):585–93.Google Scholar
Tracey, KJ. Reflex control of immunity. Nat Rev Immunol. 2009 Jun;9(6):418–28.Google Scholar
Bellinger, DL, Millar, BA, Perez, S, et al. Sympathetic modulation of immunity: Relevance to disease. Cell Immunol. 2008;252(1–2):2756.Google Scholar
Sloan, EK, Capitanio, JP, Tarara, RP, et al. Social Stress Enhances Sympathetic Innervation of Primate Lymph Nodes: Mechanisms and Implications for Viral Pathogenesis. Journal of Neuroscience. 2007 Aug 15;27(33):8857–65.Google Scholar
Pacheco, R, Riquelme, E, Kalergis, AM. Emerging evidence for the role of neurotransmitters in the modulation of T cell responses to cognate ligands. Cent Nerv Syst Agents Med Chem. 2010 Mar;10(1):6583.Google Scholar
Ilani, T, Ben-Shachar, D, Strous, RD, et al. A peripheral marker for schizophrenia: Increased levels of D3 dopamine receptor mRNA in blood lymphocytes. Proc Natl Acad Sci USA. National Acad Sciences. 2001 Jan 16;98(2):625–8.Google Scholar
Brito-Melo, GEA, Nicolato, R, de Oliveira, ACP, et al. Increase in dopaminergic, but not serotoninergic, receptors in T-cells as a marker for schizophrenia severity. Journal of Psychiatric Research. 2012 Jun 1;46(6):738–42.Google Scholar
Nagai, Y, Ueno, S, Saeki, Y, et al. Decrease of the D3 dopamine receptor mRNA expression in lymphocytes from patients with Parkinson’s disease. Neurology. 1996 Mar 1;46(3):791–5.Google Scholar
Thomson, CA, McColl, A, Cavanagh, J, Graham, GJ. Peripheral inflammation is associated with remote global gene expression changes in the brain. J Neuroinflammation. 2014 Apr 8;11(1):73.Google Scholar
Miller, AH, Raison, CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nature. 2016 Jan;16(1):2234.Google Scholar
Harrison, NA. Brain Structures Implicated in Inflammation-Associated Depression. Curr Top Behav Neurosci. 2017;31:221–48.Google Scholar
Capuron, L, Ravaud, A, Neveu, PJ, et al. Association between decreased serum tryptophan concentrations and depressive symptoms in cancer patients undergoing cytokine therapy. Mol Psychiatry. 2002 Jun 18;7(5):468–73.CrossRefGoogle ScholarPubMed
Capuron, L, Gumnick, JF, Musselman, DL, et al. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology. 2002 May;26(5):643–52.Google Scholar
Krause, D, Myint, A-M, Schuett, C, et al. High Kynurenine (a Tryptophan Metabolite) Predicts Remission in Patients with Major Depression to Add-on Treatment with Celecoxib. Front Psychiatry. 2017;8:16.Google Scholar
Abbasi, S-H, Hosseini, F, Modabbernia, A, Ashrafi, M, Akhondzadeh, S. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: randomized double-blind placebo-controlled study. J Affect Disord. 2012 Dec 10;141(2–3):308–14.Google Scholar
O’Connor, JC, Lawson, MA, André, C, et al. Induction of IDO by bacille Calmette-Guérin is responsible for development of murine depressive-like behaviour. J Immunol. 2009 Mar 1;182(5):3202–12.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
×