The evolutionary significance of depression in Pathogen Host Defense (PATHOS-D)

C L Raison, A H Miller, C L Raison, A H Miller

Abstract

Given the manifold ways that depression impairs Darwinian fitness, the persistence in the human genome of risk alleles for the disorder remains a much debated mystery. Evolutionary theories that view depressive symptoms as adaptive fail to provide parsimonious explanations for why even mild depressive symptoms impair fitness-relevant social functioning, whereas theories that suggest that depression is maladaptive fail to account for the high prevalence of depression risk alleles in human populations. These limitations warrant novel explanations for the origin and persistence of depression risk alleles. Accordingly, studies on risk alleles for depression were identified using PubMed and Ovid MEDLINE to examine data supporting the hypothesis that risk alleles for depression originated and have been retained in the human genome because these alleles promote pathogen host defense, which includes an integrated suite of immunological and behavioral responses to infection. Depression risk alleles identified by both candidate gene and genome-wide association study (GWAS) methodologies were found to be regularly associated with immune responses to infection that were likely to enhance survival in the ancestral environment. Moreover, data support the role of specific depressive symptoms in pathogen host defense including hyperthermia, reduced bodily iron stores, conservation/withdrawal behavior, hypervigilance and anorexia. By shifting the adaptive context of depression risk alleles from relations with conspecifics to relations with the microbial world, the Pathogen Host Defense (PATHOS-D) hypothesis provides a novel explanation for how depression can be nonadaptive in the social realm, whereas its risk alleles are nonetheless represented at prevalence rates that bespeak an adaptive function.

Conflict of interest statement

The authors declare no conflict of interest. Charles L Raison serves as a consultant for Pamlab, Biolex Therapeutics and Bristol Myers Squibb. Andrew H Miller has served as a consultant for Abbott Laboratories, AstraZeneca, GlaxoSmithKline, Lundbeck Research USA, F. Hoffmann-La Roche, Schering-Plough Research Institute and Wyeth/Pfizer and has received research support from Centocor, GlaxoSmithKline and Schering-Plough Research Institute.

Figures

Figure 1
Figure 1
The integrated suite of immunological and behavioral responses to infection and wounding that comprise pathogen host defense. Upon encountering a pathogen or cellular debris from tissue damage or destruction, the body reacts with an orchestrated local and systemic response that recruits both immunological and nervous system elements. The response is initiated by interaction of pathogens and/or cellular debris with pattern recognition receptors such as Toll-like receptors on relevant immune cells including macrophages that in turn are linked to inflammatory signaling pathways such as nuclear factor-κB (NF-κB), a lynchpin transcription factor in the host defense cascade. Release of cytokines (including tumor necrosis factor-α (TNF-α), interleukin (IL)-1, IL-6 and interferon-α (IFN-α)) and chemokines as well as the induction of adhesion molecules attracts and activates cells such as T cells at the site of infection/wounding, leading to the cardinal signs of inflammation (redness, heat, swelling and pain) and ultimately promoting local pathogen elimination and wound healing. Cytokines and cells in the peripheral circulation mediate the systemic host response that engages neurocircuits in the brain that mediate hypervigilance (dorsal anterior cingulate cortex (dACC)) to avoid further wounding and pathogen exposure and conservation/withdrawal (basal ganglia), which promotes the shunting of energy resources to pathogen elimination and wound healing. PowerPoint slide

References

    1. Crespi B, Summers K, Dorus S. Adaptive evolution of genes underlying schizophrenia. Proc Biol Sci. 2007;274:2801–2810. doi: 10.1098/rspb.2007.0876.
    1. Kuningas M, May L, Tamm R, van Bodegom D, van den Biggelaar AH, Meij JJ. Selection for genetic variation inducing pro-inflammatory responses under adverse environmental conditions in a Ghanaian population. PLoS One. 2009;4:e7795. doi: 10.1371/journal.pone.0007795.
    1. van Dissel JT, van Langevelde P, Westendorp RG, Kwappenberg K, Frolich M. Anti-inflammatory cytokine profile and mortality in febrile patients. Lancet. 1998;351:950–953. doi: 10.1016/S0140-6736(05)60606-X.
    1. Bermejo-Martin JF, Martin-Loeches I, Rello J, Anton A, Almansa R, Xu L. Host adaptive immunity deficiency in severe pandemic influenza. Crit Care. 2010;14:R167. doi: 10.1186/cc9259.
    1. Westendorp RG, Langermans JA, Huizinga TW, Elouali AH, Verweij CL, Boomsma DI. Genetic influence on cytokine production and fatal meningococcal disease. Lancet. 1997;349:170–173. doi: 10.1016/S0140-6736(96)06413-6.
    1. McDade Thomas W. Life history theory and the immune system: Steps toward a human ecological immunology. American Journal of Physical Anthropology. 2003;122(S37):100–125. doi: 10.1002/ajpa.10398.
    1. Pedron B, Guerin V, Cordeiro DJ, Masmoudi S, Dalle JH, Sterkers G. Development of cytomegalovirus and adenovirus-specific memory CD4 T-cell functions from birth to adulthood. Pediatr Res. 2011;69:106–111. doi: 10.1203/PDR.0b013e318204e469.
    1. Nwachuku N, Gerba CP. Health risks of enteric viral infections in children. Rev Environ Contam Toxicol. 2006;186:1–56.
    1. Chen LC, Rahman M, Sarder AM. Epidemiology and causes of death among children in a rural area of Bangladesh. Int J Epidemiol. 1980;9:25–33. doi: 10.1093/ije/9.1.25.
    1. Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry. 2009;65:732–741. doi: 10.1016/j.biopsych.2008.11.029.
    1. Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71:171–186. doi: 10.1097/PSY.0b013e3181907c1b.
    1. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–457. doi: 10.1016/j.biopsych.2009.09.033.
    1. Zorrilla EP, Luborsky L, McKay JR, Rosenthal R, Houldin A, Tax A. The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav Immun. 2001;15:199–226. doi: 10.1006/brbi.2000.0597.
    1. Gimeno D, Kivimaki M, Brunner EJ, Elovainio M, De Vogli R, Steptoe A. Associations of C-reactive protein and interleukin-6 with cognitive symptoms of depression: 12-year follow-up of the Whitehall II study. Psychol Med. 2008;39:1–11.
    1. van den Biggelaar AH, Gussekloo J, de Craen AJ, Frolich M, Stek ML, van der Mast RC. Inflammation and interleukin-1 signaling network contribute to depressive symptoms but not cognitive decline in old age. Exp Gerontol. 2007;42:693–701. doi: 10.1016/j.exger.2007.01.011.
    1. Pasco JA, Nicholson GC, Williams LJ, Jacka FN, Henry MJ, Kotowicz MA. Association of high-sensitivity C-reactive protein with de novo major depression. Br J Psychiatry. 2010;197:372–377. doi: 10.1192/bjp.bp.109.076430.
    1. Finch CE. Evolution in health and medicine Sackler colloquium: Evolution of the human lifespan and diseases of aging: roles of infection, inflammation, and nutrition. Proc Natl Acad Sci USA. 2010;107(Suppl 1):1718–1724. doi: 10.1073/pnas.0909606106.
    1. Dobson AP, Carper ER. Infectious diseases and human population history. Biosci. 1996;46:115–126. doi: 10.2307/1312814.
    1. Wolfe ND, Dunavan CP, Diamond J. Origins of major human infectious diseases. Nature. 2007;447:279–283. doi: 10.1038/nature05775.
    1. Lovel H. Targeted interventions and infant mortality. Trans R Soc Trop Med Hyg. 1989;83:10–18. doi: 10.1016/0035-9203(89)90689-5.
    1. Kavaliers M, Colwell DD, Choleris E. Parasites and behavior: an ethopharmacological analysis and biomedical implications. Neurosci Biobehav Rev. 1999;23:1037–1045. doi: 10.1016/S0149-7634(99)00035-4.
    1. Goncalves GM, Zamboni DS, Camara NO. The role of innate immunity in septic acute kidney injuries. Shock. 2010;34(Suppl 1):22–26. doi: 10.1097/SHK.0b013e3181e7e69e.
    1. Oberholzer A, Oberholzer C, Moldawer LL. Sepsis syndromes: understanding the role of innate and acquired immunity. Shock. 2001;16:83–96. doi: 10.1097/00024382-200116020-00001.
    1. Klein SL. Parasite manipulation of the proximate mechanisms that mediate social behavior in vertebrates. Physiol Behav. 2003;79:441–449. doi: 10.1016/S0031-9384(03)00163-X.
    1. Kinney DK, Tanaka M. An evolutionary hypothesis of depression and its symptoms, adaptive value, and risk factors. J Nerv Ment Dis. 2009;197:561–567. doi: 10.1097/NMD.0b013e3181b05fa8.
    1. Gershon ES, Alliey-Rodriguez N, Liu C. After GWAS: searching for genetic risk for schizophrenia and bipolar disorder. Am J Psychiatry. 2011;168:253–256. doi: 10.1176/appi.ajp.2010.10091340.
    1. Licinio J, Dong C, Wong ML. Novel sequence variations in the brain-derived neurotrophic factor gene and association with major depression and antidepressant treatment response. Arch Gen Psychiatry. 2009;66:488–497. doi: 10.1001/archgenpsychiatry.2009.38.
    1. Keller MC, Miller G. Resolving the paradox of common, harmful, heritable mental disorders: which evolutionary genetic models work best? Behav Brain Sci. 2006;29:385–404. doi: 10.1017/S0140525X06009095.
    1. Detera-Wadleigh SD, Akula N. A systems approach to the biology of mood disorders through network analysis of candidate genes. Pharmacopsychiatry. 2011;44(Suppl 1):S35–S42. doi: 10.1055/s-0031-1275275.
    1. Wong ML, Dong C, Maestre-Mesa J, Licinio J. Polymorphisms in inflammation-related genes are associated with susceptibility to major depression and antidepressant response. Mol Psychiatry. 2008;13:800–812. doi: 10.1038/mp.2008.59.
    1. Cerri AP, Arosio B, Viazzoli C, Confalonieri R, Vergani C, Annoni G. The −308 (G/A) single nucleotide polymorphism in the TNF-alpha gene and the risk of major depression in the elderly. Int J Geriatr Psychiatry. 2010;25:219–223. doi: 10.1002/gps.2323.
    1. Clerici M, Arosio B, Mundo E, Cattaneo E, Pozzoli S, Dell'osso B. Cytokine polymorphisms in the pathophysiology of mood disorders. CNS Spectr. 2009;14:419–425. doi: 10.1017/S1092852900020393.
    1. Jun TY, Pae CU, Hoon H, Chae JH, Bahk WM, Kim KS. Possible association between -G308A tumour necrosis factor-alpha gene polymorphism and major depressive disorder in the Korean population. Psychiatr Genet. 2003;13:179–181. doi: 10.1097/00041444-200309000-00008.
    1. Pae CU, Lee KU, Han H, Serretti A, Jun TY. Tumor necrosis factor alpha gene-G308A polymorphism associated with bipolar I disorder in the Korean population. Psychiatry Res. 2004;125:65–68. doi: 10.1016/j.psychres.2003.06.002.
    1. Bosker FJ, Hartman CA, Nolte IM, Prins BP, Terpstra P, Posthuma D. Poor replication of candidate genes for major depressive disorder using genome-wide association data. Mol Psychiatry. 2011;16:516–532. doi: 10.1038/mp.2010.38.
    1. Wheway J, Herzog H, Mackay F. NPY and receptors in immune and inflammatory diseases. Curr Top Med Chem. 2007;7:1743–1752. doi: 10.2174/156802607782341046.
    1. Ferreira R, Xapelli S, Santos T, Silva AP, Cristovao A, Cortes L. Neuropeptide Y modulation of interleukin-1{beta} (IL-1{beta})-induced nitric oxide production in microglia. J Biol Chem. 2010;285:41921–41934. doi: 10.1074/jbc.M110.164020.
    1. Wheway J, Herzog H, Mackay F. The Y1 receptor for NPY: a key modulator of the adaptive immune system. Peptides. 2007;28:453–458. doi: 10.1016/j.peptides.2006.09.030.
    1. Heilig M, Zachrisson O, Thorsell A, Ehnvall A, Mottagui-Tabar S, Sjogren M. Decreased cerebrospinal fluid neuropeptide Y (NPY) in patients with treatment refractory unipolar major depression: preliminary evidence for association with preproNPY gene polymorphism. J Psychiatr Res. 2004;38:113–121. doi: 10.1016/S0022-3956(03)00101-8.
    1. Sjoholm LK, Melas PA, Forsell Y, Lavebratt C. PreproNPY Pro7 protects against depression despite exposure to environmental risk factors. J Affect Disord. 2009;118:124–130. doi: 10.1016/j.jad.2009.02.009.
    1. Mickey BJ, Zhou Z, Heitzeg MM, Heinz E, Hodgkinson CA, Hsu DT. Emotion processing, major depression, and functional genetic variation of neuropeptide Y. Arch Gen Psychiatry. 2011;68:158–166. doi: 10.1001/archgenpsychiatry.2010.197.
    1. Morales-Medina JC, Dumont Y, Quirion R. A possible role of neuropeptide Y in depression and stress. Brain Res. 2010;1314:194–205. doi: 10.1016/j.brainres.2009.09.077.
    1. Kallio J, Pesonen U, Kaipio K, Karvonen MK, Jaakkola U, Heinonen OJ. Altered intracellular processing and release of neuropeptide Y due to leucine 7 to proline 7 polymorphism in the signal peptide of preproneuropeptide Y in humans. FASEB J. 2001;15:1242–1244. doi: 10.1096/fj.00-0436fje.
    1. Wilson AG, di Giovine FS, Blakemore AI, Duff GW. Single base polymorphism in the human tumour necrosis factor alpha (TNF alpha) gene detectable by NcoI restriction of PCR product. Hum Mol Genet. 1992;1:353. doi: 10.1093/hmg/1.5.353.
    1. Lotrich FE, Ferrell RE, Rabinovitz M, Pollock BG. Labile anger during interferon Alfa treatment is associated with a polymorphism in tumor necrosis factor alpha. Clin Neuropharmacol. 2010;33:191–197. doi: 10.1097/WNF.0b013e3181de8966.
    1. Correa PA, Gomez LM, Cadena J, Anaya JM. Autoimmunity and tuberculosis. Opposite association with TNF polymorphism. J Rheumatol. 2005;32:219–224.
    1. Kerr JR, McCoy M, Burke B, Mattey DL, Pravica V, Hutchinson IV. Cytokine gene polymorphisms associated with symptomatic parvovirus B19 infection. J Clin Pathol. 2003;56:725–727. doi: 10.1136/jcp.56.10.725.
    1. Chen DQ, Zeng Y, Zhou J, Yang L, Jiang S, Huang JD. Association of candidate susceptible loci with chronic infection with hepatitis B virus in a Chinese population. J Med Virol. 2010;82:371–378. doi: 10.1002/jmv.21716.
    1. Surbatovic M, Grujic K, Cikota B, Jevtic M, Filipovic N, Romic P. Polymorphisms of genes encoding tumor necrosis factor-alpha, interleukin-10, cluster of differentiation-14 and interleukin-1ra in critically ill patients. J Crit Care. 2010;25:542. doi: 10.1016/j.jcrc.2009.12.003.
    1. Larcombe LA, Orr PH, Lodge AM, Brown JS, Dembinski IJ, Milligan LC. Functional gene polymorphisms in Canadian aboriginal populations with high rates of tuberculosis. J Infect Dis. 2008;198:1175–1179. doi: 10.1086/592049.
    1. Masuda M, Senju S, Fujii Si S, Terasaki Y, Takeya M, Hashimoto Si S. Identification and immunocytochemical analysis of DCNP1, a dendritic cell-associated nuclear protein. Biochem Biophys Res Commun. 2002;290:1022–1029. doi: 10.1006/bbrc.2001.6202.
    1. Zhou T, Wang S, Ren H, Qi XR, Luchetti S, Kamphuis W. Dendritic cell nuclear protein-1, a novel depression-related protein, upregulates corticotropin-releasing hormone expression. Brain. 2010;133:3069–3079. doi: 10.1093/brain/awq207.
    1. Kim Y, Park CS, Shin HD, Choi JW, Cheong HS, Park BL. A promoter nucleotide variant of the dendritic cell-specific DCNP1 associates with serum IgE levels specific for dust mite allergens among the Korean asthmatics. Genes Immun. 2007;8:369–378. doi: 10.1038/sj.gene.6364394.
    1. Lopez-Leon S, Janssens AC, Hofman A, Claes S, Breteler MM, Tiemeier H. No association between the angiotensin-converting enzyme gene and major depression: a case-control study and meta-analysis. Psychiatr Genet. 2006;16:225–226. doi: 10.1097/01.ypg.0000242191.51397.3d.
    1. Virchow S, Ansorge N, Rosskopf D, Rubben H, Siffert W. The G protein beta3 subunit splice variant Gbeta3-s causes enhanced chemotaxis of human neutrophils in response to interleukin-8. Naunyn Schmiedebergs Arch Pharmacol. 1999;360:27–32. doi: 10.1007/s002109900040.
    1. Lindemann M, Virchow S, Ramann F, Barsegian V, Kreuzfelder E, Siffert W. The G protein beta3 subunit 825T allele is a genetic marker for enhanced T cell response. FEBS Lett. 2001;495:82–86. doi: 10.1016/S0014-5793(01)02339-0.
    1. Romundstad S, Melien O, Holmen J. The G protein beta3 subunit C825T polymorphism is associated with microalbuminuria in hypertensive women and cardiovascular disease in hypertensive men. Am J Hypertens. 2010;23:1114–1120. doi: 10.1038/ajh.2010.131.
    1. Holmen OL, Romundstad S, Melien O. Association between the G protein beta3 subunit C825T polymorphism and the occurrence of cardiovascular disease in hypertensives: The Nord-Trondelag Health Study (HUNT) Am J Hypertens. 2010;23:1121–1127. doi: 10.1038/ajh.2010.121.
    1. Hauge Opdal S, Melien O, Rootwelt H, Vege A, Arnestad M, Ole Rognum T. The G protein beta3 subunit 825C allele is associated with sudden infant death due to infection. Acta Paediatr. 2006;95:1129–1132. doi: 10.1080/08035250600580529.
    1. Bagos PG, Elefsinioti AL, Nikolopoulos GK, Hamodrakas SJ. The GNB3 C825T polymorphism and essential hypertension: a meta-analysis of 34 studies including 14,094 cases and 17,760 controls. J Hypertens. 2007;25:487–500. doi: 10.1097/HJH.0b013e328011db24.
    1. Ahlenstiel G, Nischalke HD, Bueren K, Berg T, Vogel M, Biermer M. The GNB3 C825T polymorphism affects response to HCV therapy with pegylated interferon in HCV/HIV co-infected but not in HCV mono-infected patients. J Hepatol. 2007;47:348–355. doi: 10.1016/j.jhep.2007.04.008.
    1. Sarrazin C, Berg T, Weich V, Mueller T, Frey UH, Zeuzem S. GNB3 C825T polymorphism and response to interferon-alfa/ribavirin treatment in patients with hepatitis C virus genotype 1 (HCV-1) infection. J Hepatol. 2005;43:388–393. doi: 10.1016/j.jhep.2005.03.020.
    1. Brockmeyer NH, Potthoff A, Kasper A, Nabring C, Jockel KH, Siffert W. GNB3 C825T polymorphism and response to anti-retroviral combination therapy in HIV-1-infected patients--a pilot study. Eur J Med Res. 2005;10:489–494.
    1. Lindemann M, Barsegian V, Siffert W, Ferencik S, Roggendorf M, Grosse-Wilde H. Role of G protein beta3 subunit C825T and HLA class II polymorphisms in the immune response after HBV vaccination. Virology. 2002;297:245–252. doi: 10.1006/viro.2002.1467.
    1. Fodil-Cornu N, Kozij N, Wu Q, Rozen R, Vidal SM. Methylenetetrahydrofolate reductase (MTHFR) deficiency enhances resistance against cytomegalovirus infection. Genes Immun. 2009;10:662–666. doi: 10.1038/gene.2009.50.
    1. Imamura A, Murakami R, Takahashi R, Cheng XW, Numaguchi Y, Murohara T. Low folate levels may be an atherogenic factor regardless of homocysteine levels in young healthy nonsmokers. Metabolism. 2010;59:728–733. doi: 10.1016/j.metabol.2009.09.017.
    1. Fujimaki C, Hayashi H, Tsuboi S, Matsuyama T, Kosuge K, Yamada H. Plasma total homocysteine level and methylenetetrahydrofolate reductase 677C>T genetic polymorphism in Japanese patients with rheumatoid arthritis. Biomarkers. 2009;14:49–54. doi: 10.1080/13547500902730664.
    1. Dedoussis GV, Panagiotakos DB, Pitsavos C, Chrysohoou C, Skoumas J, Choumerianou D. An association between the methylenetetrahydrofolate reductase (MTHFR) C677T mutation and inflammation markers related to cardiovascular disease. Int J Cardiol. 2005;100:409–414. doi: 10.1016/j.ijcard.2004.08.038.
    1. Chen AR, Zhang HG, Wang ZP, Fu SJ, Yang PQ, Ren JG. C-reactive protein, vitamin B12 and C677T polymorphism of N-5,10-methylenetetrahydrofolate reductase gene are related to insulin resistance and risk factors for metabolic syndrome in Chinese population. Clin Invest Med. 2010;33:E290–E297. doi: 10.25011/cim.v33i5.14354.
    1. Hammons AL, Summers CM, Woodside JV, McNulty H, Strain JJ, Young IS. Folate/homocysteine phenotypes and MTHFR 677C>T genotypes are associated with serum levels of monocyte chemoattractant protein-1. Clin Immunol. 2009;133:132–137. doi: 10.1016/j.clim.2009.06.008.
    1. Lu ZY, Morales M, Khartulyari S, Mei M, Murphy KM, Stanislawska-Sachadyn A. Genetic and biochemical determinants of serum concentrations of monocyte chemoattractant protein-1, a potential neural tube defect risk factor. Birth Defects Res A Clin Mol Teratol. 2008;82:736–741. doi: 10.1002/bdra.20507.
    1. Bronowicki JP, Abdelmouttaleb I, Peyrin-Biroulet L, Venard V, Khiri H, Chabi N. Methylenetetrahydrofolate reductase 677T allele protects against persistent HBV infection in West Africa. J Hepatol. 2008;48:532–539. doi: 10.1016/j.jhep.2007.11.017.
    1. Simhan HN, Himes KP, Venkataramanan R, Bodnar LM. Maternal serum folate species in early pregnancy and lower genital tract inflammatory milieu. Am J Obstet Gynecol. 2011;205:61. e1–e7. doi: 10.1016/j.ajog.2011.03.039.
    1. Chillemi R, Zappacosta B, Simpore J, Persichilli S, Musumeci M, Musumeci S. Hyperhomocysteinemia in acute Plasmodium falciparum malaria: an effect of host-parasite interaction. Clin Chim Acta. 2004;348:113–120. doi: 10.1016/j.cccn.2004.05.007.
    1. Maeda M, Yamamoto I, Fukuda M, Motomura T, Nishida M, Nonen S. MTHFR gene polymorphism is susceptible to diabetic retinopathy but not to diabetic nephropathy in Japanese type 2 diabetic patients. J Diabetes Complications. 2008;22:119–125. doi: 10.1016/j.jdiacomp.2006.12.002.
    1. Khandanpour N, Willis G, Meyer FJ, Armon MP, Loke YK, Wright AJ. Peripheral arterial disease and methylenetetrahydrofolate reductase (MTHFR) C677T mutations: a case-control study and meta-analysis. J Vasc Surg. 2009;49:711–718. doi: 10.1016/j.jvs.2008.10.004.
    1. Ferrara F, Novo S, Grimaudo S, Raimondi F, Meli F, Amato C. Methylenetetrahydrofolate reductase mutation in subjects with abdominal aortic aneurysm subdivided for age. Clin Hemorheol Microcirc. 2006;34:421–426.
    1. Pollex RL, Mamakeesick M, Zinman B, Harris SB, Hanley AJ, Hegele RA. Methylenetetrahydrofolate reductase polymorphism 677C>T is associated with peripheral arterial disease in type 2 diabetes. Cardiovasc Diabetol. 2005;4:17. doi: 10.1186/1475-2840-4-17.
    1. Chen J, Ma J, Stampfer MJ, Palomeque C, Selhub J, Hunter DJ. Linkage disequilibrium between the 677C>T and 1298A>C polymorphisms in human methylenetetrahydrofolate reductase gene and their contributions to risk of colorectal cancer. Pharmacogenetics. 2002;12:339–342. doi: 10.1097/00008571-200206000-00011.
    1. Movva S, Alluri RV, Venkatasubramanian S, Vedicherla B, Vattam KK, Ahuja YR. Association of methylene tetrahydrofolate reductase C677T genotype with type 2 diabetes mellitus patients with and without renal complications. Genet Test Mol Biomarkers. 2011;15:257–261. doi: 10.1089/gtmb.2010.0118.
    1. Reyes-Engel A, Munoz E, Gaitan MJ, Fabre E, Gallo M, Dieguez JL. Implications on human fertility of the 677C → T and 1298A → C polymorphisms of the MTHFR gene: consequences of a possible genetic selection. Mol Hum Reprod. 2002;8:952–957. doi: 10.1093/molehr/8.10.952.
    1. Gerdes LU. The common polymorphism of apolipoprotein E: geographical aspects and new pathophysiological relations. Clin Chem Lab Med. 2003;41:628–631. doi: 10.1515/CCLM.2003.094.
    1. Urosevic N, Martins RN. Infection and Alzheimer's disease: the APOE epsilon4 connection and lipid metabolism. J Alzheimers Dis. 2008;13:421–435. doi: 10.3233/JAD-2008-13407.
    1. Mahley RW, Rall SC., Jr Apolipoprotein E: far more than a lipid transport protein. Annu Rev Genomics Hum Genet. 2000;1:507–537. doi: 10.1146/annurev.genom.1.1.507.
    1. Jofre-Monseny L, Minihane AM, Rimbach G. Impact of apoE genotype on oxidative stress, inflammation and disease risk. Mol Nutr Food Res. 2008;52:131–145. doi: 10.1002/mnfr.200700322.
    1. Wozniak MA, Maude RJ, Innes JA, Hawkey PM, Itzhaki RF. Apolipoprotein E-epsilon2 confers risk of pulmonary tuberculosis in women from the Indian subcontinent--a preliminary study. J Infect. 2009;59:219–222. doi: 10.1016/j.jinf.2009.07.003.
    1. Roy A, Karoum F, Pollack S. Marked reduction in indexes of dopamine metabolism among patients with depression who attempt suicide. Arch Gen Psychiatry. 1992;49:447–450. doi: 10.1001/archpsyc.1992.01820060027004.
    1. Meyer JH, Kruger S, Wilson AA, Christensen BK, Goulding VS, Schaffer A. Lower dopamine transporter binding potential in striatum during depression. NeuroReport. 2001;12:4121–4125. doi: 10.1097/00001756-200112210-00052.
    1. Lambert G, Johansson M, Agren H, Friberg P. Reduced brain norepinephrine and dopamine release in treatment-refractory depressive illness: evidence in support of the catecholamine hypothesis of mood disorders. Arch Gen Psychiatry. 2000;57:787–793. doi: 10.1001/archpsyc.57.8.787.
    1. Klimek V, Schenck JE, Han H, Stockmeier CA, Ordway GA. Dopaminergic abnormalities in amygdaloid nuclei in major depression: a postmortem study. Biol Psychiatry. 2002;52:740–748. doi: 10.1016/S0006-3223(02)01383-5.
    1. Kavelaars A, Cobelens PM, Teunis MA, Heijnen CJ. Changes in innate and acquired immune responses in mice with targeted deletion of the dopamine transporter gene. J Neuroimmunol. 2005;161:162–168. doi: 10.1016/j.jneuroim.2005.01.004.
    1. Alaniz RC, Thomas SA, Perez-Melgosa M, Mueller K, Farr AG, Palmiter RD. Dopamine beta-hydroxylase deficiency impairs cellular immunity. Proc Natl Acad Sci USA. 1999;96:2274–2278. doi: 10.1073/pnas.96.5.2274.
    1. Cobat A, Gallant CJ, Simkin L, Black GF, Stanley K, Hughes J. Two loci control tuberculin skin test reactivity in an area hyperendemic for tuberculosis. J Exp Med. 2009;206:2583–2591. doi: 10.1084/jem.20090892.
    1. Stein CM, Zalwango S, Malone LL, Won S, Mayanja-Kizza H, Mugerwa RD. Genome scan of M. tuberculosis infection and disease in Ugandans. PLoS One. 2008;3:e4094. doi: 10.1371/journal.pone.0004094.
    1. Mill J, Asherson P, Browes C, D'Souza U, Craig I. Expression of the dopamine transporter gene is regulated by the 3′ UTR VNTR: evidence from brain and lymphocytes using quantitative RT-PCR. Am J Med Genet. 2002;114:975–979. doi: 10.1002/ajmg.b.10948.
    1. van de Giessen E, de Win MM, Tanck MW, van den Brink W, Baas F, Booij J. Striatal dopamine transporter availability associated with polymorphisms in the dopamine transporter gene SLC6A3. J Nucl Med. 2009;50:45–52. doi: 10.2967/jnumed.108.053652.
    1. Opdal SH, Vege A, Rognum TO. Serotonin transporter gene variation in sudden infant death syndrome. Acta Paediatr. 2008;97:861–865. doi: 10.1111/j.1651-2227.2008.00813.x.
    1. Narita N, Narita M, Takashima S, Nakayama M, Nagai T, Okado N. Serotonin transporter gene variation is a risk factor for sudden infant death syndrome in the Japanese population. Pediatrics. 2001;107:690–692. doi: 10.1542/peds.107.4.690.
    1. Weese-Mayer DE, Berry-Kravis EM, Maher BS, Silvestri JM, Curran ME, Marazita ML. Sudden infant death syndrome: association with a promoter polymorphism of the serotonin transporter gene. Am J Med Genet A. 2003;117A:268–274. doi: 10.1002/ajmg.a.20005.
    1. Prandota J. Possible pathomechanisms of sudden infant death syndrome: key role of chronic hypoxia, infection/inflammation states, cytokine irregularities, and metabolic trauma in genetically predisposed infants. Am J Ther. 2004;11:517–546. doi: 10.1097/.
    1. Fredericks CA, Drabant EM, Edge MD, Tillie JM, Hallmayer J, Ramel W. Healthy young women with serotonin transporter SS polymorphism show a pro-inflammatory bias under resting and stress conditions. Brain Behav Immun. 2010;24:350–357. doi: 10.1016/j.bbi.2009.10.014.
    1. Su S, Zhao J, Bremner JD, Miller AH, Tang W, Bouzyk M. Serotonin transporter gene, depressive symptoms, and interleukin-6. Circ Cardiovasc Genet. 2009;2:614–620. doi: 10.1161/CIRCGENETICS.109.870386.
    1. Murray DR, Schaller M. Historical prevalence of infectious diseases within 230 geopolitical regions: a tool for investigating origins of culture. J Cross Cult Psychol. 2010;41:99–108. doi: 10.1177/0022022109349510.
    1. Chiao JY, Blizinsky KD. Culture-gene coevolution of individualism-collectivism and the serotonin transporter gene. Proc Biol Sci. 2010;277:529–537. doi: 10.1098/rspb.2009.1650.
    1. Wray NR, Pergadia ML, Blackwood DH, Penninx BW, Gordon SD, Nyholt DR. Genome-wide association study of major depressive disorder: new results, meta-analysis, and lessons learned. Mol Psychiatry. 2012;17:36–48. doi: 10.1038/mp.2010.109.
    1. Cattaneo M, Otsu M, Fagioli C, Martino S, Lotti LV, Sitia R. SEL1L and HRD1 are involved in the degradation of unassembled secretory Ig-mu chains. J Cell Physiol. 2008;215:794–802. doi: 10.1002/jcp.21364.
    1. Schelhaas M, Malmstrom J, Pelkmans L, Haugstetter J, Ellgaard L, Grunewald K. Simian Virus 40 depends on ER protein folding and quality control factors for entry into host cells. Cell. 2007;131:516–529. doi: 10.1016/j.cell.2007.09.038.
    1. Oresic K, Ng CL, Tortorella D. TRAM1 participates in human cytomegalovirus US2- and US11-mediated dislocation of an endoplasmic reticulum membrane glycoprotein. J Biol Chem. 2009;284:5905–5914. doi: 10.1074/jbc.M807568200.
    1. Ban Y, Taniyama M, Tozaki T, Yanagawa T, Tomita M. SEL1L microsatellite polymorphism in Japanese patients with autoimmune thyroid diseases. Thyroid. 2001;11:335–338. doi: 10.1089/10507250152039064.
    1. Song J, Duncan MJ, Li G, Chan C, Grady R, Stapleton A. A novel TLR4-mediated signaling pathway leading to IL-6 responses in human bladder epithelial cells. Plos Pathog. 2007;3:e60. doi: 10.1371/journal.ppat.0030060.
    1. Abera AB, Sales KJ, Catalano RD, Katz AA, Jabbour HN. EP2 receptor mediated cAMP release is augmented by PGF 2 alpha activation of the FP receptor via the calcium-calmodulin pathway. Cell Signal. 2010;22:71–79. doi: 10.1016/j.cellsig.2009.09.012.
    1. Klempan TA, Rujescu D, Merette C, Himmelman C, Sequeira A, Canetti L. Profiling brain expression of the spermidine/spermine N1-acetyltransferase 1 (SAT1) gene in suicide. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:934–943. doi: 10.1002/ajmg.b.30920.
    1. Tabeta K, Hoebe K, Janssen EM, Du X, Georgel P, Crozat K. The Unc93b1 mutation 3d disrupts exogenous antigen presentation and signaling via Toll-like receptors 3, 7 and 9. Nat Immunol. 2006;7:156–164. doi: 10.1038/ni1297.
    1. Koehn J, Huesken D, Jaritz M, Rot A, Zurini M, Dwertmann A. Assessing the function of human UNC-93B in Toll-like receptor signaling and major histocompatibility complex II response. Hum Immunol. 2007;68:871–878. doi: 10.1016/j.humimm.2007.07.007.
    1. Pifer R, Benson A, Sturge CR, Yarovinsky F. UNC93B1 is essential for TLR11 activation and IL-12-dependent host resistance to Toxoplasma gondii. J Biol Chem. 2011;286:3307–3314. doi: 10.1074/jbc.M110.171025.
    1. Crozat K, Vivier E, Dalod M. Crosstalk between components of the innate immune system: promoting anti-microbial defenses and avoiding immunopathologies. Immunol Rev. 2009;227:129–149. doi: 10.1111/j.1600-065X.2008.00736.x.
    1. Lang R, Kofler B. The galanin peptide family in inflammation. Neuropeptides. 2011;45:1–8. doi: 10.1016/j.npep.2010.10.005.
    1. Matkowskyj KA, Danilkovich A, Marrero J, Savkovic SD, Hecht G, Benya RV. Galanin-1 receptor up-regulation mediates the excess colonic fluid production caused by infection with enteric pathogens. Nat Med. 2000;6:1048–1051. doi: 10.1038/79563.
    1. McDonald AC, Schuijers JA, Gundlach AL, Grills BL. Galanin treatment offsets the inhibition of bone formation and downregulates the increase in mouse calvarial expression of TNFalpha and GalR2 mRNA induced by chronic daily injections of an injurious vehicle. Bone. 2007;40:895–903. doi: 10.1016/j.bone.2006.10.018.
    1. Su Y, Ganea D, Peng X, Jonakait GM. Galanin down-regulates microglial tumor necrosis factor-alpha production by a post-transcriptional mechanism. J Neuroimmunol. 2003;134:52–60. doi: 10.1016/S0165-5728(02)00397-1.
    1. Christiansen SH, Olesen MV, Wortwein G, Woldbye DP. Fluoxetine reverts chronic restraint stress-induced depression-like behaviour and increases neuropeptide Y and galanin expression in mice. Behav Brain Res. 2011;216:585–591. doi: 10.1016/j.bbr.2010.08.044.
    1. Wardi Le Maitre T, Xia S, Le Maitre E, Dun XP, Lu J, Theodorsson E. Galanin receptor 2 overexpressing mice display an antidepressive-like phenotype: possible involvement of the subiculum. Neurosci. 2011;190:270–288. doi: 10.1016/j.neuroscience.2011.05.015.
    1. Davidson S, Lear M, Shanley L, Hing B, Baizan-Edge A, Herwig A. Differential activity by polymorphic variants of a remote enhancer that supports galanin expression in the hypothalamus and amygdala: implications for obesity, depression and alcoholism. Neuropsychopharmacology. 2011;36:2211–2221. doi: 10.1038/npp.2011.93.
    1. Rauch I, Lundstrom L, Hell M, Sperl W, Kofler B. Galanin message-associated peptide suppresses growth and the budded-to-hyphal-form transition of Candida albicans. Antimicrob Agents Chemother. 2007;51:4167–4170. doi: 10.1128/AAC.00166-07.
    1. Ikegami K, Sato S, Nakamura K, Ostrowski LE, Setou M. Tubulin polyglutamylation is essential for airway ciliary function through the regulation of beating asymmetry. Proc Natl Acad Sci USA. 2010;107:10490–10495. doi: 10.1073/pnas.1002128107.
    1. Chen C, Han YH, Yang Z, Rodrigues AD. Effect of interferon-alpha2b on the expression of various drug-metabolizing enzymes and transporters in co-cultures of freshly prepared human primary hepatocytes. Xenobiotica. 2011;41:476–485. doi: 10.3109/00498254.2011.560971.
    1. Crossland H, Constantin-Teodosiu D, Greenhaff PL, Gardiner SM. Low-dose dexamethasone prevents endotoxaemia-induced muscle protein loss and impairment of carbohydrate oxidation in rat skeletal muscle. J Physiol. 2010;588:1333–1347. doi: 10.1113/jphysiol.2009.183699.
    1. Palomer X, Alvarez-Guardia D, Rodriguez-Calvo R, Coll T, Laguna JC, Davidson MM. TNF-alpha reduces PGC-1alpha expression through NF-kappaB and p38 MAPK leading to increased glucose oxidation in a human cardiac cell model. Cardiovasc Res. 2009;81:703–712. doi: 10.1093/cvr/cvn327.
    1. Zapata-Gonzalez F, Rueda F, Petriz J, Domingo P, Villarroya F, Diaz-Delfin J. Human dendritic cell activities are modulated by the omega-3 fatty acid, docosahexaenoic acid, mainly through PPAR(gamma):RXR heterodimers: comparison with other polyunsaturated fatty acids. J Leukoc Biol. 2008;84:1172–1182. doi: 10.1189/jlb.1007688.
    1. Nawa Y, Kawahara K, Tancharoen S, Meng X, Sameshima H, Ito T. Nucleophosmin may act as an alarmin: implications for severe sepsis. J Leukoc Biol. 2009;86:645–653. doi: 10.1189/jlb.1008644.
    1. Sarek G, Jarviluoma A, Moore HM, Tojkander S, Vartia S, Biberfeld P. Nucleophosmin phosphorylation by v-cyclin-CDK6 controls KSHV latency. Plos Pathog. 2010;6:e1000818. doi: 10.1371/journal.ppat.1000818.
    1. Johnson JS, Samulski RJ. Enhancement of adeno-associated virus infection by mobilizing capsids into and out of the nucleolus. J Virol. 2009;83:2632–2644. doi: 10.1128/JVI.02309-08.
    1. Lee JJ, Seah JB, Chow VT, Poh CL, Tan EL. Comparative proteome analyses of host protein expression in response to Enterovirus 71 and Coxsackievirus A16 infections. J Proteomics. 2011;74:2018–2024. doi: 10.1016/j.jprot.2011.05.022.
    1. Zeng Y, Ye L, Zhu S, Zheng H, Zhao P, Cai W. The nucleocapsid protein of SARS-associated coronavirus inhibits B23 phosphorylation. Biochem Biophys Res Commun. 2008;369:287–291. doi: 10.1016/j.bbrc.2008.01.096.
    1. Fankhauser C, Izaurralde E, Adachi Y, Wingfield P, Laemmli UK. Specific complex of human immunodeficiency virus type 1 rev and nucleolar B23 proteins: dissociation by the Rev response element. Mol Cell Biol. 1991;11:2567–2575. doi: 10.1128/MCB.11.5.2567.
    1. Lymberopoulos MH, Bourget A, Abdeljelil NB, Pearson A. Involvement of the UL24 protein in herpes simplex virus 1-induced dispersal of B23 and in nuclear egress. Virology. 2011;412:341–348. doi: 10.1016/j.virol.2011.01.016.
    1. Garcia MA, Meurs EF, Esteban M. The dsRNA protein kinase PKR: virus and cell control. Biochimie. 2007;89:799–811. doi: 10.1016/j.biochi.2007.03.001.
    1. Burrows JF, McGrattan MJ, Johnston JA. The DUB/USP17 deubiquitinating enzymes, a multigene family within a tandemly repeated sequence. Genomics. 2005;85:524–529. doi: 10.1016/j.ygeno.2004.11.013.
    1. Burrows JF, McGrattan MJ, Rascle A, Humbert M, Baek KH, Johnston JA. DUB-3, a cytokine-inducible deubiquitinating enzyme that blocks proliferation. J Biol Chem. 2004;279:13993–14000. doi: 10.1074/jbc.M311291200.
    1. Chen R, Zhang L, Zhong B, Tan B, Liu Y, Shu HB. The ubiquitin-specific protease 17 is involved in virus-triggered type I IFN signaling. Cell Res. 2010;20:802–811. doi: 10.1038/cr.2010.41.
    1. Liu Y, Blackwood DH, Caesar S, de Geus EJ, Farmer A, Ferreira MA. Meta-analysis of genome-wide association data of bipolar disorder and major depressive disorder. Mol Psychiatry. 2011;16:2–4. doi: 10.1038/mp.2009.107.
    1. Green EK, Grozeva D, Jones I, Jones L, Kirov G, Caesar S. The bipolar disorder risk allele at CACNA1C also confers risk of recurrent major depression and of schizophrenia. Mol Psychiatry. 2010;15:1016–1022. doi: 10.1038/mp.2009.49.
    1. Erk S, Meyer-Lindenberg A, Schnell K, Opitz von Boberfeld C, Esslinger C, Kirsch P. Brain function in carriers of a genome-wide supported bipolar disorder variant. Arch Gen Psychiatry. 2010;67:803–811. doi: 10.1001/archgenpsychiatry.2010.94.
    1. Bigos KL, Mattay VS, Callicott JH, Straub RE, Vakkalanka R, Kolachana B. Genetic variation in CACNA1C affects brain circuitries related to mental illness. Arch Gen Psychiatry. 2010;67:939–945. doi: 10.1001/archgenpsychiatry.2010.96.
    1. Perrier E, Pompei F, Ruberto G, Vassos E, Collier D, Frangou S. Initial evidence for the role of CACNA1C on subcortical brain morphology in patients with bipolar disorder. Eur Psychiatry. 2011;26:135–137. doi: 10.1016/j.eurpsy.2010.10.004.
    1. Suzuki Y, Inoue T, Ra C. L-type Ca2+ channels: a new player in the regulation of Ca2+ signaling, cell activation and cell survival in immune cells. Mol Immunol. 2010;47:640–648. doi: 10.1016/j.molimm.2009.10.013.
    1. Badou A, Jha MK, Matza D, Mehal WZ, Freichel M, Flockerzi V. Critical role for the beta regulatory subunits of Cav channels in T lymphocyte function. Proc Natl Acad Sci USA. 2006;103:15529–15534. doi: 10.1073/pnas.0607262103.
    1. Matza D, Badou A, Kobayashi KS, Goldsmith-Pestana K, Masuda Y, Komuro A. A scaffold protein, AHNAK1, is required for calcium signaling during T cell activation. Immunity. 2008;28:64–74. doi: 10.1016/j.immuni.2007.11.020.
    1. Gupta S, Salam N, Srivastava V, Singla R, Behera D, Khayyam KU. Voltage gated calcium channels negatively regulate protective immunity to Mycobacterium tuberculosis. PLoS One. 2009;4:e5305. doi: 10.1371/journal.pone.0005305.
    1. Radermacher AN, Crabtree GR. Monster protein controls calcium entry and fights infection. Immunity. 2008;28:13–14. doi: 10.1016/j.immuni.2007.12.009.
    1. Suzuki Y, Yoshimaru T, Inoue T, Ra C. Ca v 1.2L-type Ca2+ channel protects mast cells against activation-induced cell death by preventing mitochondrial integrity disruption. Mol Immunol. 2009;46:2370–2380. doi: 10.1016/j.molimm.2009.03.017.
    1. Das R, Burke T, Van Wagoner DR, Plow EF. L-type calcium channel blockers exert an antiinflammatory effect by suppressing expression of plasminogen receptors on macrophages. Circ Res. 2009;105:167–175. doi: 10.1161/CIRCRESAHA.109.200311.
    1. Matza D, Badou A, Jha MK, Willinger T, Antov A, Sanjabi S. Requirement for AHNAK1-mediated calcium signaling during T lymphocyte cytolysis. Proc Natl Acad Sci USA. 2009;106:9785–9790. doi: 10.1073/pnas.0902844106.
    1. Liao P, Soong TW. CaV1.2 channelopathies: from arrhythmias to autism, bipolar disorder, and immunodeficiency. Pflugers Arch. 2010;460:353–359. doi: 10.1007/s00424-009-0753-0.
    1. Das R, Plow EF. A new function for old drugs. Cell Cycle. 2010;9:638–639. doi: 10.4161/cc.9.4.11016.
    1. Balog Z, Kiss I, Keri S. CACNA1C risk allele for psychotic disorders is related to the activation of the AKT-pathway. Am J Psychiatry. 2010;167:1276–1277. doi: 10.1176/appi.ajp.2010.10040635.
    1. Li XQ, Cao W, Li T, Zeng AG, Hao LL, Zhang XN. Amlodipine inhibits TNF-alpha production and attenuates cardiac dysfunction induced by lipopolysaccharide involving PI3K/Akt pathway. Int Immunopharmacol. 2009;9:1032–1041. doi: 10.1016/j.intimp.2009.04.010.
    1. Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell. 2004;119:19–31. doi: 10.1016/j.cell.2004.09.011.
    1. Okazaki R, Iwasaki YK, Miyauchi Y, Hirayama Y, Kobayashi Y, Katoh T. Lipopolysaccharide induces atrial arrhythmogenesis via down-regulation of L-type Ca2+ channel genes in rats. Int Heart J. 2009;50:353–363. doi: 10.1536/ihj.50.353.
    1. Azenabor AA, Chaudhry AU. Effective macrophage redox defense against Chlamydia pneumoniae depends on L-type Ca2+ channel activation. Med Microbiol Immunol. 2003;192:99–106.
    1. Wieland H, Hechtel N, Faigle M, Neumeister B. Efficient intracellular multiplication of Legionella pneumophila in human monocytes requires functional host cell L-type calcium channels. FEMS Immunol Med Microbiol. 2006;47:296–301. doi: 10.1111/j.1574-695X.2006.00092.x.
    1. Reichenberg A, Yirmiya R, Schuld A, Kraus T, Haack M, Morag A. Cytokine-associated emotional and cognitive disturbances in humans. Arch Gen Psychiatry. 2001;58:445–452.
    1. Dantzer R, O'Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:46–56. doi: 10.1038/nrn2297.
    1. Musselman DL, Lawson DH, Gumnick JF, Manatunga AK, Penna S, Goodkin RS. Paroxetine for the prevention of depression induced by high-dose interferon alfa. New Engl J Med. 2001;344:961–966. doi: 10.1056/NEJM200103293441303.
    1. Raison CL, Borisov AS, Broadwell SD, Capuron L, Woolwine BJ, Jacobson IM. Depression during pegylated interferon-alpha plus ribavirin therapy: prevalence and prediction. J Clin Psychiatry. 2005;66:41–48. doi: 10.4088/JCP.v66n0106.
    1. Kraus MR, Schafer A, Csef H, Scheurlen M. Psychiatric side effects of pegylated interferon alfa-2b as compared to conventional interferon alfa-2b in patients with chronic hepatitis C. World J Gastroenterol. 2005;11:1769–1774. doi: 10.3748/wjg.v11.i12.1769.
    1. Reichenberg A, Gorman JM, Dieterich DT. Interferon-induced depression and cognitive impairment in hepatitis C virus patients: a 72 week prospective study. AIDS. 2005;19(Suppl 3):S174–S178. doi: 10.1097/.
    1. Andreasson A, Arborelius L, Erlanson-Albertsson C, Lekander M. A putative role for cytokines in the impaired appetite in depression. Brain Behav Immun. 2007;21:147–152. doi: 10.1016/j.bbi.2006.08.002.
    1. Maier SF, Watkins LR. Cytokines for psychologists: implications of bidirectional immune-to-brain communication for understanding behavior, mood, and cognition. Psychol Rev. 1998;105:83–107. doi: 10.1037/0033-295X.105.1.83.
    1. Yirmiya R, Weidenfeld J, Pollak Y, Morag M, Morag A, Avitsur R. Cytokines, “depression due to a general medical condition,” and antidepressant drugs. Adv Exper Med Biol. 1999;461:283–316. doi: 10.1007/978-0-585-37970-8_16.
    1. Hayley S, Merali Z, Anisman H. Stress and cytokine-elicited neuroendocrine and neurotransmitter sensitization: implications for depressive illness. Stress. 2003;6:19–32. doi: 10.1080/1025389031000091167.
    1. Gibb J, Audet MC, Hayley S, Anisman H. Neurochemical and behavioral responses to inflammatory immune stressors. Front Biosci (Schol Ed) 2009;1:275–295. doi: 10.2741/s26.
    1. Maes M, Yirmyia R, Noraberg J, Brene S, Hibbeln J, Perini G. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression. Metab Brain Dis. 2009;24:27–53. doi: 10.1007/s11011-008-9118-1.
    1. Simon GE, Ludman EJ, Linde JA, Operskalski BH, Ichikawa L, Rohde P. Association between obesity and depression in middle-aged women. Gen Hosp Psychiatry. 2008;30:32–39. doi: 10.1016/j.genhosppsych.2007.09.001.
    1. Simon GE, Von Korff M, Saunders K, Miglioretti DL, Crane PK, van Belle G. Association between obesity and psychiatric disorders in the US adult population. Arch Gen Psychiatry. 2006;63:824–830. doi: 10.1001/archpsyc.63.7.824.
    1. Miller GE, Freedland KE, Carney RM, Stetler CA, Banks WA. Pathways linking depression, adiposity, and inflammatory markers in healthy young adults. Brain Behav Immun. 2003;17:276–285. doi: 10.1016/S0889-1591(03)00057-6.
    1. Lampert R, Bremner JD, Su S, Miller A, Lee F, Cheema F. Decreased heart rate variability is associated with higher levels of inflammation in middle-aged men. Am Heart J. 2008;156:759. doi: 10.1016/j.ahj.2008.07.009.
    1. Ranjit N, Diez-Roux AV, Shea S, Cushman M, Seeman T, Jackson SA. Psychosocial factors and inflammation in the multi-ethnic study of atherosclerosis. Arch Int Med. 2007;167:174–181. doi: 10.1001/archinte.167.2.174.
    1. Kloiber S, Ising M, Reppermund S, Horstmann S, Dose T, Majer M. Overweight and obesity affect treatment response in major depression. Biol Psychiatry. 2007;62:321–326. doi: 10.1016/j.biopsych.2006.10.001.
    1. Himmerich H, Fulda S, Linseisen J, Seiler H, Wolfram G, Himmerich S. TNF-alpha, soluble TNF receptor and interleukin-6 plasma levels in the general population. Eur Cytokine Network. 2006;17:196–201.
    1. Suarez EC. C-reactive protein is associated with psychological risk factors of cardiovascular disease in apparently healthy adults. Psychosom Med. 2004;66:684–691. doi: 10.1097/01.psy.0000138281.73634.67.
    1. Douglas KM, Taylor AJ, O'Malley PG. Relationship between depression and C-reactive protein in a screening population. Psychosom Med. 2004;66:679–683. doi: 10.1097/01.psy.0000138132.66332.85.
    1. Roberts RE, Deleger S, Strawbridge WJ, Kaplan GA. Prospective association between obesity and depression: evidence from the Alameda County Study. Int J Obes Relat Metab Disord. 2003;27:514–521. doi: 10.1038/sj.ijo.0802204.
    1. Pine DS, Goldstein RB, Wolk S, Weissman MM. The association between childhood depression and adulthood body mass index. Pediatrics. 2001;107:1049–1056. doi: 10.1542/peds.107.5.1049.
    1. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab. 2001;280:E745–E751. doi: 10.1152/ajpendo.2001.280.5.E745.
    1. Katz JR, Taylor NF, Goodrick S, Perry L, Yudkin JS, Coppack SW. Central obesity, depression and the hypothalamo-pituitary-adrenal axis in men and postmenopausal women. Int J Obesity Related Metab Disord. 2000;24:246–251. doi: 10.1038/sj.ijo.0801122.
    1. Haack M, Hinze-Selch D, Fenzel T, Kraus T, Kuhn M, Schuld A. Plasma levels of cytokines and soluble cytokine receptors in psychiatric patients upon hospital admission: effects of confounding factors and diagnosis. J Psychiatr Res. 1999;33:407–418. doi: 10.1016/S0022-3956(99)00021-7.
    1. Vieira VJ, Valentine RJ, McAuley E, Evans E, Woods JA. Independent relationship between heart rate recovery and C-reactive protein in older adults. J Am Geriat Soc. 2007;55:747–751. doi: 10.1111/j.1532-5415.2007.01160.x.
    1. Moyna NM, Bodnar JD, Goldberg HR, Shurin MS, Robertson RJ, Rabin BS. Relation between aerobic fitness level and stress induced alterations in neuroendocrine and immune function. Int J Sports Med. 1999;20:136–141. doi: 10.1055/s-2007-971107.
    1. Kohut ML, McCann DA, Russell DW, Konopka DN, Cunnick JE, Franke WD. Aerobic exercise, but not flexibility/resistance exercise, reduces serum IL-18, CRP, and IL-6 independent of beta-blockers, BMI, and psychosocial factors in older adults. Brain Behav Immun. 2006;20:201–209. doi: 10.1016/j.bbi.2005.12.002.
    1. Balducci S, Zanuso S, Nicolucci A, Fernando F, Cavallo S, Cardelli P. Anti-inflammatory effect of exercise training in subjects with type 2 diabetes and the metabolic syndrome is dependent on exercise modalities and independent of weight loss. Nutr Metab Cardiovasc Dis. 2010;20:608–617. doi: 10.1016/j.numecd.2009.04.015.
    1. Ghosh S, Khazaei M, Moien-Afshari F, Ang LS, Granville DJ, Verchere CB. Moderate exercise attenuates caspase-3 activity, oxidative stress, and inhibits progression of diabetic renal disease in db/db mice. Am J Physiol Renal Physiol. 2009;296:F700–F708. doi: 10.1152/ajprenal.90548.2008.
    1. Anton SD, Newton RL, Jr, Sothern M, Martin CK, Stewart TM, Williamson DA. Association of depression with body mass index, sedentary behavior, and maladaptive eating attitudes and behaviors in 11 to 13-year old children. Eat Weight Disord. 2006;11:e102–e108. doi: 10.1007/BF03327566.
    1. Colditz GA. Economic costs of obesity and inactivity. Med Sci Sports Exerc. 1999;31:S663–S667. doi: 10.1097/00005768-199911001-00026.
    1. Rajala U, Uusimaki A, Keinanen-Kiukaanniemi S, Kivela SL. Prevalence of depression in a 55-year-old Finnish population. Soc Psychiatry Psychiatr Epidemiol. 1994;29:126–130.
    1. Tanskanen A, Hibbeln JR, Tuomilehto J, Uutela A, Haukkala A, Viinamaki H. Fish consumption and depressive symptoms in the general population in Finland. Psychiatr Serv. 2001;52:529–531. doi: 10.1176/appi.ps.52.4.529.
    1. Maes M, Christophe A, Delanghe J, Altamura C, Neels H, Meltzer HY. Lowered omega3 polyunsaturated fatty acids in serum phospholipids and cholesteryl esters of depressed patients. Psychiatry Res. 1999;85:275–291. doi: 10.1016/S0165-1781(99)00014-1.
    1. Maes M, Smith R, Christophe A, Cosyns P, Desnyder R, Meltzer H. Fatty acid composition in major depression: decreased omega 3 fractions in cholesteryl esters and increased C20: 4 omega 6/C20:5 omega 3 ratio in cholesteryl esters and phospholipids. J Affect Disord. 1996;38:35–46. doi: 10.1016/0165-0327(95)00092-5.
    1. Williams LL, Kiecolt-Glaser JK, Horrocks LA, Hillhouse JT, Glaser R. Quantitative association between altered plasma esterified omega-6 fatty acid proportions and psychological stress. Prostaglandins Leukot Essent Fatty Acids. 1992;47:165–170. doi: 10.1016/0952-3278(92)90155-C.
    1. Dai J, Miller AH, Bremner JD, Goldberg J, Jones L, Shallenberger L. Adherence to the mediterranean diet is inversely associated with circulating interleukin-6 among middle-aged men: a twin study. Circulation. 2008;117:169–175. doi: 10.1161/CIRCULATIONAHA.107.710699.
    1. Zampelas A, Panagiotakos DB, Pitsavos C, Das UN, Chrysohoou C, Skoumas Y. Fish consumption among healthy adults is associated with decreased levels of inflammatory markers related to cardiovascular disease: the ATTICA study. J Am Coll Cardiol. 2005;46:120–124. doi: 10.1016/j.jacc.2005.03.048.
    1. Chrysohoou C, Panagiotakos DB, Pitsavos C, Das UN, Stefanadis C. Adherence to the Mediterranean diet attenuates inflammation and coagulation process in healthy adults: The ATTICA Study. J Am Coll Cardiol. 2004;44:152–158. doi: 10.1016/j.jacc.2004.03.039.
    1. Westover AN, Marangell LB. A cross-national relationship between sugar consumption and major depression? Depress Anxiety. 2002;16:118–120. doi: 10.1002/da.10054.
    1. O'Keefe JH, Gheewala NM, O'Keefe JO. Dietary strategies for improving post-prandial glucose, lipids, inflammation, and cardiovascular health. J Am Coll Cardiol. 2008;51:249–255. doi: 10.1016/j.jacc.2007.10.016.
    1. Schulze MB, Hoffmann K, Manson JE, Willett WC, Meigs JB, Weikert C. Dietary pattern, inflammation, and incidence of type 2 diabetes in women. Am J Clin Nutr. 2005;82:675–684. doi: 10.1093/ajcn/82.3.675.
    1. Lee O, Bruce WR, Dong Q, Bruce J, Mehta R, O'Brien PJ. Fructose and carbonyl metabolites as endogenous toxins. Chem Biol Interact. 2009;178:332–339. doi: 10.1016/j.cbi.2008.10.011.
    1. O'Connor MF, Bower JE, Cho HJ, Creswell JD, Dimitrov S, Hamby ME. To assess, to control, to exclude: effects of biobehavioral factors on circulating inflammatory markers. Brain Behav Immun. 2009;23:887–897. doi: 10.1016/j.bbi.2009.04.005.
    1. Ford DE, Kamerow DB. Epidemiologic study of sleep disturbances and psychiatric disorders. An opportunity for prevention? JAMA. 1989;262:1479–1484. doi: 10.1001/jama.1989.03430110069030.
    1. Livingston G, Blizard B, Mann A. Does sleep disturbance predict depression in elderly people? A study in inner London. Brit J Gen Pract. 1993;43:445–448.
    1. Breslau N, Roth T, Rosenthal L, Andreski P. Sleep disturbance and psychiatric disorders: a longitudinal epidemiological study of young adults. Biol Psychiatry. 1996;39:411–418. doi: 10.1016/0006-3223(95)00188-3.
    1. Gillin JC. Are sleep disturbances risk factors for anxiety, depressive and addictive disorders? Acta Psychiatr Scand Suppl. 1998;393:39–43. doi: 10.1111/j.1600-0447.1998.tb05965.x.
    1. Irwin MR, Wang M, Ribeiro D, Cho HJ, Olmstead R, Breen EC. Sleep loss activates cellular inflammatory signaling. Biol Psychiatry. 2008;64:538–540. doi: 10.1016/j.biopsych.2008.05.004.
    1. McDade TW, Hawkley LC, Cacioppo JT. Psychosocial and behavioral predictors of inflammation in middle-aged and older adults: the Chicago health, aging, and social relations study. Psychosom Med. 2006;68:376–381. doi: 10.1097/01.psy.0000221371.43607.64.
    1. Irwin MR, Wang M, Campomayor CO, Collado-Hidalgo A, Cole S. Sleep deprivation and activation of morning levels of cellular and genomic markers of inflammation. Arch Int Med. 2006;166:1756–1762. doi: 10.1001/archinte.166.16.1756.
    1. Meier-Ewert HK, Ridker PM, Rifai N, Regan MM, Price NJ, Dinges DF. Effect of sleep loss on C-reactive protein, an inflammatory marker of cardiovascular risk. J Am Coll Cardiol. 2004;43:678–683. doi: 10.1016/j.jacc.2003.07.050.
    1. Friedman EM, Hayney MS, Love GD, Urry HL, Rosenkranz MA, Davidson RJ. Social relationships, sleep quality, and interleukin-6 in aging women. Proc Natl Acad Sci USA. 2005;102:18757–18762. doi: 10.1073/pnas.0509281102.
    1. Irwin M, Rinetti G, Redwine L, Motivala S, Dang J, Ehlers C. Nocturnal proinflammatory cytokine-associated sleep disturbances in abstinent African American alcoholics. Brain Behav Immun. 2004;18:349–360. doi: 10.1016/j.bbi.2004.02.001.
    1. Vgontzas AN, Zoumakis M, Papanicolaou DA, Bixler EO, Prolo P, Lin HM. Chronic insomnia is associated with a shift of interleukin-6 and tumor necrosis factor secretion from nighttime to daytime. Metab Clin Exper. 2002;51:887–892. doi: 10.1053/meta.2002.33357.
    1. Redwine L, Hauger RL, Gillin JC, Irwin M. Effects of sleep and sleep deprivation on interleukin-6, growth hormone, cortisol, and melatonin levels in humans. J Clin Endocrinol Metab. 2000;85:3597–3603.
    1. Saules KK, Pomerleau CS, Snedecor SM, Mehringer AM, Shadle MB, Kurth C. Relationship of onset of cigarette smoking during college to alcohol use, dieting concerns, and depressed mood: results from the Young Women's Health Survey. Addict Behav. 2004;29:893–899. doi: 10.1016/j.addbeh.2004.02.015.
    1. Lenz BK. Tobacco, depression, and lifestyle choices in the pivotal early college years. J Am Coll Health. 2004;52:213–219. doi: 10.3200/JACH.52.5.213-220.
    1. Handwerker WP. Cultural diversity, stress, and depression: working women in the Americas. J Women Health Gend Based Med. 1999;8:1303–1311. doi: 10.1089/jwh.1.1999.8.1303.
    1. Moussavi S, Chatterji S, Verdes E, Tandon A, Patel V, Ustun B. Depression, chronic diseases, and decrements in health: results from the World Health Surveys. Lancet. 2007;370:851–858. doi: 10.1016/S0140-6736(07)61415-9.
    1. Brown GW, Harris TO, Hepworth C. Life events and endogenous depression. A puzzle reexamined. Arch Gen Psychiat. 1994;51:525–534. doi: 10.1001/archpsyc.1994.03950070017006.
    1. Kendler KS, Thornton LM, Gardner CO. Stressful life events and previous episodes in the etiology of major depression in women: an evaluation of the “kindling” hypothesis. Am J Psychiatry. 2000;157:1243–1251. doi: 10.1176/appi.ajp.157.8.1243.
    1. DiPietro LA. Wound healing: the role of the macrophage and other immune cells. Shock. 1995;4:233–240. doi: 10.1097/00024382-199510000-00001.
    1. Ross SR, Bloomsmith MA, Bettinger TL, Wagner KE. The influence of captive adolescent male chimpanzees on wounding: management and welfare implications. Zoo Biol. 2009;28:623–634. doi: 10.1002/zoo.20220.
    1. Eshed V, Gopher A, Pinhasi R, Hershkovitz I. Paleopathology and the origin of agriculture in the Levant. Am J Phys Anthropol. 2010;143:121–133. doi: 10.1002/ajpa.21301.
    1. Pinker S. The Better Angels of Our Nature: Why Violence Has Declined. Viking Adult: New York, NY; 2011.
    1. Dhabhar FS. Enhancing versus suppressive effects of stress on immune function: implications for immunoprotection and immunopathology. Neuroimmunomodulation. 2009;16:300–317. doi: 10.1159/000216188.
    1. Herbert TB, Cohen S. Stress and immunity in humans: a meta-analytic review. Psychosom Med. 1993;55:364–379. doi: 10.1097/00006842-199307000-00004.
    1. Bailey MT, Kinsey SG, Padgett DA, Sheridan JF, Leblebicioglu B. Social stress enhances IL-1beta and TNF-alpha production by Porphyromonas gingivalis lipopolysaccharide-stimulated CD11b+ cells. Physiol Behav. 2009;98:351–358. doi: 10.1016/j.physbeh.2009.06.013.
    1. Pace TW, Mletzko TC, Alagbe O, Musselman DL, Nemeroff CB, Miller AH. Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. Am J Psychiatry. 2006;163:1630–1633. doi: 10.1176/ajp.2006.163.9.1630.
    1. Powell ND, Mays JW, Bailey MT, Hanke ML, Sheridan JF. Immunogenic dendritic cells primed by social defeat enhance adaptive immunity to influenza A virus. Brain Behav Immun. 2011;25:46–52. doi: 10.1016/j.bbi.2010.07.243.
    1. Mays JW, Bailey MT, Hunzeker JT, Powell ND, Papenfuss T, Karlsson EA. Influenza virus-specific immunological memory is enhanced by repeated social defeat. J Immunol. 2010;184:2014–2025. doi: 10.4049/jimmunol.0900183.
    1. Steptoe A, Hamer M, Chida Y. The effect of acute psychological stress on circulating inflammatory factors in humans: a review and meta-analysis. Brain Behav Immun. 2007;7:901–912. doi: 10.1016/j.bbi.2007.03.011.
    1. Bierhaus A, Wolf J, Andrassy M, Rohleder N, Humpert PM, Petrov D. A mechanism converting psychosocial stress into mononuclear cell activation. Proc Natl Acad Sci USA. 2003;100:1920–1925. doi: 10.1073/pnas.0438019100.
    1. Avitsur R, Kavelaars A, Heijnen C, Sheridan JF. Social stress and the regulation of tumor necrosis factor-alpha secretion. Brain Behav Immun. 2005;19:311–317. doi: 10.1016/j.bbi.2004.09.005.
    1. Quan N, Avitsur R, Stark JL, He L, Shah M, Caligiuri M. Social stress increases the susceptibility to endotoxic shock. J Neuroimmunol. 2001;115:36–45. doi: 10.1016/S0165-5728(01)00273-9.
    1. Rosenberger PH, Ickovics JR, Epel E, Nadler E, Jokl P, Fulkerson JP. Surgical stress-induced immune cell redistribution profiles predict short-term and long-term postsurgical recovery. A prospective study. J Bone Joint Surg Am. 2009;91:2783–2794. doi: 10.2106/JBJS.H.00989.
    1. Joachim RA, Handjiski B, Blois SM, Hagen E, Paus R, Arck PC. Stress-induced neurogenic inflammation in murine skin skews dendritic cells towards maturation and migration: key role of intercellular adhesion molecule-1/leukocyte function-associated antigen interactions. Am J Pathol. 2008;173:1379–1388. doi: 10.2353/ajpath.2008.080105.
    1. Viswanathan K, Daugherty C, Dhabhar FS. Stress as an endogenous adjuvant: augmentation of the immunization phase of cell-mediated immunity. Int Immunol. 2005;17:1059–1069. doi: 10.1093/intimm/dxh286.
    1. Miller GE, Chen E. Harsh family climate in early life presages the emergence of a proinflammatory phenotype in adolescence. Psychol Sci. 2010;21:848–856. doi: 10.1177/0956797610370161.
    1. Danese A, Moffitt TE, Pariante CM, Ambler A, Poulton R, Caspi A. Elevated inflammation levels in depressed adults with a history of childhood maltreatment. Arch Gen Psychiatry. 2008;65:409–415. doi: 10.1001/archpsyc.65.4.409.
    1. Miller GE, Rohleder N, Cole SW. Chronic interpersonal stress predicts activation of pro- and anti-inflammatory signaling pathways 6 months later. Psychosom Med. 2009;71:57–62. doi: 10.1097/PSY.0b013e318190d7de.
    1. Miller GE, Chen E, Sze J, Marin T, Arevalo JM, Doll R. A functional genomic fingerprint of chronic stress in humans: blunted glucocorticoid and increased NF-kappaB signaling. Biol Psychiatry. 2008;64:266–272. doi: 10.1016/j.biopsych.2008.03.017.
    1. Miller GE, Chen E, Parker KJ. Psychological stress in childhood and susceptibility to the chronic diseases of aging: moving toward a model of behavioral and biological mechanisms. Psychol Bull. 2011;137:959–997. doi: 10.1037/a0024768.
    1. Kiecolt-Glaser JK, Preacher KJ, MacCallum RC, Atkinson C, Malarkey WB, Glaser R. Chronic stress and age-related increases in the proinflammatory cytokine IL-6. Proc Natl Acad Sci USA. 2003;100:9090–9095. doi: 10.1073/pnas.1531903100.
    1. Nesse RM. The smoke detector principle. Natural selection and the regulation of defensive responses. Ann NY Acad Sci. 2001;935:75–85. doi: 10.1111/j.1749-6632.2001.tb03472.x.
    1. Kiecolt-Glaser JK. Stress, food, and inflammation: psychoneuroimmunology and nutrition at the cutting edge. Psychosom Med. 2010;72:365–369. doi: 10.1097/PSY.0b013e3181dbf489.
    1. Haroon E, Raison CL, Miller AH. Psychoneuroimmunology meets neuropsychopharmacol: translational implications of the impact of inflammation on behavior. Neuropsychopharmacology. 2012;37:137–162. doi: 10.1038/npp.2011.205.
    1. Irwin MR, Cole SW. Reciprocal regulation of the neural and innate immune systems. Nat Rev Immunol. 2011;11:625–632. doi: 10.1038/nri3042.
    1. Leutscher PD, Lagging M, Buhl MR, Pedersen C, Norkrans G, Langeland N. Evaluation of depression as a risk factor for treatment failure in chronic hepatitis C. Hepatology. 2010;52:430–435. doi: 10.1002/hep.23699.
    1. Raison CL, Broadwell SD, Borisov AS, Manatunga AK, Woolwine BJ, Jacobson IM. Depressive symptoms and viral clearance in patients receiving interferon-alpha and ribavirin for hepatitis C. Brain Behav Immun. 2005;19:23–27. doi: 10.1016/j.bbi.2004.05.001.
    1. Doering LV, Martinez-Maza O, Vredevoe DL, Cowan MJ. Relation of depression, natural killer cell function, and infections after coronary artery bypass in women. Eur J Cardiovasc Nurs. 2008;7:52–58. doi: 10.1016/j.ejcnurse.2007.07.004.
    1. Faulkner S, Smith A. A longitudinal study of the relationship between psychological distress and recurrence of upper respiratory tract infections in chronic fatigue syndrome. Br J Health Psychol. 2008;13:177–186. doi: 10.1348/135910706X171469.
    1. Cruess DG, Petitto JM, Leserman J, Douglas SD, Gettes DR, Ten Have TR. Depression and HIV infection: impact on immune function and disease progression. CNS Spectr. 2003;8:52–58. doi: 10.1017/S1092852900023452.
    1. Leserman J. Role of depression, stress, and trauma in HIV disease progression. Psychosom Med. 2008;70:539–545. doi: 10.1097/PSY.0b013e3181777a5f.
    1. Evans DL, Ten Have TR, Douglas SD, Gettes DR, Morrison M, Chiappini MS. Association of depression with viral load, CD8T lymphocytes, and natural killer cells in women with HIV infection. Am J Psychiatry. 2002;159:1752–1759. doi: 10.1176/appi.ajp.159.10.1752.
    1. Zorrilla EP, McKay JR, Luborsky L, Schmidt K. Relation of stressors and depressive symptoms to clinical progression of viral illness. Am J Psychiatry. 1996;153:626–635. doi: 10.1176/ajp.153.5.626.
    1. Herbert TB, Cohen S. Depression and immunity: a meta-analytic review. Psychol Bull. 1993;113:472–486. doi: 10.1037/0033-2909.113.3.472.
    1. Castilla-Cortazar I, Castilla A, Gurpegui M. Opioid peptides and immunodysfunction in patients with major depression and anxiety disorders. J Physiol Biochem. 1998;54:203–215.
    1. Blume J, Douglas SD, Evans DL. Immune suppression and immune activation in depression. Brain Behav Immun. 2011;25:221–229. doi: 10.1016/j.bbi.2010.10.008.
    1. Vaknin I, Blinder L, Wang L, Gazit R, Shapira E, Genina O. A common pathway mediated through Toll-like receptors leads to T- and natural killer-cell immunosuppression. Blood. 2008;111:1437–1447. doi: 10.1182/blood-2007-07-100404.
    1. Moraska A, Campisi J, Nguyen KT, Maier SF, Watkins LR, Fleshner M. Elevated IL-1beta contributes to antibody suppression produced by stress. J Appl Physiol. 2002;93:207–215. doi: 10.1152/japplphysiol.01151.2001.
    1. Cope AP, Liblau RS, Yang XD, Congia M, Laudanna C, Schreiber RD. Chronic tumor necrosis factor alters T cell responses by attenuating T cell receptor signaling. J Exp Med. 1997;185:1573–1584. doi: 10.1084/jem.185.9.1573.
    1. Cope AP. Exploring the reciprocal relationship between immunity and inflammation in chronic inflammatory arthritis. Rheumatology (Oxford) 2003;42:716–731. doi: 10.1093/rheumatology/keg262.
    1. Eleftheriadis T, Kartsios C, Yiannaki E, Antoniadi G, Kazila P, Pliakos K. Decreased CD3+CD16+ natural killer-like T-cell percentage and zeta-chain expression accompany chronic inflammation in haemodialysis patients. Nephrology (Carlton) 2009;14:471–475. doi: 10.1111/j.1440-1797.2008.01041.x.
    1. Muller AJ, Sharma MD, Chandler PR, Duhadaway JB, Everhart ME, Johnson BA., III Chronic inflammation that facilitates tumor progression creates local immune suppression by inducing indoleamine 2,3 dioxygenase. Proc Natl Acad Sci USA. 2008;105:17073–17078. doi: 10.1073/pnas.0806173105.
    1. Clark J, Vagenas P, Panesar M, Cope AP. What does tumour necrosis factor excess do to the immune system long term? Ann Rheum Dis. 2005;64(Suppl 4):iv70–iv76.
    1. Doran MF, Crowson CS, Pond GR, O'Fallon WM, Gabriel SE. Frequency of infection in patients with rheumatoid arthritis compared with controls: a population-based study. Arthritis Rheum. 2002;46:2287–2293. doi: 10.1002/art.10524.
    1. Armelagos GJ, Brown PJ, Turner B. Evolutionary, historical and political economic perspectives on health and disease. Soc Sci Med. 2005;61:755–765. doi: 10.1016/j.socscimed.2004.08.066.
    1. May L, van den Biggelaar AH, van Bodegom D, Meij HJ, de Craen AJ, Amankwa J. Adverse environmental conditions influence age-related innate immune responsiveness. Immun Ageing. 2009;6:7. doi: 10.1186/1742-4933-6-7.
    1. Drenos F, Westendorp RG, Kirkwood TB. Trade-off mediated effects on the genetics of human survival caused by increasingly benign living conditions. Biogerontology. 2006;7:287–295. doi: 10.1007/s10522-006-9027-9.
    1. Raison CL, Lowry CA, Rook GA. Inflammation, sanitation, and consternation: loss of contact with coevolved, tolerogenic microorganisms and the pathophysiology and treatment of major depression. Arch Gen Psychiatry. 2010;67:1211–1224. doi: 10.1001/archgenpsychiatry.2010.161.
    1. Joosten KF, de Kleijn ED, Westerterp M, de Hoog M, Eijck FC, Hop WCJ. Endocrine and metabolic responses in children with meningoccocal sepsis: striking differences between survivors and nonsurvivors. J Clin Endocrinol Metab. 2000;85:3746–3753. doi: 10.1210/jcem.85.10.6901.
    1. Pinto RA, Arredondo SM, Bono MR, Gaggero AA, Diaz PV. T helper 1/T helper 2 cytokine imbalance in respiratory syncytial virus infection is associated with increased endogenous plasma cortisol. Pediatrics. 2006;117:e878–e886. doi: 10.1542/peds.2005-2119.
    1. Gallagher PM, Lowe G, Fitzgerald T, Bella A, Greene CM, McElvaney NG. Association of IL-10 polymorphism with severity of illness in community acquired pneumonia. Thorax. 2003;58:154–156. doi: 10.1136/thorax.58.2.154.
    1. Li J, Cowden LG, King JD, Briles DA, Schroeder HW, Jr, Stevens AB. Effects of chronic stress and interleukin-10 gene polymorphisms on antibody response to tetanus vaccine in family caregivers of patients with Alzheimer's disease. Psychosom Med. 2007;69:551–559. doi: 10.1097/PSY.0b013e3180cc2c61.
    1. Hohler T, Reuss E, Freitag CM, Schneider PM. A functional polymorphism in the IL-10 promoter influences the response after vaccination with HBsAg and hepatitis A. Hepatology. 2005;42:72–76. doi: 10.1002/hep.20740.
    1. Corsini E, Vismara L, Lucchi L, Viviani B, Govoni S, Galli CL. High interleukin-10 production is associated with low antibody response to influenza vaccination in the elderly. J Leukoc Biol. 2006;80:376–382. doi: 10.1189/jlb.0306190.
    1. Forte GI, Scola L, Misiano G, Milano S, Mansueto P, Vitale G. Relevance of gamma interferon, tumor necrosis factor alpha, and interleukin-10 gene polymorphisms to susceptibility to Mediterranean spotted fever. Clin Vaccine Immunol. 2009;16:811–815. doi: 10.1128/CVI.00121-09.
    1. Pravica V, Asderakis A, Perrey C, Hajeer A, Sinnott PJ, Hutchinson IV. In vitro production of IFN-gamma correlates with CA repeat polymorphism in the human IFN-gamma gene. Eur J Immunogenet. 1999;26:1–3. doi: 10.1046/j.1365-2370.1999.00122.x.
    1. Pravica V, Perrey C, Stevens A, Lee JH, Hutchinson IV. A single nucleotide polymorphism in the first intron of the human IFN-gamma gene: absolute correlation with a polymorphic CA microsatellite marker of high IFN-gamma production. Hum Immunol. 2000;61:863–866. doi: 10.1016/S0198-8859(00)00167-1.
    1. Pacheco AG, Cardoso CC, Moraes MO. IFNG +874T/A, IL10 −1082G/A and TNF −308G/A polymorphisms in association with tuberculosis susceptibility: a meta-analysis study. Hum Genet. 2008;123:477–484. doi: 10.1007/s00439-008-0497-5.
    1. Ansari A, Talat N, Jamil B, Hasan Z, Razzaki T, Dawood G. Cytokine gene polymorphisms across tuberculosis clinical spectrum in Pakistani patients. PLoS One. 2009;4:e4778. doi: 10.1371/journal.pone.0004778.
    1. Cardoso CC, Pereira AC, Brito-de-Souza VN, Dias-Baptista IM, Maniero VC, Venturini J. IFNG +874 T>A single nucleotide polymorphism is associated with leprosy among Brazilians. Hum Genet. 2010;128:481–490. doi: 10.1007/s00439-010-0872-x.
    1. Chong WP, Ip WK, Tso GH, Ng MW, Wong WH, Law HK. The interferon gamma gene polymorphism +874 A/T is associated with severe acute respiratory syndrome. BMC Infect Dis. 2006;6:82. doi: 10.1186/1471-2334-6-82.
    1. Torres OA, Calzada JE, Beraun Y, Morillo CA, Gonzalez A, Gonzalez CI. Role of the IFNG +874T/A polymorphism in Chagas disease in a Colombian population. Infect Genet Evol. 2010;10:682–685. doi: 10.1016/j.meegid.2010.03.009.
    1. Gao QJ, Liu DW, Zhang SY, Jia M, Wang LM, Wu LH. Polymorphisms of some cytokines and chronic hepatitis B and C virus infection. World J Gastroenterol. 2009;15:5610–5619. doi: 10.3748/wjg.15.5610.
    1. Yirmiya R, Pollak Y, Barak O, Avitsur R, Ovadia H, Bette M. Effects of antidepressant drugs on the behavioral and physiological responses to lipopolysaccharide (LPS) in rodents. Neuropsychopharmacology. 2001;24:531–544. doi: 10.1016/S0893-133X(00)00226-8.
    1. Merali Z, Brennan K, Brau P, Anisman H. Dissociating anorexia and anhedonia elicited by interleukin-1beta: antidepressant and gender effects on responding for “free chow” and “earned” sucrose intake. Psychopharmacology (Berl) 2003;165:413–418. doi: 10.1007/s00213-002-1273-1.
    1. Dunn AJ, Swiergiel AH. The reductions in sweetened milk intake induced by interleukin-1 and endotoxin are not prevented by chronic antidepressant treatment. Neuroimmunomodulation. 2001;9:163–169. doi: 10.1159/000049021.
    1. Castanon N, Bluthe RM, Dantzer R. Chronic treatment with the atypical antidepressant tianeptine attenuates sickness behavior induced by peripheral but not central lipopolysaccharide and interleukin-1beta in the rat. Psychopharmacology. 2001;154:50–60. doi: 10.1007/s002130000595.
    1. Shen Y, Connor TJ, Nolan Y, Kelly JP, Leonard BE. Differential effect of chronic antidepressant treatments on lipopolysaccharide-induced depressive-like behavioural symptoms in the rat. Life Sci. 1999;65:1773–1786. doi: 10.1016/S0024-3205(99)00430-0.
    1. Raison CL, Demetrashvili M, Capuron L, Miller AH. Neuropsychiatric adverse effects of interferon-alpha: recognition and management. CNS Drugs. 2005;19:105–123. doi: 10.2165/00023210-200519020-00002.
    1. Capuron L, Fornwalt FB, Knight BT, Harvey PD, Ninan PT, Miller AH. Does cytokine-induced depression differ from idiopathic major depression in medically healthy individuals? J Affect Disord. 2009;119:181–185. doi: 10.1016/j.jad.2009.02.017.
    1. Raison CL, Woolwine BJ, Demetrashvili MF, Borisov AS, Weinreib R, Staab JP. Paroxetine for prevention of depressive symptoms induced by interferon-alpha and ribavirin for hepatitis C. Aliment Pharm Ther. 2007;25:1163–1174. doi: 10.1111/j.1365-2036.2007.03316.x.
    1. Kraus MR, Schafer A, Al-Taie O, Scheurlen M. Prophylactic SSRI during interferon alpha re-therapy in patients with chronic hepatitis C and a history of interferon-induced depression. J Viral Hepatitis. 2005;12:96–100. doi: 10.1111/j.1365-2893.2005.00554.x.
    1. Kraus MR, Schafer A, Faller H, Csef H, Scheurlen M. Paroxetine for the treatment of interferon-alpha-induced depression in chronic hepatitis C. Aliment Pharm Ther. 2002;16:1091–1099. doi: 10.1046/j.1365-2036.2002.01265.x.
    1. Hauser P, Khosla J, Aurora H, Laurin J, Kling MA, Hill J. A prospective study of the incidence and open-label treatment of interferon-induced major depressive disorder in patients with hepatitis C. Mol Psychiatry. 2002;7:942–947. doi: 10.1038/sj.mp.4001119.
    1. Wichers MC, Koek GH, Robaeys G, Praamstra AJ, Maes M. Early increase in vegetative symptoms predicts IFN-alpha-induced cognitive-depressive changes. Psychol Med. 2005;35:433–441. doi: 10.1017/S0033291704003526.
    1. Eisenberger NI, Berkman ET, Inagaki TK, Rameson LT, Mashal NM, Irwin MR. Inflammation-induced anhedonia: endotoxin reduces ventral striatum responses to reward. Biol Psychiatry. 2010;68:748–754. doi: 10.1016/j.biopsych.2010.06.010.
    1. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol Psychiatry. 2009;66:407–414. doi: 10.1016/j.biopsych.2009.03.015.
    1. Raison CL, Rye DB, Woolwine BJ, Vogt GJ, Bautista BM, Spivey JR. Chronic interferon-alpha administration disrupts sleep continuity and depth in patients with Hepatitis C: association with fatigue, motor slowing, and increased evening cortisol. Biol Psychiatry. 2010;68:942–949. doi: 10.1016/j.biopsych.2010.04.019.
    1. Raison CL, Dantzer R, Kelley KW, Lawson MA, Woolwine BJ, Vogt G. CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFN-alpha: relationship to CNS immune responses and depression. Mol Psychiatry. 2010;15:393–403. doi: 10.1038/mp.2009.116.
    1. Raison CL, Borisov AS, Woolwine BJ, Massung B, Vogt G, Miller AH. Interferon-alpha effects on diurnal hypothalamic-pituitary-adrenal axis activity: relationship with proinflammatory cytokines and behavior. Mol Psychiatry. 2010;15:535–547. doi: 10.1038/mp.2008.58.
    1. Raison CL, Borisov AS, Majer M, Drake DF, Pagnoni G, Woolwine BJ. Activation of central nervous system inflammatory pathways by interferon-alpha: relationship to monoamines and depression. Biol Psychiatry. 2009;65:296–303. doi: 10.1016/j.biopsych.2008.08.010.
    1. Capuron L, Pagnoni G, Demetrashvili MF, Lawson DH, Fornwalt FB, Woolwine BJ. Basal ganglia hypermetabolism and symptoms of fatigue during interferon-alpha therapy. Neuropsychopharmacology. 2007;32:2384–2392. doi: 10.1038/sj.npp.1301362.
    1. Capuron L, Pagnoni G, Demetrashvili M, Woolwine BJ, Nemeroff CB, Berns GS. Anterior cingulate activation and error processing during interferon-alpha treatment. Biol Psychiatry. 2005;58:190–196. doi: 10.1016/j.biopsych.2005.03.033.
    1. Hanff TC, Furst SJ, Minor TR. Biochemical and anatomical substrates of depression and sickness behavior. Isr J Psychiatry Relat Sci. 2010;47:64–71.
    1. Hart BL. Biological basis of the behavior of sick animals. Neurosci Biobehav Rev. 1988;12:123–137. doi: 10.1016/S0149-7634(88)80004-6.
    1. Hart BL. Behavioral adaptations to pathogens and parasites: five strategies. Neurosci Biobehav Rev. 1990;14:273–294. doi: 10.1016/S0149-7634(05)80038-7.
    1. Kluger MJ. Phylogeny of fever. Fed Proc. 1979;38:30–34.
    1. Kluger MJ, Kozak W, Conn CA, Leon LR, Soszynski D. Role of fever in disease. Ann NY Acad Sci. 1998;856:224–233. doi: 10.1111/j.1749-6632.1998.tb08329.x.
    1. Sweet C, Cavanagh D, Collie MH, Smith H. Sensitivity to pyrexial temperatures: a factor contributing to virulence differences between two clones of influenza virus. Br J Exp Pathol. 1978;59:373–380.
    1. Dixon G, Booth C, Price E, Westran R, Turner M, Klein N. Fever as nature's engine. Part of beneficial host response? BMJ. 2010;340:c450. doi: 10.1136/bmj.c450.
    1. Tyrrell D, Barrow I, Arthur J. Local hyperthermia benefits natural and experimental common colds. BMJ. 1989;298:1280–1283. doi: 10.1136/bmj.298.6683.1280.
    1. Ostberg JR, Taylor SL, Baumann H, Repasky EA. Regulatory effects of fever-range whole-body hyperthermia on the LPS-induced acute inflammatory response. J Leukoc Biol. 2000;68:815–820.
    1. Jiang Q, Detolla L, Singh IS, Gatdula L, Fitzgerald B, van Rooijen N. Exposure to febrile temperature upregulates expression of pyrogenic cytokines in endotoxin-challenged mice. Am J Physiol. 1999;276:R1653–R1660. doi: 10.1152/ajpcell.1999.276.1.C152.
    1. Ostberg JR, Dayanc BE, Yuan M, Oflazoglu E, Repasky EA. Enhancement of natural killer (NK) cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells. J Leukoc Biol. 2007;82:1322–1331. doi: 10.1189/jlb.1106699.
    1. Ostberg JR, Repasky EA. Emerging evidence indicates that physiologically relevant thermal stress regulates dendritic cell function. Cancer Immunol Immunother. 2006;55:292–298. doi: 10.1007/s00262-005-0689-y.
    1. Evans SS, Wang WC, Bain MD, Burd R, Ostberg JR, Repasky EA. Fever-range hyperthermia dynamically regulates lymphocyte delivery to high endothelial venules. Blood. 2001;97:2727–2733. doi: 10.1182/blood.V97.9.2727.
    1. Swenson BR, Hedrick TL, Popovsky K, Pruett TL, Sawyer RG. Is fever protective in surgical patients with bloodstream infection? J Am Coll Surg. 2007;204:815–821. doi: 10.1016/j.jamcollsurg.2007.01.033.
    1. Mizushima Y, Ueno M, Idoguchi K, Ishikawa K, Matsuoka T. Fever in trauma patients: friend or foe? J Trauma. 2009;67:1062–1065. doi: 10.1097/TA.0b013e3181b848fc.
    1. Ohsugi Y. Recent advances in immunopathophysiology of interleukin-6: an innovative therapeutic drug, tocilizumab (recombinant humanized anti-human interleukin-6 receptor antibody), unveils the mysterious etiology of immune-mediated inflammatory diseases. Biol Pharm Bull. 2007;30:2001–2006. doi: 10.1248/bpb.30.2001.
    1. Kung'u JK, Wright VJ, Haji HJ, Ramsan M, Goodman D, Tielsch JM. Adjusting for the acute phase response is essential to interpret iron status indicators among young Zanzibari children prone to chronic malaria and helminth infections. J Nutr. 2009;139:2124–2131. doi: 10.3945/jn.108.104026.
    1. Cartwright GE, Lauritsen MA. The anemia of infection; hypoferremia, hypercupremia, and alterations in porphyrin metabolism in patients. J Clin Invest. 1946;25:65–80. doi: 10.1172/JCI101690.
    1. Kochan I, Wagner SK, Wasynczuk J. Effect of iron on antibacterial immunity in vaccinated mice. Infect Immun. 1984;43:543–548.
    1. Weinberg ED. Survival advantage of the hemochromatosis C282Y mutation. Perspect Biol Med. 2008;51:98–102. doi: 10.1353/pbm.2008.0001.
    1. Foster SL, Richardson SH, Failla ML. Elevated iron status increases bacterial invasion and survival and alters cytokine/chemokine mRNA expression in Caco-2 human intestinal cells. J Nutr. 2001;131:1452–1458. doi: 10.1093/jn/131.5.1452.
    1. Wander K, Shell-Duncan B, McDade TW. Evaluation of iron deficiency as a nutritional adaptation to infectious disease: an evolutionary medicine perspective. Am J Hum Biol. 2009;21:172–179. doi: 10.1002/ajhb.20839.
    1. Mitra AK, Akramuzzaman SM, Fuchs GJ, Rahman MM, Mahalanabis D. Long-term oral supplementation with iron is not harmful for young children in a poor community of Bangladesh. J Nutr. 1997;127:1451–1455. doi: 10.1093/jn/127.8.1451.
    1. Smith AW, Hendrickse RG, Harrison C, Hayes RJ, Greenwood BM. The effects on malaria of treatment of iron-deficiency anaemia with oral iron in Gambian children. Ann Trop Paediatr. 1989;9:17–23. doi: 10.1080/02724936.1989.11748589.
    1. van den Hombergh J, Dalderop E, Smit Y. Does iron therapy benefit children with severe malaria-associated anaemia? A clinical trial with 12 weeks supplementation of oral iron in young children from the Turiani Division, Tanzania. J Trop Pediatr. 1996;42:220–227. doi: 10.1093/tropej/42.4.220.
    1. Tielsch JM, Khatry SK, Stoltzfus RJ, Katz J, LeClerq SC, Adhikari R. Effect of routine prophylactic supplementation with iron and folic acid on preschool child mortality in southern Nepal: community-based, cluster-randomised, placebo-controlled trial. Lancet. 2006;367:144–152. doi: 10.1016/S0140-6736(06)67963-4.
    1. Sazawal S, Black RE, Ramsan M, Chwaya HM, Stoltzfus RJ, Dutta A. Effects of routine prophylactic supplementation with iron and folic acid on admission to hospital and mortality in preschool children in a high malaria transmission setting: community-based, randomised, placebo-controlled trial. Lancet. 2006;367:133–143. doi: 10.1016/S0140-6736(06)67962-2.
    1. McEnany GW, Lee KA. Effects of light therapy on sleep, mood, and temperature in women with nonseasonal major depression. Issues Ment Health Nurs. 2005;26:781–794. doi: 10.1080/01612840591008410.
    1. Rausch JL, Johnson ME, Corley KM, Hobby HM, Shendarkar N, Fei Y. Depressed patients have higher body temperature: 5-HT transporter long promoter region effects. Neuropsychobiology. 2003;47:120–127. doi: 10.1159/000070579.
    1. Szuba MP, Guze BH, Baxter LR., Jr Electroconvulsive therapy increases circadian amplitude and lowers core body temperature in depressed subjects. Biol Psychiatry. 1997;42:1130–1137. doi: 10.1016/S0006-3223(97)00046-2.
    1. Daimon K, Yamada N, Tsujimoto T, Takahashi S. Circadian rhythm abnormalities of deep body temperature in depressive disorders. J Affect Disord. 1992;26:191–198. doi: 10.1016/0165-0327(92)90015-X.
    1. Avery DH, Shah SH, Eder DN, Wildschiodtz G. Nocturnal sweating and temperature in depression. Acta Psychiatr Scand. 1999;100:295–301. doi: 10.1111/j.1600-0447.1999.tb10864.x.
    1. Avery DH, Wildschiodtz G, Smallwood RG, Martin D, Rafaelsen OJ. REM latency and core temperature relationships in primary depression. Acta Psychiatr Scand. 1986;74:269–280. doi: 10.1111/j.1600-0447.1986.tb06244.x.
    1. Avery DH, Wildschiodtz G, Rafaelsen OJ. Nocturnal temperature in affective disorder. J Affect Disord. 1982;4:61–71. doi: 10.1016/0165-0327(82)90020-9.
    1. Sugahara H, Akamine M, Kondo T, Fujisawa K, Yoshimasu K, Tokunaga S. Somatic symptoms most often associated with depression in an urban hospital medical setting in Japan. Psychiatry Res. 2004;128:305–311. doi: 10.1016/j.psychres.2004.06.015.
    1. Rangan AM, Blight GD, Binns CW. Iron status and non-specific symptoms of female students. J Am Coll Nutr. 1998;17:351–355. doi: 10.1080/07315724.1998.10718774.
    1. Maes M, Van de Vyvere J, Vandoolaeghe E, Bril T, Demedts P, Wauters A. Alterations in iron metabolism and the erythron in major depression: further evidence for a chronic inflammatory process. J Affect Disord. 1996;40:23–33. doi: 10.1016/0165-0327(96)00038-9.
    1. Maes M, Vandewoude M, Scharpe S, De Clercq L, Stevens W, Lepoutre L. Anthropometric and biochemical assessment of the nutritional state in depression: evidence for lower visceral protein plasma levels in depression. J Affect Disord. 1991;23:25–33. doi: 10.1016/0165-0327(91)90032-N.
    1. Maes M, Scharpe S, Bosmans E, Vandewoude M, Suy E, Uyttenbroeck W. Disturbances in acute phase plasma proteins during melancholia: additional evidence for the presence of an inflammatory process during that illness. Prog Neuropsychopharmacol Biol Psychiatry. 1992;16:501–515. doi: 10.1016/0278-5846(92)90056-K.
    1. Albacar G, Sans T, Martin-Santos R, Garcia-Esteve L, Guillamat R, Sanjuan J. An association between plasma ferritin concentrations measured 48 h after delivery and postpartum depression. J Affect Disord. 2011;131:136–142. doi: 10.1016/j.jad.2010.11.006.
    1. Engel GL, Schmale AH. Conservation-withdrawal: a primary regulatory process for organismic homeostasis. Ciba Found Symp. 1972;8:57–75.
    1. Majer M, Wellberg LAM, Capuron L, Pagnoni G, Raison CL, Miller AH. IFN-alpha-induced motor slowing is associated with increased depression and fatigue in patients with chronic hepatitis C. Brain Behav Immun. 2008;25:870–880. doi: 10.1016/j.bbi.2007.12.009.
    1. Capuron L, Gumnick JF, Musselman DL, Lawson DH, Reemsnyder A, Nemeroff CB. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology. 2002;26:643–652. doi: 10.1016/S0893-133X(01)00407-9.
    1. Schaller M, Murray DR. Pathogens, personality, and culture: disease prevalence predicts worldwide variability in sociosexuality, extraversion, and openness to experience. J Pers Soc Psychol. 2008;95:212–221. doi: 10.1037/0022-3514.95.1.212.
    1. Cole SW. Complex Systems Science in Biomedicine. Springer: New York, NY; 2006. The complexity of dynamic host networks; pp. 605–629.
    1. Eisenberger NI, Inagaki TK, Mashal NM, Irwin MR. Inflammation and social experience: an inflammatory challenge induces feelings of social disconnection in addition to depressed mood. Brain Behav Immun. 2010;24:558–563. doi: 10.1016/j.bbi.2009.12.009.
    1. Slavich GM, Way BM, Eisenberger NI, Taylor SE. Neural sensitivity to social rejection is associated with inflammatory responses to social stress. Proc Natl Acad Sci USA. 2010;107:14817–14822. doi: 10.1073/pnas.1009164107.
    1. Nguyen KB, Biron CA. Synergism for cytokine-mediated disease during concurrent endotoxin and viral challenges: roles for NK and T cell IFN-gamma production. J Immunol. 1999;162:5238–5246.
    1. Jakab GJ, Dick EC. Synergistic effect in viral-bacterial infection: combined infection of the murine respiratory tract with Sendai virus and Pasteurella pneumotropica. Infect Immun. 1973;8:762–768.
    1. Degre M, Glasgow LA. Synergistic effect in viral-bacterial infection. I. Combined infection of the respiratory tract in mice with parainfluenza virus and Hemophilus influenza. J Infect Dis. 1968;118:449–462. doi: 10.1093/infdis/118.5.449.
    1. Jones WT, Menna JH, Wennerstrom DE. Lethal synergism induced in mice by influenza type A virus and type Ia group B streptococci. Infect Immun. 1983;41:618–623.
    1. Beadling C, Slifka MK. How do viral infections predispose patients to bacterial infections? Curr Opin Infect Dis. 2004;17:185–191. doi: 10.1097/00001432-200406000-00003.
    1. Molyneux EM, Tembo M, Kayira K, Bwanaisa L, Mweneychanya J, Njobvu A. The effect of HIV infection on paediatric bacterial meningitis in Blantyre, Malawi. Arch Dis Child. 2003;88:1112–1118. doi: 10.1136/adc.88.12.1112.
    1. McCullers JA, McAuley JL, Browall S, Iverson AR, Boyd KL, Henriques Normark B. Influenza enhances susceptibility to natural acquisition of and disease due to Streptococcus pneumoniae in ferrets. J Infect Dis. 2010;202:1287–1295. doi: 10.1086/656333.
    1. Thornhill R, Fincher CL, Aran D. Parasites, democratization, and the liberalization of values across contemporary countries. Biol Rev Camb Philos Soc. 2009;84:113–131. doi: 10.1111/j.1469-185X.2008.00062.x.
    1. Diagnostic and Statistical Manual of Mental Disorders, 4th edn Text Revision: DSM-IV--4--TR. American Psychiatric Association: Washington, DC, 2000.
    1. Schmidt WD, O'Connor PJ, Cochrane JB, Cantwell M. Resting metabolic rate is influenced by anxiety in college men. J Appl Physiol. 1996;80:638–642. doi: 10.1152/jappl.1996.80.2.638.
    1. Blaza SE, Garrow JS. The effect of anxiety on metabolic rate. Proc Nutr Soc. 1980;39:13A. doi: 10.1079/PNS19800003.
    1. Bonnet MH, Arand DL. Insomnia, metabolic rate and sleep restoration. J Intern Med. 2003;254:23–31. doi: 10.1046/j.1365-2796.2003.01176.x.
    1. Bonnet MH, Arand DL. 24-h metabolic rate in insomniacs and matched normal sleepers. Sleep. 1995;18:581–588. doi: 10.1093/sleep/18.7.581.
    1. Miller GE, Cohen S. Human Psychoneuroimmunology. Oxford University Press: Oxford, UK; 2005. Infectious disease and psychoneuroimmunology; pp. 219–242.
    1. Constant A, Castera L, Dantzer R, Couzigou P, de Ledinghen V, Demotes-Mainard J. Mood alterations during interferon-alfa therapy in patients with chronic hepatitis C: evidence for an overlap between manic/hypomanic and depressive symptoms. J Clin Psychiatry. 2005;66:1050–1057. doi: 10.4088/JCP.v66n0814.
    1. Critchley HD, Mathias CJ, Josephs O, O'Doherty J, Zanini S, Dewar BK. Human cingulate cortex and autonomic control: converging neuroimaging and clinical evidence. Brain. 2003;126:2139–2152. doi: 10.1093/brain/awg216.
    1. Eisenberger NI, Lieberman MD, Williams KD. Does rejection hurt? An FMRI study of social exclusion. Science. 2003;302:290–292. doi: 10.1126/science.1089134.
    1. Brydon L, Harrison NA, Walker C, Steptoe A, Critchley HD. Peripheral inflammation is associated with altered substantia nigra activity and psychomotor slowing in humans. Biol Psychiatry. 2008;63:1022–1029. doi: 10.1016/j.biopsych.2007.12.007.
    1. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Dolan RJ. Neural origins of human sickness in interoceptive responses to inflammation. Biol Psychiatry. 2009;66:415–422. doi: 10.1016/j.biopsych.2009.03.007.
    1. Wing EJ, Barczynski LK, Boehmer SM. Effect of acute nutritional deprivation on immune function in mice. I. Macrophages. Immunol. 1983;48:543–550.
    1. Murray MJ, Murray AB. Anorexia of infection as a mechanism of host defense. Am J Clin Nutr. 1979;32:593–596. doi: 10.1093/ajcn/32.3.593.
    1. Heuer JG, Bailey DL, Sharma GR, Zhang T, Ding C, Ford A. Cecal ligation and puncture with total parenteral nutrition: a clinically relevant model of the metabolic, hormonal, and inflammatory dysfunction associated with critical illness. J Surg Res. 2004;121:178–186. doi: 10.1016/j.jss.2004.04.018.
    1. Sena MJ, Utter GH, Cuschieri J, Maier RV, Tompkins RG, Harbrecht BG. Early supplemental parenteral nutrition is associated with increased infectious complications in critically ill trauma patients. J Am Coll Surg. 2008;207:459–467. doi: 10.1016/j.jamcollsurg.2008.04.028.
    1. Heyland DK. Parenteral nutrition in the critically-ill patient: more harm than good? Proc Nutr Soc. 2000;59:457–466. doi: 10.1017/S002966510000063X.
    1. Casaer MP, Mesotten D, Hermans G, Wouters PJ, Schetz M, Meyfroidt G. Early versus late parenteral nutrition in critically ill adults. N Engl J Med. 2011;365:506–517. doi: 10.1056/NEJMoa1102662.
    1. Carter JD, Joyce PR, Mulder RT, Luty SE, McKenzie J. Gender differences in the presentation of depressed outpatients: a comparison of descriptive variables. J Affect Disord. 2000;61:59–67. doi: 10.1016/S0165-0327(00)00151-8.
    1. Adamo SA, Fidler TL, Forestell CA. Illness-induced anorexia and its possible function in the caterpillar, Manduca sexta. Brain Behav Immun. 2007;21:292–300. doi: 10.1016/j.bbi.2006.10.006.
    1. Heyland DK, MacDonald S, Keefe L, Drover JW. Total parenteral nutrition in the critically ill patient: a meta-analysis. JAMA. 1998;280:2013–2019. doi: 10.1001/jama.280.23.2013.
    1. Amprey JL, Spath GF, Porcelli SA. Inhibition of CD1 expression in human dendritic cells during intracellular infection with Leishmania donovani. Infect Immun. 2004;72:589–592. doi: 10.1128/IAI.72.1.589-592.2004.
    1. Roura-Mir C, Wang L, Cheng TY, Matsunaga I, Dascher CC, Peng SL. Mycobacterium tuberculosis regulates CD1 antigen presentation pathways through TLR-2. J Immunol. 2005;175:1758–1766. doi: 10.4049/jimmunol.175.3.1758.
    1. Raison CL, Miller AH. Is depression an inflammatory disorder? Curr Psychiatry Rep. 2011;13:467–475. doi: 10.1007/s11920-011-0232-0.
    1. Nesse RM. Is depression an adaptation? Arch Gen Psychiatry. 2000;57:14–20. doi: 10.1001/archpsyc.57.1.14.
    1. Sloman L, Gilbert P, Hasey G. Evolved mechanisms in depression: the role and interaction of attachment and social rank in depression. J Affect Disord. 2003;74:107–121. doi: 10.1016/S0165-0327(02)00116-7.
    1. Andrews PW, Thomson JA., Jr The bright side of being blue: depression as an adaptation for analyzing complex problems. Psychol Rev. 2009;116:620–654. doi: 10.1037/a0016242.
    1. Raison CL, Miller AH. When not enough is too much: the role of insufficient glucocorticoid signaling in the pathophysiology of stress-related disorders. Am J Psychiatry. 2003;160:1554–1565. doi: 10.1176/appi.ajp.160.9.1554.
    1. Norbiato G, Bevilacqua M, Vago T, Taddei A, Clerici M. Glucocorticoids and the immune function in the human immunodeficiency virus infection: a study in hypercortisolemic and cortisol-resistant patients. J Clin Endocrinol Metab. 1997;82:3260–3263.
    1. Huff GR, Huff WE, Balog JM, Rath NC. The effects of behavior and environmental enrichment on disease resistance of turkeys. Brain Behav Immun. 2003;17:339–349. doi: 10.1016/S0889-1591(03)00035-7.
    1. Schaller M. The behavioural immune system and the psychology of human sociality. Philos Trans R Soc London B Biol Sci. 2011;366:3418–3426. doi: 10.1098/rstb.2011.0029.
    1. Fincher CL, Thornhill R, Murray DR, Schaller M. Pathogen prevalence predicts human cross-cultural variability in individualism/collectivism. Proc Biol Sci. 2008;275:1279–1285. doi: 10.1098/rspb.2008.0094.
    1. Kim YK, Suh IB, Kim H, Han CS, Lim CS, Choi SH. The plasma levels of interleukin-12 in schizophrenia, major depression, and bipolar mania: effects of psychotropic drugs. Mol Psychiatry. 2002;7:1107–1114. doi: 10.1038/sj.mp.4001084.
    1. Foster R, Kandanearatchi A, Beasley C, Williams B, Khan N, Fagerhol MK. Calprotectin in microglia from frontal cortex is up-regulated in schizophrenia: evidence for an inflammatory process? Eur J Neurosci. 2006;24:3561–3566. doi: 10.1111/j.1460-9568.2006.05219.x.
    1. Weigelt K, Carvalho LA, Drexhage RC, Wijkhuijs A, de Wit H, van Beveren NJ. TREM-1 and DAP12 expression in monocytes of patients with severe psychiatric disorders. EGR3, ATF3 and PU.1 as important transcription factors. Brain Behav Immun. 2011;25:1162–1169. doi: 10.1016/j.bbi.2011.03.006.
    1. Maes M, Delange J, Ranjan R, Meltzer HY, Desnyder R, Cooremans W. Acute phase proteins in schizophrenia, mania and major depression: modulation by psychotropic drugs. Psychiatry Res. 1997;66:1–11. doi: 10.1016/S0165-1781(96)02915-0.

Source: PubMed

3
Předplatit