Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-cjp7w Total loading time: 0 Render date: 2024-06-27T21:43:23.898Z Has data issue: false hasContentIssue false

Chapter 3 - Stress, Immune System and the Brain

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

Stress encompasses the psychological perception of pressure from the environment, and the body’s physiological response to it. The sources of stress have evolved over time, from predation and natural disasters, to things like interpersonal conflicts and economic insecurities. While in the past, stressors evoked a very acute physical ‘fight or flight’ response, these events are rare in today’s terms. In contrast, the stressors we experience in the modern world are arguably more trivial – they are not often immediately life threatening – however they are more persistent, manifesting as a chronic, low level source of anxiety in our daily lives. The natural stress response involves multiple systems and is designed to provide short-term beneficial effects to the individual to help see them through a threatening situation. It is thought this response is mediated largely through glucocorticoid (GC) production and will rapidly normalize following the stressful event. In the event of chronic exposure to stress, some of these short-term physiological changes fail to return to ‘normality’, and as a result, the nature of our homeostasis is altered. This chapter will focus on the changes to the immune system and brain mediated through exposure to stress, with particular emphasis on the detrimental effects of chronic stress.

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

Ulrich-Lai, YM, Herman, JP. Neural regulation of endocrine and autonomic stress responses. Nature Reviews Neuroscience. 2009;10:397.Google Scholar
Morey, JN, Boggero, IA, Scott, AB, Segerstrom, SC. Current directions in stress and human immune function. Current Opinion in Psychology. 2015;5:13–7.Google Scholar
Dhabhar, FS, Malarkey, WB, Neri, E, McEwen, BS. Stress-induced redistribution of immune cells – From barracks to boulevards to battlefields: a tale of three hormones – Curt Richter Award Winner. Psychoneuroendocrinology. 2012;37(9):1345–68.Google Scholar
Sternberg, EM. Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nature Reviews Immunology. 2006;6:318.Google Scholar
McEwen, BS. Physiology and neurobiology of stress and adaptation: central role of the brain. Physiological Reviews. 2007;87(3):873904.CrossRefGoogle ScholarPubMed
Bisht, K, Sharma, K, Tremblay, M-È. Chronic stress as a risk factor for Alzheimer’s disease: roles of microglia-mediated synaptic remodeling, inflammation, and oxidative stress. Neurobiology of Stress. 2018;9:921.Google Scholar
Brachman, RA, Lehmann, ML, Maric, D, Herkenham, M. Lymphocytes from chronically stressed mice confer antidepressant-like effects to naive mice. The Journal of Neuroscience. 2015;35(4):1530.Google Scholar
American Psychological Association. From stress to inflammation and major depressive disorder: a social signal transduction theory of depression (press release). 2014.Google Scholar
Kudryavtseva, NN, Bakshtanovskaya, IV, Koryakina, LA. Social model of depression in mice of C57BL/6J strain. Pharmacology Biochemistry and Behavior. 1991;38(2):315–20.Google Scholar
Weber, MD, Godbout, JP, Sheridan, JF. Repeated social defeat, neuroinflammation, and behavior: monocytes carry the signal. Neuropsychopharmacology. 2017;42(1):4661.Google Scholar
Reader, BF, Jarrett, BL, McKim, DB, et al. Peripheral and central effects of repeated social defeat stress: monocyte trafficking, microglial activation, and anxiety. Neuroscience. 2015;289:429–42.CrossRefGoogle ScholarPubMed
Willner, P. The chronic mild stress (CMS) model of depression: history, evaluation and usage. Neurobiology of Stress. 2016;6:7893.Google Scholar
Willner, P, Towell, A, Sampson, D, Sophokleous, S, Muscat, R. Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology. 1987;93(3):358–64.Google Scholar
Muscat, R, Willner, P. Suppression of sucrose drinking by chronic mild unpredictable stress: a methodological analysis. Neuroscience & Biobehavioral Reviews. 1992;16(4):507–17.CrossRefGoogle Scholar
Willner, P. Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. Psychopharmacology. 1997;134(4):319–29.Google Scholar
Murthy, S, Gould, E. Early life stress in rodents: animal models of illness or resilience? Frontiers in Behavioral Neuroscience. 2018;12:157.Google Scholar
Walker, C-D, Bath, KG, Joels, M, et al. Chronic early life stress induced by limited bedding and nesting (LBN) material in rodents: critical considerations of methodology, outcomes and translational potential. Stress. 2017;20(5):421–48.CrossRefGoogle ScholarPubMed
George, ED, Bordner, KA, Elwafi, HM, Simen, AA. Maternal separation with early weaning: a novel mouse model of early life neglect. BMC Neuroscience. 2010;11(1):123.CrossRefGoogle ScholarPubMed
Engler, H, Bailey, MT, Engler, A, Sheridan, JF. Effects of repeated social stress on leukocyte distribution in bone marrow, peripheral blood and spleen. Journal of Neuroimmunology. 2004;148(1):106–15.CrossRefGoogle ScholarPubMed
Powell, ND, Sloan, EK, Bailey, MT, et al. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via β-adrenergic induction of myelopoiesis. Proceedings of the National Academy of Sciences. 2013;110(41):16574.CrossRefGoogle ScholarPubMed
Wohleb, ES, Powell, ND, Godbout, JP, Sheridan, JF. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. The Journal of Neuroscience. 2013;33(34):13820.CrossRefGoogle Scholar
Heidt, T, Sager, HB, Courties, G, et al. Chronic variable stress activates hematopoietic stem cells. Nature Medicine. 2014;20:754.Google Scholar
Wohleb, ES, Hanke, ML, Corona, AW, et al. β-adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. The Journal of Neuroscience. 2011;31(17):6277.Google Scholar
Hanke, ML, Powell, ND, Stiner, LM, Bailey, MT, Sheridan, JF. Beta adrenergic blockade decreases the immunomodulatory effects of social disruption stress. Brain, Behavior, and Immunity. 2012;26(7):1150–9.Google Scholar
Bailey, MT, Engler, H, Powell, ND, Padgett, DA, Sheridan, JF. Repeated social defeat increases the bactericidal activity of splenic macrophages through a Toll-like receptor-dependent pathway. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2007;293(3):R1180–R90.Google ScholarPubMed
Powell, ND, Bailey, MT, Mays, JW, et al. Repeated social defeat activates dendritic cells and enhances Toll-like receptor dependent cytokine secretion. Brain, Behavior, and Immunity. 2009;23(2):225–31.Google Scholar
Gjerstad, JK, Lightman, SL, Spiga, F. Role of glucocorticoid negative feedback in the regulation of HPA axis pulsatility. Stress (Amsterdam, Netherlands). 2018;21(5):403–16.CrossRefGoogle ScholarPubMed
Avitsur, R, Stark, JL, Dhabhar, FS, Padgett, DA, Sheridan, JF. Social disruption-induced glucocorticoid resistance: kinetics and site specificity. Journal of Neuroimmunology. 2002;124(1):5461.Google Scholar
Wohleb, ES, McKim, DB, Shea, DT, et al. Re-establishment of anxiety in stress-sensitized mice is caused by monocyte trafficking from the spleen to the brain. Biological Psychiatry. 2014;75(12):970–81.CrossRefGoogle ScholarPubMed
Sawicki, CM, McKim, DB, Wohleb, ES, et al. Social defeat promotes a reactive endothelium in a brain region-dependent manner with increased expression of key adhesion molecules, selectins and chemokines associated with the recruitment of myeloid cells to the brain. Neuroscience. 2015;302:151–64.Google Scholar
O’Carroll, SJ, Kho, DT, Wiltshire, R, et al. Pro-inflammatory TNFα and IL-1β differentially regulate the inflammatory phenotype of brain microvascular endothelial cells. Journal of Neuroinflammation. 2015;12:131.CrossRefGoogle ScholarPubMed
Lakhan, SE, Kirchgessner, A, Tepper, D, Leonard, A. Matrix metalloproteinases and blood-brain barrier disruption in acute ischemic stroke. Frontiers in Neurology. 2013;4:32.CrossRefGoogle ScholarPubMed
Könnecke, H, Bechmann, I. The role of microglia and matrix metalloproteinases involvement in neuroinflammation and gliomas. Clin Dev Immunol. 2013;2013:914104.CrossRefGoogle ScholarPubMed
Ramirez, K, Niraula, A, Sheridan, JF. GABAergic modulation with classical benzodiazepines prevent stress-induced neuro-immune dysregulation and behavioral alterations. Brain, Behavior, and Immunity. 2016;51:154–68.Google Scholar
Frick, LR, Arcos, ML, Rapanelli, M, et al. Chronic restraint stress impairs T-cell immunity and promotes tumor progression in mice. Stress. 2009;12(2):134–43.Google Scholar
Budiu, RA, Vlad, AM, Nazario, L, et al. Restraint and social isolation stressors differentially regulate adaptive immunity and tumor angiogenesis in a breast cancer mouse model. Cancer Clin Oncol. 2017;6(1):1224.Google Scholar
Sommershof, A, Scheuermann, L, Koerner, J, Groettrup, M. Chronic stress suppresses anti-tumor TCD8+ responses and tumor regression following cancer immunotherapy in a mouse model of melanoma. Brain, Behavior, and Immunity. 2017;65:140–9.Google Scholar
Li, Q, Barres, BA. Microglia and macrophages in brain homeostasis and disease. Nature Reviews Immunology. 2017;18:225.Google Scholar
Frank, M, Fonken, L, Watkins, L, Maier, S. Microglia: Neuroimmune-sensors of stress. Seminars in Cell & Developmental Biology. 2019;94:176–85.Google Scholar
Hinwood, M, Tynan, RJ, Charnley, JL, et al. Chronic stress induced remodeling of the prefrontal cortex: structural re-organization of microglia and the inhibitory effect of minocycline. Cerebral Cortex. 2012;23(8):1784–97.Google Scholar
Walker, FR, Morandini, J, Day, TA, Hinwood, M. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cerebral Cortex. 2012;22(6):1442–54.Google Scholar
Churchward, MA, Michaud, ER, Todd, KG. Supporting microglial niches for therapeutic benefit in psychiatric disorders. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2019;94:109648.Google Scholar
Garrido-Mesa, N, Zarzuelo, A, Gálvez, J. Minocycline: far beyond an antibiotic. British Journal of Pharmacology. 2013;169(2):337–52.Google Scholar
McKim, DB, Patterson, JM, Wohleb, ES, et al. Sympathetic release of splenic monocytes promotes recurring anxiety following repeated social defeat. Biological Psychiatry. 2016;79(10):803–13.CrossRefGoogle ScholarPubMed
Cohen, S, Janicki-Deverts, D, Doyle, WJ, et al. Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(16):5995–9.Google Scholar
Kelly, KA, Michalovicz, LT, Miller, JV, et al. Prior exposure to corticosterone markedly enhances and prolongs the neuroinflammatory response to systemic challenge with LPS. PLoS ONE. 2018;13(1):e0190546.Google Scholar
Markham, JA, Greenough, WT. Experience-driven brain plasticity: beyond the synapse. Neuron Glia Biology. 2004;1(4):351–63.Google Scholar
Lara, AH, Wallis, JD. The role of prefrontal cortex in working memory: a mini review. Frontiers in Systems Neuroscience. 2015;9:173.Google Scholar
Domenech, P, Koechlin, E. Executive control and decision-making in the prefrontal cortex. Current Opinion in Behavioral Sciences. 2015;1:101–6.Google Scholar
Hosokawa, T, Nakamura, S, Matsui, Y, et al. The effect of inactivation of prefrontal cortex on immediate behavioral adaptation in group reversal task by offline repetitive transcranial magnetic stimulation (rTMS) in monkeys. Brain Stimulation: Basic, Translational, and Clinical Research in Neuromodulation. 2015;8(2):321.Google Scholar
Radley, JJ, Rocher, AB, Miller, M, et al. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cerebral Cortex. 2006;16(3):313–20.Google Scholar
Radley, JJ, Sisti, HM, Hao, J, et al. Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex. Neuroscience. 2004;125(1):16.Google Scholar
Cook, SC, Wellman, CL. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. Journal of Neurobiology. 2004;60(2):236–48.Google Scholar
Rempel-Clower, NL. Role of orbitofrontal cortex connections in emotion. Annals of the New York Academy of Sciences. 2007;1121(1):7286.Google Scholar
Liston, C, Miller, MM, Goldwater, DS, et al. Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting. The Journal of Neuroscience. 2006;26(30):7870–4.Google Scholar
Frederick Rohan, W, Michael, N, Kimberley, J. Acute and chronic stress-induced disturbances of microglial plasticity, phenotype and function. Current Drug Targets. 2013;14(11):1262–76.Google Scholar
Wohleb, ES, Terwilliger, R, Duman, CH, Duman, RS. Stress-induced neuronal colony stimulating Factor 1 provokes microglia-mediated neuronal remodeling and depressive-like behavior. Biological Psychiatry. 2018;83(1):3849.Google Scholar
Phelps, EA, LeDoux, JE. Contributions of the amygdala to emotion processing: from animal models to human behavior. Neuron. 2005;48(2):175–87.Google Scholar
Vyas, A, Mitra, R, Shankaranarayana Rao, BS, Chattarji, S. Chronic stress induces contrasting patterns of dendritic remodeling in hippocampal and amygdaloid neurons. The Journal of Neuroscience. 2002;22(15):6810–8.Google Scholar
Vyas, A, Pillai, AG, Chattarji, S. Recovery after chronic stress fails to reverse amygdaloid neuronal hypertrophy and enhanced anxiety-like behavior. Neuroscience. 2004;128(4):667–73.Google Scholar
Johnson, FK, Delpech, J-C, Thompson, GJ, et al. Amygdala hyper-connectivity in a mouse model of unpredictable early life stress. Translational Psychiatry. 2018;8(1):49.Google Scholar
Lee, T, Jarome, T, Li, S-J, Kim, JJ, Helmstetter, FJ. Chronic stress selectively reduces hippocampal volume in rats: a longitudinal magnetic resonance imaging study. Neuroreport. 2009;20(17):1554–8.Google Scholar
Schoenfeld, TJ, McCausland, HC, Morris, HD, Padmanaban, V, Cameron, HA. Stress and Loss of Adult Neurogenesis Differentially Reduce Hippocampal Volume. Biological Psychiatry. 2017;82(12):914–23.Google Scholar
McEwen, BS. Stress and hippocampal plasticity. Annual Review of Neuroscience. 1999;22(1):105–22.Google Scholar
Magarin˜os, AM, McEwen, BS. Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: Involvement of glucocorticoid secretion and excitatory amino acid receptors. Neuroscience. 1995;69(1):8998.Google Scholar
Brunson, KL, Kramár, E, Lin, B, et al. Mechanisms of late-onset cognitive decline after early-life stress. The Journal of Neuroscience. 2005;25(41):9328–38.Google Scholar
Christian, KM, Miracle, AD, Wellman, CL, Nakazawa, K. Chronic stress-induced hippocampal dendritic retraction requires CA3 NMDA receptors. Neuroscience. 2011;174:2636.Google Scholar
Pham, K, Nacher, J, Hof, PR, McEwen, BS. Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus. European Journal of Neuroscience. 2003;17(4):879–86.Google Scholar
Gould, E, McEwen, BS, Tanapat, P, Galea, LAM, Fuchs, E. Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. The Journal of Neuroscience. 1997;17(7):2492–8.CrossRefGoogle ScholarPubMed
Milior, G, Lecours, C, Samson, L, et al. Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress. Brain, Behavior, and Immunity. 2016;55:114–25.Google Scholar
Duman, RS, Aghajanian, GK, Sanacora, G, Krystal, JH. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nature Medicine. 2016;22:238.Google Scholar
Xiong, W, Wei, H, Xiang, X, et al. The effect of acute stress on LTP and LTD induction in the hippocampal CA1 region of anesthetized rats at three different ages. Brain Research. 2004;1005(1):187–92.Google Scholar
Lakshminarasimhan, H, Chattarji, S. Stress leads to contrasting effects on the levels of brain derived neurotrophic factor in the hippocampus and amygdala. PLoS ONE. 2012;7(1):e30481.Google Scholar
Halassa, MM, Fellin, T, Haydon, PG. The tripartite synapse: roles for gliotransmission in health and disease. Trends in Molecular Medicine. 2007;13(2):5463.Google Scholar
Volterra, A, Meldolesi, J. Astrocytes, from brain glue to communication elements: the revolution continues. Nature Reviews Neuroscience. 2005;6:626.Google Scholar
Segerstrom, SC, Miller, GE. Psychological stress and the human immune system: a meta-analytic study of 30 years of inquiry. Psychological Bulletin. 2004;130(4):601–30.Google Scholar
Miller, GE, Chen, E, Sze, J, et al. A functional genomic fingerprint of chronic stress in humans: blunted glucocorticoid and increased NF-kappaB signaling. Biological Psychiatry. 2008;64(4):266–72.CrossRefGoogle ScholarPubMed
Miller, AH, Haroon, E, Raison, CL, Felger, JC. Cytokine targets in the brain: impact on neurotransmitters and neurocircuits. Depression and Anxiety. 2013;30(4):297306.Google Scholar
Muscatell, KA, Dedovic, K, Slavich, GM, et al. Greater amygdala activity and dorsomedial prefrontal-amygdala coupling are associated with enhanced inflammatory responses to stress. Brain, Behavior, and Immunity. 2015;43:4653.Google Scholar
Evans, GW, Swain, JE, King, AP, et al. Childhood cumulative risk exposure and adult amygdala volume and function. Journal of Neuroscience Research. 2016;94(6):535–43.Google Scholar
Hanson, JL, Nacewicz, BM, Sutterer, MJ, et al. Behavioral problems after early life stress: contributions of the hippocampus and amygdala. Biological Psychiatry. 2015;77(4):314–23.Google Scholar
Kim, Y-K, Amidfar, M, Won, E. A review on inflammatory cytokine-induced alterations of the brain as potential neural biomarkers in post-traumatic stress disorder. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2019;91:103–12.Google Scholar
Hantsoo, L, Kornfield, S, Anguera, MC, Epperson, CN. Inflammation: a proposed intermediary between maternal stress and offspring neuropsychiatric risk. Biological Psychiatry. 2019;85(2):97106.Google Scholar
Lou, HC, Hansen, D, Nordentoft, M, et al. Prenatal stressors of human life affect fetal brain development. Developmental Medicine & Child Neurology. 1994;36(9):826–32.Google Scholar
Obel, C, Hedegaard, M, Brink Henriksen, T, Jørgen, N. Stressful life events in pregnancy and head circumference at birth. Developmental Medicine & Child Neurology. 2003;45(12):802–6.Google Scholar
Clark, A. Whatever next? Predictive brains, situated agents, and the future of cognitive science. Behav Brain Sci. 2013;36(3):181204.Google Scholar
Friston, K, Kilner, J, Harrison, L. A free energy principle for the brain. J Physiol Paris. 2006;100(1–3):7087.CrossRefGoogle ScholarPubMed
Hohwy, J. The Predictive Mind. First edition. Oxford University Press, Oxford; 2013.Google Scholar
Seth, AK, Friston, KJ. Active interoceptive inference and the emotional brain. Philos Trans R Soc Lond B Biol Sci. 2016;371:(1708).Google Scholar
Friston, K. The free-energy principle: a rough guide to the brain? Trends Cogn Sci. 2009;13(7):293301.Google Scholar
Seth, AK. Interoceptive inference, emotion, and the embodied self. Trends Cogn Sci. 2013;17(11):565–73.CrossRefGoogle ScholarPubMed
Peters, A, McEwen, BS, Friston, K. Uncertainty and stress: why it causes diseases and how it is mastered by the brain. Prog Neurobiol. 2017;156:164–88.Google Scholar
Mason, J. Hormones and metabolism: psychological influences on the pituitary-adrenal cortical. In Pincus, C, ed. Recent Progress In Hormone Research: Proceedings of the Laurentian Hormone Conference 1958. Academic Press, New York and London; 1959.Google Scholar
McEwen, BS. Stress, adaptation, and disease. Allostasis and allostatic load. Ann N Y Acad Sci. 1998;840:3344.Google Scholar
Barrett, LF, Simmons, WK. Interoceptive predictions in the brain. Nat Rev Neurosci. 2015;16(7):419–29.Google Scholar
Craig, AD. How do you feel? Interoception: the sense of the physiological condition of the body. Nat Rev Neurosci. 2002;3(8):655–66.CrossRefGoogle Scholar
Craig, AD. Interoception: the sense of the physiological condition of the body. Curr Opin Neurobiol. 2003;13(4):500–5.Google Scholar
Critchley, HD, Harrison, NA. Visceral influences on brain and behavior. Neuron. 2013;77(4):624–38.Google Scholar
Tracey, KJ. The inflammatory reflex. Nature. 2002;420(6917):853–9.Google Scholar
Tracey, KJ. Reflex control of immunity. Nat Rev Immunol. 2009;9(6):418–28.Google Scholar
Stephan, KE, Manjaly, ZM, Mathys, CD, et al. Allostatic self-efficacy: a metacognitive theory of dyshomeostasis-induced fatigue and depression. Front Hum Neurosci. 2016;10:550.Google Scholar
Lind, RW. Bi-directional, chemically specified neural connections between the subfornical organ and the midbrain raphe system. Brain Research. 1986;384(2):250–61.Google Scholar
Shepard, JD, Barron, KW, Myers, DA. Corticosterone delivery to the amygdala increases corticotropin-releasing factor mRNA in the central amygdaloid nucleus and anxiety-like behavior. Brain Research. 2000;861(2):288–95.CrossRefGoogle Scholar
Roozendaal, B. Glucocorticoids and the regulation of memory consolidation. Psychoneuroendocrinology. 2000;25(3):213–38.Google Scholar
Diamond, DM, Fleshner, M, Ingersoll, N, Rose, G. Psychological stress impairs spatial working memory: Relevance to electrophysiological studies of hippocampal function. Behavioral Neuroscience. 1996;110(4):661–72.Google Scholar
McNelis, JC, Manolopoulos, KN, Gathercole, LL, et al. Dehydroepiandrosterone exerts antiglucocorticoid action on human preadipocyte proliferation, differentiation, and glucose uptake. American Journal of Physiology Endocrinology and Metabolism. 2013;305(9):E1134-E44.Google Scholar
Kalimi, M, Shafagoj, Y, Loria, R, Padgett, D, Regelson, W. Anti-glucocorticoid effects of dehydroepiandrosterone (DHEA). Molecular and Cellular Biology. 1994;131(2): 99104.Google Scholar
Prough, RA, Clark, BJ, Klinge, CM. Novel mechanisms for DHEA action. Journal of Molecular Endocrinology. 2016;56(3):R139–55.Google Scholar
Cardounel, A, Regelson, W, Kalimi, M. Dehydroepiandrosterone protects hippocampal neurons against neurotoxin-induced cell death: mechanism of action (44437). Proceedings of the Society for Experimental Biology and Medicine. 1999;222(2):145–9.Google Scholar
Kaminska, M, Harris, J, Gijsbers, K, Dubrovsky, B. Dehydroepiandrosterone sulfate (DHEAS) counteracts decremental effects of corticosterone on dentate gyrus LTP. Implications for depression.Brain Research Bulletin. 2000;52(3):229–34.Google Scholar
Young, AH, Gallagher, P, Porter, RJ. Elevation of the cortisol-dehydroepiandrosterone ratio in drug-free depressed patients. American Journal of Psychiatry. 2002;159(7):1237–9.Google Scholar
Wolkowitz, OM, Reus, VI, Keebler, A, et al. Double-blind treatment of major depression with dehydroepiandrosterone. American Journal of Psychiatry. 1999;156(4):646–9.Google Scholar
Krzystyniak, A, Baczynska, E, Magnowska, M, et al. Prophylactic ketamine treatment promotes resilience to chronic stress and accelerates recovery: correlation with changes in synaptic plasticity in the CA3 subregion of the hippocampus. International Journal of Molecular Sciences. 2019;20(7):1726.Google Scholar
Wang, J, Hodes, GE, Zhang, H, et al. Epigenetic modulation of inflammation and synaptic plasticity promotes resilience against stress in mice. Nature Communications. 2018;9(1):477.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
×