A prospective study of leukocyte telomere length and risk of type 2 diabetes in postmenopausal women

Nai-Chieh Y You, Brian H Chen, Yiqing Song, XuYang Lu, Yilin Chen, JoAnn E Manson, Mo Kang, Barbara V Howard, Karen L Margolis, J David Curb, Lawrence S Phillips, Marcia L Stefanick, Lesley F Tinker, Simin Liu, Nai-Chieh Y You, Brian H Chen, Yiqing Song, XuYang Lu, Yilin Chen, JoAnn E Manson, Mo Kang, Barbara V Howard, Karen L Margolis, J David Curb, Lawrence S Phillips, Marcia L Stefanick, Lesley F Tinker, Simin Liu

Abstract

Telomere length (TL) has been implicated in the pathogenesis of age-related disorders. However, there are no prospective studies directly investigating the role of TL and relevant genes in diabetes development. In the multiethnic Women's Health Initiative, we identified 1,675 incident diabetes case participants in 6 years of follow-up and 2,382 control participants matched by age, ethnicity, clinical center, time of blood draw, and follow-up duration. Leukocyte TL at baseline was measured using quantitative PCR, and Mendelian randomization analysis was conducted to test whether TL is causally associated with diabetes risk. After adjustment for matching and known diabetes risk factors, odds ratios per 1-kilobase increment were 1.00 (95% CI 0.90-1.11) in whites, 0.95 (0.85-1.06) in blacks, 0.96 (0.79-1.17) in Hispanics, and 0.88 (0.70-1.10) in Asians. Of the 80 single nucleotide polymorphisms (SNPs) in nine genes involved in telomere regulation, 14 SNPs were predictive of TL, but none were significantly associated with diabetes risk. Using ethnicity-specific SNPs as randomization instruments, we observed no statistically significant association between TL and diabetes risk (P = 0.52). Although leukocyte TL was weakly associated with diabetes risk, this association was not independent of known risk factors. These prospective findings indicate limited clinical utility of TL in diabetes risk stratification among postmenopausal women.

Figures

FIG. 1.
FIG. 1.
TL (kb) by clinical diabetes status and race/ethnicity. The boxplots show the mean (+), median, and interquartile range. The vertical line indicates the minimum and maximum value of TL in each group. (A high-quality color representation of this figure is available in the online issue.)
FIG. 2.
FIG. 2.
TL by GRS according to race/ethnicity. The boxplots show the mean (◇), median, and interquartile range. The vertical line indicates the minimum and maximum value >1.5 times the upper quartile of TL in each group. Outliers (○) are displayed when the individual TL is >1.5 times the interquartile range of the upper quartile value. GRS represents the unweighted sum of alleles from the selected SNPs. The selected SNPs for each of the ethnic groups are as follows: 1) whites, rs34368910, rs4888444, rs4975605, rs938886, and rs12880583; 2) blacks, rs872072, rs938886, rs1713458, and rs4387287; 3) Hispanics, rs35276863, rs729421, rs11972248, rs4635969, rs2853669, rs2736098, and rs2853676; and 4) Asians, rs11556640 and rs2297613. (A high-quality color representation of this figure is available in the online issue.)

References

    1. von Zglinicki T. Role of oxidative stress in telomere length regulation and replicative senescence. Ann N Y Acad Sci 2000;908:99–110
    1. von Zglinicki T. Oxidative stress shortens telomeres. Trends Biochem Sci 2002;27:339–344
    1. Cawthon RM, Smith KR, O’Brien E, Sivatchenko A, Kerber RA. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 2003;361:393–395
    1. Vasa-Nicotera M, Brouilette S, Mangino M, et al. Mapping of a major locus that determines telomere length in humans. Am J Hum Genet 2005;76:147–151
    1. Takubo K, Izumiyama-Shimomura N, Honma N, et al. Telomere lengths are characteristic in each human individual. Exp Gerontol 2002;37:523–531
    1. Slagboom PE, Droog S, Boomsma DI. Genetic determination of telomere size in humans: a twin study of three age groups. Am J Hum Genet 1994;55:876–882
    1. Adaikalakoteswari A, Balasubramanyam M, Mohan V. Telomere shortening occurs in Asian Indian type 2 diabetic patients. Diabet Med 2005;22:1151–1156
    1. Sampson MJ, Winterbone MS, Hughes JC, Dozio N, Hughes DA. Monocyte telomere shortening and oxidative DNA damage in type 2 diabetes. Diabetes Care 2006;29:283–289
    1. Fitzpatrick AL, Kronmal RA, Gardner JP, et al. Leukocyte telomere length and cardiovascular disease in the cardiovascular health study. Am J Epidemiol 2007;165:14–21
    1. Demissie S, Levy D, Benjamin EJ, et al. Insulin resistance, oxidative stress, hypertension, and leukocyte telomere length in men from the Framingham Heart Study. Aging Cell 2006;5:325–330
    1. Nakajima T, Moriguchi M, Katagishi T, et al. Premature telomere shortening and impaired regenerative response in hepatocytes of individuals with NAFLD. Liver Int 2006;26:23–31
    1. Aviv A, Valdes A, Gardner JP, Swaminathan R, Kimura M, Spector TD. Menopause modifies the association of leukocyte telomere length with insulin resistance and inflammation. J Clin Endocrinol Metab 2006;91:635–640
    1. Gardner JP, Li S, Srinivasan SR, et al. Rise in insulin resistance is associated with escalated telomere attrition. Circulation 2005;111:2171–2177
    1. Ding EL, Song Y, Manson JE, et al. Sex hormone-binding globulin and risk of type 2 diabetes in women and men. N Engl J Med 2009;361:1152–1163
    1. Liu S, Tinker L, Song Y, et al. A prospective study of inflammatory cytokines and diabetes mellitus in a multiethnic cohort of postmenopausal women. Arch Intern Med 2007;167:1676–1685
    1. Song Y, Manson JE, Tinker L, et al. Insulin sensitivity and insulin secretion determined by homeostasis model assessment and risk of diabetes in a multiethnic cohort of women: the Women’s Health Initiative Observational Study. Diabetes Care 2007;30:1747–1752
    1. Song Y, Manson JE, Tinker L, et al. Circulating levels of endothelial adhesion molecules and risk of diabetes in an ethnically diverse cohort of women. Diabetes 2007;56:1898–1904
    1. The Women’s Health Initiative Study Group Design of the Women’s Health Initiative clinical trial and observational study. Control Clin Trials 1998;19:61–109
    1. O’Callaghan N, Dhillon V, Thomas P, Fenech M. A quantitative real-time PCR method for absolute telomere length. Biotechniques 2008;44:807–809
    1. Hao K, Liu S, Niu T. A sparse marker extension tree algorithm for selecting the best set of haplotype tagging single nucleotide polymorphisms. Genet Epidemiol 2005;29:336–352
    1. Lee CC, You NC, Song Y, et al. Relation of genetic variation in the gene coding for C-reactive protein with its plasma protein concentrations: findings from the Women’s Health Initiative Observational Cohort. Clin Chem 2009;55:351–360
    1. International HapMap Consortium A haplotype map of the human genome. Nature 2005;437:1299–1320
    1. Wang L, Liu S, Niu T, Xu X. SNPHunter: a bioinformatic software for single nucleotide polymorphism data acquisition and management. BMC Bioinformatics 2005;6:60.
    1. Levy D, Neuhausen SL, Hunt SC, et al. Genome-wide association identifies OBFC1 as a locus involved in human leukocyte telomere biology. Proc Natl Acad Sci USA 2010;107:9293–9298
    1. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res 2001;125:279–284
    1. Thomas DC, Conti DV. Commentary: the concept of “Mendelian Randomization”. Int J Epidemiol 2004;33:21–25
    1. de Lange T. Shelterin: the protein complex that shapes and safeguards human telomeres. Genes Dev 2005;19:2100–2110
    1. Wu X, Zhao H, Suk R, Christiani DC. Genetic susceptibility to tobacco-related cancer. Oncogene 2004;23:6500–6523
    1. van Steensel B, de Lange T. Control of telomere length by the human telomeric protein TRF1. Nature 1997;385:740–743
    1. van Steensel B, Smogorzewska A, de Lange T. TRF2 protects human telomeres from end-to-end fusions. Cell 1998;92:401–413
    1. Lei M, Podell ER, Cech TR. Structure of human POT1 bound to telomeric single-stranded DNA provides a model for chromosome end-protection. Nat Struct Mol Biol 2004;11:1223–1229
    1. Colgin LM, Baran K, Baumann P, Cech TR, Reddel RR. Human POT1 facilitates telomere elongation by telomerase. Curr Biol 2003;13:942–946
    1. Kelleher C, Kurth I, Lingner J. Human protection of telomeres 1 (POT1) is a negative regulator of telomerase activity in vitro. Mol Cell Biol 2005;25:808–818
    1. Loayza D, De Lange T. POT1 as a terminal transducer of TRF1 telomere length control. Nature 2003;423:1013–1018
    1. O’Connor MS, Safari A, Xin H, Liu D, Songyang Z. A critical role for TPP1 and TIN2 interaction in high-order telomeric complex assembly. Proc Natl Acad Sci USA 2006;103:11874–11879
    1. Liu D, Safari A, O’Connor MS, et al. PTOP interacts with POT1 and regulates its localization to telomeres. Nat Cell Biol 2004;6:673–680
    1. Serrano AL, Andrés V. Telomeres and cardiovascular disease: does size matter? Circ Res 2004;94:575–584
    1. Codd V, Mangino M, van der Harst P, et al. Wellcome Trust Case Control Consortium Common variants near TERC are associated with mean telomere length. Nat Genet 2010;42:197–199
    1. Jeanclos E, Krolewski A, Skurnick J, et al. Shortened telomere length in white blood cells of patients with IDDM. Diabetes 1998;47:482–486
    1. Shen Q, Zhao X, Yu L, et al. Association of leukocyte telomere length with type 2 diabetes in mainland chinese populations. J Clin Endocrinol Metab 2012;97:1371–1374
    1. Monickaraj F, Aravind S, Gokulakrishnan K, et al. Accelerated aging as evidenced by increased telomere shortening and mitochondrial DNA depletion in patients with type 2 diabetes. Mol Cell Biochem 13 March 2012 [Epub ahead of print]
    1. Allsopp RC, Vaziri H, Patterson C, et al. Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci USA 1992;89:10114–10118
    1. Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature 1990;345:458–460
    1. Pierce BL, Ahsan H, Vanderweele TJ. Power and instrument strength requirements for Mendelian randomization studies using multiple genetic variants. Int J Epidemiol 2011;40:740–752

Source: PubMed

3
Abonnieren