HIV-1 Infection Accelerates Age According to the Epigenetic Clock

Steve Horvath, Andrew J Levine, Steve Horvath, Andrew J Levine

Abstract

Background: Infection with human immunodeficiency virus type 1 (HIV) is associated with clinical symptoms of accelerated aging, as evidenced by the increased incidence and diversity of age-related illnesses at relatively young ages and supporting findings of organ and cellular pathologic analyses. But it has been difficult to detect an accelerated aging effect at a molecular level.

Methods: Here, we used an epigenetic biomarker of aging based on host DNA methylation levels to study accelerated aging effects due to HIV infection. DNA from brain and blood tissue was assayed via the Illumina Infinium Methylation 450 K platform.

Results: Using 6 novel DNA methylation data sets, we show that HIV infection leads to an increase in epigenetic age both in brain tissue (7.4 years) and blood (5.2 years). While the observed accelerated aging effects in blood may reflect changes in blood cell composition (notably exhausted cytotoxic T cells), it is less clear what explains the observed accelerated aging effects in brain tissue.

Conclusions: Overall, our results demonstrate that the epigenetic clock is a useful biomarker for detecting accelerated aging effects due to HIV infection. This tool can be used to accurately determine the extent of age acceleration in individual tissues and cells.

Keywords: DNA methylation; HIV-1; aging; biomarker; epigenetics.

© The Author 2015. Published by Oxford University Press on behalf of the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Discovery brain data from human immunodeficiency virus (HIV)–infected subjects (cases) and HIV-uninfected subjects (controls). AD, DNA methylation (DNAm) age versus chronological age in all brain samples (A) and occipital cortex (B), cerebellum (C), and frontal lobe (D) samples. Red squares and black triangles in the scatterplot denote samples from cases and controls, respectively. The orange line depicts a linear regression line through case samples. Similarly, the black solid line corresponds to a regression line of DNAm age on chronological age in control samples. For each subject (point), age acceleration is defined as the vertical distance to the black regression line (ie, DNAm age minus the expected value based on control samples). E, Mean age acceleration versus control status in all brain samples. Each bar plot depicts 1 standard error around the mean value and reports the Kruskal–Wallis (nonparametric) group comparison test P value. Analogous results can be obtained when restricting the analysis to occipital cortex (B and F) or cerebellar (C and G) samples but not for samples from the frontal lobe (D and G). EH, The group comparisons in panels EH involved a total of 130, 72, 24, and 24, samples respectively. The number of samples in each group is reported by the rotated number under each bar. IL, HIV load (log10 transformed) in cerebrospinal fluid (CSF) of cases versus age acceleration in all brain samples (I) and occipital cortex (J), cerebellum (K), and frontal lobe (L) samples. Abbreviation: cor, correlation.
Figure 2.
Figure 2.
Validation brain data from human immunodeficiency virus (HIV)–infected subjects (cases) and HIV-uninfected subjects (controls). Analogous to Figure 1, we used independent data sets (data 2 and 3) to relate HIV status to epigenetic age acceleration in the frontal lobe and cerebellum of cases and controls. A and B, DNA methylation (DNAm) age versus chronological age in cerebellum (A) and frontal lobe (B) samples. Points (subjects) are colored by HIV status: cases correspond to red squares. The orange and black lines depict regression lines case and control samples, respectively. The measure of age acceleration is the same as used in Figure 1. C and D, Age acceleration versus HIV status in cerebellum (C) and frontal lobe (D) specimens. Each bar plot reports the Kruskal–Wallis (nonparametric) group comparison test P value and 1 standard error around the mean. Abbreviation: cor, correlation.
Figure 3.
Figure 3.
Epigenetic age in blood tissue versus human immunodeficiency virus (HIV) status. The first column (A and C) and second column (B and D) report findings for blood data sets 4 and 5, respectively. A and B, DNA methylation (DNAm) age versus chronological age. Samples from HIV-infected patients (cases; points) are colored in red. The black solid line and the orange line correspond to regression lines through HIV-uninfected control samples and case samples, respectively. For each subject (point), age acceleration is defined as the vertical distance to the black regression line. C and D, Mean age acceleration versus HIV status. The rotated numbers on the x-axis report the group sizes. The bar graphs report 1 standard error and a Kruskal–Wallis test P value. Abbreviation: cor, correlation.
Figure 4.
Figure 4.
Age acceleration versus blood cell counts in human immunodeficiency virus (HIV)–infected subjects (cases). Here we used DNA methylation (DNAm) data from peripheral blood mononuclear cells from cases (data set 6). A, DNAm age versus age. The red line indicates the regression line. The black line corresponds to y = x. Epigenetic age acceleration (defined as the residual) versus the percentage of natural killer (NK) cells (B), CD14+ monocytes (C), granulocytes (D), CD4+ T cells (E), CD8+ T cells (F), naive CD8+ T cells (G), and exhausted (CD28−CD45RA−) CD8+ T cells (H).
Figure 5.
Figure 5.
Age acceleration versus blood cell counts in HIV-uninfected control subjects (controls). The first 2 rows (AH) and the last 2 rows (IP) present results for controls from blood data sets 5 and 7, respectively. A and I, DNA methylation (DNAm) age versus chronological age. Age acceleration was defined as the residual resulting from the regression line (red line). Age acceleration versus natural killer (NK) cells (B and J), CD14+ monocytes (C and K), granulocytes (D and L), CD4+ T cells (E and M), CD8+ T cells (F and N), naive CD8+ T cells (G and O), and exhausted (CD28−CD45RA−) CD8+ T cells (H and P). The blood cell abundance measures were estimated on the basis of DNAm data (“Methods” section).

References

    1. Fausto A, Bongiovanni M, Cicconi P et al. . Potential predictive factors of osteoporosis in HIV-positive subjects. Bone 2006; 38:893–7.
    1. Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrinol Metab 2007; 92:2506–12.
    1. Lucas GM, Mehta SH, Atta MG et al. . End-stage renal disease and chronic kidney disease in a cohort of African-American HIV-infected and at-risk HIV-seronegative participants followed between 1988 and 2004. AIDS 2007; 21:2435–43.
    1. Silverberg M, Chao C, Leyden W et al. . HIV infection and the risk of cancers with and without a known infectious cause. AIDS 2009; 23:2337–45.
    1. Martin J, Volberding P. HIV and premature aging: a field still in its infancy. Ann Intern Med 2010; 153:477–9.
    1. Deeks SG. HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med 2011; 62:141–55.
    1. Desquilbet L, Jacobson LP, Fried LP et al. . A frailty-related phenotype before HAART initiation as an independent risk factor for AIDS or death after HAART among HIV-infected men. J Gerontol A Biol Sci Med Sci 2011; 66A:1030–8.
    1. Silverberg MJ, Chao C, Leyden WA et al. . HIV infection, immunodeficiency, viral replication, and the risk of cancer. Cancer Epidemiol Biomarkers Prev 2011; 20:2551–9.
    1. Womack JA, Goulet JL, Gibert C et al. . Increased risk of fragility fractures among HIV infected compared to uninfected male veterans. PLoS One 2011; 6:e17217.
    1. Wendelken L, Valcour V. Impact of HIV and aging on neuropsychological function. J Neurovirol 2012; 18:256–63.
    1. Kirk GD, Mehta SH, Astemborski J et al. . HIV, age, and the severity of hepatitis C virus–related liver disease A cohort study. Ann Intern Med 2013; 158:658–66.
    1. Greene M, Justice A, Lampiris H, Valcour V. Management of human immunodeficiency virus infection in advanced age. JAMA 2013; 309:1397–405.
    1. High KP, Brennan-Ing M, Clifford DB et al. . HIV and aging: state of knowledge and areas of critical need for research. A report to the NIH Office of AIDS Research by the HIV and Aging Working Group. J Acquir Immune Defic Syndr 2012; 60(Suppl 1):S1–18.
    1. Christensen B, Houseman E, Marsit C et al. . Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG Island context. PLoS Genet 2009; 5:e1000602.
    1. Bollati V, Schwartz J, Wright R et al. . Decline in genomic DNA methylation through aging in a cohort of elderly subjects. Mech Ageing Dev 2009; 130:234–9.
    1. Rakyan VK, Down TA, Maslau S et al. . Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res 2010; 20:434–9.
    1. Teschendorff AE, Menon U, Gentry-Maharaj A et al. . Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res 2010; 20:440–6.
    1. Vivithanaporn P, Heo G, Gamble J et al. . Neurologic disease burden in treated HIV/AIDS predicts survival. Neurology 2010; 75:1150–8.
    1. Horvath S, Zhang Y, Langfelder P et al. . Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol 2012; 13:R97.
    1. Numata S, Ye T, Hyde TM et al. . DNA methylation signatures in development and aging of the human prefrontal cortex. Am J Hum Genet 2012; 90:260–72.
    1. Alisch RS, Barwick BG, Chopra P et al. . Age-associated DNA methylation in pediatric populations. Genome Res 2012; 22:623–32.
    1. Johansson A, Enroth S, Gyllensten U. Continuous aging of the human DNA methylome throughout the human lifespan. PLoS One 2013; 8:e67378.
    1. Day K, Waite L, Thalacker-Mercer A et al. . Differential DNA methylation with age displays both common and dynamic features across human tissues that are influenced by CpG landscape. Genome Biol 2013; 14:R102.
    1. Bocklandt S, Lin W, Sehl ME et al. . Epigenetic predictor of age. PLoS One 2011; 6:e14821.
    1. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol 2013; 14:R115.
    1. Boks MP, Derks EM, Weisenberger DJ et al. . The relationship of DNA methylation with age, gender and genotype in twins and healthy controls. PLoS One 2009; 4:e6767.
    1. Horvath S, Erhart W, Brosch M et al. . Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci U S A 2014; 111:15538–43.
    1. Marioni R, Shah S, McRae A et al. . DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol 2015; 16:25.
    1. Marioni RE, Shah S, McRae AF et al. . The epigenetic clock is correlated with physical and cognitive fitness in the Lothian Birth Cohort 1936. Int J Epidemiol 2015; pii:dyu277.
    1. Horvath S, Garagnani P, Bacalini M et al. . Accelerated epigenetic aging in down syndrome. Aging Cell 2015; 14:491–5.
    1. Morgello S, Gelman B, Kozlowski P et al. . The National NeuroAIDS Tissue Consortium: a new paradigm in brain banking with an emphasis on infectious disease. Neuropathol Appl Neurobiol 2001; 27:326–35.
    1. Heaton R, Kirson D, Velin R, Grant I, Martin A. The utility of clinical ratings for detecting cognitive change in HIV infection. New York: Oxford University Press, 1994. (Grant I, Martin A, eds. Neuropsychology of HIV infection).
    1. Dana Consortium Clinical confirmation of the American Academy of Neurology algorithm for HIV-1-associated cognitive/motor disorder. The Dana Consortium on Therapy for HIV Dementia and Related Cognitive Disorders. Neurology 1996; 47:1247–53.
    1. Antinori A, Arendt G, Becker JT et al. . Updated research nosology for HIV-associated neurocognitive disorders. Neurology 2007; 69:1789–99.
    1. Soontornniyomkij V, Soontornniyomkij B, Moore DJ et al. . Antioxidant sestrin-2 redistribution to neuronal soma in human immunodeficiency virus-associated neurocognitive disorders. J Neuroimmune Pharmacol 2012; 7:579–90.
    1. Soontornniyomkij V, Risbrough VB, Young JW et al. . Short-term recognition memory impairment is associated with decreased expression of FK506 binding protein 51 in the aged mouse brain. Age (Dordr) 2010; 32:309–22.
    1. Liu Y, Aryee MJ, Padyukov L et al. . Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat Biotech 2013; 31:142–7.
    1. Houseman E, Accomando W, Koestler D et al. . DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics 2012; 13:86.
    1. Accomando WP, Wiencke JK, Houseman EA et al. . Decreased NK cells in patients with head and neck cancer determined in archival DNA. Clin Cancer Res 2012; 18:6147–54.
    1. Reinius LE, Acevedo N, Joerink M et al. . Differential DNA methylation in purified human blood cells: implications for cell lineage and studies on disease susceptibility. PLoS One 2012; 7:e41361.
    1. Zilbauer M, Rayner TF, Clark C et al. . Genome-wide methylation analyses of primary human leukocyte subsets identifies functionally important cell-type-specific hypomethylated regions. Blood 2013; 122:e52–60.
    1. Ibarrondo FJ, Yang OO, Chodon T et al. . Natural killer T cells in advanced melanoma patients treated with tremelimumab. PLoS One 2013; 8:e76829.
    1. Levine AJ, Panos SE, Horvath S. Genetic, transcriptomic, and epigenetic studies of HIV-associated neurocognitive disorder. J Acquir Immune Defic Syndr 2014; 65:481–503.
    1. Boks MP, Mierlo HCV, Rutten BPF et al. . Longitudinal changes of telomere length and epigenetic age related to traumatic stress and post-traumatic stress disorder. Psychoneuroendocrinology 2015; 51:506–12.
    1. Tachiwana H, Shimura M, Nakai-Murakami C et al. . HIV-1 Vpr induces DNA double-strand breaks. Cancer Res 2006; 66:627–31.
    1. Lau A, Swinbank KM, Ahmed PS et al. . Suppression of HIV-1 infection by a small molecule inhibitor of the ATM kinase. Nat Cell Biol 2005; 7:493–500.
    1. Zimmerman ES, Chen J, Andersen JL et al. . Human immunodeficiency virus type 1 Vpr-mediated G2 arrest requires rad17 and Hus1 and induces nuclear BRCA1 and γ-H2AX focus formation. Mol Cell Biol 2004; 24:9286–94.
    1. Nath AA. HIV and aging. J Neurovirol 2012; 18:245–6.

Source: PubMed

3
Předplatit