Hostname: page-component-76fb5796d-2lccl Total loading time: 0 Render date: 2024-04-27T03:03:05.433Z Has data issue: false hasContentIssue false

Childhood adversity predicts black young adults’ DNA methylation-based accelerated aging: A dual pathway model

Published online by Cambridge University Press:  20 December 2021

Steven R. H. Beach*
Affiliation:
Department of Psychology, University of Georgia, Athens, GA, USA Center for Family Research, University of Georgia, Athens, GA, USA
Frederick X. Gibbons
Affiliation:
Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA
Sierra E. Carter
Affiliation:
Department of Psychology, Georgia State University, Atlanta, GA, USA
Mei Ling Ong
Affiliation:
Center for Family Research, University of Georgia, Athens, GA, USA
Justin A. Lavner
Affiliation:
Department of Psychology, University of Georgia, Athens, GA, USA Center for Family Research, University of Georgia, Athens, GA, USA
Man-Kit Lei
Affiliation:
Department of Sociology, University of Georgia, Athens, GA, USA
Ronald L. Simons
Affiliation:
Department of Sociology, University of Georgia, Athens, GA, USA
Meg Gerrard
Affiliation:
Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA
Robert A. Philibert
Affiliation:
Department of Psychiatry, University of Iowa, Iowa City, IA, USA Behavioral Diagnostics, Coralville, IA, USA
*
Corresponding author: Steven R. H. Beach, email: srhbeach@uga.edu

Abstract

We expand upon prior work (Gibbons et al., 2012) relating childhood stressor effects, particularly harsh childhood environments, to risky behavior and ultimately physical health by adding longer-term outcomes – deoxyribonucleic acid (DNA) methylation-based measures of accelerated aging (DNAm-aging). Further, following work on the effects of early exposure to danger (McLaughlin et al., 2014), we also identify an additional pathway from harsh childhood environments to DNAm-aging that we label the danger/FKBP5 pathway, which includes early exposure to dangerous community conditions that are thought to impact glucocorticoid regulation and pro-inflammatory mechanisms. Because different DNAm-aging indices provide different windows on accelerated aging, we contrast effects on early indices of DNAm-aging based on chronological age with later indices that focused on predicting biological outcomes. We utilize data from Family and Community Health Study participants (N = 449) from age 10 to 29. We find that harshness influences parenting, which, in turn, influences accelerated DNAm-aging through the risky cognitions and substance use (i.e., behavioral) pathway outlined by Gibbons et al. (2012). Harshness is also associated with increased exposure to threat/danger, which, in turn, leads to accelerated DNAm-aging through effects on FKBP5 activity and enhanced pro-inflammatory tendencies (i.e., the danger/FKBP5 pathway).

Type
Special Issue Article
Copyright
© The Author(s), 2021. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Abbas, B. K., Lichtman, A. H., & Pillai, S. (2022). Cellular and molecular immunology (10th ed.). Elsevier, SBN-10: 0323757480.Google Scholar
Bachmann, C. J., Beecham, J., O’Connor, T. G., Briskman, J., & Scott, S. (2021). A good investment: Longer-term cost savings of sensitive parenting in childhood. Journal of Child Psychology and Psychiatry. Advance online publication. https://doi.org/10.1111/jcpp.13461 Google ScholarPubMed
Beach, S. R. H., Lei, M. K., Simons, R. L., Barr, A. B., Simons, L. G., Ehrlich, K.Philibert, R. A. (2017). When inflammation and depression go together: The longitudinal effects of parent-child relationships. Development and Psychopathology, 29, 19691986. https://doi.org/10.1017/S0954579417001523 CrossRefGoogle ScholarPubMed
Belsky, J., Steinberg, L., & Draper, P. (1991). Childhood experience, interpersonal development, and reproductive strategy: An evolutionary theory of socialization. Child Development, 62(4), 647670. https://doi.org/10.2307/1131166 CrossRefGoogle ScholarPubMed
Benner, A. D., Wang, Y., Shen, Y., Boyle, A. E., Polk, R., & Cheng, Y. P. (2018). Racial/ethnic discrimination and well-being during adolescence: A meta-analytic review. American Psychologist, 73(7), 855883. https://doi.org/10.1037/amp0000204 CrossRefGoogle ScholarPubMed
Blair, L. J., Nordhues, B. A., Hill, S. E., Scaglione, K. M., O’Leary, J. C., Fontaine, S. N.Dickey, C. A. (2013). Accelerated neurodegeneration through chaperone-mediated oligomerization of tau. Journal of Clinical Investigation, 123(10), 41584169. https://doi.org/10.1172/jci69003 CrossRefGoogle ScholarPubMed
Bleich, S., Findling, M., Casey, L., Blendon, R., Benson, J., SteelFisher, G, Sayde, J., & Miller, C. (2019). Discrimination in the United States: Experiences of black Americans. Health Services Research, 54(S2), 13991408. https://doi.org/10.1111/1475-6773.13220 CrossRefGoogle ScholarPubMed
Bocklandt, S., Lin, W., Sehl, M. E., Sánchez, F. J., Sinsheimer, J. S., Horvath, S.Vilain, E. (2011). Epigenetic predictor of age. PLoS One, 6(6), e14821. https://doi.org/10.1371/journal.pone.0014821 CrossRefGoogle ScholarPubMed
Brody, G. H., Miller, G. E., Yu, T., Beach, S. R. H., & Chen, E. (2016). Supportive family environments ameliorate the link between racial discrimination and epigenetic aging. Psychological Science, 27(4), 530541. https://doi.org/10.1177/0956797615626703 CrossRefGoogle ScholarPubMed
Carnethon, M. R., Pu, J., Howard, G., Albert, M. A., Anderson, C. A., Bertoni, A. G.Yancy, C. W. (2017). Cardiovascular health in African Americans: A scientific statement from the American Heart Association. Circulation, 136, e393e423. https://doi.org/10.1161/cir.0000000000000534 CrossRefGoogle Scholar
Carter, S. E., Gibbons, F. X., & Beach, S. R. H. (in press). Measuring the biological embedding of racial trauma among Black Americans utilizing the RDoC approach. Development and Psychopathology.Google Scholar
Carter, S. E., Ong, M. L., Simons, R. L., Gibbons, F. X., Lei, M. K., & Beach, S. R. H. (2019). The effect of early discrimination on accelerated aging among African Americans. Health Psychology, 38(11), 10101013. https://doi.org/10.1037/hea0000788 CrossRefGoogle ScholarPubMed
Chen, E., Matthews, K. A., & Boyce, W. T. (2002). Socioeconomic differences in children’s health: How and why do these relationships change with age? Psychological Bulletin, 128(2), 295329.CrossRefGoogle ScholarPubMed
Clark, L. A., & Watson, D. The Mini Mood and Anxiety Symptom Questionnaire (mini-MASQ) 1995. Unpublished manuscript, University of Iowa, Iowa City.Google Scholar
Cohen, S., Janicki-Deverts, D., Doyle, W. J., Miller, G. E., Frank, E., Rabin, B. S.Turner, R. B. (2012). Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proceedings of the National Academy of Sciences of the United States of America, 109(16), 59955999. https://doi.org/10.1073/pnas.1118355109 CrossRefGoogle ScholarPubMed
Cole, S. W. (2014). Human social genomics. PLoS Genetics, 10(8), e1004601. https://doi.org/10.1371/journal.pgen.1004601 CrossRefGoogle ScholarPubMed
Cosmides, L., & Tooby, J. (1987). From evolution to behavior: Evolutionary psychology as the missing link. In Dupre, J. (Ed.), The latest on the best essays on evolution and optimality (pp. 276–306). MIT Press.Google Scholar
Dogan, M. V., Lei, M. K., Beach, S. R., Brody, G. H., & Philibert, R. A. (2016). Alcohol and tobacco consumption alter hypothalamic pituitary adrenal axis DNA methylation. Psychoneuroendocrinology, 66, 176184. https://doi.org/10.1016/j.psyneuen.2016.01.018 CrossRefGoogle ScholarPubMed
Ellis, B. J., del Giudice, M., Dishion, T. J., Figueredo, A. J., Gray, P., Griskevicius, V.Wilson, D. S. (2012). The evolutionary basis of risky adolescent behavior: Implications for science, policy, and practice. Developmental Psychology, 48(3), 598623. https://doi.org/10.1037/a0026220 CrossRefGoogle ScholarPubMed
Ellis, B. J., Figueredo, A. J., Brumbach, B. H., & Schlomer, G. L. (2009). Fundamental dimensions of environmental risk. Human Nature, 20(2), 204268. https://doi.org/10.1007/s12110-009-9063-7 CrossRefGoogle ScholarPubMed
English, D., Lambert, S. F., Tynes, B. M., Bowleg, L., Zea, M. C., & Howard, L. C. (2020). Daily multidimensional racial discrimination among Black U.S. American adolescents. Journal of Applied Developmental Psychology, 66, 101068. https://doi.org/10.1016/j.appdev.2019.101068 CrossRefGoogle ScholarPubMed
Erlejman, A. G., de Leo, S. A., Mazaira, G. I., Molinari, A. M., Camisay, M. F., Fontana, V.Galigniana, M. D. (2014). NF-κB transcriptional activity is modulated by FK506-binding proteins FKBP51 and FKBP52. Journal of Biological Chemistry, 289(38), 2626326276. https://doi.org/10.1074/jbc.m114.582882 CrossRefGoogle ScholarPubMed
Ferrucci, L., & Fabbri, E. (2018). Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nature Reviews Cardiology, 15(9), 505522. https://doi.org/10.1038/s41569-018-0064-2 CrossRefGoogle ScholarPubMed
Figueredo, A. J., Vásquez, G., Brumbach, B. H., Sefcek, J. A., Kirsner, B. R., & Jacobs, W. J. (2005). The K-factor: Individual differences in life history strategy. Personality and Individual Differences, 39(8), 13491360. https://doi.org/10.1016/j.paid.2005.06.009 CrossRefGoogle Scholar
Franceschi, C., & Campisi, J. (2014). Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, 69(Suppl 1), S4S9. https://doi.org/10.1093/gerona/glu057 CrossRefGoogle ScholarPubMed
Fransquet, P. D., Wrigglesworth, J., Woods, R. L., Ernst, M. E., & Ryan, J. (2019). The epigenetic clock as a predictor of disease and mortality risk: A systematic review and meta-analysis. Clinical Epigenetics, 11(1), 62. https://doi.org/10.1186/s13148-019-0656-7 CrossRefGoogle ScholarPubMed
Geronimus, A. T., Hicken, M., Keene, D., & Bound, J. (2006). Weathering, and age patterns of allostatic load scores among Blacks and Whites in the United States. American Journal of Public Health, 96(5), 826833. https://doi.org/10.2105/ajph.2004.060749 CrossRefGoogle ScholarPubMed
Geronimus, A. T., Hicken, M. T., Pearson, J. A., Seashols, S. J., Brown, K. L., & Cruz, T. D. (2010). Do US Black women experience stress-related accelerated biological aging? Human Nature, 21(1), 1938. https://doi.org/10.1007/s12110-010-9078-0 CrossRefGoogle ScholarPubMed
Geronimus, A. T., James, S. A., Destin, M., Graham, L. F., Hatzenbuehler, M. L., Murphy, M. C.Thompson, J. P. (2016). Jedi public health: Co-creating an identity-safe culture to promote health equity. SSM - Population Health, 2, 105116. https://doi.org/10.1016/j.ssmph.2016.02.008 CrossRefGoogle ScholarPubMed
Gerrard, M., Gibbons, F. X., Houlihan, A. E., Stock, M. L., & Pomery, E. A. (2008). A dual-process approach to health risk decision making: The prototype willingness model. Developmental Review, 28(1), 2961.CrossRefGoogle Scholar
Gibbons, F. X., Gerrard, M., & Boney McCoy, S. (1995). Prototype perception predicts (lack of) pregnancy prevention. Personality and Social Psychology Bulletin, 21, 8593. https://doi.org/10.1177/0146167295211009 CrossRefGoogle Scholar
Gibbons, F. X., Gerrard, M., Cleveland, M. J., Wills, T. A., & Brody, G. H. (2004). Perceived discrimination and substance use in African American parents and their children: A panel study. Journal of Personality and Social Psychology, 86(4), 517529.CrossRefGoogle ScholarPubMed
Gibbons, F. X., Gerrard, M., & Lane, D. J. (2003). A social reaction model of adolescent health risk. In Suls, J. M. & Wallston, K. A. (Eds.), Social psychological foundations of health and illness (pp. 107–136). Wiley-Blackwell.Google Scholar
Gibbons, F. X., Roberts, M. E., Gerrard, M., Li, Z., Beach, S. R. H., Simons, R. L.Philibert, R. A. (2012). The impact of stress on the life history strategies of African American adolescents: Cognitions, genetic moderation, and the role of discrimination. Developmental Psychology, 48(3), 722739. https://doi.org/10.1037/a0026599 CrossRefGoogle ScholarPubMed
Goosby, B. J., Cheadle, J. E., & Mitchell, C. (2018). Stress-related biosocial mechanisms of discrimination and African American health inequities. Annual Review of Sociology, 44(1), 319340. https://doi.org/10.1146/annurev-soc-060116-053403 CrossRefGoogle Scholar
Greene, M. L., Way, N., & Pahl, K. (2006). Trajectories of perceived adult and peer discrimination among Black, Latino, and Asian American adolescents: Patterns and psychological correlates. Developmental Psychology, 42(2), 218236. https://doi.org/10.1037/0012-1649.42.2.218 CrossRefGoogle ScholarPubMed
Gunnar, M., & Quevedo, K. (2007). The neurobiology of stress and development. Annual Review of Psychology, 58(1), 145173. https://doi.org/10.1146/annurev.psych.58.110405.085605 CrossRefGoogle ScholarPubMed
Hannum, G., Guinney, J., Zhao, L., Zhang, L., Hughes, G., Sadda, S.Zhang, K. (2013). Genome-wide methylation profiles reveal quantitative views of human aging rates. Molecular Cell, 49(2), 359367. https://doi.org/10.1016/j.molcel.2012.10.016 CrossRefGoogle ScholarPubMed
Hart, A. R., Lavner, J. A., Carter, S. E., & Beach, S. R. H. (2021). Racial discrimination, depressive symptoms, and sleep problems among Blacks in the rural South. Cultural Diversity and Ethnic Minority Psychology, 27(1), 123134. https://doi.org/10.1037/cdp0000365 CrossRefGoogle ScholarPubMed
Hayflick, L. (2007). Biological aging is no longer an unsolved problem. Annals of the New York Academy of Sciences, 1100(1), 113. https://doi.org/10.1196/annals.1395.001 CrossRefGoogle ScholarPubMed
Hillary, R. F., Stevenson, A. J., McCartney, D. L., Campbell, A., Walker, R. M., Howard, D. M.Marioni, R. E. (2020). Epigenetic clocks predict prevalence and incidence of leading causes of death and disease burden. BioRxiv, 10(1), 429, https://doi.org/10.1101/2020.01.31.928648,Google Scholar
Horvath, S. (2013). DNA methylation age of human tissues and cell types. Genome Biology, 14(10), R115. https://doi.org/10.1186/gb-2013-14-10-r115 CrossRefGoogle ScholarPubMed
Horvath, S., & Raj, K. (2018). DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nature Reviews Genetics, 19(6), 371384. https://doi.org/10.1038/s41576-018-0004-3 CrossRefGoogle ScholarPubMed
Houseman, E. A., Accomando, W. P., Koestler, D. C., Christensen, B. C., Marsit, C. J., Nelson, H. H.Kelsey, K. T. (2012). DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics, 13(1), 86. https://doi.org/10.1186/1471-2105-13-86 CrossRefGoogle ScholarPubMed
Howard, G., Safford, M. M., Moy, C. S., Howard, V. J., Kleindorder, D. O., Unverzagt, F. W.Cushman, M. (2017). Racial differences in the incidence of cardiovascular risk factors in older Black and White adults. Journal of the American Geriatrics Society, 65(1), 8390.CrossRefGoogle ScholarPubMed
Ishikawa, S., Saito, Y., Yanagawa, Y., Otani, S., Hiraide, S., Shimamura, K. I.Togashi, H. (2011). Early postnatal stress alters extracellular signal-regulated kinase signaling in the corticolimbic system modulating emotional circuitry in adult rats. European Journal of Neuroscience, 35(1), 135145. https://doi.org/10.1111/j.1460-9568.2011.07921.x CrossRefGoogle ScholarPubMed
Jinwal, U. K., Koren, J., Borysov, S. I., Schmid, A. B., Abisambra, J. F., Blair, L. J.Dickey, C. A. (2010). The hsp90 cochaperone, FKBP51, increases tau stability and polymerizes microtubules. Journal of Neuroscience, 30(2), 591599. https://doi.org/10.1523/jneurosci.4815-09.2010 CrossRefGoogle ScholarPubMed
Jylhävä, J., Pedersen, N. L., & Hägg, S. (2017). Biological age predictors. EBioMedicine, 21, 2936. https://doi.org/10.1016/j.ebiom.2017.03.046 CrossRefGoogle ScholarPubMed
Keppel, K. G., Pearcy, J. N., & Wagener, D. K. (2002). Trends in racial and ethnic-specific rates for the health status indicators: United States, 1990-98. Healthy People 2000 Statistical Notes, 23, 116.Google Scholar
Kremer, T., Braun, U., Chen, J., Harneit, A., Zang, Z., Schweiger, J. I.Tost, H. (2020). Epigenetic signature of FKBP5 is associated with structural variance in emotion regulatory brain circuits. Biological Psychiatry, 87(9), S391. https://doi.org/10.1016/j.biopsych.2020.02.1001 CrossRefGoogle Scholar
Kruger, D. J., Reischl, T., & Zimmerman, M. A. (2008). Time perspective as a mechanism for functional developmental adaptation. Journal of Social, Evolutionary, and Cultural Psychology, 2(1), 122. https://doi.org/10.1037/h0099336 CrossRefGoogle Scholar
Landrine, H., & Klonoff, E. A. (1996). The schedule of racist events: A measure of racial discrimination and a study of its negative physical and mental health consequences. Journal of Black Psychology, 22(2), 144168. https://doi.org/10.1177/00957984960222002 CrossRefGoogle Scholar
Lavner, J. A., Hart, A. R., Carter, S. E., & Beach, S. R. (in press). Longitudinal effects of racial discrimination on depressive symptoms among Black youth: Between- and within-person effects. Journal of the American Academy of Child & Adolescent Psychiatry, https://doi.org/10.1016/j.jaac.2021.04.020 Google Scholar
Lei, M. K., Gibbons, F. X., Simons, R. L., Philibert, R. A., & Beach, S. R. H. (2020). The effect of tobacco smoking differs across indices of DNA methylation-based aging in an African American sample: DNA methylation-based indices of smoking capture these effects. Genes, 11(3), 311. https://doi.org/10.3390/genes11030311 CrossRefGoogle Scholar
Lei, M.-K., Simons, R. L., Beach, S. R., & Philibert, R. A. (2019). Neighborhood disadvantage and biological aging: Using marginal structural models to assess the link between neighborhood census variables and epigenetic aging. The Journals of Gerontology: Series B, 74(7), 5059. https://doi.org/10.1093/geronb/gbx015 CrossRefGoogle ScholarPubMed
Levine, M. E., Lu, A. T., Quach, A., Chen, B. H., Assimes, T. L., Bandinelli, S.Horvath, S. (2018). An epigenetic biomarker of aging for lifespan and healthspan. Sedentary Life and Nutrition, 10(4), 573591. https://doi.org/10.18632/aging.101414 Google ScholarPubMed
Lim, E., Gandhi, K., Davis, J., & Chen, J. J. (2016). Prevalence of chronic conditions and multimorbidities in a geographically defined geriatric population with diverse races and ethnicities. Journal of Aging and Health, 30(3), 421444. https://doi.org/10.1177/0898264316680903 CrossRefGoogle Scholar
Link, B. G., & Phelan, J. (1995). Social conditions as fundamental causes of disease. Journal of Health and Social Behavior, 35, 8094. https://doi.org/10.2307/2626958 CrossRefGoogle Scholar
Liu, Y.-Z., Wang, Y.-X., & Jiang, C.-L. (2017). Inflammation: The common pathway of stress-related diseases. Frontiers in Human Neuroscience, 11, 316. https://doi.org/10.3389/fnhum.2017.00316 CrossRefGoogle ScholarPubMed
Low, B. S., Hazel, A., Parker, N., & Welch, K. B. (2008). Influences on women’s reproductive lives: Unexpected ecological underpinnings. Cross-Cultural Research, 42(3), 201219.CrossRefGoogle Scholar
Lu, A. T., Quach, A., Wilson, J. G., Reiner, A. P., Aviv, A., Raj, K.Horvath, S. (2019). DNA methylation GrimAge strongly predicts lifespan and healthspan. Sedentary Life and Nutrition, 11(2), 303327. https://doi.org/10.18632/aging.101684 Google ScholarPubMed
Lu, A. T., Seeboth, A., Tsai, P., Sun, D., Quach, A., Reiner, A. P.Horvath, S. (2019). DNA methylation-based estimator of telomere length. Sedentary Life and Nutrition, 11(16), 58955923. https://doi.org/10.18632/aging.102173 Google ScholarPubMed
Lupien, S. J., McEwen, B. S., Gunnar, M. R., & Heim, C. (2009). Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nature Reviews: Neuroscience, 10(6), 434445. https://doi.org/10.1038/nrn2639 CrossRefGoogle ScholarPubMed
Lynch, S. M. (2003). Cohort and life-course patterns in the relationship between education and health: A hierarchical approach. Demography, 40(2), 309331. https://doi.org/10.2307/3180803 CrossRefGoogle ScholarPubMed
Maiarù, M., Tochiki, K. K., Cox, M. B., Annan, L. V., Bell, C. G., Feng, X.Géranton, S. M. (2016). The stress regulator FKBP51 drives chronic pain by modulating spinal glucocorticoid signa ling. Science Translational Medicine, 8(325), 325ra19. https://doi.org/10.1126/scitranslmed.aab3376 CrossRefGoogle Scholar
Marzi, S. J., Sugden, K., Arseneault, L., Belsky, D. W., Burrage, J., Corcoran, D. L.Caspi, A. (2018). Analysis of DNA methylation in young people: Limited evidence for an association between victimization stress and epigenetic variation in blood. American Journal of Psychiatry, 175(6), 517529. https://doi.org/10.1176/appi.ajp.2017.17060693 CrossRefGoogle ScholarPubMed
Matsumoto, M., Togashi, H., Konno, K., Koseki, H., Hirata, R., Izumi, T.Yoshioka, M. (2008). Early postnatal stress alters the extinction of context-dependent conditioned fear in adult rats. Pharmacology, Biochemistry and Behavior, 89(3), 247252.CrossRefGoogle ScholarPubMed
McArdle, N., Osypuk, T. L., & Acevedo-Garcia, D.. Disparities in neighborhood poverty of poor Black and White children . 2007. http://diversitydata.sph.harvard.edu/Publications/brief7.pdf.Google Scholar
McLaughlin, K. A., & Sheridan, M. A. (2016). Beyond cumulative risk: A dimensional approach to childhood adversity. Current Directions in Psychological Science, 25(4), 239245. https://doi.org/10.1177/0963721416655883 CrossRefGoogle ScholarPubMed
McLaughlin, K. A., Sheridan, M. A., & Lambert, H. K. (2014). Childhood adversity and neural development: Deprivation and threat as distinct dimensions of early experience. Neuroscience and Biobehavioral Review, 47, 578591. https://doi.org/10.1016/j.neubiorev.2014.10.012 CrossRefGoogle ScholarPubMed
Miller, G. E., Chen, E., & Parker, K. J. (2011). Psychological stress in childhood and susceptibility to the chronic diseases of aging: Moving toward a model of behavioral and biological mechanisms. Psychological Bulletin, 137(6), 959997. https://doi.org/10.1037/a0024768 CrossRefGoogle Scholar
Nordfjäll, K., Svenson, U., Norrback, K., Adolfsson, R., & Roos, G. (2010). Large-scale parent-child comparison confirms a strong paternal influence on telomere length. European Journal of Human Genetics, 18(3), 385389.CrossRefGoogle Scholar
Nowakowski, A. C. H., & Sumerau, J. E. (2015). Swell foundations: Fundamental causes and chronic inflammation. Sociological Spectrum, 35(2), 161178.CrossRefGoogle Scholar
Paalani, M., Lee, J. W., Haddad, E., & Tonstad, S. (2011). Determinants of inflammatory markers in a bi-ethnic population. Ethnicity & Disease, 21(2), 142149.Google Scholar
Pascoe, E. A., & Smart Richman, L. (2009). Perceived discrimination and health: A meta-analytic review. Psychological Bulletin, 135, 531554. https://doi.org/10.1037/a0016059 CrossRefGoogle ScholarPubMed
Phelan, J. C., & Link, B. G. (2015). Is racism a fundamental cause of inequalities in health? Annual Review of Sociology, 41(1), 3113130.CrossRefGoogle Scholar
Philibert, R. A., & Beach, S. R. H. (2018). Would addressing alcohol consumption further account for variance in methylation? The American Journal of Psychiatry, 175(7), 684685.CrossRefGoogle ScholarPubMed
Philibert, R. A., Long, J. D., Mills, J. A. Dogan, Beach, M., S.R., H., Gibbons, F. X.Dogan, M. V. (2021). A simple, rapid, interpretable, actionable and implementable digital PCR based mortality index. Epigenetics, 16(10), 11351149. https://doi.org/10.1080/15592294.2020.1841874 CrossRefGoogle ScholarPubMed
Powell, N. D., Sloan, E. K., Bailey, M. T., Arevalo, J. M., Miller, G. E., Chen., E.Cole, S. W. (2013). Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via β-adrenergic induction of myclopoiesis. Proceedings of the National Academy of Sciences of the United States of America, 41, 1657416579.CrossRefGoogle Scholar
Schneider, D., Hastings, O. P., & LaBriola, J. (2018). Income inequality and class divides in parental investments. American Sociological Review, 83(3), 475507. https://doi.org/10.1177/0003122418772034 CrossRefGoogle Scholar
Simons, R. L., Lei, M. K, Klopack, E., Beach, S. R. H., Gibbons, F. X., & Philibert, R. A. (in press). The effects of social adversity, discrimination, and health risk behaviors on the accelerated aging of African Americans: Further support for the weathering hypothesis. Social Science and Medicine, 282, 113169. https://doi.org/10.1016/j.socscimed.2020.113169 CrossRefGoogle Scholar
Simons, R. L., Lei, M. K., Beach, S. H. R., Philibert, R. A., Cutrona, C., Gibbons, F. X.Barr, A. (2016). Economic hardship and biological weathering: The epigenetics of aging in a U.S. sample of black women. Social Science and Medicine, 150(8), 192200.CrossRefGoogle Scholar
Simons, R. L., Lei, M. K., Beach, S. R. H., Barr, A. B., Cutrona, C. E., Gibbons, F. X.Philibert, R. A. (2017). An index of the ratio of inflammatory to antiviral cell types mediates effects of social adversity and age on chronic illness. Social Science & Medicine, 185, 158165. https://doi.org/10.1016/j.socscimed.2017.03.005 CrossRefGoogle ScholarPubMed
Slavich, G. M., & Cole, S. W. (2013). The emerging field of human social genomics. Clinical Psychological Science, 1(3), 331348.CrossRefGoogle ScholarPubMed
Thornberry, T., Huzinga, D., & Loeber, R. (2004). The causes and correlates studies: Findings and policy implications. Juvenile Justice, 9(1), 319, https://www.ojp.gov/pdffiles1/ojjdp/203555.pdf Google Scholar
Thorpe, R. J., Gesahazion, R. G., Parker, L., Wilder, T., Rooks, R. N., Bowie, J. V.LaVeist, T. A. (2016). Accelerated health declines among African Americans in the USA. Journal of Urban Health, 93(5), 808819.CrossRefGoogle ScholarPubMed
van Marle, H. J. F., Hermans, E. J., Qin, S., & Fernandez, G. (2009). From specificity to sensitivity: How acute stress affects amygdala processing of biologically salient stimuli. Biological Psychiatry, 66(7), 649655.CrossRefGoogle ScholarPubMed
Williams, D. R. (2012). Miles to go before we sleep: Racial inequities in health. Journal of Health and Social Behavior., 53(3), 279295.CrossRefGoogle ScholarPubMed
Williams, D. R., Neighbors, H. W., & Jackson, J. S. (2003). Racial/ethnic discrimination and health: Findings from community studies. American Journal of Public Health, 93, 200208. https://doi.org/10.2105/AJPH.93.2.200 CrossRefGoogle ScholarPubMed
Zannas, A. S., Arloth, J., Carrillo-Roa, T., Lurato, S., Roh, S., Ressler, K. J.Mehta, D. (2015). Lifetime stress accelerates epigenetic aging in an urban, African American cohort: Relevance of glucocorticoid signaling. Genome Biology, 16, 266. https://doi.org/10.1186/s13059-015-0828-5 CrossRefGoogle Scholar
Zannas, A. S., Jia, M., Hafner, K., Baumert, J., Wiechmann, T. Pape, J., C.Binder, E. B. (2019). Epigenetic upregulation of FKBP5 by aging and stress contributes to NF-κB-driven inflammation and cardiovascular risk. Proceedings of the National Academy of Sciences of the United States of America, 116(23), 1137011379.CrossRefGoogle ScholarPubMed
Zapolski, T. C., Fisher, S., Hsu, W. W., & Barnes, J. (2016). What can parents do? Examining the role of parental support on the negative relationship among racial discrimination, depression, and drug use among African American youth. Clinical Psychological Science, 4(4), 718731.CrossRefGoogle ScholarPubMed
Supplementary material: File

Beach et al. supplementary material

Beach et al. supplementary material

Download Beach et al. supplementary material(File)
File 448.1 KB