Increased epigenetic age and granulocyte counts in the blood of Parkinson's disease patients

Steve Horvath, Beate R Ritz, Steve Horvath, Beate R Ritz

Abstract

It has been a long standing hypothesis that blood tissue of PD Parkinson's disease (PD) patients may exhibit signs of accelerated aging. Here we use DNA methylation based biomarkers of aging ("epigenetic clock") to assess the aging rate of blood in two ethnically distinct case-control data sets. Using n=508 Caucasian and n=84 Hispanic blood samples, we assess a) the intrinsic epigenetic age acceleration of blood (IEAA), which is independent of blood cell counts, and b) the extrinsic epigenetic age acceleration rate of blood (EEAA) which is associated with age dependent changes in blood cell counts. Blood of PD subjects exhibits increased age acceleration according to both IEAA (p=0.019) and EEAA (p=6.1 x 10(-3)). We find striking differences in imputed blood cell counts between PD cases and controls. Compared to control subjects, PD subjects contains more granulocytes (p=1.0 x 10(-9) in Caucasians, p=0.00066 in Hispanics) but fewer T helper cells (p=1.4 x 10(-6) in Caucasians, p=0.0024 in Hispanics) and fewer B cells (p=1.6 x 10(-5) in Caucasians, p=4.5 x 10(-5) in Hispanics). Overall, this study shows that the epigenetic age of the immune system is significantly increased in PD patients and that granulocytes play a significant role.

Keywords: DNA methylation; Parkinson's disease; epigenetic clock; epigenetics; granulocyte; neutrophil.

Conflict of interest statement

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1. Epigenetic age analysis of PD
Figure 1. Epigenetic age analysis of PD
(A-C) DNA methylation age (y-axis) versus chronological age (x-axis) in (A) all subjects, (B) Caucasians only, and (C) Hispanics only. Dots corresponds to subjects and are colored by PD disease status (red=PD, black=control). We define three measures of epigenetic age acceleration. (D-F) presents results for the “universal” measure of epigenetic age acceleration, which is defined as residual to a regression line through the control samples, i.e. the vertical distance of a point from the line. By definition, the mean age acceleration in controls is zero. (G-I) The bar plots relate measures of intrinsic epigenetic age acceleration to PD status. This measure is independent of blood cell counts. The fourth row (panels J-L) reports findings for the measure of extrinsic epigenetic age acceleration, which does relate to changes in cell composition. Each bar plot depicts the mean value (y-axis), 1 standard error, and the group size (underneath the bar). The p-value results from a Student T-test.
Figure 2. Levodopa medication (x-axis) versus epigenetic…
Figure 2. Levodopa medication (x-axis) versus epigenetic age acceleration in PD subjects
Each scatter plot depicts the amount of levodopa medication (milligram per day) versus (A,B,C) universal epigenetic age acceleration, (D,E,F) intrinsic epigenetic age acceleration (G,H,I), extrinsic epigenetic age acceleration. The first, second, and third column correspond to all subjects, Caucasians only, and Hispanics only, respectively. Each dot (PD patient) is colored in red for the sake of consistency with Figure 1. The heading of each plot reports a robust correlation coefficient (biweight midcorrelation and a corresponding p-value).
Figure 3. Levodopa medication status versus epigenetic…
Figure 3. Levodopa medication status versus epigenetic age acceleration in PD patients
The first, second, and third column correspond to all subjects, Caucasians only, and Hispanics only, respectively. Levodopa medication status versus (A,B,C) universal epigenetic age acceleration, (D,E,F) intrinsic epigenetic age acceleration (G,H,I), extrinsic epigenetic age acceleration. Each bar plot depicts the mean value (y-axis), 1 standard error, and the group size (underneath the bar). The p-value results from a non-parametric group comparison test (Kruskal Wallis).
Figure 4. Blood cell counts versus PD…
Figure 4. Blood cell counts versus PD status
As indicated in the heading of each panel, the panels alternate between the two data sets. PD status (x-axis) versus (A,B) proportion of cytotoxic CD8+ T cells, (C,D) naïve CD8+ T cell count, (E,F) percentage of exhausted CD8+ T cells (defined as CD8+CD28-CD45RA-), (G,H) proportion of helper CD4+ T cells, (I,J) naïve CD4+ T cell count, (K,L) proportion of natural killer cells, (M,N) proportion of monocytes, (O,P) granulocytes, (Q,R) B cells, (S,T) plasma blasts (activated B cells). The abundance measures of blood cell counts were estimated based on DNA methylation levels using the epigenetic clock software. The y-axis of (E,F) reports a percentage, that of (C,D,I,J) a cell counts but it is best to interpret these measures as ordinal abundance measures. The y-axis of the other panels reports estimated proportions based on the Houseman method [45]. Each bar plot depicts the mean value (y-axis), 1 standard error, and the group size (underneath the bar). The p-value results from a non-parametric group comparison test (Kruskal Wallis).
Figure 5. Amount of medication (x-axis) versus…
Figure 5. Amount of medication (x-axis) versus epigenetic age acceleration in PD subjects
As indicated in the heading of each panel, the panels alternate between the two data sets. PD status (x-axis) versus (A,B) proportion of CD8+ T cells, (C,D) naïve CD8+ T cell count, (E,F) exhausted CD4+T cell counts (defined as CD8+CD28-CD45RA-), (G,H) proportion of CD4+ T cells, (I,J) naïve CD4 T cell count, (K,L) proportion of natural killer cells, (M,N) proportion of monocytes, (O,P) granulocytes, (Q,R) B cells, (S,T) plasma blasts (activated B cells). All cell types were estimated based on DNA methylation levels as described in Methods. The heading of each plot reports a robust correlation coefficient (biweight midcorrelation and a corresponding p-value).
Figure 6. Medication status versus blood cell…
Figure 6. Medication status versus blood cell counts in PD patients
As indicated in the heading of each panel, the panels alternate between the two data sets. Levodopa medication status (x-axis) versus (A,B) proportion of CD8+ T cells, (C,D) naïve CD8+ T cell count, (E,F) exhausted CD+T cell counts (defined as CD8+CD28-CD45RA-), (G,H) proportion of CD4+ T cells, (I,J) naïve CD4+ T cell count, (K,L) proportion of natural killer cells, (M,N) proportion of monocytes, (O,P) granulocytes, (Q,R)B cells, (S,T) plasma blasts (activated B cells). All cell types were estimated based on DNA methylation levels as described in Methods. Each bar plot depicts the mean value (y-axis), 1 standard error, and the group size (underneath the bar). The p-value results from a non-parametric group comparison test (Kruskal Wallis).

References

    1. Schrag A, Horsfall L, Walters K, Noyce A, Petersen I. Prediagnostic presentations of Parkinson's disease in primary care: a case-control study. The Lancet Neurology. 14:57–64.
    1. Schapira AH, Olanow CW, Greenamyre JT, Bezard E. Slowing of neurodegeneration in Parkinson's disease and Huntington's disease: future therapeutic perspectives. Lancet. 2014;384:545–555.
    1. Hirsch EC, Hunot S. Neuroinflammation in Parkinson's disease: a target for neuroprotection? Lancet Neurol. 2009;8:382–397.
    1. Marttila RJ, Eskola J, Soppi E, Rinne UK. Immune functions in Parkinson's disease lymphocyte subsets, concanavalin A-induced suppressor cell activity and in vitro immunoglobulin production. J Neurol Sci. 1985;69:121–131.
    1. Guan JZ, Maeda T, Sugano M, Oyama J, Higuchi Y, Suzuki T, Makino N. A percentage analysis of the telomere length in Parkinson's disease patients. J Gerontol A Biol Sci Med Sci. 2008;63:467–473.
    1. Wang H, Chen H, Gao X, McGrath M, Deer D, De Vivo I, Schwarzschild MA, Ascherio A. Telomere length and risk of Parkinson's disease. Mov Disord. 2008;23:302–305.
    1. Maeda T, Guan JZ, Oyama J, Higuchi Y, Makino N. Aging-associated alteration of subtelomeric methylation in Parkinson's disease. J Gerontol A Biol Sci Med Sci. 2009;64:949–955.
    1. Eerola J, Kananen L, Manninen K, Hellstrom O, Tienari PJ, Hovatta I. No evidence for shorter leukocyte telomere length in Parkinson's disease patients. J Gerontol A Biol Sci Med Sci. 2010;65:1181–1184.
    1. Hudson G, Faini D, Stutt A, Eccles M, Robinson L, Burn DJ, Chinnery PF. No evidence of substantia nigra telomere shortening in Parkinson's disease. Neurobiol Aging. 2011;32:2107, e2103–2105.
    1. Watfa G, Dragonas C, Brosche T, Dittrich R, Sieber CC, Alecu C, Benetos A, Nzietchueng R. Study of telomere length and different markers of oxidative stress in patients with Parkinson's disease. J Nutr Health Aging. 2011;15:277–281.
    1. Maeda T, Guan JZ, Koyanagi M, Higuchi Y, Makino N. Aging-associated alteration of telomere length and subtelomeric status in female patients with Parkinson's disease. J Neurogenet. 2012;26:245–251.
    1. Schurks M, Buring J, Dushkes R, Gaziano JM, Zee RY, Kurth T. Telomere length and Parkinson's disease in men: a nested case-control study. Eur J Neurol. 2014;21:93–99.
    1. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan J-B Y. Genome-wide methylation profiles reveal quantitative views of human aging rates. Molecular cell. 2013;49:359–367.
    1. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(R115)
    1. Weidner CI, Lin Q, Koch CM, Eisele L, Beier F, Ziegler P, Bauerschlag DO, Jockel KH, Erbel R, Muhleisen TW, Zenke M, Brummendorf TH, Wagner W. Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol. 2014;15:R24.
    1. Spiers H, Hannon E, Schalkwyk LC, Smith R, Wong CC, O'Donovan MC, Bray NJ, Mill J. Methylomic trajectories across human fetal brain development. Genome research. 2015;25:338–352.
    1. Marioni R, Shah S, McRae A, Chen B, Colicino E, Harris S, Gibson J, Henders A, Redmond P, Cox S, Pattie A, Corley J, Murphy L, 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, Ritchie SJ, Muniz-Terrera G, Harris SE, Gibson J, Redmond P, Cox SR, Pattie A. The epigenetic clock is correlated with physical and cognitive fitness in the Lothian Birth Cohort 1936. International journal of epidemiology. 2015 dyu277.
    1. Horvath S, Erhart W, Brosch M, Ammerpohl O, von Schönfels W, Ahrens M, Heits N, Bell JT, Tsai P-C, Spector TD, Deloukas P, Siebert R, Sipos B, et al. Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci U S A. 2014;111:15538–15543.
    1. Horvath S, Garagnani P, Bacalini M, Pirazzini C, Salvioli S, Gentilini D, DiBlasio A, Giuliani C, Tung S, Vinters H, Franceschi C. Accelerated Epigenetic Aging in Down Syndrome. Aging Cell. 2015;14:491–495.
    1. Horvath S, Levine AJ. HIV-1 infection accelerates age according to the epigenetic clock. J Infect Dis. 2015;212:1563–1573.
    1. Levine ME, Hosgood HD, Chen B, Absher D, Assimes T, Horvath S. DNA methylation age of blood predicts future onset of lung cancer in the women's health initiative. Aging (Albany NY) 2015;7:690–700.
    1. Horvath S, Mah V, Lu AT, Woo JS, Choi OW, Jasinska AJ, Riancho JA, Tung S, Coles NS, Braun J, Vinters HV, Coles LS. The cerebellum ages slowly according to the epigenetic clock. Aging (Albany NY) 2015;7:294–306.
    1. Walker RF, Liu JS, Peters BA, Ritz BR, Wu T, Ophoff RA, Horvath S. Epigenetic age analysis of children who seem to evade aging. Aging (Albany NY) 2015;7:334–339.
    1. Lin Q, Wagner W. Epigenetic Aging Signatures Are Coherently Modified in Cancer. PLoS Genet. 2015;11:e1005334.
    1. Kananen L, Nevalainen T, Jylhävä J, Marttila S, Hervonen A, Jylhä M, Hurme M. Cytomegalovirus infection accelerates epigenetic aging. Experimental Gerontology. 2015;72:227–229.
    1. Miller GE, Yu T, Chen E, Brody GH. Self-control forecasts better psychosocial outcomes but faster epigenetic aging in low-SES youth. Proceedings of the National Academy of Sciences. 2015;112:10325–10330.
    1. Costello S, Cockburn M, Bronstein J, Zhang X, Ritz B. Parkinson's Disease and Residential Exposure to Maneb and Paraquat From Agricultural Applications in the Central Valley of California. American Journal of Epidemiology. 2009;169:919–926.
    1. Ritz B, Lee PC, Lassen CF, Arah OA. Parkinson disease and smoking revisited: ease of quitting is an early sign of the disease. Neurology. 2014;83:1396–1402.
    1. Wang A, Cockburn M, Ly TT, Bronstein JM, Ritz B. The association between ambient exposure to organophosphates and Parkinson's disease risk. Occup Environ Med. 2014;71:275–281.
    1. Fagnoni FF, Vescovini R, Mazzola M, Bologna G, Nigro E, Lavagetto G, Franceschi C, Passeri M, Sansoni P. Expansion of cytotoxic CD8+ CD28-T cells in healthy ageing people, including centenarians. Immunology. 1996;88:501–507.
    1. Fagnoni FF, Vescovini R, Passeri G, Bologna G, Pedrazzoni M, Lavagetto G, Casti A, Franceschi C, Passeri M, Sansoni P. Shortage of circulating naive CD8+ T cells provides new insights on immunodeficiency in aging. Blood. 2000;95:2860–2868.
    1. Gruver AL, Hudson LL, Sempowski GD. Immunosenescence of ageing. The Journal of Pathology. 2007;211:144–156.
    1. Bas J, Calopa M, Mestre M, Mollevi DG, Cutillas B, Ambrosio S, Buendia E. Lymphocyte populations in Parkinson's disease and in rat models of parkinsonism. J Neuroimmunol. 2001;113:146–152.
    1. Calopa M, Bas J, Callen A, Mestre M. Apoptosis of peripheral blood lymphocytes in Parkinson patients. Neurobiol Dis. 2010;38:1–7.
    1. Niwa F, Kuriyama N, Nakagawa M, Imanishi J. Effects of peripheral lymphocyte subpopulations and the clinical correlation with Parkinson's disease. Geriatr Gerontol Int. 2012;12:102–107.
    1. Saunders JA, Estes KA, Kosloski LM, Allen HE, Dempsey KM, Torres-Russotto DR, Meza JL, Santamaria PM, Bertoni JM, Murman DL, Ali HH, Standaert DG, Mosley RL, et al. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson's disease. Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology. 2012;7:927–938.
    1. Stevens CH, Rowe D, Morel-Kopp MC, Orr C, Russell T, Ranola M, Ward C, Halliday GM. Reduced T helper and B lymphocytes in Parkinson's disease. J Neuroimmunol. 2012;252:95–99.
    1. Akil E, Bulut A, Kaplan I, Ozdemir HH, Arslan D, Aluclu MU. The increase of carcinoembryonic antigen (CEA), high-sensitivity C-reactive protein, and neutrophil/lymphocyte ratio in Parkinson's disease. Neurol Sci. 2015;36:423–428.
    1. Ji KA, Eu MY, Kang SH, Gwag BJ, Jou I, Joe EH. Differential neutrophil infiltration contributes to regional differences in brain inflammation in the substantia nigra pars compacta and cortex. Glia. 2008;56:1039–1047.
    1. Jowaed A, Schmitt I, Kaut O, Wüllner U. Methylation Regulates Alpha-Synuclein Expression and Is Decreased in Parkinson's Disease Patients' Brains. The Journal of Neuroscience. 2010;30:6355–6359.
    1. Matsumoto L, Takuma H, Tamaoka A, Kurisaki H, Date H, Tsuji S, Iwata A. CpG Demethylation Enhances Alpha-Synuclein Expression and Affects the Pathogenesis of Parkinson's Disease. PLoS ONE. 2010;5:e15522.
    1. Desplats P, Spencer B, Coffee E, Patel P, Michael S, Patrick C, Adame A, Rockenstein E, Masliah E. α-Synuclein Sequesters Dnmt1 from the Nucleus. Journal of Biological Chemistry. 2011;286:9031–9037.
    1. Klemera P, Doubal S. A new approach to the concept and computation of biological age. Mech Ageing Dev. 2006;127:240–248.
    1. Houseman E, Accomando W, Koestler D, Christensen B, Marsit C, Nelson H, Wiencke J, Kelsey K. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012;13:86.

Source: PubMed

3
Prenumerera