Protective and damaging effects of stress mediators: central role of the brain

Bruce S McEwen, Bruce S McEwen

Abstract

The mind involves the whole body and two-way communication between the brain and the cardiovascular, immune, and other systems via neural and endocrine mechanisms. Stress is a condition of the mind-body interaction, and a factor in the expression of disease that differs among individuals. It is notjust the dramatic stressful events that exact their toll, but rather the many events of daily life that elevate and sustain activities of physiological systems and cause sleep deprivation, overeating, and other health-damaging behaviors, producing the feeling of being "stressed out." Over time, this results in wear and tear on the body which is called "allostatic load," and it reflects not only the impact of life experiences but also of genetic load, individual lifestyle habits reflecting items such as diet, exercise, and substance abuse, and developmental experiences that set life-long patterns of behavior and physiological reactivity. Hormones associated with stress and allostatic load protect the body in the short run and promote adaptation by the process known as allostasis, but in the long run allostatic load causes changes in the body that can lead to disease. The brain is the key organ of stress, allostasis, and allostatic load, because it determines what is threatening and therefore stressful, and also determines the physiological and behavioral responses. Brain regions such as the hippocampus, amygdala, and prefrontal cortex respond to acute and chronic stress by undergoing structural remodeling, which alters behavioral and physiological responses. Translational studies in humans with structural and functional imaging reveal smaller hippocampal volume in stress-related conditions, such as mild cognitive impairment in aging and prolonged major depressive illness, as well as in individuals with low self-esteem. Alterations in amygdala and prefrontal cortex are also reported. Besides pharmaceuticals, approaches to alleviate chronic stress and reduce allostatic load and the incidence of diseases of modern life include lifestyle change, and policies of government and business that would improve the ability of individuals to reduce their own chronic stress burden.

Figures

Figure 1.. Four types of allostatic load.…
Figure 1.. Four types of allostatic load. The top panel illustrates the normal allostatic response, in which a response is initiated by a stressor, sustained for an appropriate interval, and then turned off. The remaining panels illustrate four conditions that lead to allostatic load: top left- repeated “hits” from multiple stressors; top right- lack of adaptation; bottom left- prolonged response due to delayed shut down; and bottom right - inadequate response that leads to compensatory hyperactivity of other mediators (eg, inadequate secretion of glucocorticoid, resulting in increased levels of cytokines that are normally counter-regulated by glucocorticoids). Reproduced from reference 1 ; McEwen BS. Protective and damaging effects of stress mediators. N Engl J Med. 1998; 338:171-179. Copyright © Massachusetts Medical Society 1998.
Figure 2.. Nonlinear network of mediators of…
Figure 2.. Nonlinear network of mediators of allostasis involved in the stress response. Arrows indicate that each system regulates the others in a reciprocal manner, creating a nonlinear network. Moreover, there are multiple pathways for regulation- eg, inflammatory cytokine production is negatively regulated via anti-inflammatory cytokines as well as via parasympathetic and glucocorticoid pathways, whereas sympathetic activity increases inflammatory cytokine production. Parasympathetic activity in turn, restrains sympathetic activity DHEA, dehydroepiandrosterone
Figure 3.. Central role of the brain…
Figure 3.. Central role of the brain in allostasis and the behavioral and physiological response to stressors. Reproduced from reference 1: McEwen BS. Protective and damaging effects of stress mediators. N Engl J Med. 1998;338:171-179. Copyright © Massachusetts Medical Society 1998.
Figure 4.. Brain regions that are involved…
Figure 4.. Brain regions that are involved in perception and response ress, and which show structural remodeling as a result of stress.

References

    1. McEwen BS. Protective and damaging effects of stress mediators. N Engl J Med. 1998;338:171–179.
    1. Sterling P., Eyer J. Allostasis: a new paradigm to explain arousal pathology. In: Fisher S, Reason J, eds. Handbook of Life Stress, Cognition and Health. New York, NY: John Wiley & Sons; 1988:629–649.
    1. McEwen BS., Stellar E. Stress and the individual: mechanisms leading to disease. Arch Int Med. 1993;153:2093–2101.
    1. McEwen BS., Wingfield JC. The concept of allostasis in biology and bio-medicine. Horm Behav. 2003;43:2–15.
    1. Sapolsky RM., Romero LM., Munck AU. How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions. Endocr Rev. 2000;21:55–89.
    1. Dinkel K., MacPherson A., Sapolsky RM. Novel glucocorticoid effects on acute inflammation in the CNS. J Neurochem. 2003;84:705–716.
    1. MacPherson A., Dinkel K., Sapolsky R. Glucocorticoids worsen excitotoxin-induced expression of pro-inflammatory cytokines in hippocampal cultures. Exp Neurol. 2005;194:376–383.
    1. Borovikova LV., Ivanova S., Zhang M., et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–462.
    1. Thayer JF., Lane RD. A model of neurovisceral integration in emotion regulation and dysregulation. J Affect Disord. 2000;61:201–216.
    1. Wingfield JC., Romero LM. Adrenocortical responses to stress and their modulation in free-living vertebrates. In: Coping with the Environment: Neural and Endocrine Mechanisms. Vol. IV. New York, NY: Oxford University Press. 2000:211–234.
    1. Nelson RA. Protein and fat metabolism in hibernating bears. Fed Proc. 1980;39:2955–2958.
    1. Farrell AP. Coronary arteriosclerosis in salmon: growing old or growing fast? Comp Biochem Physiol. 2002;132:723–735.
    1. Maule AG., Tripp RA., Kaattari SL., Schreck CB. Stress alters immune function and disease resistance in chinook salmon (Oncorhynchus tshawytscha). J Endocrinol. 1989;120:135–142.
    1. Gotz ME., Malz CR., Dirr A., et al. Brain aging phenomena in migrating sockeye salmon Oncorhynchus nerka nerka. J Neural Transm. 2005;112:1177–1199.
    1. Cockburn A., Lee AK. Marsupial femmes fatales. Natural History. 1988;97:40–47.
    1. Leproult R., Copinschi G., Buxton O., VanCauter E. Sleep loss results in an elevation of Cortisol levels the next evening. Sleep. 1997;20:865–870.
    1. Spiegel K., Leproult R., Van Cauter E. Impact of sleep debt on metabolic and endocrine function. Lancet. 1999;354:1435–1439.
    1. Spiegel K., Tasali E., Penev P., Van Cauter E. Brief communication: sleep curtailment in healthy young men is associated with decreased leptin levels, elevated ghrelin levels, and increased hunger and appetite. Ann Intern Med. 2004;141:846–850.
    1. Vgontzas AN., Zoumakis E., Bixler EO., et al. Adverse effects of modest sleep restriction on sleepiness, performance, and inflammatory cytokines. J Clin Endocrin Metab. 2004;89:2119–2126.
    1. Gangwisch JE., Malaspina D., Boden-AIbala B., Heymsfield SB. Inadequate sleep as a risk factor for obesity: analyses of the NHANES I. Sleep. 2005;28:1289–1296.
    1. McEwen BS., Chattarji S. Molecular mechanisms of neuroplasticity and pharmacological implications: the example of tianeptine. Eur Neuropsycho-pharmacol. 2004;14:S497–S502.
    1. Taishi P., Chen Z., Obal F Jr., et al. Sleep-associated changes in interleukin1, mRNA in the brain. J Interferon Cytokine Res. 1998;18:793–798.
    1. Clark RA., Valente AJ. Nuclear factor kappa B activation by NADPH oxidases. Mech Ageing Dev. 2004;125:799–810.
    1. Tang J., Liu J., Zhou C., et al. Role of NADPH oxidase in the brain injury of intracerebral hemorrhage. J Neurochem. 2005;94:1342–1350.
    1. Silva RH., Abilio VC., Takatsu AL., et al. Role of hippocampal oxidative stress in memory deficits induced by sleep deprivation in mice. Neuropharmacology. 2004;46:895–903.
    1. Kong J., Shepel PN., Holden CP., Mackiewicz M., Pack AI., Geiger JD. Brain glycogen decreases with increased periods of wakefulness: implications for homeostatic drive to sleep. J Neurosci. 2002;22:5581–5587.
    1. Brown AM. Brain glycogen re-awakened. J Neurochem. 2004;89:537–552.
    1. Wender R., Brown AM., Fern R., Swanson RA., Farrell K., Ransom BR. Astrocytic glycogen influences axon function and survival during glucose deprivation in central white matter. J Neurosci. 2000;20:6804–6810.
    1. Guzman-Marin R., Suntsova N., Stewart DR., Gong H., Szymusiak R., McGinty D. Sleep deprivation reduces proliferation of cells in the dentate gyrus of the hippocampus in rats. J Physiol. 2003;549. 2:563–571.
    1. Roman V., Van der Borght K., Leemburg SA., Van der Zee EA., Meerlo P. Sleep restriction by forced activity reduces hippocampal cell proliferation. Brain Res. 2005;1065:53–59.
    1. Hairston IS., Little MTM., Scanlon MD., et al. Sleep restriction suppresses neurogenesis induced by hippocampus-dependent learning. J Neurophysiol. 2005;94:4224–4233.
    1. Youngblood BD., Zhou J., Smagin GN., Ryan DH., Harris RBS. Sleep deprivation by the “flower pot” technique and spatial reference memory. Physiol Behav. 1997;61:249–256.
    1. Kim EY., Mahmoud GS., Grover LM. REM sleep deprivation inhibits LTP in vivo in area CA1 of rat hippocampus. Neurosci Lett. 2005;388:163–167.
    1. Graves LA., Heller EA., Pack AI., Abel T. Sleep deprivation selectively impairs memory consolidation for contextual fear conditioning. Learn Mem. 2003;10:168–176.
    1. Guan Z., Peng X., Fang J. Sleep deprivation impairs spatial memory and decreases extracellular signal-regulated kinase phosphorylation in the hippocampus. Brain Res. 2004;1018:38–47.
    1. Romcy-Pereira R., Pavlides C. Distinct modulatory effects of sleep on the maintenance of hippocampal and medial prefrontal cortex LTP. Eur J Neurosci. 2004;20:3453–3462.
    1. de Paula HMG., Hoshino K. Correlation between the fighting rates of REM sleep-deprived rats and susceptibility to the 'wild running' of audiogenic seizures. Brain Res. 2002;926:80–85.
    1. Musty RE., Consroe PF. Phencyclidine produces aggressive behavior in rapid eye movement sleep-deprived rats. Life Sciences. 1982;30:1733–1738.
    1. Wood GE., Young LT., Reagan LP., McEwen BS. Acute and chronic restraint stress alter the incidence of social conflict in male rats. Horm Behav. 2003;43:205–213.
    1. Meerlo P., Overkamp GJF., Benning MA., Koolhaas JM., Van Den Hoofdakker RH. Long-term changes in open field behaviour following a single social defeat in rats can be reversed by sleep deprivation. Physiol Behav. 1996;60:115–119.
    1. Felitti VJ., Anda RF., Nordenberg D., et al. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The adverse childhood experiences (ACE) study. Am J Prev Med. 1998;14:245–258.
    1. Heim C., Nemeroff CB. The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biol Psychiatry. 2001;49:1023–1039.
    1. Repetti RL., Taylor SE., Seeman TE. Risky families: family social environments and the mental and physical health of offspirng. Psychol Bull. 2002;128:330–366.
    1. Kaufman J., Charney DS. Neurobiological correlates of child abuse. Biol Psychiatry. 1999;45:1235–1236.
    1. Kaufman J., Plotsky PM., Nemeroff CB., Charney DS. Effects of early adverse experiences on brain structure and function: clinical implications. Biol Psychiatry. 2000;48:778–790.
    1. Vermetten E., Schmahl C., Lindner S., Loewenstein RJ., Bremner JD. Hippocampal and amygdalar volumes in dissociative identity disorder. Am J Psychiatry. 2006;163:630–636.
    1. Francis D., Diorio J., Liu D., Meaney MJ. Nongenomic transmission across generations of maternal behavior and stress responses in the rat. Science. 1999;286:1155–1158.
    1. Cavigelli SA., McClintock MK. Fear of novelty in infant rats predicts adult corticosterone dynamics and an early death. Proc Natl Acad Sci USA. 2003;100:16131–16136.
    1. Weaver ICG., Cervoni N., Champagne FA., et al. Epigenetic programming by maternal behavior. Nature Neurosci. 2004;7:847–854.
    1. Sullivan RM., Landers M., Yeaman B., Wilson DA. Good memories of bad events in infancy. Nature. 2000;407:38–39.
    1. Coplan JD., Smith ELP., Altemus M., et al. Variable foraging demand rearing: sustained elevations in cisternal cerebrospinal fluid corticotropin-releasing factor concentrations in adult primates. Biol Psychiatry. 2001;50:200–204.
    1. Caspi A., Sugden K., Moffitt TE., et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301:386–389.
    1. Caspi A., McClay J., Moffitt TE., et al. Role of genotype in the cycle of violence in maltreated children. Science. 2002;297:851–854.
    1. Szeszko PR., Lipsky R., Mentschel C., et al. Brain-derived neurotrophic factor val66met polymorphism and volume of the hippocampal formation. Mol Psychiatry. 2005;10:631–636.
    1. Hariri AR., Goldberg TE., Mattay VS., et al. Brain-derived neurotrophic factor val66met polymorphism affects human memory-related hippocampal activity and predicts memory performance. J Neurosci. 2003;23:6690–6694.
    1. Pezawas L., Verchinski BA., Mattay VS., et al. The brain-derived neurotrophic factor val66met polymorphism and variation in human cortical morphology. J Neurosci. 2004;24:10099–10102.
    1. Jiang X., Xu K., Hoberman J., et al. BDNF variation and mood disorders: a novel functional promoter polymorphism and Val66Met are associated with anxiety but have opposing effects. Neuropsychopharmacology. 2005;30:1353–1361.
    1. McEwen BS. Stress and hippocampal plasticity. Annu Rev Neurosci. 1999;22:105–122.
    1. Lisman JE., Otmakhova NA. Storage, recall, and novelty detection of sequences by the hippocampus: elaborating on the SOCRATIC model to account for normal and aberrant effects of dopamine. Hippocampus. 2001;11:551–568.
    1. Popov VI., Bocharova LS. Hibernation-induced structural changes in synaptic contacts between mossy fibres and hippocampal pyramidal neurons. Neuroscience. 1992;48:53–62.
    1. Popov VI., Bocharova LS., Bragin AG. Repeated changes of dendritic morphology in the hippocampus of ground squirrels in the course of hibernation. Neuroscience. 1992;48:45–51.
    1. Seri B., Garcia-Verdugo JM., McEwen BS., AIvarez-Buylla A. Astrocytes give rise to new neurons in the adult mammalian hippocampus. J Neurosci. 2001;21:7153–7160.
    1. Kempermann G., Gage FH. New nerve cells for the adult brain. Sci Am. 1999;280:48–53.
    1. Cameron HA., McKay RDG. Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus. J Comp Neurol. 2001;435:406–417.
    1. Czeh B., Michaelis T., Watanabe T., et al. Stress-induced changes in cerebral metabolites, hippocampal volume and cell proliferation are prevented by antidepressant treatment with tianeptine. Proc Natl Acad Sci USA. 2001;98:12796–12801.
    1. Aberg MA., Aberg ND., Hedbacker H., Oscarsson J., Eriksson PS. Peripheral infusion of IGF-1 selectively induces neurogenesis in the adult rat hippocampus. J Neurosci. 2000;20:2896–2903.
    1. Trejo JL., Carro E., Torres-AIeman I. Circulating insulin-like growth factor I mediates exercise-induced increases in the number of new neurons in the adult hippocampus. J Neurosci. 2001;21:1628–1634.
    1. 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. J Neurosci. 1997;17:2492–2498.
    1. Sousa N., Lukoyanov NV., Madeira MD., Almeida OFX., Paula-Barbosa MM. Reorganization of the morphology of hippocampal neurites and synapses after stress-induced damage correlates with behavioral improvement. Neuroscience. 2000;97:253–266.
    1. McKittrick CR., Magarinos AM., Blanchard DC., Blanchard RJ., McEwen BS., Sakai RR. Chronic social stress reduces dendritic arbors in CA3 of hippocampus and decreases binding to serotonin transporter sites. Synapse. 2000;36:85–94.
    1. Arendt T., Stieler J., Strijkstra AM., et al. Reversible paired helical filament-like phosphorylation of tau is an adaptive process associated with neuronal plasticity in hibernating animals. J Neurosci. 2003;23:6972–6981.
    1. Wood GE., Young LT., Reagan LP., Chen B., McEwen BS. Stress-induced structural remodeling in hippocampus: prevention by lithium treatment. Proc Natl Acad Sci U S A. 2004;101:3973–3978.
    1. Magarinos AM., Verdugo Garcia JM., McEwen BS. Chronic restraint stress alters synaptic terminal structure in hippocampus. Proc Natl Acad Sci U S A. 1997;94:14002–14008.
    1. Gao Y., Bezchlibnyk YB., Sun X., Wang JF., McEwen BS., Young LT. Effects of restraint stress on the expression of proteins involved in synaptic vesicle exocytosis in the hippocampus. Neuroscience. 2006;14:1139–1148.
    1. Grillo CA., Piroli GG., Wood GE., Reznikov LR., McEwen BS., Reagan LP. Immunocytochemical analysis of synaptic proteins provides new insights into diabetes-mediated plasticity in the rat hippocampus. Neuroscience. 2005;136:477–486.
    1. Sandi C. Stress, cognitive impairment and cell adhesion molecules. Nat Rev Neurosci. 2004;5:917–930.
    1. Pawlak R., Rao BSS., Melchor JP., Chattarji S., McEwen B., Strickland S. Tissue plasminogen activator and plasminogen mediate stress-induced decline of neuronal and cognitive functions in the mouse hippocampus. Proc Natl Acad Sci U S A. 2005;102:18201–18206.
    1. Bianchi M., Heidbreder C., Crespi F. Cytoskeletal changes in the hippocampus following restraint stress: role of serotonin and microtubules. Synapse. 2003;49:188–194.
    1. Smith MA., Cizza G. Stress-induced changes in brain-dervied neurotrophic factor expression are attenuated in aged Fischer 344/N rats. Neurobiol Aging. 1996;17:859–864.
    1. Kuroda Y., McEwen BS. Effect of chronic restraint stress and tianeptine on growth factors, GAP-43 and MAP2 mRNA expression in the rat hippocampus. Mol Brain Res. 1998;59:35–39.
    1. Isgor C., Kabbaj M., Akil H., Watson SJ. Delayed effects of chronic variable stress during peripubertal-juvenile period on hippocampal morphology and on cognitive and stress axis functions in rats. Hippocampus. 2004;14:636–468.
    1. Marmigere F., Givalois L., Rage F., Arancibia S., Tapia-Arancibia L. Rapid induction of BDNF expression in the hippocampus during immobilization stress challenge in adult rats. Hippocampus. 2003;13:646–655.
    1. Hansson AC., Sommer WH., Metsis M., Stromberg I., Agnati LF., Fuxe K. Corticosterone actions on the hippocampal brain-derived neurotrophic factor expression are mediated by Exon IV promoter. J Neuroendocrinol. 2006;18:104–114.
    1. Koob GF. Corticotropin-releasing factor, norepinephrine, and stress. Biol Psychiatry. 1999;46:1167–1180.
    1. Matys T., Pawlak R., Matys E., Pavlides C., McEwen BS., Strickland S. Tissue plasminogen activator promotes the effects of corticotropin releasing factor on the amygdala and anxiety-like behavior. Proc Natl Acad Sci U S A. 2004;101:16345–16350.
    1. Brunson KL., Eghbal-Ahmadi M., Bender R., Chen Y., Baram TZ. Longterm, progressive hippocampal cell loss and dysfunction induced by early-life administration of corticotropin-releasing hormone reproduce the effects of early-life stress. Proc Natl Acad Sci U S A. 2001;98:8856–8861.
    1. Brunson KL., Kramar E., Lin B., et al. Mechanisms of late-onset cognitive decline after early-life stress. J Neurosci. 2005;25:9328–9338.
    1. Chen Y., Bender RA., Brunson KL., et al. Modulation of dendritic differentiation by corticotropin-releasing factor in the developing hippocampus. Proc Natl Acad Sci USA. 2004; 101:15782–15787.
    1. Radley JJ., Rocher AB., Miller M., et al. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cereb Cortex. 2006;16:313–320.
    1. 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;125:1–6.
    1. Wellman CL. Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration. J Neurobiol. 2001;49:245–253.
    1. Cook SC., Wellman CL. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. J Neurobiol. 2004;60:236–248.
    1. Brown SM., Henning S., Wellman CL. Mild, short-term stress alters dendritic morphology in rat medial prefrontal cortex. Cereb Cortex. 2005;30:1–9.
    1. Kreibich AS., Blendy JA. cAMP response element-binding protein is required for stress but not cocaine-induced reinstatement. J Neurosci. 2004;24:6686–6692.
    1. Vyas A., Mitra R., Rao BSS., Chattarji S. Chronic stress induces contrasting patterns of dendritic remodeling in hippocampal and amygdaloid neurons. J Neurosci. 2002;22:6810–6818.
    1. Liston C., Miller MM., Goldwater DS., et al. Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting. J Neurosci. 2003;23:8867–8871.
    1. Melchor JP., Pawlak R., Strickland S. The tissue plasminogen activator plasminogen proteolytic cascade accelerates amyloid-p (Ap) degradation and inhibits Ap-induced neurodegeneration. J. Neurosci. 2003;23:8867–8871.
    1. Shors TJ., Chua C., Falduto J. Sex differences and opposite effects of stress on dendritic spine density in the male versus female hippocampus. J Neurosci. 2001;21:6292–6297.
    1. Mitra R., Vyas A., Chattarji S. Contrasting effects of chronic immobilization and unpredictable stress on spine density in the hippocampus and amygdala. Proc Natl Acad Sci U S A. 2005;102:9371–9376.
    1. Conrad CD., Magarinos AM., LeDoux JE., McEwen BS. Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy. Behav Neurosci. 1999;113:902–913.
    1. Wang J., Rao H., Wetmore GS., et al. Perfusion functional MRI reveals cerebral blood flow pattern under psychological stress. Proc Natl Acad Sci USA. 2005;102:17804–17809.
    1. KessIer RC. The effects of stressful life events on depression. Annu Rev Psychol. 1997;48:191–214.
    1. KendIer KS. Major depression and the environment: a psychiatric genetic perspective. Pharmacopsychiatry. 1998;31:5–9.
    1. SheIine Yl., Sanghavi M., Mintun MA., Gado MH. Depression duration but not age predicts hippocampal volume loss in medically healthy women with recurrent major depression. J Neurosci. 1999;19:5034–5043.
    1. Drevets WC., Price JL., Simpson JR Jr., et al. Subgenual prefrontal cortex abnormalities in mood disorders. Nature. 1997;386:824–827.
    1. Sheline Yl., Gado MH., Kraemer HC. Untreated depression and hippocampal volume loss. Am J Psychiatry. 2003;160:1516–1518.
    1. Frodl T., Meisenzahl EM., Zetzsche T., et al. Larger amygdala volumes in first depressive episode as compared to recurrent major depression and healthy control subjects. Biol Psychiatry. 2003;53:338–344.
    1. MacQueen GM., Campbell S., McEwen BS., et al. Course of illness, hippocampal function, and hippocampal volume in major depression. Proc Natl Acad Sci U S A. 2003;100:1387–1392.
    1. Sachar EJ., Hellman L., Roffwarg HP., Halpern FS., Fukushima DK., Gallagher TF. Disrupted 24-hour patterns of Cortisol secretion in psychotic depression. Arch Gen Psychiatry. 1973;28:19–24.
    1. Murphy BEP. Treatment of major depression with steroid suppressive drugs. J Steroid Biochem Mol Biol. 1991;39:239–244.
    1. Starkman MN., Schteingart DE. Neuropsychiatrie manifestations of patients with Cushing's syndrome. Arch Intern Med. 1981;141:215–219.
    1. Starkman MN., Gebarski SS., Berent S., Schteingart DE. Hippocampal formation volume, memory dysfunction, and Cortisol levels in patients with Cushing's syndrome. Biol Psychiatry. 1992;32:756–765.
    1. Bremner JD. Neuroimaging studies in post-traumatic stress disorder. Curr Psychiat Reports. 2002;4:254–263.
    1. Pitman RK. Hippocampal diminution in PTSD: more (or less?) than meets the eye. Hippocampus. 2001;11:73–74.
    1. Driessen M., Hermann J., Stahl K., et al. Magnetic resonance imaging volumes of the hippocampus and the amygdala in women with borderline personality disorder and early traumatization. Arch Gen Psychiatry. 2000;57:1115–1122.
    1. Convit A., Wolf OT., Tarshish C., de Leon MJ. Reduced glucose tolerance is associated with poor memory performance and hippocampal atrophy among normal elderly. Proc Natl Acad Sci U S A. 2003;100:2019–2022.
    1. Ott A., Stolk RP., Hofman A., van Harskamp F., Grobbee DE., Breteler MMB. Association of diabetes mellitus and dementia: the Rotterdam study. Diabetologia. 1996;39:1392–1397.
    1. Haan MN. Therapy insight: type 2 diabetes mellitus and the risk of lateonset Alzheimer's disease. Nat Clin Pract Neurol. 2006;2:159–166.
    1. de Leon MJ., Convit A., Wolf OT., et al. Prediction of cognitive decline in normal elderly subjects with 2-[18F]fIuoro-2-deoxy-D-gIucose/positron-emission tomography (FDG/PET). Proc Natl Acad Sci USA. 2001;98:10966–10971.
    1. Pressman SD., Cohen S. Does positive affect influence health? Psychol Bull. 2005;131:925–971.
    1. Seeman TE., Singer BH., Ryff CD., Dienberg G., Levy-Storms L. Social relationships, gender, and allostatic load across two age cohorts. Psychosom Med. 2002;64:395–406.
    1. Steptoe A., Wardle J., Marmot M. Positive affect and health-related neuroendocrine, cardiovascular, and inflammatory processes. Proc Natl Acad Sci USA. 2005;102:6508–6512.
    1. Kirschbaum C., Prussner JC., Stone AA., et al. Persistent high Cortisol responses to repeated psychological stress in a subpopulation of healthy men. Psychosom Med. 1995;57:468–474.
    1. Pruessner JC., Baldwin MW., Dedovic K., et al. Self-esteem, locus of control, hippocampal volume, and Cortisol regulation in young and old adulthood. Neuroimage. 2005;28:815–826.
    1. Pruessner JC., Hellhammer DH., Kirschbaum C. Low self-esteem, induced failure and the adrenocortical stress response. Pers Individ Diff. 1999;27:477–489.
    1. Steptoe A., Owen N., Kunz-Ebrecht SR., Brydon L. Loneliness and neuroendocrine, cardiovascular, and inflammatory stress responses in middleaged men and women. Psychoneuroendocrinology. 2004;29:593–611.
    1. Bernadet P. Benefits of physical activity in the prevention of cardiovascular disease. J Cardiovasc Pharmacol. 1995;25(suppl 1):S3–S8.
    1. Rovio S., Kareholt I., Helkala E-L., et al. Leisure-time physical activity at midlife and the risk of dementia and Alzheimer's disease. Lancet. 2005;4:705–711.
    1. Acheson SD. Independent Inquiry into Inequalities in Health Report. London: The Stationery Office; 1998
    1. Sampson RJ., Raudenbush SW., Earls F. Neighborhoods and violent crime: a multilevel study of collective effects. Science. 1997;277:918–924.
    1. Whitmer RW., Pelletier KR., Anderson DR., Baase CM., Frost GJ. A wakeup call for corporate America. J Occup Environ Med. 2003;45:916–925.
    1. Pelletier KR. A review and analysis of the clinical- and cost-effectiveness studies of comprehensive health promotion and disease management programs at the worksite: 1998-2000 update. Am J Health Promotion. 2001;16:107–115.
    1. Aldana SG. Financial impact of health promotion programs: a comprehensive review of the literature. Am J Health Promotion. 2001;15:296–320.
    1. Frick KD., Carlson MC., Glass TA., et al. Modeled cost-effectiveness of the Experience Corps Baltimore based on a pilot randomized trial. J Urban Health Bull NY Acad Med. 2004;81:106–117.
    1. Fried LP., Carlson MC., Freedman M., et al. Asocial model for health promotion for an aging population: initial evidence on the experience corps model. J Urban Health Bull NY Acad Med. 2004;81:64–78.
    1. Abbruzzese R. O'Malley and Brown release detailed plan to support Maryland's aging population. Press Release. Jan. 24, 2006

Source: PubMed

3
Suscribir