Developmental pathways to adiposity begin before birth and are influenced by genotype, prenatal environment and epigenome

Xinyi Lin, Ives Yubin Lim, Yonghui Wu, Ai Ling Teh, Li Chen, Izzuddin M Aris, Shu E Soh, Mya Thway Tint, Julia L MacIsaac, Alexander M Morin, Fabian Yap, Kok Hian Tan, Seang Mei Saw, Michael S Kobor, Michael J Meaney, Keith M Godfrey, Yap Seng Chong, Joanna D Holbrook, Yung Seng Lee, Peter D Gluckman, Neerja Karnani, GUSTO study group, Pratibha Agarwal, Arijit Biswas, Choon Looi Bong, Birit F P Broekman, Shirong Cai, Jerry Kok Yen Chan, Yiong Huak Chan, Cornelia Yin Ing Chee, Helen Chen, Yin Bun Cheung, Amutha Chinnadurai, Chai Kiat Chng, Mary Foong-Fong Chong, Yap-Seng Chong, Shang Chee Chong, Mei Chien Chua, Doris Fok, Marielle V Fortier, Peter D Gluckman, Keith M Godfrey, Anne Eng Neo Goh, Yam Thiam Daniel Goh, Joshua J Gooley, Wee Meng Han, Mark Hanson, Christiani Jeyakumar Henry, Joanna D Holbrook, Chin-Ying Hsu, Neerja Karnani, Jeevesh Kapur, Kenneth Kwek, Ivy Yee-Man Lau, Bee Wah Lee, Yung Seng Lee, Ngee Lek, Sok Bee Lim, Iliana Magiati, Lourdes Mary Daniel, Michael Meaney, Cheryl Ngo, Krishnamoorthy Niduvaje, Wei Wei Pang, Anqi Qiu, Boon Long Quah, Victor Samuel Rajadurai, Mary Rauff, Salome A Rebello, Jenny L Richmond, Anne Rifkin-Graboi, Seang-Mei Saw, Lynette Pei-Chi Shek, Allan Sheppard, Borys Shuter, Leher Singh, Shu-E Soh, Walter Stunkel, Lin Lin Su, Kok Hian Tan, Oon Hoe Teoh, Mya Thway Tint, Hugo P S van Bever, Rob M van Dam, Inez Bik Yun Wong, P C Wong, Fabian Yap, George Seow Heong Yeo, Xinyi Lin, Ives Yubin Lim, Yonghui Wu, Ai Ling Teh, Li Chen, Izzuddin M Aris, Shu E Soh, Mya Thway Tint, Julia L MacIsaac, Alexander M Morin, Fabian Yap, Kok Hian Tan, Seang Mei Saw, Michael S Kobor, Michael J Meaney, Keith M Godfrey, Yap Seng Chong, Joanna D Holbrook, Yung Seng Lee, Peter D Gluckman, Neerja Karnani, GUSTO study group, Pratibha Agarwal, Arijit Biswas, Choon Looi Bong, Birit F P Broekman, Shirong Cai, Jerry Kok Yen Chan, Yiong Huak Chan, Cornelia Yin Ing Chee, Helen Chen, Yin Bun Cheung, Amutha Chinnadurai, Chai Kiat Chng, Mary Foong-Fong Chong, Yap-Seng Chong, Shang Chee Chong, Mei Chien Chua, Doris Fok, Marielle V Fortier, Peter D Gluckman, Keith M Godfrey, Anne Eng Neo Goh, Yam Thiam Daniel Goh, Joshua J Gooley, Wee Meng Han, Mark Hanson, Christiani Jeyakumar Henry, Joanna D Holbrook, Chin-Ying Hsu, Neerja Karnani, Jeevesh Kapur, Kenneth Kwek, Ivy Yee-Man Lau, Bee Wah Lee, Yung Seng Lee, Ngee Lek, Sok Bee Lim, Iliana Magiati, Lourdes Mary Daniel, Michael Meaney, Cheryl Ngo, Krishnamoorthy Niduvaje, Wei Wei Pang, Anqi Qiu, Boon Long Quah, Victor Samuel Rajadurai, Mary Rauff, Salome A Rebello, Jenny L Richmond, Anne Rifkin-Graboi, Seang-Mei Saw, Lynette Pei-Chi Shek, Allan Sheppard, Borys Shuter, Leher Singh, Shu-E Soh, Walter Stunkel, Lin Lin Su, Kok Hian Tan, Oon Hoe Teoh, Mya Thway Tint, Hugo P S van Bever, Rob M van Dam, Inez Bik Yun Wong, P C Wong, Fabian Yap, George Seow Heong Yeo

Abstract

Background: Obesity is an escalating health problem worldwide, and hence the causes underlying its development are of primary importance to public health. There is growing evidence that suboptimal intrauterine environment can perturb the metabolic programing of the growing fetus, thereby increasing the risk of developing obesity in later life. However, the link between early exposures in the womb, genetic susceptibility, and perturbed epigenome on metabolic health is not well understood. In this study, we shed more light on this aspect by performing a comprehensive analysis on the effects of variation in prenatal environment, neonatal methylome, and genotype on birth weight and adiposity in early childhood.

Methods: In a prospective mother-offspring cohort (N = 987), we interrogated the effects of 30 variables that influence the prenatal environment, umbilical cord DNA methylation, and genotype on offspring weight and adiposity, over the period from birth to 48 months. This is an interim analysis on an ongoing cohort study.

Results: Eleven of 30 prenatal environments, including maternal adiposity, smoking, blood glucose and plasma unsaturated fatty acid levels, were associated with birth weight. Polygenic risk scores derived from genetic association studies on adult adiposity were also associated with birth weight and child adiposity, indicating an overlap between the genetic pathways influencing metabolic health in early and later life. Neonatal methylation markers from seven gene loci (ANK3, CDKN2B, CACNA1G, IGDCC4, P4HA3, ZNF423 and MIRLET7BHG) were significantly associated with birth weight, with a subset of these in genes previously implicated in metabolic pathways in humans and in animal models. Methylation levels at three of seven birth weight-linked loci showed significant association with prenatal environment, but none were affected by polygenic risk score. Six of these birth weight-linked loci continued to show a longitudinal association with offspring size and/or adiposity in early childhood.

Conclusions: This study provides further evidence that developmental pathways to adiposity begin before birth and are influenced by environmental, genetic and epigenetic factors. These pathways can have a lasting effect on offspring size, adiposity and future metabolic outcomes, and offer new opportunities for risk stratification and prevention of obesity.

Clinical trial registration: This birth cohort is a prospective observational study, designed to study the developmental origins of health and disease, and was retrospectively registered on 1 July 2010 under the identifier NCT01174875 .

Keywords: Birth weight; DNA methylation; Epigenome-wide association study; Offspring adiposity; Prenatal environment.

Figures

Fig. 1
Fig. 1
Prenatal environment influences on birth weight. a Scatterplots of birth weight (vertical axis) against significantly associated continuous prenatal environment variables (horizontal axis). b Boxplots of birth weight (vertical axis) against significantly associated binary prenatal environment variables (horizontal axis). c Univariate association between birth weight and each significantly associated prenatal environment variable, adjusted for infant sex, ethnicity and gestational age. Point estimates (height of bars) and 95% confidence intervals (top and bottom whiskers), show percentage change in birth weight for two standard deviations increase in continuous prenatal environment variable, or for comparing the two categories of binary prenatal environment variables. d Multivariate association between birth weight and significantly associated prenatal environment variables, adjusted for infant sex, ethnicity, gestational age and for each other. Point estimates (height of bars) and 95% confidence intervals (top and bottom whiskers), show percentage change in birth weight, for two standard deviations increase in a continuous prenatal environment variable, or for comparing the two categories of binary prenatal environment variables
Fig. 2
Fig. 2
Genetic influences on birth weight: Associations of child weight (a and b) and body mass index (c and d) at different time points with best-fit polygenic risk score (PRS). Best-fit PRS for Chinese, Malay and Indian ethnic groups used clumping P value thresholds pT = 0.5, 0.1 and 10–4, respectively. PRS was standardised to mean zero and unit variance within each ethnic group. Left panel (a and c) shows point estimates (height of bars) and 95% confidence intervals (top and bottom whiskers), for percentage change in child outcome, for a 2 SD increase in PRS, adjusted for child sex, gestational age and ethnicity. Analysis was done by linear regression of log-transformed child anthropometric outcome at each time point against PRS, adjusted for child sex, gestational age and ethnicity. Right panel (b and d) shows scatterplot of standardised (mean zero and unit variance) log-transformed child outcome (vertical axis) against PRS (horizontal axis)
Fig. 3
Fig. 3
Influence of prenatal environment on methylome at birth. a Associations of DNA methylation at birth with prenatal environment. Colour in heatmap represents regression coefficients for associations between methylation and each prenatal environment variable. Each row represents a CpG and each column represents a prenatal environment variable. With increasing magnitudes, colour changes from white to red (for negative coefficients) or from white to blue (for positive coefficients). Asterisks within each square represent P values for associations between methylation and each prenatal environment variable (P < 5 × 10–8 is represented with eight asterisks, 5 × 10–8 ≤ P < 5 × 10–7 is represented with seven asterisks, 5 × 10–3 ≤ P < 5 × 10–2 is represented with two asterisks, P ≥ 5 × 10–2 is represented with a blank square). Analysis was done by linear regression of methylation at each CpG site against each prenatal environment variable, adjusted for child sex, gestational age, ethnicity, cellular proportions and interactions between ethnicity and cellular proportions. Regression coefficients and P values are reported as an increase in percent methylation for a 2 SD increase in continuous prenatal environment variable, or for comparing the two categories of binary prenatal environment variables. b Flow chart summarises associations between birth weight, methylation and prenatal environment for three CpGs (three loci) influenced by the prenatal environment. A CpG was defined to be influenced by the prenatal environment if the most significant association between the CpG and prenatal environment attained a P value of < 5 × 10–4 the Bonferroni threshold to maintain a family-wise Type 1 error rate of 0.05 across approximately 100 tests (8 CpGs x 11 prenatal environment variables). Directions in arrows indicate temporal sequence, measurements obtained at the same time are indicated with two-headed arrows
Fig. 4
Fig. 4
Influence of methylome at birth on adiposity outcomes in early childhood: Associations of child weight (a) and body mass index (b) at different time points with DNA methylation at birth. Colour in heatmap represents regression coefficients for associations between child anthropometric outcome and methylation. Each row represents a CpG and each column represents a time point. With increasing magnitudes, colour changes from white to red (for negative coefficients) or from white to grey (for positive coefficients). Asterisks within each square represent P values for associations between child anthropometric outcome and methylation (P < 5 × 10–8 is represented with eight asterisks, 5 × 10–8 ≤ P < 5 × 10–7 is represented with seven asterisks, 5 × 10–3 ≤ P < 5 × 10–2 is represented with two asterisks, P ≥ 5 × 10–2 is represented with a blank square). Analysis was done by linear regression of log-transformed child anthropometric outcome at each time point against methylation at each CpG site, adjusted for child sex, gestational age, ethnicity, cellular proportions and interactions between ethnicity and cellular proportions. Regression coefficients and P values are reported as percentage change in child anthropometric outcome for 10% increase in percent methylation

References

    1. Stein Z, Susser M, Saenger G, Marolla F. Famine and Human Development. The Dutch Hunger Winter of 1944-1945. New York: Oxford University Press; 1975.
    1. Desai M, Hales CN. Role of fetal and infant growth in programming metabolism in later life. Biol Rev. 1997;72:329–348. doi: 10.1017/S0006323196005026.
    1. Gluckman PD, Hanson M, Zimmet P, Forrester T. Losing the war against obesity: the need for a developmental perspective. Sci Transl Med. 2011;3(93):93cm19. doi: 10.1126/scitranslmed.3002554.
    1. Godfrey KM, Reynolds RM, Prescott SL, Nyirenda M, Jaddoe VW, Eriksson JG, Broekman BF. Influence of maternal obesity on the long-term health of offspring. Lancet Diabetes Endocrinol. 2017;5(1):53–64. doi: 10.1016/S2213-8587(16)30107-3.
    1. Forsen T, Eriksson JG, Tuomilehto J, Teramo K, Osmond C, Barker DJ. Mother’s weight in pregnancy and coronary heart disease in a cohort of Finnish men: follow up study. BMJ. 1997;315(7112):837–840. doi: 10.1136/bmj.315.7112.837.
    1. Gluckman PD, Hanson MA, Buklijas T, Low FM, Beedle AS. Epigenetic mechanisms that underpin metabolic and cardiovascular diseases. Nat Rev Endocrinol. 2009;5(7):401–408. doi: 10.1038/nrendo.2009.102.
    1. Lawlor DA, Relton C, Sattar N, Nelson SM. Maternal adiposity - a determinant of perinatal and offspring outcomes? Nat Rev Endocrinol. 2012;8(11):679–688. doi: 10.1038/nrendo.2012.176.
    1. Lillycrop KA, Burdge GC. Epigenetic changes in early life and future risk of obesity. Int J Obes (Lond) 2011;35(1):72–83. doi: 10.1038/ijo.2010.122.
    1. Fraser HB, Lam LL, Neumann SM, Kobor MS. Population-specificity of human DNA methylation. Genome Biol. 2012;13(2):R8. doi: 10.1186/gb-2012-13-2-r8.
    1. Gibbs JR, van der Brug MP, Hernandez DG, Traynor BJ, Nalls MA, Lai SL, Arepalli S, Dillman A, Rafferty IP, Troncoso J, et al. Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain. PLoS Genet. 2010;6(5):e1000952. doi: 10.1371/journal.pgen.1000952.
    1. Bell JT, Pai AA, Pickrell JK, Gaffney DJ, Pique-Regi R, Degner JF, Gilad Y, Pritchard JK. DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol. 2011;12(1):R10. doi: 10.1186/gb-2011-12-1-r10.
    1. Tobi EW, Lumey LH, Talens RP, Kremer D, Putter H, Stein AD, Slagboom PE, Heijmans BT. DNA methylation differences after exposure to prenatal famine are common and timing- and sex-specific. Hum Mol Genet. 2009;18(21):4046–4053. doi: 10.1093/hmg/ddp353.
    1. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, Slagboom PE, Lumey LH. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A. 2008;105(44):17046–17049. doi: 10.1073/pnas.0806560105.
    1. Teh AL, Pan H, Chen L, Ong ML, Dogra S, Wong J, MacIsaac JL, Mah SM, McEwen LM, Saw SM, et al. The effect of genotype and in utero environment on interindividual variation in neonate DNA methylomes. Genome Res. 2014;24(7):1064–1074. doi: 10.1101/gr.171439.113.
    1. Rakyan VK, Down TA, Balding DJ, Beck S. Epigenome-wide association studies for common human diseases. Nat Rev Genet. 2011;12(8):529–541. doi: 10.1038/nrg3000.
    1. Hanson MA, Gluckman PD. Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiol Rev. 2014;94(4):1027–1076. doi: 10.1152/physrev.00029.2013.
    1. Kramer MS. Determinants of low birth weight: methodological assessment and meta-analysis. Bull World Health Organ. 1987;65(5):663–737.
    1. Bulik-Sullivan B, Finucane HK, Anttila V, Gusev A, Day FR, Loh PR, Duncan L, Perry JR, Patterson N, Robinson EB, et al. An atlas of genetic correlations across human diseases and traits. Nat Genet. 2015;47(11):1236–1241. doi: 10.1038/ng.3406.
    1. Engel SM, Joubert BR, Wu MC, Olshan AF, Haberg SE, Ueland PM, Nystad W, Nilsen RM, Vollset SE, Peddada SD, et al. Neonatal genome-wide methylation patterns in relation to birth weight in the Norwegian Mother and Child Cohort. Am J Epidemiol. 2014;179(7):834–842. doi: 10.1093/aje/kwt433.
    1. Simpkin AJ, Suderman M, Gaunt TR, Lyttleton O, McArdle WL, Ring SM, Tilling K, Davey Smith G, Relton CL. Longitudinal analysis of DNA methylation associated with birth weight and gestational age. Hum Mol Genet. 2015;24(13):3752–3763.
    1. Sharp GC, Lawlor DA, Richmond RC, Fraser A, Simpkin A, Suderman M, Shihab HA, Lyttleton O, McArdle W, Ring SM, et al. Maternal pre-pregnancy BMI and gestational weight gain, offspring DNA methylation and later offspring adiposity: findings from the Avon Longitudinal Study of Parents and Children. Int J Epidemiol. 2015;44(4):1288–1304. doi: 10.1093/ije/dyv042.
    1. Haworth KE, Farrell WE, Emes RD, Ismail KM, Carroll WD, Hubball E, Rooney A, Yates AM, Mein C, Fryer AA. Methylation of the FGFR2 gene is associated with high birth weight centile in humans. Epigenomics. 2014;6:477–491. doi: 10.2217/epi.14.40.
    1. Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC, McLean C, Rodford J, Slater-Jefferies JL, Garratt E, Crozier SR, et al. Epigenetic gene promoter methylation at birth is associated with child’s later adiposity. Diabetes. 2011;60(5):1528–1534. doi: 10.2337/db10-0979.
    1. Dick KJ, Nelson CP, Tsaprouni L, Sandling JK, Aissi D, Wahl S, Meduri E, Morange PE, Gagnon F, Grallert H, et al. DNA methylation and body-mass index: a genome-wide analysis. Lancet. 2014;383(9933):1990–1998. doi: 10.1016/S0140-6736(13)62674-4.
    1. Pan H, Lin X, Wu Y, Chen L, Teh AL, Soh SE, Lee YS, Tint MT, MacIsaac JL, Morin AM, et al. HIF3A association with adiposity: the story begins before birth. Epigenomics. 2015;7(6):937–950. doi: 10.2217/epi.15.45.
    1. Soh S-E, Tint MT, Gluckman PD, Godfrey KM, Rifkin-Graboi A, Chan YH, Stünkel W, Holbrook JD, Kwek K, Chong Y-S, et al. Cohort Profile: Growing Up in Singapore Towards healthy Outcomes (GUSTO) birth cohort study. Int J Epidemiol. 2014;43(5):1401–1409. doi: 10.1093/ije/dyt125.
    1. Rolland-Cachera MF. Childhood obesity: current definitions and recommendations for their use. Int J Pediatr Obes. 2011;6(5-6):325–331. doi: 10.3109/17477166.2011.607458.
    1. Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression. Development of the 10-item Edinburgh Postnatal Depression Scale. Br J Psychiatry. 1987;150:782–786. doi: 10.1192/bjp.150.6.782.
    1. Chee CYI, Lee DTS, Chong YS, Tan LK, Ng TP, Fones CSL. Confinement and other psychosocial factors in perinatal depression: A transcultural study in Singapore. J Affect Disord. 2005;89(1):157–166. doi: 10.1016/j.jad.2005.09.004.
    1. Chen H, Wang J, Ch’ng YC, Mingoo R, Lee T, Ong J. Identifying mothers with postpartum depression early: integrating perinatal mental health care into the obstetric setting. ISRN Obstet Gynecol. 2011;2011:309189. doi: 10.5402/2011/309189.
    1. Spielberger CD, Gorsuch RL, Lushene R, Vagg PR, Jacobs GA. Manual for the State-Trait Anxiety Inventory. Palo Alto: Consulting Psychologists Press; 1983.
    1. Chong S-C, Broekman BF, Qiu A, Aris IM, Chan YH, Rifkin-Graboi A, Law E, Chee CYI, Chong Y-S, Kwek KYC, et al. Anxiety and depression during pregnancy and temperament in early infancy: findings from a multi-ethnic, Asian prospective birth cohort study. Infant Mental Health J. 2016;37(5):584–598. doi: 10.1002/imhj.21582.
    1. Pan H, Chen L, Dogra S, Teh AL, Tan JH, Lim YI, Lim YC, Jin S, Lee YK, Ng PY, et al. Measuring the methylome in clinical samples: improved processing of the Infinium Human Methylation450 BeadChip Array. Epigenetics. 2012;7(10):1173–1187. doi: 10.4161/epi.22102.
    1. Johnson WE, Rabinovic A, Li C. Adjusting batch effects in microarray expression data using Empirical Bayes methods. Biostatistics. 2007;8(1):118–127. doi: 10.1093/biostatistics/kxj037.
    1. Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, Gallinger S, Hudson TJ, Weksberg R. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray. Epigenetics. 2013;8(2):203–209. doi: 10.4161/epi.23470.
    1. Price ME, Cotton AM, Lam LL, Farre P, Emberly E, Brown CJ, Robinson WP, Kobor MS. Additional annotation enhances potential for biologically-relevant analysis of the Illumina Infinium HumanMethylation450 BeadChip array. Epigenetics Chromatin. 2013;6(1):4. doi: 10.1186/1756-8935-6-4.
    1. Daca-Roszak P, Pfeifer A, Zebracka-Gala J, Rusinek D, Szybinska A, Jarzab B, Witt M, Zietkiewicz E. Impact of SNPs on methylation readouts by Illumina Infinium HumanMethylation450 BeadChip Array: implications for comparative population studies. BMC Genomics. 2015;16:1003. doi: 10.1186/s12864-015-2202-0.
    1. Xu Z, Niu L, Li L, Taylor JA. ENmix: a novel background correction method for Illumina HumanMethylation450 BeadChip. Nucleic Acids Res. 2016;44(3):e20. doi: 10.1093/nar/gkv907.
    1. Gelman A. Scaling regression inputs by dividing by two standard deviations. Stat Med. 2008;27(15):2865–2873. doi: 10.1002/sim.3107.
    1. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, Powell C, Vedantam S, Buchkovich ML, Yang J, et al. Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197–206. doi: 10.1038/nature14177.
    1. Houseman EA, Accomando WP, Koestler DC, Christensen BC, Marsit CJ, Nelson HH, Wiencke JK, Kelsey KT. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012;13:86. doi: 10.1186/1471-2105-13-86.
    1. Gutierrez-Arcelus M, Lappalainen T, Montgomery SB, Buil A, Ongen H, Yurovsky A, Bryois J, Giger T, Romano L, Planchon A, et al. Passive and active DNA methylation and the interplay with genetic variation in gene regulation. Elife. 2013;2:e00523.
    1. Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28(6):882–883. doi: 10.1093/bioinformatics/bts034.
    1. Leek JT, Scharpf RB, Bravo HC, Simcha D, Langmead B, Johnson WE, Geman D, Baggerly K, Irizarry RA. Tackling the widespread and critical impact of batch effects in high-throughput data. Nat Rev Genet. 2010;11(10):733–739. doi: 10.1038/nrg2825.
    1. Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet. 2007;3(9):1724–1735. doi: 10.1371/journal.pgen.0030161.
    1. Agha G, Houseman EA, Kelsey KT, Eaton CB, Buka SL, Loucks EB. Adiposity is associated with DNA methylation profile in adipose tissue. Int J Epidemiol. 2014;44(4):1277–1287. doi: 10.1093/ije/dyu236.
    1. Yoav B, Yosef H. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc Series B. 1995;57(1):289–300.
    1. Benjamini Y, Yekutieli D. The control of the false discovery rate in multiple testing under dependency. Annals Statistics. 2001;29(4):1165–1188. doi: 10.1214/aos/1013699998.
    1. Clarke S, Hall P. Robustness of multiple testing procedures against dependence. Annals Statistics. 2009;37(1):332–358. doi: 10.1214/07-AOS557.
    1. Lin X, Aris IM, Tint MT, Soh SE, Godfrey KM, Yeo GS, Kwek K, Chan JK, Gluckman PD, Chong YS, et al. Ethnic differences in effects of maternal pre-pregnancy and pregnancy adiposity on offspring size and adiposity. J Clin Endocrinol Metab. 2015;100(10):3641–3650. doi: 10.1210/jc.2015-1728.
    1. Aris IM, Soh SE, Tint MT, Saw SM, Rajadurai VS, Godfrey KM, Gluckman PD, Yap F, Chong YS, Lee YS. Associations of gestational glycemia and prepregnancy adiposity with offspring growth and adiposity in an Asian population. Am J Clin Nutr. 2015;102(5):1104–1112. doi: 10.3945/ajcn.115.117614.
    1. Chawla R, Badon SE, Rangarajan J, Reisetter AC, Armstrong LL, Lowe LP, Urbanek M, Metzger BE, Hayes MG, Scholtens DM, et al. Genetic risk score for prediction of newborn adiposity and large-for-gestational-age birth. J Clin Endocrinol Metab. 2014;99(11):E2377–E2386. doi: 10.1210/jc.2013-4221.
    1. Lorenzo DN, Healy JA, Hostettler J, Davis J, Yang J, Wang C, Hohmeier HE, Zhang M, Bennett V. Ankyrin-B metabolic syndrome combines age-dependent adiposity with pancreatic beta cell insufficiency. J Clin Invest. 2015;125(8):3087–3102. doi: 10.1172/JCI81317.
    1. Svensson PA, Wahlstrand B, Olsson M, Froguel P, Falchi M, Bergman RN, McTernan PG, Hedner T, Carlsson LM, Jacobson P. CDKN2B expression and subcutaneous adipose tissue expandability: possible influence of the 9p21 atherosclerosis locus. Biochem Biophys Res Commun. 2014;446(4):1126–1131. doi: 10.1016/j.bbrc.2014.03.075.
    1. Hannou SA, Wouters K, Paumelle R, Staels B. Functional genomics of the CDKN2A/B locus in cardiovascular and metabolic disease: what have we learned from GWASs? Trends Endocrinol Metab. 2015;26(4):176–184. doi: 10.1016/j.tem.2015.01.008.
    1. Uebele VN, Gotter AL, Nuss CE, Kraus RL, Doran SM, Garson SL, Reiss DR, Li Y, Barrow JC, Reger TS, et al. Antagonism of T-type calcium channels inhibits high-fat diet-induced weight gain in mice. J Clin Invest. 2009;119(6):1659–1667. doi: 10.1172/JCI36954.
    1. McGregor RA, Choi MS. microRNAs in the regulation of adipogenesis and obesity. Curr Mol Med. 2011;11(4):304–316. doi: 10.2174/156652411795677990.
    1. Sun T, Fu M, Bookout AL, Kliewer SA, Mangelsdorf DJ. MicroRNA let-7 regulates 3 T3-L1 adipogenesis. Mol Endocrinol. 2009;23(6):925–931. doi: 10.1210/me.2008-0298.
    1. Deiuliis JA. MicroRNAs as regulators of metabolic disease: pathophysiologic significance and emerging role as biomarkers and therapeutics. Int J Obes. 2016;40(1):88–101. doi: 10.1038/ijo.2015.170.
    1. McCann SE, Liu S, Wang D, Shen J, Hu Q, Hong CC, Newman VA, Zhao H. Reduction of dietary glycaemic load modifies the expression of microRNA potentially associated with energy balance and cancer pathways in pre-menopausal women. Br J Nutr. 2013;109(4):585–592. doi: 10.1017/S0007114512001936.
    1. Ogden CL, Carroll MD, Fryar CD, Flegal KM. Prevalence of Obesity Among Adults and Youth: United States, 2011–2014. Hyattsville: National Center for Health Statistics; 2015.
    1. Hayward CJ, Fradette J, Galbraith T, Remy M, Guignard R, Gauvin R, Germain L, Auger FA. Harvesting the potential of the human umbilical cord: isolation and characterisation of four cell types for tissue engineering applications. Cells Tissues Organs. 2013;197(1):37–54. doi: 10.1159/000341254.
    1. Lim IJ, Phan TT. Epithelial and mesenchymal stem cells from the umbilical cord lining membrane. Cell Transplant. 2014;23(4-5):497–503. doi: 10.3727/096368914X678346.
    1. Guo SS, Chumlea WC. Tracking of body mass index in children in relation to overweight in adulthood. Am J Clin Nutr. 1999;70(1):145S–148S.
    1. Freedman DS, Khan LK, Serdula MK, Dietz WH, Srinivasan SR, Berenson GS. The relation of childhood BMI to adult adiposity: the Bogalusa Heart Study. Pediatrics. 2005;115(1):22–27. doi: 10.1542/peds.2004-0220.
    1. Deshmukh-Taskar P, Nicklas TA, Morales M, Yang SJ, Zakeri I, Berenson GS. Tracking of overweight status from childhood to young adulthood: the Bogalusa Heart Study. Eur J Clin Nutr. 2006;60(1):48–57. doi: 10.1038/sj.ejcn.1602266.
    1. Biro FM, Wien M. Childhood obesity and adult morbidities. Am J Clin Nutr. 2010;91(5):1499S–1505S. doi: 10.3945/ajcn.2010.28701B.
    1. Kaplowitz PB, Slora EJ, Wasserman RC, Pedlow SE, Herman-Giddens ME. Earlier onset of puberty in girls: relation to increased body mass index and race. Pediatrics. 2001;108(2):347–353. doi: 10.1542/peds.108.2.347.
    1. Adair LS, Gordon-Larsen P. Maturational timing and overweight prevalence in US adolescent girls. Am J Public Health. 2001;91(4):642–644. doi: 10.2105/AJPH.91.4.642.
    1. Daniels SR, Arnett DK, Eckel RH, Gidding SS, Hayman LL, Kumanyika S, Robinson TN, Scott BJ, St Jeor S, Williams CL. Overweight in children and adolescents: pathophysiology, consequences, prevention, and treatment. Circulation. 2005;111(15):1999–2012. doi: 10.1161/01.CIR.0000161369.71722.10.
    1. Dietz WH. Overweight in childhood and adolescence. N Engl J Med. 2004;350(9):855–857. doi: 10.1056/NEJMp048008.

Source: PubMed

3
Abonnieren