Epigenetic Pathways in Human Disease: The Impact of DNA Methylation on Stress-Related Pathogenesis and Current Challenges in Biomarker Development

M Austin Argentieri, Sairaman Nagarajan, Bobak Seddighzadeh, Andrea A Baccarelli, Alexandra E Shields, M Austin Argentieri, Sairaman Nagarajan, Bobak Seddighzadeh, Andrea A Baccarelli, Alexandra E Shields

Abstract

HPA axis genes implicated in glucocorticoid regulation play an important role in regulating the physiological impact of social and environmental stress, and have become a focal point for investigating the role of glucocorticoid regulation in the etiology of disease. We conducted a systematic review to critically assess the full range of clinical associations that have been reported in relation to DNA methylation of CRH, CRH-R1/2, CRH-BP, AVP, POMC, ACTH, ACTH-R, NR3C1, FKBP5, and HSD11β1/2 genes in adults. A total of 32 studies were identified. There is prospective evidence for an association between HSD11β2 methylation and hypertension, and functional evidence of an association between NR3C1 methylation and both small cell lung cancer (SCLC) and breast cancer. Strong associations have been reported between FKBP5 and NR3C1 methylation and PTSD, and biologically-plausible associations have been reported between FKBP5 methylation and Alzheimer's Disease. Mixed associations between NR3C1 methylation and mental health outcomes have been reported according to different social and environmental exposures, and according to varying gene regions investigated. We conclude by highlighting key challenges and future research directions that will need to be addressed in order to develop both clinically meaningful prognostic biomarkers and an evidence base that can inform public policy practice.

Keywords: Adverse childhood experiences (ACE); Alzheimer's; Cancer; Depression; FKBP5; Glucocorticoids; HPA axis; HSD11β2; Hypertension; Methylation; NR3C1; PTSD; Stress.

Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Overview of the hypothalamus-pituitary-adrenal (HPA) axis. Activation of the HPA axis leads to the production of corticotrophin-releasing hormone (CRH) and arginine vasopressin (AVP) in the hypothalamus. CRH is transported via the hypophyseal portal system of blood vessels to the anterior pituitary gland, which causes the pituitary gland to secrete adrenocorticotropic hormone (ACTH) into the bloodstream. ACTH then stimulates the production of glucocorticoids (e.g., cortisol) by the adrenal cortex. Glucocorticoids (GCs) produced by the adrenal cortex bind to glucocorticoid receptors in the anterior pituitary, hypothalamus, and the hippocampus to regulate production of CRH and ACTH, creating a negative feedback loop that stabilizes circulating levels of stress hormones within an appropriate physiological range.
Fig. 2
Fig. 2
Genomic actions of glucocorticoids (GCs). When bound to GCs, the glucocorticoid receptor (GR) complex translocates to the cell nucleus and modifies the synthesis of a number of immune, inflammatory, and metabolic proteins. This is done through directly binding to glucocorticoid response elements (GREs) in the DNA of genes that code for these proteins (transactivation), and through influencing the activity of transcription factors without contacting the DNA itself (transrepression). Transactivation leads to up-regulated synthesis of immune- and metabolic-related proteins, while transrepression leads to down-regulated synthesis of immunosuppressive and pro-inflammatory proteins.
Fig. 3
Fig. 3
Flow diagram of study selection.
Fig. 4
Fig. 4
Mean and CpG site-specific methylation results reported for exon 1F and its promoter in NR3C1. First exon variants in red represent the proximal promoter region. ▼ or ▲ denote hypomethylation or hypermethylation, respectively. ↓ or ↑ arrows denote a correlation between observed methylation at a specific CpG site and decreased or increased NR3C1 expression levels, respectively. “Null” denotes no methylation found, or no difference found in methylation between healthy controls and those with the disease under investigation. Boxes around CpG site numbers represent NGFI-A transcription factor binding sites according to McGowan et al. (2009). CpG numbering taken from Palma-Gudiel et al. (2015). Figure is not to scale.
Fig. 5
Fig. 5
Significant methylation results reported across the entire NR3C1 proximal promoter. Methylation results reported only for clinical outcomes that have been significantly associated with methylation status in at least one study. Diseases for which there have only been non-significant findings or for which the authors concluded that NR3C1 was not a likely biomarker are not included. First exon variants in red represent the proximal promoter region. ▼ or ▲ denote hypomethylation or hypermethylation, respectively. ↓ or ↑ arrows denote a correlation between observed methylation and decreased or increased NR3C1 expression levels, respectively. Methylation shown here represents either mean or CpG site-specific methylation, depending on the study method used, and arrows shown here are not meant to indicate methylation at specific CpG loci. Figure is not to scale. *Mean methylation levels only reported across all 4 first exon promoters, and not within each promoter.
Fig. 6
Fig. 6
Human tissue-specific gene expression of non-coding first exons within the NR3C1 proximal promoter, as reported by Turner and Muller (2005) and Presul et al. (2007). Dark blue to light blue gradients represent strong to weak expression, respectively. Note: exon 1J was discovered by Presul et al., and thus was not measured in Turner and Muller. Presul et al. did not measure exons 1E and 1H.
Fig. 7
Fig. 7
Model of (A) epigenetic and (B) subsequent systems-level PTSD pathogenesis proposed by Klengel et al. (2013). Note: Klengel et al. observed that FKBP5 methylation only leads to higher levels of FKBP5 expression in the presence of GR complex activation.

References

    1. Adalsteinsson B.T., Gudnason H., Aspelund T. Heterogeneity in white blood cells has potential to confound DNA methylation measurements. PLoS One. 2012;7(10):e46705.
    1. Ahlquist T., Lind G.E., Costa V.L. Gene methylation profiles of normal mucosa, and benign and malignant colorectal tumors identify early onset markers. Mol. Cancer. 2008;7:94.
    1. Alikhani-Koopaei R., Fouladkou F., Frey F.J., Frey B.M. Epigenetic regulation of 11 beta-hydroxysteroid dehydrogenase type 2 expression. J. Clin. Invest. 2004;114(8):1146–1157.
    1. Alt S.R., Turner J.D., Klok M.D. Differential expression of glucocorticoid receptor transcripts in major depressive disorder is not epigenetically programmed. Psychoneuroendocrinology. 2010;35(4):544–556.
    1. Baylin S.B., Esteller M., Rountree M.R., Bachman K.E., Schuebel K., Herman J.G. Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer. Hum. Mol. Genet. 2001;10(7):687–692.
    1. Bech P., Rasmussen N.A., Olsen L.R., Noerholm V., Abildgaard W. The sensitivity and specificity of the major depression inventory, using the present state examination as the index of diagnostic validity. J. Affect. Disord. 2001;66(2–3):159–164.
    1. Binder E.B. The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology. 2009;34(Suppl. 1):S186–S195.
    1. Binder E.B., Bradley R.G., Liu W. Association of FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symptoms in adults. JAMA. 2008;299(11):1291–1305.
    1. Birgisdottir V., Stefansson O.A., Bodvarsdottir S.K., Hilmarsdottir H., Jonasson J.G., Eyfjord J.E. Epigenetic silencing and deletion of the BRCA1 gene in sporadic breast cancer. Breast Cancer Res. 2006;8(4):R38.
    1. Blair L.J., Nordhues B.A., Hill S.E. Accelerated neurodegeneration through chaperone-mediated oligomerization of tau. J. Clin. Invest. 2013;123(10):4158–4169.
    1. Bock C. Analysing and interpreting DNA methylation data. Nat. Rev. Genet. 2012;13(10):705–719.
    1. Braak H., Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–259.
    1. Bremner J.D., Vermetten E., Mazure C.M. Development and preliminary psychometric properties of an instrument for the measurement of childhood trauma: the early trauma inventory. Depress Anxiety. 2000;12(1):1–12.
    1. Burris H.H., Braun J.M., Byun H.M. Association between birth weight and DNA methylation of IGF2, glucocorticoid receptor and repetitive elements LINE-1 and Alu. Epigenomics. 2013;5(3):271–281.
    1. Cameron O.G. Anxious-depressive comorbidity: effects on HPA axis and CNS noradrenergic functions. Essent. Psychopharmacol. 2006;7(1):24–34.
    1. Cao-Lei L., Leija S.C., Kumsta R. Transcriptional control of the human glucocorticoid receptor: identification and analysis of alternative promoter regions. Hum. Genet. 2011;129(5):533–543.
    1. Ceccato F., Lombardi G., Manara R. Temozolomide and pasireotide treatment for aggressive pituitary adenoma: expertise at a tertiary care center. J. Neuro-Oncol. 2015;122(1):189–196.
    1. Chen H., Fan J., Shou Q., Zhang L., Ma H., Fan Y. Hypermethylation of glucocorticoid receptor gene promoter results in glucocorticoid receptor gene low expression in peripheral blood mononuclear cells of patients with systemic lupus erythematosus. Rheumatol. Int. 2015;35(8):1335–1342.
    1. Cohen S., Kamarck T., Mermelstein R. A global measure of perceived stress. J. Health Soc. Behav. 1983;24(4):385–396.
    1. Conradt E., Lester B.M., Appleton A.A., Armstrong D.A., Marsit C.J. The roles of DNA methylation of NR3C1 and 11beta-HSD2 and exposure to maternal mood disorder in utero on newborn neurobehavior. Epigenetics. 2013;8(12):1321–1329.
    1. Crujeiras A.B., Campion J., Diaz-Lagares A. Association of weight regain with specific methylation levels in the NPY and POMC promoters in leukocytes of obese men: a translational study. Regul. Pept. 2013;186:1–6.
    1. Dammann G., Teschler S., Haag T., Altmuller F., Tuczek F., Dammann R.H. Increased DNA methylation of neuropsychiatric genes occurs in borderline personality disorder. Epigenetics. 2011;6(12):1454–1462.
    1. Daskalakis N.P., Yehuda R. Site-specific methylation changes in the glucocorticoid receptor exon 1F promoter in relation to life adversity: systematic review of contributing factors. Front. Neurosci. 2014;8:369.
    1. de Calignon A., Fox L.M., Pitstick R. Caspase activation precedes and leads to tangles. Nature. 2010;464(7292):1201–1204.
    1. de Calignon A., Polydoro M., Suarez-Calvet M. Propagation of tau pathology in a model of early Alzheimer's disease. Neuron. 2012;73(4):685–697.
    1. Eades L.E., Thiagarajah A.S., Harris J., Jones S.A., Morand E.F., Leech M. GILZ: a new link between the hypothalamic pituitary adrenal axis and rheumatoid arthritis? Immunol. Cell Biol. 2014;92(9):747–751.
    1. Edelman S., Shalev I., Uzefovsky F. Epigenetic and genetic factors predict women's salivary cortisol following a threat to the social self. PLoS One. 2012;7(11):e48597.
    1. Ehrlich S., Weiss D., Burghardt R. Promoter specific DNA methylation and gene expression of POMC in acutely underweight and recovered patients with anorexia nervosa. J. Psychiatr. Res. 2010;44(13):827–833.
    1. Falini B., Sportoletti P., Brunetti L., Martelli M.P. Perspectives for therapeutic targeting of gene mutations in acute myeloid leukaemia with normal cytogenetics. Br. J. Haematol. 2015
    1. Falsetti S.A., Resnick H.S., Resick P.A., Kilpatrick D. The Modified PTSD Symptom Scale: a brief self-report measure of posttraumatic stress disorder. Behav. Ther. 1993;16:161–162.
    1. Ferrari P., Sansonnens A., Dick B., Frey F.J. In vivo 11beta-HSD-2 activity: variability, salt-sensitivity, and effect of licorice. Hypertension. 2001;38(6):1330–1336.
    1. Fink L.A., Bernstein D., Handelsman L., Foote J., Lovejoy M. Initial reliability and validity of the childhood trauma interview: a new multidimensional measure of childhood interpersonal trauma. Am. J. Psychiatry. 1995;152(9):1329–1335.
    1. First M., Gibbon M., Spitzer R., Williams J.B.W., Smith B.L. American Psychiatric Association; New York: 1994. Structured Clinical Interview for DSM-IV Personality Disorders (SCID-II)
    1. Foa E.B., Tolin D.F. Comparison of the PTSD symptom scale-interview version and the clinician-administered PTSD scale. J. Trauma. Stress. 2000;13(2):181–191.
    1. Foa E., Cashman L., Jaycox L., Perry K. The validation of a self-report measure of PTSD: The Posttraumatic Diagnostic Scale. Psychol. Assess. 1997;9:445–451.
    1. Fries G.R., Vasconcelos-Moreno M.P., Gubert C. Hypothalamic-pituitary-adrenal axis dysfunction and illness progression in bipolar disorder. Int. J. Neuropsychopharmacol. 2015;18(1)
    1. Friso S., Pizzolo F., Choi S.W. Epigenetic control of 11 beta-hydroxysteroid dehydrogenase 2 gene promoter is related to human hypertension. Atherosclerosis. 2008;199(2):323–327.
    1. Fukuda K., Straus S.E., Hickie I., Sharpe M.C., Dobbins J.G., Komaroff A. The chronic fatigue syndrome: a comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann. Intern. Med. 1994;121(12):953–959.
    1. Furukawa H., Oka S., Matsui T. Genome, epigenome and transcriptome analyses of a pair of monozygotic twins discordant for systemic lupus erythematosus. Hum. Immunol. 2013;74(2):170–175.
    1. Gillum R.F. Epidemiology of hypertension in African American women. Am. Heart J. 1996;131(2):385–395.
    1. Gómez-Beneyto M., Villar M., Renovell M. The diagnosis of personality disorder with a modified version of the SCID-II in a Spanish clinical sample. J. Personal. Disord. 1994;8(2):104–110.
    1. Hahn O.S., Ahn J.H., Song S.H. Development of Korean version of structured clinical interview schedule for DSM-IV axis I disorder: interrater reliability. J. Korean Neuropsychiatr. Assoc. 2000;39(2):362–372.
    1. Hiraki M., Kitajima Y., Koga Y. Aberrant gene methylation is a biomarker for the detection of cancer cells in peritoneal wash samples from advanced gastric cancer patients. Ann. Surg. Oncol. 2011;18(10):3013–3019.
    1. Hochberg M.C. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.
    1. Höhne N., Poidinger M., Merz F. FKBP5 genotype-dependent DNA methylation and mRNA regulation after psychosocial stress in remitted depression and healthy controls. Int J Neuropsychopharmacol. 2015;18(4)
    1. Houseman E.A., Molitor J., Marsit C.J. Reference-free cell mixture adjustments in analysis of DNA methylation data. Bioinformatics. 2014;30(10):1431–1439.
    1. Houseman E.A., Kile M.L., Christiani D.C., Ince T.A., Kelsey K.T., Marsit C.J. Reference-free deconvolution of DNA methylation data and mediation by cell composition effects. BMC Bioinforma. 2016;17:259.
    1. Hunter R.W., Ivy J.R., Bailey M.A. Glucocorticoids and renal Na(+) transport: implications for hypertension and salt sensitivity. J. Physiol. 2014;592(8):1731–1744.
    1. Jessop D.S. Review: central non-glucocorticoid inhibitors of the hypothalamo-pituitary-adrenal axis. J. Endocrinol. 1999;160(2):169–180.
    1. Kadmiel M., Cidlowski J.A. Glucocorticoid receptor signaling in health and disease. Trends Pharmacol. Sci. 2013;34(9):518–530.
    1. Kalsbeek A., van der Spek R., Lei J., Endert E., Buijs R.M., Fliers E. Circadian rhythms in the hypothalamo-pituitary-adrenal (HPA) axis. Mol. Cell. Endocrinol. 2012;349(1):20–29.
    1. Kang G.H., Lee S., Cho N.Y. DNA methylation profiles of gastric carcinoma characterized by quantitative DNA methylation analysis. Lab. Investig. 2008;88(2):161–170.
    1. Kay P., Schlossmacher G., Matthews L. Loss of glucocorticoid receptor expression by DNA methylation prevents glucocorticoid induced apoptosis in human small cell lung cancer cells. PLoS One. 2011;6(10):e24839.
    1. Khorram N.M., Magee T.R., Wang C., Desai M., Ross M., Khorram O. Maternal undernutrition programs offspring adrenal expression of steroidogenic enzymes. Reprod. Sci. (Thousand Oaks, Calif) 2011;18(10):931–940.
    1. Kim D., Kubzansky L.D., Baccarelli A.A. Psychological factors and DNA methylation of genes related to immune/inflammatory system markers: the VA Normative Aging Study. BMJ Open. 2016;6(1):e009790.
    1. Kirschbaum C., Kudielka B.M., Gaab J., Schommer N.C., Hellhammer D.H. Impact of gender, menstrual cycle phase, and oral contraceptives on the activity of the hypothalamus-pituitary-adrenal axis. Psychosom. Med. 1999;61(2):154–162.
    1. Klengel T., Mehta D., Anacker C. Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nat. Neurosci. 2013;16(1):33–41.
    1. Kunzel H.E., Binder E.B., Nickel T. Pharmacological and nonpharmacological factors influencing hypothalamic-pituitary-adrenocortical axis reactivity in acutely depressed psychiatric in-patients, measured by the Dex-CRH test. Neuropsychopharmacology. 2003;28(12):2169–2178.
    1. Labonte B., Azoulay N., Yerko V., Turecki G., Brunet A. Epigenetic modulation of glucocorticoid receptors in posttraumatic stress disorder. Transl. Psychiatry. 2014;4
    1. Lam L.L., Emberly E., Fraser H.B. Factors underlying variable DNA methylation in a human community cohort. Proc. Natl. Acad. Sci. U. S. A. 2012;109(Suppl. 2):17253–17260.
    1. Lasagna-Reeves C.A., Castillo-Carranza D.L., Sengupta U. Identification of oligomers at early stages of tau aggregation in Alzheimer's disease. FASEB J. 2012;26(5):1946–1959.
    1. Lee A.G., Buckmaster C.L., Yi E., Schatzberg A.F., Lyons D.M. Coping and glucocorticoid receptor regulation by stress inoculation. Psychoneuroendocrinology. 2014;49:272–279.
    1. Li S., Rong M., Iacopetta B. DNA hypermethylation in breast cancer and its association with clinicopathological features. Cancer Lett. 2006;237(2):272–280.
    1. Liberati A., Altman D.G., Tetzlaff J. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700.
    1. Lien H.C., Lu Y.S., Cheng A.L. Differential expression of glucocorticoid receptor in human breast tissues and related neoplasms. J. Pathol. 2006;209(3):317–327.
    1. Lind G.E., Kleivi K., Meling G.I. ADAMTS1, CRABP1, and NR3C1 identified as epigenetically deregulated genes in colorectal tumorigenesis. Cell. Oncol. 2006;28(5–6):259–272.
    1. Madrigano J., Baccarelli A., Mittleman M.A. Air pollution and DNA methylation: interaction by psychological factors in the VA normative aging study. Am. J. Epidemiol. 2012;176(3):224–232.
    1. Martin-Blanco A., Ferrer M., Soler J. Association between methylation of the glucocorticoid receptor gene, childhood maltreatment, and clinical severity in borderline personality disorder. J. Psychiatr. Res. 2014;57:34–40.
    1. McGowan P.O., Sasaki A., D'Alessio A.C. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat. Neurosci. 2009;12(3):342–348.
    1. Melas P.A., Wei Y., Wong C.C. Genetic and epigenetic associations of MAOA and NR3C1 with depression and childhood adversities. Int. J. Neuropsychopharmacol. 2013;16(7):1513–1528.
    1. Mizoguchi Y., Kajiume T., Miyagawa S., Okada S., Nishi Y., Kobayashi M. Steroid-dependent ACTH-produced thymic carcinoid: regulation of POMC gene expression by cortisol via methylation of its promoter region. Horm. Res. 2007;67(5):257–262.
    1. Moisiadis V.G., Matthews S.G. Glucocorticoids and fetal programming part 1: outcomes. Nat. Rev. Endocrinol. 2014;10(7):391–402.
    1. Moser D., Molitor A., Kumsta R., Tatschner T., Riederer P., Meyer J. The glucocorticoid receptor gene exon 1-F promoter is not methylated at the NGFI-A binding site in human hippocampus. World J. Biol. Psychiatry. 2007;8(4):262–268.
    1. Muschler M.A., Hillemacher T., Kraus C., Kornhuber J., Bleich S., Frieling H. DNA methylation of the POMC gene promoter is associated with craving in alcohol dependence. J Neural Transm. (Vienna) 2010;117(4):513–519.
    1. Na K.S., Chang H.S., Won E. Association between glucocorticoid receptor methylation and hippocampal subfields in major depressive disorder. PLoS One. 2014;9(1)
    1. NCI Cancer Health Disparities. 2008.
    1. Needham B.L., Smith J.A., Zhao W. Life course socioeconomic status and DNA methylation in genes related to stress reactivity and inflammation: the multi-ethnic study of atherosclerosis. Epigenetics. 2015;10(10):958–969.
    1. Nesset K.A., Perri A.M., Mueller C.R. Frequent promoter hypermethylation and expression reduction of the glucocorticoid receptor gene in breast tumors. Epigenetics. 2014;9(6):851–859.
    1. Newell-Price J. Proopiomelanocortin gene expression and DNA methylation: implications for Cushing's syndrome and beyond. J. Endocrinol. 2003;177(3):365–372.
    1. Nwankwo T., Yoon S.S., Burt V., Gu Q. Hypertension among adults in the United States: National Health and Nutrition Examination Survey, 2011-2012. NCHS Data Brief. 2013;(133):1–8.
    1. Paakinaho V., Makkonen H., Jaaskelainen T., Palvimo J.J. Glucocorticoid receptor activates poised FKBP51 locus through long-distance interactions. Mol. Endocrinol. 2010;24(3):511–525.
    1. Palma-Gudiel H., Cordova-Palomera A., Leza J.C., Fananas L. Glucocorticoid receptor gene (NR3C1) methylation processes as mediators of early adversity in stress-related disorders causality: a critical review. Neurosci. Biobehav. Rev. 2015;55:520–535.
    1. Papadopoulou N.G., Oleson L., Kempuraj D., Donelan J., Cetrulo C.L., Theoharides T.C. Regulation of corticotropin-releasing hormone receptor-2 expression in human cord blood-derived cultured mast cells. J. Mol. Endocrinol. 2005;35(3):R1–R8.
    1. Parrella P., Poeta M.L., Gallo A.P. Nonrandom distribution of aberrant promoter methylation of cancer-related genes in sporadic breast tumors. Clin. Cancer Res. 2004;10(16):5349–5354.
    1. Perroud N., Paoloni-Giacobino A., Prada P. Increased methylation of glucocorticoid receptor gene (NR3C1) in adults with a history of childhood maltreatment: a link with the severity and type of trauma. Transl. Psychiatry. 2011;1:e59.
    1. Perroud N., Paoloni-Giacobino A., Prada P. Increased methylation of glucocorticoid receptor gene (NR3C1) in adults with a history of childhood maltreatment: a link with the severity and type of trauma. Transl. Psychiatry. 2011;1(12)
    1. Pizzolo F., Friso S., Morandini F. Apparent mineralocorticoid excess by a novel mutation and epigenetic modulation by HSD11B2 promoter methylation. J. Clin. Endocrinol. Metab. 2015;100(9):E1234–E1241.
    1. Prados J., Stenz L., Courtet P. Borderline personality disorder and childhood maltreatment: a genome-wide methylation analysis. Genes Brain Behav. 2015;14(2):177–188.
    1. Preisig M., Fenton B.T., Matthey M.-L., Berney A., Ferrero F. Diagnostic interview for genetic studies (DIGS): inter-rater and test-retest reliability of the French version. Eur. Arch. Psychiatry Clin. Neurosci. 1999;249(4):174–179.
    1. Presul E., Schmidt S., Kofler R., Helmberg A. Identification, tissue expression, and glucocorticoid responsiveness of alternative first exons of the human glucocorticoid receptor. J. Mol. Endocrinol. 2007;38(1–2):79–90.
    1. Quax R.A., Manenschijn L., Koper J.W. Glucocorticoid sensitivity in health and disease. Nat. Rev. Endocrinol. 2013;9(11):670–686.
    1. Ratman D., Vanden Berghe W., Dejager L. How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering. Mol. Cell. Endocrinol. 2013;380(1–2):41–54.
    1. Ronco A.M., Llaguno E., Epunan M.J., Llanos M.N. Effect of cadmium on cortisol production and 11beta-hydroxysteroid dehydrogenase 2 expression by cultured human choriocarcinoma cells (JEG-3) Toxicol. in Vitro. 2010;24(6):1532–1537.
    1. Sanchez-Vega B., Gandhi V. Glucocorticoid resistance in a multiple myeloma cell line is regulated by a transcription elongation block in the glucocorticoid receptor gene (NR3C1) Br. J. Haematol. 2009;144(6):856–864.
    1. Santacruz K., Lewis J., Spires T. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309(5733):476–481.
    1. Schloesser R.J., Martinowich K., Manji H.K. Mood-stabilizing drugs: mechanisms of action. Trends Neurosci. 2012;35(1):36–46.
    1. Shannon C.P., Balshaw R., Chen V. Enumerateblood – an R package to estimate the cellular composition of whole blood from Affymetrix Gene ST gene expression profiles. BMC Genomics. 2017;18:43.
    1. Shields A.E., Wise L.A., Ruiz-Narvaez E.A. Childhood abuse, promoter methylation of leukocyte NR3C1 and the potential modifying effect of emotional support. Epigenomics. 2016;8(11):1507–1517.
    1. Siegel R.L., Miller K.D., Jemal A. Cancer statistics, 2015. CA Cancer J. Clin. 2015;65(1):5–29.
    1. Smith S.M., Vale W.W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin. Neurosci. 2006;8(4):383–395.
    1. Smyth L.J., McKay G.J., Maxwell A.P., McKnight A.J. DNA hypermethylation and DNA hypomethylation is present at different loci in chronic kidney disease. Epigenetics. 2014;9(3):366–376.
    1. Steiger H., Labonte B., Groleau P., Turecki G., Israel M. Methylation of the glucocorticoid receptor gene promoter in bulimic women: associations with borderline personality disorder, suicidality, and exposure to childhood abuse. Int. J. Eat. Disord. 2013;46(3):246–255.
    1. Strehl C., Buttgereit F. Optimized glucocorticoid therapy: teaching old drugs new tricks. Mol. Cell. Endocrinol. 2013;380(1–2):32–40.
    1. Tasker J.G., Herman J.P. Mechanisms of rapid glucocorticoid feedback inhibition of the hypothalamic-pituitary-adrenal axis. Stress. 2011;14(4):398–406.
    1. Tomlinson J.W., Walker E.A., Bujalska I.J. 11β-Hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Endocr. Rev. 2004;25(5):831–866.
    1. Tseng E., Prica A., Zhang L. Monthly blood transfusions decrease after four months of azacitidine. Vox Sang. 2015
    1. Turner J.D., Alt S.R., Cao L. Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more. Biochem. Pharmacol. 2010;80(12):1860–1868.
    1. Turecki G. The molecular bases of the suicidal brain. Nat. Rev. Neurosci. 2014;15(12):802–816.
    1. Turecki G., Meaney M.J. Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review. Biol. Psychiatry. 2014
    1. Turner J.D., Muller C.P. Structure of the glucocorticoid receptor (NR3C1) gene 5′ untranslated region: identification, and tissue distribution of multiple new human exon 1. J. Mol. Endocrinol. 2005;35(2):283–292.
    1. Turner J.D., Vernocchi S., Schmitz S., Muller C.P. Role of the 5′-untranslated regions in post-transcriptional regulation of the human glucocorticoid receptor. Biochim. Biophys. Acta. 2014;1839(11):1051–1061.
    1. Tyrka A.R., Ridout K.K., Parade S.H., Paquette A., Marsit C.J., Seifer R. Childhood maltreatment and methylation of FK506 binding protein 5 gene (FKBP5) Dev. Psychopathol. 2015;27(4 Pt 2):1637–1645.
    1. Udali S., Guarini P., Moruzzi S., Choi S.W., Friso S. Cardiovascular epigenetics: from DNA methylation to microRNAs. Mol. Asp. Med. 2013;34(4):883–901.
    1. van der Goes M.C., Jacobs J.W., Bijlsma J.W. The value of glucocorticoid co-therapy in different rheumatic diseases - positive and adverse effects. Arthritis Res. Ther. 2014;16(Suppl. 2):S2.
    1. Vangeel E., Van Den Eede F., Hompes T. Chronic fatigue syndrome and DNA hypomethylation of the glucocorticoid receptor gene promoter 1F region: associations with HPA Axis hypofunction and childhood trauma. Psychosom. Med. 2015;77(8):853–862.
    1. Veenendaal M.V., Costello P.M., Lillycrop K.A. Prenatal famine exposure, health in later life and promoter methylation of four candidate genes. J. Dev. Orig. Health Dis. 2012;3(6):450–457.
    1. Vermeer H., Hendriks-Stegeman B.I., van der Burg B., van Buul-Offers S.C., Jansen M. Glucocorticoid-induced increase in lymphocytic FKBP51 messenger ribonucleic acid expression: a potential marker for glucocorticoid sensitivity, potency, and bioavailability. J. Clin. Endocrinol. Metab. 2003;88(1):277–284.
    1. Vilasco M., Communal L., Mourra N., Courtin A., Forgez P., Gompel A. Glucocorticoid receptor and breast cancer. Breast Cancer Res. Treat. 2011;130(1):1–10.
    1. Vukojevic V., Kolassa I.T., Fastenrath M. Epigenetic modification of the glucocorticoid receptor gene is linked to traumatic memory and post-traumatic stress disorder risk in genocide survivors. J. Neurosci. 2014;34(31):10274–10284.
    1. Wan Q., Gao K., Rong H. Histone modifications of the Crhr1 gene in a rat model of depression following chronic stress. Behav. Brain Res. 2014;271:1–6.
    1. Weathers F.W., Keane T.M., Davidson J.R. Clinician-administered PTSD scale: a review of the first ten years of research. Depress Anxiety. 2001;13(3):132–156.
    1. Westphal N.J., Seasholtz A.F. CRH-BP: the regulation and function of a phylogenetically conserved binding protein. Front. Biosci. 2006;11:1878–1891.
    1. Wittchen H.U., Lachner G., Wunderlich U., Pfister H. Test-retest reliability of the computerized DSM-IV version of the Munich-Composite International Diagnostic Interview (M-CIDI) Soc. Psychiatry Psychiatr. Epidemiol. 1998;33(11):568–578.
    1. Wochnik G.M., Ruegg J., Abel G.A., Schmidt U., Holsboer F., Rein T. FK506-binding. Proteins 51 and 52 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor in mammalian cells. J. Biol. Chem. 2005;280(6):4609–4616.
    1. Wu Q., Lothe R.A., Ahlquist T. DNA methylation profiling of ovarian carcinomas and their in vitro models identifies HOXA9, HOXB5, SCGB3A1, and CRABP1 as novel targets. Mol. Cancer. 2007;6:45.
    1. Yang X., Ewald E.R., Huo Y. Glucocorticoid-induced loss of DNA methylation in non-neuronal cells and potential involvement of DNMT1 in epigenetic regulation of Fkbp5. Biochem. Biophys. Res. Commun. 2012;420(3):570–575.
    1. Yehuda R., Daskalakis N.P., Desarnaud F. Epigenetic biomarkers as predictors and correlates of symptom improvement following psychotherapy in combat veterans with PTSD. Front. Psychiatry. 2013;4:118.
    1. Yehuda R., Flory J.D., Bierer L.M. Lower methylation of glucocorticoid receptor gene promoter 1 in peripheral blood of veterans with posttraumatic stress disorder. Biol. Psychiatry. 2015;77(4):356–364.
    1. Yehuda R., Daskalakis N.P., Bierer L.M. Holocaust exposure induced intergenerational effects on FKBP5 methylation. Biol. Psychiatry. 2015
    1. Yoon S.S., Carroll M.D., Fryar C.D. Hypertension prevalence and control among adults: United States, 2011–2014. NCHS Data Brief. 2015;(220):1–8.
    1. Zannas A.S., Wiechmann T., Gassen N.C., Binder E.B. Gene-stress-epigenetic regulation of FKBP5: clinical and translational implications. Neuropsychopharmacology. 2016;41(1):261–274.
    1. Zhang X., Clark A.F., Yorio T. FK506-binding protein 51 regulates nuclear transport of the glucocorticoid receptor beta and glucocorticoid responsiveness. Invest. Ophthalmol. Vis. Sci. 2008;49(3):1037–1047.
    1. Zhao J., An Q., Goldberg J., Quyyumi A.A., Vaccarino V. Promoter methylation of glucocorticoid receptor gene is associated with subclinical atherosclerosis: a monozygotic twin study. Atherosclerosis. 2015;242(1):71–76.
    1. Zigmond A.S., Snaith R.P. The hospital anxiety and depression scale. Acta Psychiatr. Scand. 1983;67(6):361–370.

Source: PubMed

3
Abonnere