Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-x5gtn Total loading time: 0 Render date: 2024-06-05T14:15:53.464Z Has data issue: false hasContentIssue false

Section II - Exposures Driving Long-Term DOHaD Effects

Published online by Cambridge University Press:  01 December 2022

Lucilla Poston
Affiliation:
King's College London
Keith M. Godfrey
Affiliation:
University of Southampton
Peter D. Gluckman
Affiliation:
University of Auckland
Mark A. Hanson
Affiliation:
University of Southampton
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2022

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

References

Stearns, SC. Evolutionary medicine: its scope, interest and potential. Proceedings of the Royal Society B: Biological Sciences. 2012;279(1746), 4305–4321.Google Scholar
Gluckman, P, , Low F. Evolutionary medicine. In Oxford Bibliographies. (ed. Futuyma, DJ), 2019. Oxford University Press: Oxford.Google Scholar
Low, FM, Gluckman, PD, Hanson, MA. Niche modification, human cultural evolution and the Anthropocene. Trends in Ecology and Evolution. 2019;34(10) 883–885.Google Scholar
Gluckman, PD, Low, FM, Hanson, MA. Anthropocene-related disease: the inevitable outcome of progressive niche modification? Evolution, Medicine, and Public Health. 2020;2020(1), 304–310.Google Scholar
Lucock, MD, Martin, CE, Yates, ZR, Veysey, M. Diet and our genetic legacy in the recent Anthropocene: a Darwinian perspective to nutritional health. Journal of Evidence-Based Complementary & Alternative Medicine. 2014;19(1), 68–83.Google Scholar
Gluckman, P, Beedle, A, Buklijas, T, Low, F, Hanson, M. Nutritional and metabolic adaptation. In Principles of Evolutionary Medicine. 2016; pp. 205–236. Oxford University Press: Oxford.Google Scholar
Ma, RCW, Chan, JCN, Tam, WH, Hanson, MA, Gluckman, PD. Gestational diabetes, maternal obesity and the NCD burden. Clinical Obstetrics and Gynecology. 2013;56(3), 633–641.Google Scholar
Nijs, H, Benhalima, K. Gestational diabetes mellitus and the long-term risk for glucose intolerance and overweight in the offspring: a narrative review. Journal of Clinical Medicine. 2020;9(2), 599.Google Scholar
Czosnykowska-Łukacka, M, Królak-Olejnik, B, Orczyk-Pawiłowicz, M. Breast milk macronutrient components in prolonged lactation. Nutrients. 2018;10(12), 1893.Google Scholar
Monasta, L, Batty, GD, Cattaneo, A, et al. Early-life determinants of overweight and obesity: a review of systematic reviews. Obesity Reviews. 2010;11(10), 695–708.Google Scholar
Belfort, MB, Anderson, PJ, Nowak, VA, et al. Breast milk feeding, brain development, and neurocognitive outcomes: a 7-year longitudinal study in infants born at less than 30 weeks’ gestation. The Journal of Pediatrics. 2016;177, 133–139.e131.Google Scholar
Gluckman, PD, Buklijas, T, Hanson, MA. Chapter 1 – The Developmental Origins of Health and Disease (DOHaD) concept: past, present, and future. In The Epigenome and Developmental Origins of Health and Disease. (ed. Rosenfeld, CS), 2016; pp. 1–15. Academic Press: Boston.Google Scholar
Gluckman, PD, Hanson, MA, Low, FM. Evolutionary and developmental mismatches are consequences of adaptive developmental plasticity in humans and have implications for later disease risk. Philosophical Transactions of the Royal Society B: Biological Sciences. 2019;374(1770), 20180109.Google Scholar
Sultan, SE. Developmental plasticity: re-conceiving the genotype. Interface Focus. 2017;7(5),20170009.Google Scholar
Karakochuk, CD, Whitfield, KC, Green, TJ, Kraemer, K. The Biology of the First 1,000 Days. 2018; p. 494. CRC Press: Boca Raton.Google Scholar
Hochberg, Z, Feil, R, Constancia, M, et al. Child health, developmental plasticity, and epigenetic programming. Endocrine Society. 2011;32(2), 159–224.Google Scholar
Pararas, MV, Skevaki, CL, Kafetzis, DA. Preterm birth due to maternal infection: causative pathogens and modes of prevention. European Journal of Clinical Microbiology & Infectious Diseases: Official Publication of the European Society of Clinical Microbiology. 2006;25(9), 562–569.Google Scholar
Bazaes, RA, Salazar, TE, Pittaluga, E, et al. Glucose and lipid metabolism in small for gestational infants at 48 hours of age. Pediatrics. 2003;111 (4), 804–809.Google Scholar
Kuzawa, CW. Beyond feast–famine: Brain evolution, human life history, and the metabolic syndrome. In Human Evolutionary Biology. (ed. Muehlenbein, MP), 2010; pp. 518–527. Cambridge University Press: Cambridge.Google Scholar
Gluckman, PD, Hanson, MA, Spencer, HG. Predictive adaptive responses and human evolution. Trends in Ecology & Evolution. 2005;20(10), 527–533.Google Scholar
Burgess, SC, Marshall, DJ. Adaptive parental effects: the importance of estimating environmental predictability and offspring fitness appropriately. Oikos. 2014;123(7), 769–776.Google Scholar
Pener, MP, Simpson, SJ. Locust phase polyphenism: an update. In Advances in Insect Physiology. (eds. Simpson, SJ, Pener, MP), 2009; pp. 1–272. Academic Press.Google Scholar
Sheriff, MJ, Krebs, CJ, Boonstra, R. The ghosts of predators past: population cycles and the role of maternal programming under fluctuating predation risk. Ecology. 2010;91(10), 2983–2994.Google Scholar
Bateson, P, Gluckman, P, Hanson, M. The biology of developmental plasticity and the Predictive Adaptive Response hypothesis. The Journal of Physiology. 2014;592(11), 2357–2368.Google Scholar
Kuzawa, CW. Fetal origins of developmental plasticity: are fetal cues reliable predictors of future nutritional environments? Am J Human Biol. 2005;17(1), 5–21.Google Scholar
Mericq, V, Martinez-Aguayo, A, Uauy, R, Iñiguez, G, Van der Steen, M, Hokken-Koelega, A. Long-term metabolic risk among children born premature or small for gestational age. Nature Reviews Endocrinology. 2017;13(1), 50–62.Google Scholar
Godfrey, KM, Sheppard, A, Gluckman, PD, et al. Epigenetic gene promoter methylation at birth is associated with child’s later adiposity. Diabetes. 2011;60(5)1528–1534.Google Scholar
Popkin, BM, Adair, LS, Ng, SW. Global nutrition transition and the pandemic of obesity in developing countries. Nutrition Reviews. 2012;70(1), 3–21.Google Scholar
Robinson, N, McKay, JA, Pearce, MS, et al. The biological and social determinants of childhood obesity: comparison of 2 cohorts 50 years apart. The Journal of Pediatrics. 2021;228, 138–146.e135.Google Scholar
Jablonka, E, Oborny, B, Molnar, I, Kisdi, E, Hofbauer, J, Czaran, T. The adaptive advantage of phenotypic memory in changing environments. Philosophical Transactions of the Royal Society of London – Series B: Biological Sciences. 1995;350(1332), 133–141.Google Scholar
Gluckman, PD, Hanson, MA. Maternal constraint of fetal growth and its consequences. Seminars in Fetal & Neonatal Medicine. 2004;9(5), 419–425.Google Scholar
Vasak, B, Koenen, SV, Koster, MPH, et al. Human fetal growth is constrained below optimal for perinatal survival. Ultrasound in Obstetrics & Gynecology. 2015;45(2)162–167.CrossRefGoogle Scholar
Lee, TM, Zucker, I. Vole infant development is influenced perinatally by maternal photoperiodic history. American Journal of Physiology. 1988;255, (5Pt2) R831–R838.Google Scholar
Bauerfeind, SS, Perlick, JEC, Fischer, K. Disentangling environmental effects on adult life span in a butterfly across the metamorphic boundary. Experimental Gerontology. 2009;44(12), 805–811.Google Scholar
Saastamoinen, M, van der Sterren, D, Vastenhout, N, Zwaan, BJ, Brakefield, PM. Predictive adaptive responses: condition-dependent impact of adult nutrition and flight in the tropical butterfly Bicyclus anynana. The American Naturalist. 2010;176(6), 686–698.Google Scholar
Sato, A, Sokabe, T, Kashio, M, Yasukochi, Y, Tominaga, M, Shiomi, K. Embryonic thermosensitive TRPA1 determines transgenerational diapause phenotype of the silkworm, Bombyx mori. PNAS. 2014;111(13), E1249–E1255.Google Scholar
Mitchell, A, Romano, GH, Groisman, B, et al. Adaptive prediction of environmental changes by microorganisms. Nature. 2009;460(7252), 220–224.Google Scholar
Petrusek, A, Tollrian, R, Schwenk, K, Haas, A, Laforsch, C. A ‘crown of thorns’ is an inducible defense that protects Daphnia against an ancient predator. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(7), 2248–2252.Google Scholar
Pham, T-P-T, Alou, MT, Golden, M, Million, M, Raoult, D. Difference between kwashiorkor and marasmus: comparative meta-analysis of pathogenic characteristics and implications for treatment. Microbial Pathogenesis. 2020;150, 104702.Google Scholar
Jahoor, F, Badaloo, A, Reid, M, Forrester, T. Unique metabolic characteristics of the major syndromes of severe childhood malnutrition. In The Tropical Metabolism Research Unit, The University of the West Indies, Jamaica 1956–2006: The House that John Built. (eds. Forrester, T, Picou, D, Walker, S), 2006; pp. 23–60. Ian Randle Publishers: Kingston.Google Scholar
Kumar, D, Rao, S, Singh, T. Clinico-biochemical profile of sick children with severe acute malnutrition. Journal of Family Medicine and Primary Care. 2020;9(5) 2269–2272.Google Scholar
Forrester, TE, Badaloo, AV, Boyne, MS, et al. Prenatal factors contribute to emergence of kwashiorkor or marasmus in response to severe undernutrition: evidence for the predictive adaptation model. PLoS One. 2012;7(4), e35907.Google Scholar
Francis-Emmanuel, PM, Thompson, DS, Barnett, AT, et al. Glucose metabolism in adult survivors of severe acute malnutrition. The Journal of Clinical Endocrinology & Metabolism. 2014;99(6), 2233–2240.Google Scholar
Schulze, KV, Swaminathan, S, Howell, S, et al. Edematous severe acute malnutrition is characterized by hypomethylation of DNA. Nature Communications. 2019;10(1), 5791.Google Scholar
Sheppard, A, Ngo, S, Li, X, et al. Molecular evidence for differential long-term outcomes of early life severe acute malnutrition. EBioMedicine. 2017;18, 274–280.Google Scholar
Boyne, MS, Francis-Emmanuel, P, Tennant, IA, Thompson, DS, Forrester, TE. Cardiometabolic risk in marasmus and kwashiorkor survivors. In Handbook of Famine, Starvation, and Nutrient Deprivation: From Biology to Policy. (eds. Preedy, V, Patel, VB), 2019; pp. 1–23. Springer International Publishing: Cham.Google Scholar
Engelgau, MM, Rosenthal, JP, Newsome, BJ, Price, L, Belis, D, Mensah, GA. Noncommunicable diseases in low- and middle-income countries: a strategic approach to develop a global implementation research workforce. Global Heart. 2018;13(2), 131–137.Google Scholar
Low, FM, Gluckman, PD, Hanson, MA. Maternal and child health: is making ‘healthy choices’ an oxymoron? Global Health Promotion. 2020; doi: 10.1177/1757975920967351, 1757975920967351.Google Scholar
Richardson, SS, Daniels, CR, Gillman, MW, et al. Don’t blame the mothers. Nature. 2014;512(7513), 131–132.Google Scholar
Low, FM, Gluckman, PD. Evolutionary medicine: mismatch. In The Encyclopedia of Evolutionary Biology. (ed. Kliman, RM), 2016. Academic Press: Oxford.Google Scholar

References

Reh, RK, Dias, BG, Nelson, CA 3rd, Kaufer, D, Werker, JF, Kolb, B, Levine, JD, Hensch, TK. Critical period regulation across multiple timescales. Proc Natl Acad Sci U S A 2020 Sep 22;117(38):23242–23251.CrossRefGoogle Scholar
Godfrey, KM, Inskip, HM, Hanson, MA. The long-term effects of prenatal development on growth and metabolism. Semin Reprod Med 2011;29(3):257–265.CrossRefGoogle Scholar
Waddington, CH. Canalization of development and the inheritance of acquired characters. Nature 1942;150(3811):563–565.Google Scholar
Low, FM, Gluckman, PD, Godfrey, KM. Early life development and epigenetic mechanisms: mediators of metabolic programming and obesity risk. In Nutrigenomics and Proteomics in Health and Disease. (ed. Kussmann, M, Stover, P.), Germany: Wiley 2017, 42–63. ISBN: 978-1-1190-9883-6Google Scholar
Godfrey, KM, Costello, PM, Lillycrop, KA. Development, epigenetics and metabolic programming. Nestle Nutr Inst Workshop Ser 2016;85:71–80. doi: 10.1159/000439488. Epub 2016 Apr 18. PMID: 27088334; PMCID: PMC4880042.Google Scholar
Fleming, TP, Watkins, AJ, Velazquez, MA, Mathers, JC, Prentice, AM, Stephenson, J, Barker, M, Saffery, R, Yajnik, CS, Eckert, JJ, Hanson, MA, Forrester, T, Gluckman, PD, Godfrey, KM. Origins of lifetime health around the time of conception: causes and consequences. Lancet 2018;391(10132):1842–1852.Google Scholar
Velazquez, MA, Fleming, TP, Watkins, AJ. Periconceptional environment and the developmental origins of disease. J Endocrinol 2019;242(1):T33–T49.CrossRefGoogle Scholar
Huang, JY, Cai, S, Huang, Z, Tint, MT, Yuan, WL, Aris, IM, Godfrey, KM, Karnani, N, Lee, YS, Chan, JKY, Chong, YS, Eriksson, JG, Chan, SY. Analyses of child cardiometabolic phenotype following assisted reproductive technologies using a pragmatic trial emulation approach. Nat Commun 2021;12(1):5613.CrossRefGoogle Scholar
Kleijkers, SHM, Mantikou, E, Slappendel, E, Consten, D, van Echten-Arends, J, Wetzels, AM, van Wely, M, Smits, LJM, van Montfoort, APA, Repping, S, Dumoulin, JC, Mastenbroek, S. Influence of embryo culture medium (G5 and HTF) on pregnancy and perinatal outcome after IVF: a multicenter RCT. Hum Reprod 2016;31(10):2219–2230.Google Scholar
Wu, LL, Russell, DL, Wong, SL, Chen, M, Tsai, TS, St John, JC, Norman, RJ, Febbraio, MA, Carroll, J, Robker, RL. Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 2015;142(4):681–691.Google Scholar
Gould, JM, Smith, PJ, Airey, CJ, Mort, EJ, Airey, LE, Warricker, FDM, Pearson-Farr, JE, Weston, EC, Gould, PJW, Semmence, OG, Restall, KL, Watts, JA, McHugh, PC, Smith, SJ, Dewing, JM, Fleming, TP, Willaime-Morawek, S. Mouse maternal protein restriction during preimplantation alone permanently alters brain neuron proportion and adult short-term memory. Proc Natl Acad Sci U S A 2018;115(31):E7398–E7407.Google Scholar
Steegers-Theunissen, RP, Twigt, J, Pestinger, V, Sinclair, KD. The periconceptional period, reproduction and long-term health of offspring: the importance of one-carbon metabolism. Hum Reprod Update 2013;19(6):640–655.Google Scholar
Watkins, AJ, Sinclair, KD. Paternal low protein diet affects adult offspring cardiovascular and metabolic function in mice. Am J Physiol Heart Circ Physiol 2014;306:H1444–1452.Google Scholar
Watkins, AJ, Dias, I, Tsuro, H, Allen, D, Emes, RD, Moreton, J, Wilson, R, Ingram, RJM, Sinclair, KD. Paternal diet programs offspring health through sperm- and seminal plasma-specific pathways in mice. Proc Natl Acad Sci U S A 2018;115(10):10064–10069.Google Scholar
Viner, RM, Ross, D, Hardy, R, et al. Life course epidemiology: recognising the importance of adolescence. J Epidemiol Community Health 2015;69(40):719–720.CrossRefGoogle Scholar
Alberga, AS, Sigal, RJ, Goldfield, G, Prud’ homme, D, Kenny, GP. The critical period of adolescence. Pediatric Obesity 2012;7(4):261–273.Google Scholar
Results of the National Diet and Nutrition Survey (NDNS) rolling programme for 2014 to 2015 and 2015 to 2016. www.gov.uk/government/statistics/ndns-results-from-years-7-and-8-combinedGoogle Scholar
Johnson, W, Moore, SE. Adolescent pregnancy, nutrition, and health outcomes in low- and middle-income countries: what we know and what we don’t know. BJOG 2016; 123(10):1589–1592.CrossRefGoogle Scholar
Neufeld, LM, Andrade EB, Suleiman AB, Barker M, Beal T, Blum LS, Demmler KM, Dogra S, Hardy-Johnson P, Lahiri A, Larson N. Food choice in transition: adolescent autonomy, agency, and the food environment. The Lancet 2021.Google Scholar
Blakemore, SJ, Mills, KL. Is adolescence a sensitive period for sociocultural processing? Ann Review Psychol 2014; 65:187–207.Google Scholar
Patton, G, Temmerman, M. Evidence and evidence gaps in adolescent health. J Adolesc Health 2016;59(4S):S1–S3. doi:10.1016/j.jadohealth.2016.08.001Google Scholar
Bay, JL, Vickers, MH, Mora, HA, Sloboda, DM, Morton, SM. Adolescents as agents of healthful change through scientific literacy development: a school-university partnership program in New Zealand. Int J STEM Education 2017;4(1):15.CrossRefGoogle Scholar
Woods-Townsend, K, Leat, H, Bay, J, Bagust, L, Davey, H, Lovelock, D, Christodoulou, A, Griffiths, J, Grace, M, Godfrey, K, Hanson, M, Inskip, H. LifeLab Southampton: a programme to engage adolescents with DOHaD concepts as a tool for increasing health literacy in teenagers -a pilot cluster-randomized control trial. J Dev Orig Health Dis 2018;9(5):475–480.Google Scholar
Woods-Townsend, K, Hardy-Johnson, P, Bagust, L, Barker, M, Davey, H, Griffiths, J, et al. A cluster-randomised controlled trial of the LifeLab education intervention to improve health literacy in adolescents. PLoS ONE 2021;16(5):e0250545.Google Scholar
De-Regil, LM, Peña-Rosas, JP, Fernández-Gaxiola, AC, Rayco-Solon, P. Effects and safety of periconceptional oral folate supplementation for preventing birth defects. Cochrane Database Syst Rev 2015 Dec 14;2015(12):CD007950. doi: 10.1002/14651858.CD007950.pub3. PMID: 26662928.Google Scholar
Stephenson, J, Heslehurst, N, Hall, J, Schoenaker, DAJM, Hutchinson, J, Cade, JE, Poston, L, Barrett, G, Crozier, SR, Barker, M, Kumaran, K, Yajnik, CS, Baird, J, Mishra, GD. Before the beginning: nutrition and lifestyle in the preconception period and its importance for future health. Lancet 2018 May 5;391(10132):1830–1841.Google Scholar
Brackenridge, L, Finer, N, Batterham, RL, Pedram, K, Ding, T, Stephenson, J, Barry, J, Hardiman, P. Pre-pregnancy weight loss in women with obesity requesting removal of their intra-uterine contraceptive device in order to conceive: a pilot study of full meal replacement. Clin Obes 2018 Aug;8(4):244–249.Google Scholar
International Weight Management in Pregnancy (i-WIP) Collaborative Group. Effect of diet and physical activity based interventions in pregnancy on gestational weight gain and pregnancy outcomes: meta-analysis of individual participant data from randomised trials. BMJ 2017 Jul 19;358:j3119.Google Scholar
Ellsworth, L, Harman, E, Padmanabhan, V, Gregg, B. Lactational programming of glucose homeostasis: a window of opportunity. Reproduction 2018;156(2):R23–R42.Google Scholar
Padmanabhan, V, Cardoso, RC, Puttabyatappa, M. Developmental programming, a pathway to disease. Endocrinology 2016;157(4):1328–1340.Google Scholar
Bartol, FF, Wiley, AA, George, AF, Miller, DJ, Bagnell, CA. Postnatal reproductive development and the lactocrine hypothesis. J Anim Sci 2017;95(5):2200–2210.Google Scholar
Vickers, MH, Sloboda, DM. Strategies for reversing the effects of metabolic disorders induced as a consequence of developmental programming. Front Physiol 2012;3:242.Google Scholar
Grattan, DR. Fetal programming from maternal obesity: eating too much for two? Endocrinology 2008;149(11):5345–5347.Google Scholar
Aiken, CE, Ozanne, SE. Transgenerational developmental programming. Hum Reprod Update 2014;20(1):63–75.Google Scholar
Howie, GJ, Sloboda, DM, Vickers, MH. Maternal undernutrition during critical windows of development results in differential and sex-specific effects on postnatal adiposity and related metabolic profiles in adult rat offspring. Br J Nutr 2012;108(2):298–307.Google Scholar
Howie, GJ, Sloboda, DM, Reynolds, CM, Vickers, MH. Timing of maternal exposure to a high fat diet and development of obesity and hyperinsulinemia in male rat offspring: same metabolic phenotype, different developmental pathways? J Nutr Metab 2013;2013:517384.Google Scholar
Crump, C. Preterm birth and mortality in adulthood: a systematic review. J Perinatol 2020;40(6):833–843.Google Scholar
Crump, C. An overview of adult health outcomes after preterm birth. Early Hum Dev 2020;150:105187.Google Scholar
Andraweera, PH, Condon, B, Collett, G, Gentilcore, S, Lassi, ZS. Cardiovascular risk factors in those born preterm – systematic review and meta-analysis. J Dev Orig Health Dis 2021;12(4):539–554.Google Scholar
Crump, C, Sundquist, J, Sundquist, K. Risk of hypertension into adulthood in persons born prematurely: a national cohort study. Eur Heart J 2020;41(16):1542–1550.Google Scholar
Yoshida-Montezuma, Y, Stone, E, Iftikhar, S, De Rubeis, V, Andreacchi, A, Keown-Stoneman, C, et al. Late preterm birth as a risk-factor for cardiometabolic conditions across the life-course: a systematic review and meta-analysis. Paediatr Perinatal Epidemiol 2022;36(2):264–275.Google Scholar
Li, S, Zhang, M, Tian, H, Liu, Z, Yin, X, Xi, B. Preterm birth and risk of type 1 and type 2 diabetes: systematic review and meta-analysis. Obes Rev 2014;15(10):804–811.Google Scholar
Bloomfield, FH. Impact of prematurity for pancreatic islet and beta-cell development. J Endocrinol 2018;238(3):R161R71.Google Scholar
Bukowski, R, Smith, GC, Malone, FD, Ball, RH, Nyberg, DA, Comstock, CH, et al. Fetal growth in early pregnancy and risk of delivering low birth weight infant: prospective cohort study. BMJ 2007;334(7598):836.Google Scholar
Story, L, Zhang, T, Uus, A, Hutter, J, Egloff, A, Gibbons, D, Ho, A, Al-Adnani, M, Knight, CL, Theodoulou, I, Deprez, M, Seed, PT, Tribe, RM, Shennan, AH, Rutherford, M. Antenatal thymus volumes in fetuses that delivered <32 weeks’ gestation: An MRI pilot study. Acta Obstet Gynecol Scand 2021;100:1040–1050.Google Scholar
Cooke, RW. Conventional birth weight standards obscure fetal growth restriction in preterm infants. Arch Dis Child Fetal Neonatal Ed 2007;92(3):F189–92.Google Scholar
Abitbol, CL, Rodriguez, MM. The long-term renal and cardiovascular consequences of prematurity. Nat Rev Nephrol 2012;8(5):265–274.Google Scholar
Moschino, L, Bonadies, L, Baraldi, E. Lung growth and pulmonary function after prematurity and bronchopulmonary dysplasia. Pediatr Pulmonol 2021; Mar 17;56(11):3499–3508. doi: 10.1002/ppul.25380.Google Scholar
Bensley, JG, Moore, L, De Matteo, R, Harding, R, Black, MJ. Impact of preterm birth on the developing myocardium of the neonate. Pediatr Res 2017;83(4):880–888.Google Scholar
Lin, L, Amissah, E, Gamble, GD, Crowther, CA, Harding, JE. Impact of macronutrient supplements on later growth of children born preterm or small for gestational age: A systematic review and meta-analysis of randomised and quasirandomised controlled trials. PLoS Med 2020;17(5):e1003122.Google Scholar
Lin, L, Amissah, E, Gamble, GD, Crowther, CA, Harding, JE. Impact of macronutrient supplements for children born preterm or small for gestational age on developmental and metabolic outcomes: A systematic review and meta-analysis. PLoS Med 2019;16(10):e1002952.Google Scholar
Woo, JG, et al. Infant growth and long-term cardiometabolic health: a review of recent findings. Curr Nutr Rep 2019;8(1):29–41.Google Scholar
Andrew, SB, et al. Does the association between early life growth and later obesity differ by race/ethnicity or socioeconomic status? A systematic review. Ann Epidemiol 2017;27(9):583–592.Google Scholar
Azad, MB, et al. Infant feeding and weight gain: separating breast milk from breastfeeding and formula from food. Pediatrics 2018;142(4):e20181092.Google Scholar
Patro-Golab, B, et al. Protein concentration in milk formula, growth, and later risk of obesity: a systematic review. J Nutr 2016;146:551–564.Google Scholar
Totzauer, M, et al. Effect of lower versus higher protein content in infant formula through the first year on body composition from 1 to 6 years: follow-up of a randomized clinical trial. Obesity 2018;26:1203–1212.CrossRefGoogle Scholar
Dewey, KG, et al. Breastfeeding and risk of overweight in childhood and beyond: a systematic review with emphasis on sibling-pair and intervention studies. Am J Clin Nutr 2021;114:1774–1790.Google Scholar
Martin, R, et al. Effects of promoting long-term exclusive breastfeeding on adolescent adiposity, blood pressure, and growth trajectories. A secondary analysis of a randomized controlled trial. JAMA Pediatr 2017;171(7):e170698.Google Scholar
Wang, J, et al. Introduction of complementary feeding before 4 months of age increases the risk of childhood overweight or obesity: a meta-analysis of prospective cohort studies. Nutr Res 2016;36:759–70.Google Scholar
Taylor, RW, et al. Effect of a baby-led approach to complementary feeding on infant growth and overweight; a randomized controlled trial. JAMA Pediatr 2017;171:838–846.Google Scholar
Campoy, C et al. Complementary feeding in developed countries: the 3 Ws (when, what, and why?). Ann Nutr Metab 2018;73:27–36.Google Scholar
Godfrey, KM, Barton, SJ, El-Heis, S, Kenealy, T, Nield, H, Baker, PN, Chong, YS, Cutfield, W, Chan, SY. NiPPeR study group. Myo-Inositol, probiotics, and micronutrient supplementation from preconception for glycemia in pregnancy: NiPPeR International multicenter double-blind randomized controlled trial. Diabetes Care 2021;44:1091–1099.Google Scholar
Patel, N, Godfrey, KM, Pasupathy, D, Levin, J, Flynn, AC, Hayes, L, Briley, AL, Bell, R, Lawlor, DA, Oteng-Ntim, E, Nelson, SM, Robson, SC, Sattar, N, Singh, C, Wardle, J, White, SL, Seed, PT, Poston, L. Infant adiposity following a randomised controlled trial of a behavioural intervention in obese pregnancy. Int J Obes (Lond) 2017;41:1018–1026.Google Scholar
Poston, L, Bell, R, Briley, AL, Godfrey, KM, Nelson, SM, Oteng-Ntim, E, Sandall, J, Sanders, TAB, Sattar, N, Seed, PT, Robson, SC, Trépel, D, Wardle, J. Improving pregnancy outcome in obese women: the UK Pregnancies Better Eating and Activity randomised controlled Trial. Southampton (UK): NIHR Journals Library 2017;5(10):1–444.Google Scholar
Antoun, E, Kitaba, NT, Titcombe, P, Dalrymple, KV, Garratt, ES, Barton, SJ, Murray, R, Seed, PT, Holbrook, JD, Kobor, MS, Lin, DT, MacIsaac, JL, Burdge, GC, White, SL, Poston, L, Godfrey, KM, Lillycrop, KA. UPBEAT Consortium. Maternal dysglycaemia, changes in the infant’s epigenome modified with a diet and physical activity intervention in pregnancy: Secondary analysis of a randomised control trial. PLoS Med 2020;17(11):e1003229.Google Scholar

References

Hanson, M., The birth and future health of DOHaD. J Dev Orig Health Dis, 2015. 6(5): pp. 434–7.Google Scholar
Collaborators, G.B.D.C.o.D., Global, regional, and national age-sex specific mortality for 264 causes of death, 1980–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet, 2017. 390(10100): pp. 1151–210.Google Scholar
FAO, I., UNICEF, WFP and WHO., The State of Food Security and Nutrition in the World 2019. Safeguarding against economic slowdowns and downturns., FAO, Editor. 2019: Rome.Google Scholar
Maternal and Child Nutrition Study Group, et al., Maternal and child nutrition: building momentum for impact. Lancet, 2013. 382(9890): pp. 372–5.Google Scholar
FAO, The State of Food Insecurity in the World 2001 FAO, Editor. 2002: Rome.Google Scholar
Development Initiatives, 2018 Global Nutrition Report: Shining a light to spur action on nutrition, D. Initiatives, Editor. 2018: Bristol, UK.Google Scholar
Bailey, R.L., West, K.P., Jr., and Black, R.E., The epidemiology of global micronutrient deficiencies. Ann Nutr Metab, 2015. 66 Suppl 2: pp. 22–33.Google Scholar
in Prevention of Micronutrient Deficiencies: Tools for Policymakers and Public Health Workers, Howson, C.P., Kennedy, E.T., and Horwitz, A., Editors. 1998. National Academies Press: Washington (DC).Google Scholar
Seligman, H.K., Laraia, B.A., and Kushel, M.B., Food insecurity is associated with chronic disease among low-income NHANES participants. J Nutr, 2010. 140(2): pp. 304–10.Google Scholar
Richter, L.M., et al., Cohort profile: the consortium of health-orientated research in transitioning societies. Int J Epidemiol, 2012. 41(3): pp. 621–6.Google Scholar
Fall, C.H., et al., Association between maternal age at childbirth and child and adult outcomes in the offspring: a prospective study in five low-income and middle-income countries (COHORTS collaboration). Lancet Glob Health, 2015. 3(7): pp. e366–77.Google Scholar
Martorell, R., et al., Weight gain in the first two years of life is an important predictor of schooling outcomes in pooled analyses from five birth cohorts from low- and middle-income countries. J Nutr, 2010. 140(2): pp. 348–54.Google Scholar
Stein, A.D., et al., Growth patterns in early childhood and final attained stature: data from five birth cohorts from low- and middle-income countries. Am J Hum Biol, 2010. 22(3): pp. 353–9.Google Scholar
Victora, C.G., et al., Maternal and child undernutrition: consequences for adult health and human capital. Lancet, 2008. 371(9609): pp. 340–57.Google Scholar
Krishnaveni, G.V. and Yajnik, C.S., Developmental origins of diabetes-an Indian perspective. Eur J Clin Nutr, 2017. 71(7): pp. 865–9.Google Scholar
India State-Level Disease Burden Initiative Diabetes, C., The increasing burden of diabetes and variations among the states of India: the global burden of disease study 1990–2016. Lancet Glob Health, 2018. 6(12): pp. e135262.Google Scholar
Yajnik, C.S., et al., Neonatal anthropometry: the thin-fat Indian baby. The Pune maternal nutrition study. Int J Obes Relat Metab Disord, 2003. 27(2): pp. 173–80.Google Scholar
Rao, S., et al., Intake of micronutrient-rich foods in rural Indian mothers is associated with the size of their babies at birth: Pune maternal nutrition study. J Nutr, 2001. 131(4): pp. 1217–24.Google Scholar
Yajnik, C.S., et al., Vitamin B12 and folate concentrations during pregnancy and insulin resistance in the offspring: the Pune maternal nutrition study. Diabetologia, 2008. 51(1): pp. 29–38.Google Scholar
Norris, S.A., et al., Understanding and acting on the developmental origins of health and disease in Africa would improve health across generations. Glob Health Action, 2017. 10(1): p. 1334985.Google Scholar
Moore, S.E., Using longitudinal data to understand nutrition and health interactions in rural Gambia. Ann Hum Biol, 2020. (In Press). 47(2):125–31Google Scholar
Rayco-Solon, P., Fulford, A.J., and Prentice, A.M., Differential effects of seasonality on preterm birth and intrauterine growth restriction in rural Africans. Am J Clin Nutr, 2005. 81(1): p. 134–9.Google Scholar
Moore, S.E., et al., Season of birth predicts mortality in rural Gambia. Nature, 1997. 388(6641): p. 434.Google Scholar
Moore, S.E., et al., Prenatal or early postnatal events predict infectious deaths in young adulthood in rural Africa. Int J Epidemiol, 1999. 28(6): pp. 1088–95.Google Scholar
Moore, S.E., Early life nutritional programming of health and disease in The Gambia. J Dev Orig Health Dis, 2016. 7(2): pp. 123–31.Google Scholar
Rakyan, V.K., et al., Metastable epialleles in mammals. Trends Genet, 2002. 18(7): pp. 348–51.Google Scholar
Waterland, R.A. and Michels, K.B., Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr, 2007. 27: pp. 363–88.Google Scholar
Waterland, R.A., et al., Season of conception in rural gambia affects DNA methylation at putative human metastable epialleles. PLoS Genet, 2010. 6(12): p. e1001252.Google Scholar
Waterland, R.A. and Jirtle, R.L., Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol, 2003. 23(15): pp. 5293–300.Google Scholar
Dominguez-Salas, P., et al., DNA methylation potential: dietary intake and blood concentrations of one-carbon metabolites and cofactors in rural African women. Am J Clin Nutr, 2013. 97(6): pp. 1217–27.Google Scholar
Dominguez-Salas, P., et al., Maternal nutrition at conception modulates DNA methylation of human metastable epialleles. Nat Commun, 2014. 5: p. 3746.Google Scholar
Stein, A.D., et al., Cohort profile: the Institute of Nutrition of Central America and Panama (INCAP) Nutrition Trial Cohort Study. Int J Epidemiol, 2008. 37(4): pp. 716–20.Google Scholar
Habicht, J.P., Martorell, R., and Rivera, J.A., Nutritional impact of supplementation in the INCAP longitudinal study: analytic strategies and inferences. J Nutr, 1995. 125(4 Suppl): pp. 1042S1050S.Google Scholar
Martorell, R., Overview of long-term nutrition intervention studies in Guatemala, 1968–1989. Food Nutr Bull, 1993. 14: pp. 270–7.CrossRefGoogle Scholar
Rivera, J.A., et al., Nutritional supplementation during the preschool years influences body size and composition of Guatemalan adolescents. J Nutr, 1995. 125(4 Suppl): pp. 1068S1077S.Google Scholar
Stein, A.D., et al., Exposure to a nutrition supplementation intervention in early childhood and risk factors for cardiovascular disease in adulthood: evidence from Guatemala. Am J Epidemiol, 2006. 164(12): pp. 1160–70.Google Scholar
Hoddinott, J., et al., Effect of a nutrition intervention during early childhood on economic productivity in Guatemalan adults. Lancet, 2008. 371(9610): pp. 411–16.Google Scholar
Kinra, S., et al., Effect of integration of supplemental nutrition with public health programmes in pregnancy and early childhood on cardiovascular risk in rural Indian adolescents: long term follow-up of Hyderabad nutrition trial. BMJ, 2008. 337: p. a605.Google Scholar
Ceesay, S.M., et al., Effects on birth weight and perinatal mortality of maternal dietary supplements in rural Gambia: 5 year randomised controlled trial. BMJ, 1997. 315(7111): pp. 786–90.Google Scholar
Moore, S.E., Collinson, A.C., and Prentice, A.M., Immune function in rural Gambian children is not related to season of birth, birth size, or maternal supplementation status. Am J Clin Nutr, 2001. 74(6): pp. 840–7.Google Scholar
Hawkesworth, S., et al., Maternal protein-energy supplementation does not affect adolescent blood pressure in The Gambia. Int J Epidemiol, 2009. 38(1): pp. 119–27.Google Scholar
Hawkesworth, S., et al., Nutritional supplementation during pregnancy and offspring cardiovascular disease risk in The Gambia. Am J Clin Nutr, 2011. 94(6 Suppl): pp. 1853S1860S.Google Scholar
Alderman, H., et al., Supplemental feeding during pregnancy compared with maternal supplementation during lactation does not affect schooling and cognitive development through late adolescence. Am J Clin Nutr, 2014. 99(1): pp. 122–9.Google Scholar
Persson, L.A., et al., Effects of prenatal micronutrient and early food supplementation on maternal hemoglobin, birth weight, and infant mortality among children in Bangladesh: the MINIMat randomized trial. JAMA, 2012. 307(19): pp. 2050–9.Google Scholar
Tofail, F., et al., Effects of prenatal food and micronutrient supplementation on infant development: a randomized trial from the Maternal and Infant Nutrition Interventions, Matlab (MINIMat) study. Am J Clin Nutr, 2008. 87(3): pp. 704–11.Google Scholar
Khan, A.I., et al., Early invitation to food and/or multiple micronutrient supplementation in pregnancy does not affect body composition in offspring at 54 months: follow-up of the MINIMat randomised trial, Bangladesh. Matern Child Nutr, 2015. 11(3): pp. 385–97.Google Scholar
Hawkesworth, S., et al., Combined food and micronutrient supplements during pregnancy have limited impact on child blood pressure and kidney function in rural Bangladesh. J Nutr, 2013. 143(5): pp. 728–34.Google Scholar
Ekstrom, E.C., et al., Effects of prenatal micronutrient and early food supplementation on metabolic status of the offspring at 4.5 years of age. The MINIMat randomized trial in rural Bangladesh. Int J Epidemiol, 2016. 45(5): pp. 1656–67.Google Scholar
Belizan, J.M., et al., Long-term effect of calcium supplementation during pregnancy on the blood pressure of offspring: follow up of a randomised controlled trial. BMJ, 1997. 315(7103): pp. 281–5.Google Scholar
Hatton, D.C., et al., Gestational calcium supplementation and blood pressure in the offspring. Am J Hypertens, 2003. 16(10): pp. 801–5.Google Scholar
Hawkesworth, S., et al., Effect of maternal calcium supplementation on offspring blood pressure in 5- to 10-y-old rural Gambian children. Am J Clin Nutr, 2010. 92(4): pp. 741–7.Google Scholar
Goldberg, G.R., et al., Randomized, placebo-controlled, calcium supplementation trial in pregnant Gambian women accustomed to a low calcium intake: effects on maternal blood pressure and infant growth. Am J Clin Nutr, 2013. 98(4): pp. 972–82.Google Scholar
Hiller, J.E., et al., Calcium supplementation in pregnancy and its impact on blood pressure in children and women: follow up of a randomised controlled trial. Aust N Z J Obstet Gynaecol, 2007. 47(2): pp. 115–21.Google Scholar
Kalra, P., et al., Effect of vitamin D supplementation during pregnancy on neonatal mineral homeostasis and anthropometry of the newborn and infant. Br J Nutr, 2012. 108(6): pp. 1052–8.Google Scholar
Roth, D.E., et al., Maternal vitamin D3 supplementation during the third trimester of pregnancy: effects on infant growth in a longitudinal follow-up study in Bangladesh. J Pediatr, 2013. 163(6): pp. 1605–11 e3.Google Scholar
Goldring, S.T., et al., Prenatal vitamin d supplementation and child respiratory health: a randomised controlled trial. PLoS One, 2013. 8(6): p. e66627.Google Scholar
Darmstadt, G.L., et al., Effect of antenatal zinc supplementation on impetigo in infants in Bangladesh. Pediatr Infect Dis J, 2012. 31(4): pp. 407–9.Google Scholar
Hamadani, J.D., et al., Zinc supplementation during pregnancy and effects on mental development and behaviour of infants: a follow-up study. Lancet, 2002. 360(9329): pp. 290–4.Google Scholar
Osendarp, S.J., et al., Zinc supplementation during pregnancy and effects on growth and morbidity in low birthweight infants: a randomised placebo controlled trial. Lancet, 2001. 357(9262): pp. 1080–5.Google Scholar
Tamura, T., et al., Effect of zinc supplementation of pregnant women on the mental and psychomotor development of their children at 5 y of age. Am J Clin Nutr, 2003. 77(6): pp. 1512–6.Google Scholar
Schmidt, M.K., et al., Randomised double-blind trial of the effect of vitamin A supplementation of Indonesian pregnant women on morbidity and growth of their infants during the first year of life. Eur J Clin Nutr, 2002. 56(4): pp. 338–46.Google Scholar
Schmidt, M.K., et al., Mental and psychomotor development in Indonesian infants of mothers supplemented with vitamin A in addition to iron during pregnancy. Br J Nutr, 2004. 91(2): pp. 279–86.Google Scholar
Parsons, A.G., et al., Effect of iron supplementation during pregnancy on the behaviour of children at early school age: long-term follow-up of a randomised controlled trial. Br J Nutr, 2008. 99(5): pp. 1133–9.Google Scholar
Zhou, S.J., et al., Effect of iron supplementation during pregnancy on the intelligence quotient and behavior of children at 4 y of age: long-term follow-up of a randomized controlled trial. Am J Clin Nutr, 2006. 83(5): pp. 1112–7.Google Scholar
WHO, Recommendations on Antenatal Care for a Positive Pregnancy Experience. 2018: Geneva, Switzerland: World Health Organization.Google Scholar
WHO/UNU/UNICEF, Composition of a multi-micronutrient supplement to be used in pilot programmes among pregnany women in developing countries: report of a United Nations Children’s Fund (UNICEF), World Health Orgnanisation (WHO) and United Nations University (UNU) workshop., UNICEF, Editor. 1999: New York.Google Scholar
Keats, E.C., et al., Multiple-micronutrient supplementation for women during pregnancy. Cochrane Database Syst Rev, 2019. 3: p. CD004905.Google Scholar
Smith, E.R., et al., Modifiers of the effect of maternal multiple micronutrient supplementation on stillbirth, birth outcomes, and infant mortality: a meta-analysis of individual patient data from 17 randomised trials in low-income and middle-income countries. Lancet Glob Health, 2017. 5(11): pp. e1090e1100.Google Scholar
WHO, WHO antenatal care recommendations for a positive pregnancy experience. Nutritional interventions update: Multiple micronutrient supplements during pregnancy, World Health Organization, Editor. 2020: Geneva.Google Scholar
Devakumar, D., et al., Maternal antenatal multiple micronutrient supplementation for long-term health benefits in children: a systematic review and meta-analysis. BMC Med, 2016. 14: p. 90.Google Scholar
Potdar, R.D., et al., Improving women’s diet quality preconceptionally and during gestation: effects on birth weight and prevalence of low birth weight--a randomized controlled efficacy trial in India (Mumbai Maternal Nutrition Project). Am J Clin Nutr, 2014. 100(5): pp. 1257–68.Google Scholar
Moore, S.E., et al., A randomized trial to investigate the effects of prenatal and infant nutritional supplementation on infant immune development in rural Gambia: the ENID trial: Early Nutrition and Immune Development. BMC Pregnancy Childbirth, 2012. 12: p. 107.Google Scholar
Moore, S.E., et al., Thymic size is increased by infancy, but not pregnancy, nutritional supplementation in rural Gambian children: a randomized clinical trial. BMC Med, 2019. 17(1): p. 38.Google Scholar
Okala, S.G., et al., Impact of nutritional supplementation during pregnancy on antibody responses to diphtheria-tetanus-pertussis vaccination in infants: A randomised trial in The Gambia. PLoS Med, 2019. 16(8): p. e1002854.Google Scholar
Hawkes, C., et al., Double-duty actions: seizing programme and policy opportunities to address malnutrition in all its forms. Lancet, 2020. 395(10218): pp. 142–55.Google Scholar

References

World Health Organisation. Obesity and Overweight Fact Sheet. Geneva: 2020 01/04/2020. Report No.Google Scholar
Access Economics. The growing cost of obesity in 2008: three years on. Canberra: Report for Diabetes Australia, 2008.Google Scholar
Wang, Y, Beydoun, MA, Liang, L, Caballero, B, Kumanyika, SK. Will all Americans become overweight or obese? Estimating the progression and cost of the US obesity epidemic. Obesity. 2008;16(10):2323–30.Google Scholar
Australian Institute of Health and Welfare. Australia’s mothers and babies 2017 – in brief. Canberra: Australian Institute of Health, 2019 Contract No.: Cat. no. PER 100.Google Scholar
Chen, C, Xu, X, Yan, Y. Estimated global overweight and obesity burden in pregnant women based on panel data model. PLoS One. 2018;13(8):e0202183. Epub 2018/08/10. doi: 10.1371/journal.pone.0202183. PubMed PMID: 30092099; PubMed Central PMCID: PMCPMC6084991.Google Scholar
Dodd, JM, Grivell, RM, Nguyen, A-M, Chan, A, Robinson, JS. Maternal and perinatal health outcomes by body mass index category. ANZJOG. 2011;51(2):136–40.Google Scholar
La Coursiere, DY, Bloebaum, L, Duncan, JD, Varner, MW. Population-based trends and correlates of maternal overweight and obesity, Utah 1991–2001. Am J Obstet Gynecol. 2005;192(3);32–9.Google Scholar
Galtier-Dereure, F, Boegner, C, Bringer, J. Obesity and pregnancy: complications and cost. Am J Clin Nutr. 2000;71(5 Suppl):S1242S8.Google Scholar
Liu, P, Xu, L, Wang, Y, Zhang, Y, Du, Y, Sun, Y, et al. Association between perinatal outcomes and maternal pre-pregnancy body mass index. Obes Rev. 2016;17(11):1091–102. Epub 2016/08/19. doi: 10.1111/obr.12455. PubMed PMID: 27536879.Google Scholar
Poston, L, Caleyachetty, R, Cnattingius, S, Corvalan, C, Uauy, R, Herring, S, et al. Preconceptional and maternal obesity: epidemiology and health consequences. Lancet Diabetes Endocrinol. 2016;4(12):1025–36. Epub 2016/10/17. doi: 10.1016/S2213–8587(16)30217–0. PubMed PMID: 27743975.Google Scholar
Golab, P, Santos, S, Voerman, E, Lawlor, DA, Jaddoe, VWV, Gaillard, R, et al. Influence of maternal obesity on the association between common pregnancy complications and risk of childhood obesity: an individual participant data meta-analysis. Lancet Child Adolesc Health. 2018;2(11):812–21. Epub 2018/09/12. doi: 10.1016/S2352-4642(18)30273-6. PubMed PMID: 30201470; PubMed Central PMCID: PMCPMC6196075.Google Scholar
Kitsantas, P, Gaffney, KF. Risk profiles for overweight/obesity among preschoolers. Early Hum Dev. 2010;86(86):563–8.Google Scholar
Rooney, BL, Mathiason, MA, Schauberger, CW. Predictors of obesity in childhood, adolescence, and adulthood in a birth cohort. Maternal Child Health J. 2011;DOI10.1007/s10995-010-0689-1.Google Scholar
Dodd, JM, Catcheside, B, Scheil, W. Can shoulder dystocia be reliably predicted? Aust N Z J Obstet Gynaecol. 2012;52(3):248–52.Google Scholar
Silverman, BL, Rizzo, TA, Cho, NH, Metzger, BE. Long-term effects of the intrauterine environment. Diabetes Care. 1998;21:B142–9.Google Scholar
Dodd, JM, Crowther, CA, Antoniou, G, Baghurst, P, Robinson, JS. Screening for gestational diabetes: the effect of varying blood glucose definitions in the prediction of adverse maternal and infant health outcomes. Aust N Z J Obstet Gynaecol. 2007;47(4):307–12.Google Scholar
Catalano, PM, McIntyre, HD, Cruickshank, JK, McCance, DR, Dyer, AR, Metzger, BE, et al. The hyperglycemia and adverse pregnancy outcome study: associations of GDM and obesity with pregnancy outcomes. Diabetes Care. 2012;35(4):780–6.Google Scholar
Nelson, SM, Matthews, P, Poston, L. Maternal metabolism and obesity: modifiable determinants of pregnancy outcome. Hum Reprod Update. 2010;16(3):255–75.Google Scholar
Challier, JC, Basu, S, Bintein, T, Minium, J, Hotmire, K, Catalano, PM, et al. Obesity in pregnancy stimulates macrophage accumulation and inflammation in the placenta. Placenta. 2008;29(3):274–81.Google Scholar
Ramsay, JE, Ferrell, WR, Crawford, L, Wallace, AM, Greer, IA, Sattar, N. Maternal obesity is associated with dysregulation of metabolic, vascular, and inflammatory pathways. J Clin Endocrinol Metab. 2002;87(9):4231–7.Google Scholar
Catalano, PM, Hauguel-De, Mouzon, S. Is it time to revisit the Pedersen hypothesis in the face of the obesity epidemic? Am J Obstet Gynecol. 2011;204(6):479–87.Google Scholar
Ingvorsen, C, Brix, S, Ozanne, SE, Hellgren, LI. The effect of maternal Inflammation on foetal programming of metabolic disease. Acta Physiol (Oxf). 2015;214(4):440–9. Epub 2015/05/27. doi: 10.1111/apha.12533. PubMed PMID: 26011013.Google Scholar
Pederson, J. Weight and length at birth of infants of diabetic mothers. Acta Endocrinologica. 1954;16 (4)330–42.Google Scholar
Sattar, N, Wannamethee, SG, Forouhi, NG. Novel biochemical risk factors for type 2 diabetes: pathogenic insights or prediction possibilities? Diabetologia. 2008;51(6):926–40.Google Scholar
McLaughlin, T, Abbasi, F, Lamendola, C, al e. Differentiation between obesity and insulin resistance in the association with C-reactive protein. Circulation. 2002;106(23): 2908–12.Google Scholar
Sibai, B, Dekker, G, Kupferminc, M. Pre-eclampsia. Lancet. 2005;365(9461):785–99.Google Scholar
Rodie, VA, Freeman, DJ, Sattar, N, Greer, IA. Pre-eclampsia and cardiovascular disease: metabolic syndrome of pregnancy? Atherosclerosis. 2004;175(2):189–202.Google Scholar
Grivell, RM, Yelland, LN, Deussen, A, Crowther, CA, Dodd, JM. Antenatal dietary and lifestyle advice for women who are overweight or obese and the effect on fetal growth and adiposity: the LIMIT randomised trial. BJOG. 2016;123(2):233–43.Google Scholar
Poprzeczny, AJ, Louise, J, Deussen, AR, Dodd, JM. The mediating effects of gestational diabetes on fetal growth and adiposity in women who are overweight and obese: secondary analysis of the LIMIT randomised trial. BJOG. 2018;125(12):1558–66.Google Scholar
Schaefer-Graf, UM, Heuer, R, Kilavuz, O, Pandura, A, Henrich, W, Vetter, K. Maternal obesity not maternal glucose values correlates best with high rates of fetal macrosomia in pregnancies complicated by gestational diabetes. J Perinat Med. 2002;30(4):313–21. Epub 2002/09/19. doi: 10.1515/jpm.2002.046. PubMed PMID: 12235720.Google Scholar
Donahue, SM, Rifas-Shiman, SL, Gold, DR, Jouni, ZE, Gillman, MW, Oken, E. Prenatal fatty acid status and child adiposity at age 3 y: results from a US pregnancy cohort. Am J Clin Nutr. 2011;93(4):780–8.Google Scholar
Moon, RJ, Harvey, NC, Robinson, SM, Ntani, G, Davies, JH, Inskip, HM, et al. Maternal plasma polyunsaturated fatty acid status in late pregnancy is associated with offspring body composition in childhood. J Clin Endocrinol Metab. 2013;98(1):299–307.Google Scholar
Gaillard, R, Steegers, EA, Franco, OH, Hofman, A, Jaddoe, VW. Maternal weight gain in different periods of pregnancy and childhood cardio-metabolic outcomes. The Generation R Study. Int J Obes (Lond). 2015;39(4):677–85.Google Scholar
Gaillard, R, Welten, M, Oddy, WH, Beilin, LJ, Mori, TA, Jaddoe, VWV, et al. Associations of maternal prepregnancy body mass index and gestational weight gain with cardio-metabolic risk factors in adolescent offspring: a prospective cohort study. BJOG. 2016;123(2):207–16.Google Scholar
Beillin, L, Huang, RC. Childhood obesity, hypertension, the metabolic syndrome and adult cardiovascular disease. Clin Exp Pharmacol Physiol. 2008;35(4):409–11.Google Scholar
James, WP. The epidemiology of obesity: the size of the problem. J Intern Med. 2008;263(4):336–52.Google Scholar
Thornburg, KL. The programming of cardiovascular disease. J Dev Orig Health Dis. 2015;6(5): 366–76Google Scholar
Simmons, R. Perinatal programming of obesity. Semin Perinatol. 2008;32 (5): 371–4.Google Scholar
Nivoit, P, Morens, C, Van Assche, FA, Jansen, E, Poston, L, Remacle, C, et al. Established diet-induced obesity in female rats leads to offspring hyperphagia, adiposity and insulin resistance. Diabetologia. 2009;52(6):1133–42.Google Scholar
Nathanielsz, PW, Ford, SP, Long, NM, Vega, CC, Reyes-Castro, LA, Zambrano, E. Interventions to prevent adverse fetal programming due to maternal obesity during pregnancy. Nutr Rev. 2013;71 Suppl 1:S7887.Google Scholar
Poston, L. Maternal obesity, gestational weight gain and diet as determinants of offspring long term health. Best Pract Res Clin Endocrinol Metab. 2012 26(5):627–39.Google Scholar
Gluckman, PD, Hanson, MA, Beedle, AS. Non-genomic transgenerational inheritance of disease risk. Bioessays. 2007;29(2):145–54. Epub 2007/01/18. doi: 10.1002/bies.20522. PubMed PMID: 17226802.Google Scholar
Sharp, GC, for the PACE Consortium. Maternal BMI at the start of pregnancy and offspring epigenome-wide DNA methylation: findings from the pregnancy and childhood epigenetics consortium. Hum Mol Genet. 2017;26(20):4067–85.Google Scholar
Martin, CL, Jima, D, Sharp, GC, McCullough, LE, Park, SS, Gowdy, KM, et al. Maternal pre-pregnancy obesity, offspring cord blood DNA methylation, and offspring cardiometabolic health in early childhood: an epigenome-wide association study. Epigenetics. 2019;14(4):325–40.Google Scholar
Antoun, E, Kitaba, NT, Titcombe, P, Dalrymple, KV, Garratt, ES, Barton, SJ, et al. Maternal dysglycaemia, changes in the infant’s epigenome modified with a diet and physical activity intervention in pregnancy: Secondary analysis of a randomised control trial. PLoS Med. 2020;17(11):e1003229. Epub 2020/11/06. doi: 10.1371/journal.pmed.1003229. PubMed PMID: 33151971; PubMed Central PMCID: PMCPMC7643947Google Scholar
The International Weight Management in Pregnancy (i-WIP) Collaborative Group. Effect of diet and physical activity based interventions in pregnancy on gestational weight gain and pregnancy outcomes: meta-analysis of individual participant data from randomised trials. BMJ. 2017;358:j3119 http://dx.doi.org/10.1136/bmj.j3119.Google Scholar
Dodd, JM, Grivell, RM, Deussen, AR, Hague, W. Metformin for women who are overweight or obese during pregnancy for improving maternal and infant outcomes. Cochrane Database Syst Rev. 2018;7(7):CD010564Google Scholar
Dodd, JM, Louise, J, Deussen, AR, Grivell, RM, Dekker, G, McPhee, AJ, et al. Effect of metformin in addition to dietary and lifestyle advice for pregnant women who are overweight or obese: the GRoW randomised, double-blind placebo-controlled trial. Lancet Diabetes Endocrinol. 2019;7(1):15–24.Google Scholar
Dodd, JM, Cramp, CS, Sui, Z, Yelland, LN, Deussen, AR, Grivell, RM, et al. Effects of antenatal lifestyle advice for women who are overweight or obese on maternal diet and physical activity: the LIMIT randomised trial. BMC Med. 2014;12:161; www.biomedcentral.com/1741-7015/12/161.Google Scholar
Dodd, JM, Turnbull, DA, McPhee, AJ, Deussen, AR, Grivell, RM, Yelland, LN, et al. Antenatal lifestyle advice for women who are overweight or obese: the LIMIT randomised trial. BMJ. 2014;348:g1285.Google Scholar
Dodd, JM, McPhee, AJ, Turnbull, DA, Yelland, LN, Deussen, AR, Grivell, RM, et al. The effect of antenatal lifestyle advice for women who are overweight or obese on neonatal health: the LIMIT randomised trial. BMC Med. 2014;12:163; www.biomedcentral.com/1741-7015/12/163.Google Scholar
Dodd, JM, McPhee, AJ, Deussen, AR, Louise, J, Yelland, LN, Owens, JA, et al. Effects of an antenatal dietary intervention in overweight and obese women on 6 month infant outcomes: follow-up from the LIMIT randomised trial. Int J Obes (Lond). 2018;42(7):1326–1335.Google Scholar
Dodd, JM, Louise, J, Deussen, A. R., McPhee, AJ, Deussen, AR, Owens, JA, et al. Prenatal diet and child growth at 18 months. Pediatrics 2018 Sep;142(3). pii: e20180035. doi: 10.1542/peds.2018-0035. Epub 2018 Aug 8.Google Scholar
Dodd, JM, Deussen, AR, Louise, J. Effects of an antenatal dietary intervention in women with obesity or overweight on child outcomes at 3–5 years of age: LIMIT randomised trial follow-up. Int J Obes (Lond). 2020;44(7) 1531–35. doi: 10.1038/s41366-020-0560-4.Google Scholar
Vinter, CA, Jensen, DM, Ovesen, P, Beck-Nielsen, H, Tanvig, M, Lamont, RF, et al. Postpartum weight retention and breastfeeding among obese women from the randomized controlled Lifestyle in Pregnancy (LiP) trial. Acta Obstet Gynecol Scand. 2014;93(8):794–801.Google Scholar
Horan, MK, McGowan, CA, Gibney, ER, Byrne, J, Donnelly, JM, McAuliffe, FM. Maternal Nutrition and Glycaemic Index during Pregnancy Impacts on Offspring Adiposity at 6 Months of Age--Analysis from the ROLO Randomised Controlled Trial. Nutrients. 2016;Jan 4;8(1). pii: E7. doi: 10.3390/nu8010007.Google Scholar
Patel, N, Godfrey, KM, Pasupathy, D, Levin, J, Flynn, AC, Hayes, L, et al. Infant adiposity following a randomised controlled trial of a behavioural intervention in obese pregnancy. Int J Obes (Lond). 2017;41(7);1018–26. doi: 10.1038/ijo.2017.44.Google Scholar
Tanvig, M, Vinter, CA, Jørgensen, JS, Wehberg, S, Ovesen, PG, Beck-Nielsen, H, et al. Effects of lifestyle intervention in pregnancy and anthropometrics at birth on offspring metabolic profile at 2.8 years: results from the Lifestyle in Pregnancy and Offspring (LiPO) study. J Clin Endocrinol Metab. 2015;100(1):175–83.Google Scholar
Tanvig, M, Vinter, CA, Jørgensen, JS, Wehberg, S, Ovesen, PG, Lamont, RF, et al. Anthropometrics and body composition by dual energy X-ray in children of obese women: a follow-up of a randomized controlled trial (the Lifestyle in Pregnancy and Offspring [LiPO] study). PLoS One. 2014;Feb 24;9(2):e89590.Google Scholar
Geraghty, AA, Alberdi, G, O’Sullivan, EJ, O’Brien, EC, Crosbie, B, Twomey, PJ, et al. Maternal Blood Lipid Profile during Pregnancy and Associations with Child Adiposity: Findings from the ROLO Study. PLoS One. 2016;11(8):e0161206.Google Scholar
Horan, MK, Donnelly, JM, McGowan, CA, Gibney, ER, McAuliffe, FM. The association between maternal nutrition and lifestyle during pregnancy and 2-year-old offspring adiposity: analysis from the ROLO study. Z Gesundh Wiss. 2016;24(5):427–36.Google Scholar
Scully, H, Alberdi, G, Segurado, R, McNamara, A, Lindsay, K, Horan, M, et al. Child Care Exposure Influences Childhood Adiposity at 2 Years: Analysis from the ROLO Study. Child Obes. 2017;13(2):93–101.Google Scholar
Louise, J, Poprzeczny, AJ, Deussen, AR, Vinter, C, Tanvig, M, Jensen, DM, et al. The effects of dietary and lifestyle interventions among pregnant women with overweight or obesity on early childhood outcomes: an individual participant data meta-analysis from randomised trials. BMC Med. 2021;19(1):128. Epub 2021/06/03. doi: 10.1186/s12916-021-01995-6. PubMed PMID: 34074261; PubMed Central PMCID: PMCPMC8170974.Google Scholar
Institute of Medicine. Weight gain during pregnancy: reexamining the guidelines. Washington D.C.:2009.Google Scholar
Hanson, MA, Barker, M, Bustreo, F, Dodd, JM, Kumanyika, S, Norris, S, et al. Interventions to prevent preconception and maternal obesity. Lancet Diabetes Endocrinol 2017;5(1):65–76.Google Scholar

References

Van den Bergh, BR, Mulder, E.J.Mennes, M., et al. Antenatal maternal anxiety and stress and the neurobehavioural development of the fetus and child: links and possible mechanisms. A review. Neurosci Biobehav Rev. 2005; 29(2): pp. 237–58.Google Scholar
Van den Bergh, BRH., van den Heuvel, M.I., Lahti, M.,et al. Prenatal developmental origins of behavior and mental health: the influence of maternal stress in pregnancy. Neurosci Biobehav Rev, 2020. 117: pp. 26–64.Google Scholar
Loomans, EM, Van, Dijk, A.E., Vrijkotte, T.G.M., et al., Psychosocial stress during pregnancy is related to adverse birth outcomes: results from a large multi-ethnic community-based birth cohort. Eur J Public Health, 2012. 23(3): pp. 485–91.Google Scholar
Melville, JL, Gavin, A., Guo, Y., et al., Depressive disorders during pregnancy: prevalence and risk factors in a large urban sample. Obstetrics and Gynecology, 2010. 116(5): pp. 1064–70.Google Scholar
Stetler, C. and Miller, G.E., Depression and hypothalamic-pituitary-adrenal activation: a quantitative summary of four decades of research. Psychosom Med, 2011. 73(2): pp. 114–26.Google Scholar
Lima, S.A.M., El, Dib, R.P., Rodrigues, M.R.K., et al., Is the risk of low birth weight or preterm labor greater when maternal stress is experienced during pregnancy? A systematic review and meta-analysis of cohort studies. PLoS One, 2018. 13(7): p. e0200594.Google Scholar
Tarabulsy, G.M., Pearson, J., Vaillancourt-Morel, M.P., et al., Meta-analytic findings of the relation between maternal prenatal stress and anxiety and child cognitive outcome. J Dev Behav Pediatr, 2014. 35(1): pp. 38–43.Google Scholar
King, S., Dancause, K., Turcotte, Tremblay, A.M., et al., Using natural disasters to study the effects of prenatal maternal stress on child health and development. Birth Defects Res C Embryo Today, 2012. 96(4): pp. 273–88.Google Scholar
Laplante, D.P., Brunet, A., Schmitz, N., et al., Project Ice Storm: prenatal maternal stress affects cognitive and linguistic functioning in 5 1/2-year-old children. J Am Acad Child Adolesc Psychiatry, 2008. 47(9): pp. 1063–72.Google Scholar
Lin, B., Crnic, K.A., Luecken, L.J., et al., Maternal prenatal stress and infant regulatory capacity in Mexican Americans. Infant Behav Dev, 2014. 37(4): pp. 571–82.Google Scholar
DiPietro, J.A., Novak, M.F., Costigan, K.A., et al., Maternal psychological distress during pregnancy in relation to child development at age two. Child Development, 2006. 77(3): pp. 573–87.Google Scholar
Glover, V., O’Connor, TG, and O’Donnell, K, Prenatal stress and the programming of the HPA axis. Neurosci Biobehav Rev, 2010. 35(1): pp. 17–22.Google Scholar
Glover, V., Annual research review: prenatal stress and the origins of psychopathology: an evolutionary perspective. J Child Psychol Psychiatry, 2011. 52(4): pp. 356–67.Google Scholar
Winsper, C, Wolke, D, and Lereya, T., Prospective associations between prenatal adversities and borderline personality disorder at 11–12 years. Psychol Med, 2015. 45(5): pp. 1025–37.Google Scholar
Pearson, RM, Bornstein, M.H., Cordero, M., et al., Maternal perinatal mental health and offspring academic achievement at age 16: the mediating role of childhood executive function. J Child Psychol Psychiatry, 2016. 57(4): pp. 491–501.Google Scholar
Yildiz, P.D., Ayers, S, and Phillips, L, The prevalence of posttraumatic stress disorder in pregnancy and after birth: a systematic review and meta-analysis. J Affect Disord, 2017. 208: pp. 634–45.Google Scholar
Cook, N., Ayers, S, and Horsch, A, Maternal posttraumatic stress disorder during the perinatal period and child outcomes: a systematic review. J Affect Disord, 2018. 225: pp. 18–31.Google Scholar
Miranda, J.K., de la Osa, N., Granero, R., et al., Maternal experiences of childhood abuse and intimate partner violence: psychopathology and functional impairment in clinical children and adolescents. Child Abuse Negl, 2011. 35(9): pp. 700–11.Google Scholar
Braga, L.L., Mello, M.F., and Fiks, J.P., Transgenerational transmission of trauma and resilience: a qualitative study with Brazilian offspring of Holocaust survivors. BMC Psychiatry, 2012. 12(1): p. 134.Google Scholar
Keenan, K., Hipwell, A.E., Class, Q.A., et al., Extending the developmental origins of disease model: impact of preconception stress exposure on offspring neurodevelopment. Dev Psychobiol, 2018. 60(7): pp. 753–64.Google Scholar
Hutchens, B.F. and Kearney, J., Risk factors for postpartum depression: an umbrella review. J Midwifery Womens Health, 2020. 65(1): pp. 96–108.Google Scholar
Brummelte, S. and Galea, L.A., Postpartum depression: etiology, treatment and consequences for maternal care. Horm Behav, 2016. 77: pp. 153–66.CrossRefGoogle Scholar
Kingston, D., Tough, S., and Whitfield, H., Prenatal and postpartum maternal psychological distress and infant development: a systematic review. Child Psychiatry Hum Dev, 2012. 43(5): pp. 683–714.Google Scholar
Werner, E., Miller, M., Osborne, L., et al., Preventing postpartum depression: review and recommendations. Arch Womens Ment Health, 2015. 18(1): pp. 41–60.Google Scholar
Glover, V., O'Donnell, K.J., O'Connor, T.G., et al., Prenatal maternal stress, fetal programming, and mechanisms underlying later psychopathology-A global perspective. Dev Psychopathol, 2018. 30(3): pp. 843–54.Google Scholar
Franke, K., Van den Bergh, B.R., de Rooij, S.R., et al., Effects of maternal stress and nutrient restriction during gestation on offspring neuroanatomy in humans. Neurosci Biobehav Rev, 2020. 117: pp. 5–25.Google Scholar
Khashan, A.S., Abel, K.M., McNamee, R., et al., Higher risk of offspring schizophrenia following antenatal maternal exposure to severe adverse life events. Arch Gen Psychiatry, 2008. 65(2): pp. 146–52.Google Scholar
Class, Q.A., Abel, K.M., Khashan, A.S., et al., Offspring psychopathology following preconception, prenatal and postnatal maternal bereavement stress. Psychological Medicine, 2014. 44(1): pp. 71–84.Google Scholar
Goodman, J.H., Perinatal depression and infant mental health. Arch Psychiatr Nurs, 2019. 33(3): pp. 217–24.Google Scholar
Chan, J.C., Nugent, B.M., and Bale, T.L., Parental advisory: maternal and paternal stress can impact offspring neurodevelopment. Biol Psychiatry, 2018. 83(10): pp. 886–94.Google Scholar
Hicks, L.M., Swales, D.A., Garcia, S.E., et al., Does prenatal maternal distress contribute to sex differences in child psychopathology? Curr Psychiatry Rep, 2019. 21(2): p. 7.Google Scholar
Rakers, F., Rupprecht, S., Dreiling, M., et al., Transfer of maternal psychosocial stress to the fetus. Neurosci Biobehav Rev, 2020. 117: pp. 185–97.Google Scholar
Alcantara-Alonso, V., Panetta, P., De, Gortari, P., et al., Corticotropin-releasing hormone as the homeostatic rheostat of feto-maternal symbiosis and developmental programming in utero and neonatal life. Front Endocrinol (Lausanne), 2017. 8: p. 161.Google Scholar
Troubat, R., Barone, P., Leman, S., et al., Neuroinflammation and depression: a review. Eur J Neurosci, 2021. 53(1): pp. 151–71.Google Scholar
Rudzki, L. and Maes, M., The Microbiota-Gut-Immune-Glia (MGIG) axis in major depression. Mol Neurobiol, 2020. 57(10): pp. 4269–95.Google Scholar
Rutsch, A., Kantsjo, J.B., and Ronchi, F., The Gut-Brain Axis: how microbiota and host inflammasome influence brain physiology and pathology. Front Immunol, 2020. 11: p. 604179.Google Scholar
Jasarevic, E., Howard, C.D., Morrison, K., et al., The maternal vaginal microbiome partially mediates the effects of prenatal stress on offspring gut and hypothalamus. Nat Neurosci, 2018. 21(8): pp. 1061–71.Google Scholar
Chong, C.Y.L., Bloomfield, F.H., and O’Sullivan, J.M., Factors affecting gastrointestinal microbiome development in neonates. Nutrients, 2018. 10(3).Google Scholar
Müller, J.J., Antonow, -Schlorke, I., Kroegel, N., et al., Cardiovascular effects of prenatal stress – Are there implications for cerebrovascular, cognitive and mental health outcome? Neurosci Biobehav Rev, 2020. 117: pp. 78–97.Google Scholar
Cao-Lei, L., De, Rooij, S.R., King, S., et al., Prenatal stress and epigenetics. Neurosci Biobehav Rev, 2020. 117: pp. 198–210.Google Scholar
Rodgers, A.B. and Bale, T.L., Germ cell origins of posttraumatic stress disorder risk: the transgenerational impact of parental stress experience. Biol Psychiatry, 2015. 78(5): pp. 307–14.Google Scholar
Seckl, J.R., Nyirenda, M.J., Walker, B.R., et al., Glucocorticoids and fetal programming. Biochem Soc Trans, 1999. 27(2): pp. 74–8.Google Scholar
Braun, K., Bock, J., Wainstock, T., et al., Experience-induced transgenerational (re-)programming of neuronal structure and functions: Impact of stress prior and during pregnancy. Neurosci Biobehav Rev, 2020. 117: pp. 281–96.Google Scholar
Baker, J., Liu, J.P., Robertson, E.J., et al., Role of insulin-like growth factors in embryonic and postnatal growth. Cell, 1993. 75(1): pp. 73–82.Google Scholar
Numakawa, T., Odaka, H., and Adachi, N., Actions of brain-derived neurotrophic factor and glucocorticoid stress in neurogenesis. Int J Mol Sci, 2017. 18(11): p. 2312.Google Scholar
Kundakovic, M. and Jaric, I., The epigenetic link between prenatal adverse environments and neurodevelopmental disorders. Genes (Basel), 2017. 8(3): p. 104.Google Scholar
Liu, J., Speder, P., and Brand, A.H., Control of brain development and homeostasis by local and systemic insulin signalling. Diabetes Obes Metab, 2014. 16 Suppl 1: pp. 16–20.Google Scholar
Basta-Kaim, A., Szczesny, E., Glombik, K., et al., Prenatal stress leads to changes in IGF-1 binding proteins network in the hippocampus and frontal cortex of adult male rat. Neuroscience, 2014. 274: pp. 59–68.Google Scholar
Schepanski, S., Buss, C., Hanganu, -Opatz, I.L., et al., Prenatal immune and endocrine modulators of offspring’s brain development and cognitive functions later in life. Front Immunol, 2018. 9: p. 2186.Google Scholar
Kiss, D., Ambeskovic, M., Montina, T., et al., Stress transgenerationally programs metabolic pathways linked to altered mental health. Cell Mol Life Sci, 2016. 73(23): pp. 4547–57.Google Scholar
Bauer, S., Kerr, B.J., and Patterson, P.H., The neuropoietic cytokine family in development, plasticity, disease and injury. Nat Rev Neurosci, 2007. 8(3): pp. 221–32.Google Scholar
Stepanichev, M., Dygalo, N.N., Grigoryan, G., et al., Rodent models of depression: neurotrophic and neuroinflammatory biomarkers. Biomed Res Int, 2014. 2014: p. 932757.Google Scholar

References

Global Burden of Disease 2017 Risk Factor Collaborators. Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018; 392, 1923–1994.Google Scholar
World Health Organisation. Air pollution and child health: prescribing clean air. Geneva: World Health Organisation, 2018.Google Scholar
World Heatlh Organisation. Burden of disease from environmental noise. Quantification of healthy life years lost in Europe. Geneva: World Health Organisation, 2011.Google Scholar
Pedersen, M, Stayner, L, Slama, R, et al. Ambient air pollution and pregnancy-induced hypertensive disorders: a systematic review and meta-analysis. Hypertension. 2014; 64, 494–500.Google Scholar
National Toxicology Program. NTP monograph on the systematic review of traffic-related air pollution and hypertensive disorders of pregnancy. NTP Monogr. 2019; 7; doi: 10.22427/NTP-MGRAPH-7.Google Scholar
Hu, CY, Gao, X, Fang, Y, et al. Human epidemiological evidence about the association between air pollution exposure and gestational diabetes mellitus: systematic review and meta-analysis. Environ Res. 2020; 180, 108843.Google Scholar
Ha, S, Sundaram, R, Buck Louis, GM, et al. Ambient air pollution and the risk of pregnancy loss: a prospective cohort study. Fertil Steril. 2018; 109, 148–153.Google Scholar
Siddika, N, Balogun, HA, Amegah, AK, Jaakkola, JJ. Prenatal ambient air pollution exposure and the risk of stillbirth: systematic review and meta-analysis of the empirical evidence. Occup Environ Med. 2016; 73, 573–581.Google Scholar
Pedersen, M, Giorgis-Allemand, L, Bernard, C, et al. Ambient air pollution and low birthweight: a European cohort study (ESCAPE). Lancet Respir Med. 2013; 1, 695–704.Google Scholar
Huang, I, Mak, D, Cheung, P, Abraham, M, Clemens, T, Turner, S. A systematic review of associations between maternal exposures during pregnancy other than smoking and antenatal fetal measurements. Environ Res. 2019; 173, 528–538.Google Scholar
Malley, CS, Kuylenstierna, JC, Vallack, HW, Henze, DK, Blencowe, H, Ashmore, MR. Preterm birth associated with maternal fine particulate matter exposure: a global, regional and national assessment. Environ Int. 2017; 101, 173–182.Google Scholar
Vrijheid, M, Martinez, D, Manzanares, S, et al. Ambient air pollution and risk of congenital anomalies: a systematic review and meta-analysis. Environ Health Perspect. 2011; 119, 598–606.Google Scholar
Watts, N, Amann, M, Arnell, N, et al. The 2019 report of The Lancet Countdown on health and climate change: ensuring that the health of a child born today is not defined by a changing climate. Lancet. 2019; 394, 1836–1878.Google Scholar
Khreis, H, Nieuwenhuijsen, MJ. Traffic-related air pollution and childhood asthma: recent advances and remaining gaps in the exposure assessment methods. Int J Environ Res Public Health. 2017; 14(3), 312Google Scholar
Achakulwisut, P, Brauer, M, Hystad, P, Anenberg, SC. Global, national, and urban burdens of paediatric asthma incidence attributable to ambient NO2 pollution: estimates from global datasets. Lancet Planet Health. 2019; 3, e166e178.Google Scholar
Gehring, U, Wijga, AH, Koppelman, GH, Vonk, JM, Smit, HA, Brunekreef, B. Air pollution and the development of asthma from birth until young adulthood. Eur Respir J. 2020; 56(1), 2000147.Google Scholar
Götschi, T, Heinrich, J, Sunyer, J, Künzli, N. Long-term effects of ambient air pollution on lung function: a review. Epidemiology. 2008; 19, 690–701.Google Scholar
Gauderman, WJ, Urman, R, Avol, E, et al. Association of improved air quality with lung development in children. N Engl J Med. 2015; 372, 905–913.Google Scholar
King, C, Kirkham, J, Hawcutt, D, Sinha, I. The effect of outdoor air pollution on the risk of hospitalisation for bronchiolitis in infants: a systematic review. PeerJ. 2018; 6, e5352.Google Scholar
Nhung, NTT, Amini, H, Schindler, C, et al. Short-term association between ambient air pollution and pneumonia in children: a systematic review and meta-analysis of time-series and case-crossover studies. Environ Pollut. 2017; 230, 1000–1008.Google Scholar
Bowatte, G, Lodge, C, Lowe, AJ, et al. The influence of childhood traffic-related air pollution exposure on asthma, allergy and sensitization: a systematic review and a meta-analysis of birth cohort studies. Allergy. 2015; 70, 245–256.Google Scholar
Malmqvist, E, Larsson, HE, Jönsson, I, et al. Maternal exposure to air pollution and type 1 diabetes – accounting for genetic factors. Environ Res. 2015; 140, 268–274.Google Scholar
Jerrett, M, McConnell, R, Wolch, J, et al. Traffic-related air pollution and obesity formation in children: a longitudinal, multilevel analysis. Environ Health. 2014; 13, 49.Google Scholar
Flores-Pajot, MC, Ofner, M, Do, MT, Lavigne, E, Villeneuve, PJ. Childhood autism spectrum disorders and exposure to nitrogen dioxide, and particulate matter air pollution: a review and meta-analysis. Environ Res. 2016; 151, 763–776.Google Scholar
Aghaei, M, Janjani, H, Yousefian, F, Jamal, A, Yunesian, M. Association between ambient gaseous and particulate air pollutants and attention deficit hyperactivity disorder (ADHD) in children; a systematic review. Environ Res. 2019; 173, 135–156.Google Scholar
Sunyer, J, Suades-González, E, García-Esteban, R, et al. Traffic-related air pollution and attention in primary school children: short-term association. Epidemiology. 2017; 28, 181–189.Google Scholar
Herting, MM, Younan, D, Campbell, CE, Chen, JC. Outdoor air pollution and brain structure and function from across childhood to young adulthood: a methodological review of brain MRI studies. Front Public Health. 2019; 7, 332.Google Scholar
Filippini, T, Hatch, EE, Rothman, KJ, et al. Association between outdoor air pollution and childhood leukemia: a systematic review and dose-response meta-analysis. Environ Health Perspect. 2019; 127, 46002.Google Scholar
Grandjean, P, Landrigan, PJ. Neurobehavioural effects of developmental toxicity. Lancet Neurology. 2014; 13, 330–338.Google Scholar
Laiosa, MD, Tate, ER. Fetal hematopoietic stem cells are the canaries in the coal mine that portend later life immune deficiency. Endocrinology. 2015; 156, 3458–3465.Google Scholar
World Health Organization Inheriting a sustainable world? Atlas on children’s health and the environment. Geneva: World Health Organization, 2017 9789241511773.Google Scholar
Govarts, E, Nieuwenhuijsen, M, Schoeters, G, et al. Birth weight and prenatal exposure to polychlorinated biphenyls (PCBs) and dichlorodiphenyldichloroethylene (DDE): a meta-analysis within 12 European Birth Cohorts. Environ Health Perspect. 2012; 120, 162–170.Google Scholar
Kahn, LG, Philippat, C, Nakayama, SF, Slama, R, Trasande, L. Endocrine-disrupting chemicals: implications for human health. Lancet Diabetes Endocrinol. 2020; 8, 703–718.Google Scholar
Fenton, SE, Ducatman, A, Boobis, A, et al. Per- and polyfluoroalkyl substance toxicity and human health review: current state of knowledge and strategies for informing future research. Environ Toxicol Chem. 2021; 40, 606–630.Google Scholar
Tang, J, Zhai, JX. Distribution of polybrominated diphenyl ethers in breast milk, cord blood and placentas: a systematic review. Environ Sci Pollut Res Int. 2017; 24, 21548–21573.Google Scholar
Zhang, T, Zhou, X, Xu, A, et al. Toxicity of polybrominated diphenyl ethers (PBDEs) on rodent male reproductive system: a systematic review and meta-analysis of randomized control studies. Sci Total Environ. 2020; 720, 137419–137419.Google Scholar
Hauser, R, Skakkebaek, NE, Hass, U, et al. Male reproductive disorders, diseases, and costs of exposure to endocrine-disrupting chemicals in the European Union. J Clin Endocrinol Metab. 2015; 100, 1267–1277.Google Scholar
Ferguson, KK, O’Neill, MS, Meeker, JD. Environmental contaminant exposures and preterm birth: a comprehensive review. J Toxicol Environ Health B Crit Rev. 2013; 16, 69–113.Google Scholar
Khanam, R, Kumar, I, Oladapo-Shittu, O, et al. Prenatal environmental metal exposure and preterm birth: a scoping review. Int J Environ Res Public Health. 2021; 18(2), 573CrossRefGoogle Scholar
Bihaqi, SW. Early life exposure to lead (Pb) and changes in DNA methylation: relevance to Alzheimer’s disease. Rev Environ Health. 2019; 34, 187–195.Google Scholar
Mustieles, V, Fernández, MF. Bisphenol A shapes children’s brain and behavior: towards an integrated neurotoxicity assessment including human data. Environ Health. 2020; 19, 66–66.Google Scholar
Radke, EG, Braun, JM, Meeker, JD, Cooper, GS. Phthalate exposure and male reproductive outcomes: a systematic review of the human epidemiological evidence. Environ Int. 2018; 121, 764–793.Google Scholar
Cano-Sancho, G, Salmon, AG, Merrill, MAL. Obesity: integrated systematic review and meta-analysis. Environ Health Perspect. 2017, 1–15.Google Scholar
Bonde, JP, Flachs, EM, Rimborg, S, et al. The epidemiologic evidence linking prenatal and postnatal exposure to endocrine disrupting chemicals with male reproductive disorders: a systematic review and meta-analysis. Hum Reprod Update. 2016; 23, 104–125.Google Scholar
Kreitinger, JM, Beamer, CA, Shepherd, DM. Environmental immunology: lessons learned from exposure to a select panel of immunotoxicants. J Immunol. 2016; 196, 3217–3225.Google Scholar
Grieb, ZA, Ragan, CM. The effects of perinatal SSRI exposure on anxious behavior and neurobiology in rodent and human offspring. Eur Neuropsychopharmacol. 2019; 29, 1169–1184.Google Scholar
Andrade, C. Valproate in pregnancy. J Clin Psychiatry. 2018; 79, 13–16.Google Scholar
McCrae, JC, Morrison, EE, MacIntyre, IM, Dear, JW, Webb, DJ. Long-term adverse effects of paracetamol – a review. Br J Clin Pharmacol. 2018; 84, 2218–2230.Google Scholar
Sheffield, PE, Landrigan, PJ. Global climate change and children’s health: threats and strategies for prevention. Environ Health Perspect. 2011; 119, 291–298.Google Scholar
Xu, Z, Sheffield, PE, Su, H, Wang, X, Bi, Y, Tong, S. The impact of heat waves on children’s health: a systematic review. Int J Biometeorol. 2014; 58, 239–247.Google Scholar
Kuehn, L, McCormick, S. Heat exposure and maternal health in the face of climate change. Int J Environ Res Public Health. 2017; 14(8), 853Google Scholar
Park, CS, Vogel, E, Larson, LM, Myers, SS, Daniel, M, Biggs, BA. The global effect of extreme weather events on nutrient supply: a superposed epoch analysis. Lancet Planet Health. 2019; 3, e429e438.Google Scholar
Duff, EM, Cooper, ES. Neural tube defects in Jamaica following Hurricane Gilbert. Am J Public Health. 1994; 84, 473–476.Google Scholar
Harville, E, Xiong, X, Buekens, P. Disasters and perinatal health: a systematic review. Obstet Gynecol Surv. 2010; 65, 713–728.Google Scholar
Tees, MT, Harville, EW, Xiong, X, Buekens, P, Pridjian, G, Elkind-Hirsch, K. Hurricane Katrina-related maternal stress, maternal mental health, and early infant temperament. Matern Child Health J. 2010; 14, 511–518.Google Scholar
Laplante, DP, Hart, KJ, O’Hara, MW, Brunet, A, King, S. Prenatal maternal stress is associated with toddler cognitive functioning: the Iowa Flood Study. Early Hum Dev. 2018; 116, 84–92.Google Scholar
Dancause, KN, Laplante, DP, Hart, KJ, et al. Prenatal stress due to a natural disaster predicts adiposity in childhood: the Iowa Flood Study. J Obes. 2015; 2015, 570541.Google Scholar
Noyes, PD, McElwee, MK, Miller, HD, et al. The toxicology of climate change: environmental contaminants in a warming world. Environ Int. 2009; 35, 971–986.Google Scholar
Black, C, Tesfaigzi, Y, Bassein, JA, Miller, LA. Wildfire smoke exposure and human health: Significant gaps in research for a growing public health issue. Environ Toxicol Pharmacol. 2017; 55, 186–195.Google Scholar
Ngai, M, Weckman, AM, Erice, C, et al. Malaria in pregnancy and adverse birth outcomes: new mechanisms and therapeutic opportunities. Trends Parasitol. 2020; 36, 127–137.Google Scholar
Lawford, HLS, Lee, AC, Kumar, S, Liley, HG, Bora, S. Establishing a conceptual framework of the impact of placental malaria on infant neurodevelopment. Int J Infect Dis. 2019; 84, 54–65.Google Scholar
Christensen, SS, Eslick, GD. Cerebral malaria as a risk factor for the development of epilepsy and other long-term neurological conditions: a meta-analysis. Trans R Soc Trop Med Hyg. 2015; 109, 233–238.Google Scholar

References

Vandenplas, Y., Carnielli, V. P., Ksiazyk, J., Luna, M. S., Migacheva, N., Mosselmans, J. M., Picaud, J. C., Possner, M., Singhal, A., & Wabitsch, M., Factors affecting early-life intestinal microbiota development. Nutrition, 78 (2020) 110812. https://doi.org/10.1016/j.nut.2020.110812.Google Scholar
Moles, L. & Otaegui, D., The impact of diet on microbiota evolution and human health. Is diet an adequate tool for microbiota modulation? Nutrients, 12 (2020) 1654. https://doi.org/10.3390/nu12061654.Google Scholar
Eggesbø, M., Moen, B., Peddada, S., Baird, D., Rugtveit, J., Midtvedt, T., Bushel, P. R., Sekelja, M., & Rudi, K., Development of gut microbiota in infants not exposed to medical interventions. APMIS, 119 (2011) 17–35. https://doi.org/10.1111/j.1600-0463.2010.02688.x.Google Scholar
Kimura, I., Miyamoto, J., Ohue-Kitano, R., Watanabe, K., Yamada, T., Onuki, M., Aoki, R., Isobe, Y., Kashihara, D., Inoue, D., Inaba, A., Takamura, Y., Taira, S., Kumaki, S., Watanabe, M., Ito, M., Nakagawa, F., Irie, J., Kakuta, H., Shinohara, M., Iwatsuki, K., Tsujimoto, G., Ohno, H., Arita, M., Itoh, H., & Hase, K., Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science, 367 (2020). https://doi.org/10.1126/science.aaw8429.Google Scholar
Louis, P. & Flint, H. J., Formation of propionate and butyrate by the human colonic microbiota. Environmental Microbiology, 19 (2017) 29–41. https://doi.org/10.1111/1462-2920.13589.Google Scholar
Ma, J., Prince, A. L., Bader, D., Hu, M., Ganu, R., Baquero, K., Blundell, P., Alan Harris, R., Frias, A. E., Grove, K. L., & Aagaard, K. M., High-fat maternal diet during pregnancy persistently alters the offspring microbiome in a primate model. Nature Communications, 5 (2014) 1–11. https://doi.org/10.1038/ncomms4889.Google Scholar
Grech, A., C. E. Collins, A. Holmes, R. Lal, K. Duncanson, R. Taylor, & A. Gordon, Maternal exposures and the infant gut microbiome: a systematic review with meta-analysis. Gut Microbes, 13 (2021) 1–30. https://doi.org/10.1080/19490976.2021.1897210.Google Scholar
de Goffau, M. C., Lager, S., Sovio, U., Gaccioli, F., Cook, E., Peacock, S. J., Parkhill, J., Charnock-Jones, D. S., & Smith, G. C. S., Human placenta has no microbiome but can contain potential pathogens. Nature, (2019). https://doi.org/10.1038/s41586-019-1451-5.Google Scholar
Palmer, C., Bik, E. M., DiGiulio, D. B., Relman, D. A., & Brown, P. O., Development of the human infant intestinal microbiota. PLoS Biology, 5 (2007) 1556–1573. https://doi.org/10.1371/journal.pbio.0050177.Google Scholar
Daele, E. Van, Knol, J., & Belzer, C., Microbial transmission from mother to child: Improving infant intestinal microbiota development by identifying the obstacles. Critical Reviews in Microbiology, 45 (2019) 613–648. https://doi.org/10.1080/1040841X.2019.1680601.Google Scholar
Stewart, C. J., Ajami, N. J., O’Brien, J. L., Hutchinson, D. S., Smith, D. P., Wong, M. C., Ross, M. C., Lloyd, R. E., Doddapaneni, H. V., Metcalf, G. A., Muzny, D., Gibbs, R. A., Vatanen, T., Huttenhower, C., Xavier, R. J., Rewers, M., Hagopian, W., Toppari, J., Ziegler, A. G., She, J. X., Akolkar, B., Lernmark, A., Hyoty, H., Vehik, K., Krischer, J. P., & Petrosino, J. F., Temporal development of the gut microbiome in early childhood from the TEDDY study (Nature Publishing Group, 2018). https://doi.org/10.1038/s41586-018-0617-x.Google Scholar
Mitchell, C., Hogstrom, L., Bryant, A. M., Bergerat, A., Cher, A., Pochan, S., Herman, P., Carrigan, M., Sharp, K., Huttenhower, C., Lander, E. S., Vlamakis, H., Xavier, R. J., & Yassour, M., Delivery mode impacts newborn gut colonization efficiency. bioRxiv, (2020) 2020.01.29.919993. https://doi.org/10.1101/2020.01.29.919993.CrossRefGoogle Scholar
Mesa, M. D., Loureiro, B., Iglesia, I., Gonzalez, S. F., Olivé, E. L., Algar, O. G., Solana, M. J., Perez, M. J. C., Sainz, T., Martinez, L., Escuder-Vieco, D., Parra-Llorca, A., Sánchez-Campillo, M., Martinez, G. R., Roig, D. G., Gruz, M. P., Andreu-Fernández, V., Clotet, J., Sailer, S., Iglesias-Platas, I., López-Herce, J., Aras, R., Pallás-Alonso, C., de Pipaon, M. S., Vento, M., Gormaz, M., Daza, E. L., Calvo, C., & Cabañas, F., The evolving microbiome from pregnancy to early infancy: A comprehensive review. Nutrients, 12 (2020). https://doi.org/10.3390/nu12010133.Google Scholar
Enav, H., F. Bäckhed, & R. E. Ley, The developing infant gut microbiome: A strain-level view. Cell Host & Microbe, 30 (2022) 627–638. https://doi.org/10.1016/J.CHOM.2022.04.009.Google Scholar
Ho, N. T., Li, F., Lee-Sarwar, K. A., Tun, H. M., Brown, B. P., Pannaraj, P. S., Bender, J. M., Azad, M. B., Thompson, A. L., Weiss, S. T., Azcarate-Peril, M. A., Litonjua, A. A., Kozyrskyj, A. L., Jaspan, H. B., Aldrovandi, G. M., & Kuhn, L., Meta-analysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nature Communications, 9 (2018). https://doi.org/10.1038/s41467-018-06473-x.Google Scholar
Lagström, H., Rautava, S., Ollila, H., Kaljonen, A., Turta, O., Mäkelä, J., Yonemitsu, C., Gupta, J., & Bode, L., Associations between human milk oligosaccharides and growth in infancy and early childhood. The American Journal of Clinical Nutrition, (2020). https://doi.org/10.1093/ajcn/nqaa010.Google Scholar
Korpela, K., Salonen, A., Saxen, H., Nikkonen, A., Peltola, V., Jaakkola, T., de Vos, W., & Kolho, K. L., Antibiotics in early life associate with specific gut microbiota signatures in a prospective longitudinal infant cohort. Pediatric Research, (2020). https://doi.org/10.1038/s41390-020-0761-5.Google Scholar
Zeissig, S. & Blumberg, R. S., Life at the beginning: Perturbation of the microbiota by antibiotics in early life and its role in health and disease. Nature Immunology, 15 (2014) 307–310. https://doi.org/10.1038/ni.2847.Google Scholar
Vitetta, L., Vitetta, G., & Hall, S., Immunological tolerance and function: Associations between intestinal bacteria, probiotics, prebiotics, and phages. Frontiers in Immunology, 9 (2018). https://doi.org/10.3389/fimmu.2018.02240.Google Scholar
Azad, M. B., Konya, T., Persaud, R. R., Guttman, D. S., Chari, R. S., Field, C. J., Sears, M. R., Mandhane, P. J., Turvey, S. E., Subbarao, P., Becker, A. B., Scott, J. A., & Kozyrskyj, A. L., Impact of maternal intrapartum antibiotics, method of birth and breastfeeding on gut microbiota during the first year of life: A prospective cohort study. BJOG: An International Journal of Obstetrics and Gynaecology, 123 (2016) 983–993. https://doi.org/10.1111/1471-0528.13601.Google Scholar
Coker, M. O., Hoen, A. G., Dade, E., Lundgren, S., Li, Z., Wong, A. D., Zens, M. S., Palys, T. J., Morrison, H. G., Sogin, M. L., Baker, E. R., Karagas, M. R., & Madan, J. C., Specific class of intrapartum antibiotics relates to maturation of the infant gut microbiota: A prospective cohort study. BJOG: An International Journal of Obstetrics and Gynaecology, 127 (2020) 217–227. https://doi.org/10.1111/1471-0528.15799.Google Scholar
Ledder, O., Antibiotics in inflammatory bowel diseases: Do we know what we’re doing? Translational Pediatrics, 8 (2019) 42–55. https://doi.org/10.21037/tp.2018.11.02.Google Scholar
Dedrick, S., Sundaresh, B., Huang, Q., Brady, C., Yoo, T., Cronin, C., Rudnicki, C., Flood, M., Momeni, B., Ludvigsson, J., & Altindis, E., The role of gut microbiota and environmental factors in Type 1 diabetes pathogenesis. Frontiers in Endocrinology, 11 (2020). https://doi.org/10.3389/fendo.2020.00078.Google Scholar
Ridaura, V. K., Faith, J. J., Rey, F. E., Cheng, J., Duncan, A. E., Kau, A. L., Griffin, N. W., Lombard, V., Henrissat, B., Bain, J. R., Muehlbauer, M. J., Ilkayeva, O., Semenkovich, C. F., Funai, K., Hayashi, D. K., Lyle, B. J., Martini, M. C., Ursell, L. K., Clemente, J. C., Treuren, W. Van, Walters, W. A., Knight, R., Newgard, C. B., Heath, A. C., & Gordon, J. I., Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science, 341 (2013). https://doi.org/10.1126/science.1241214.Google Scholar
Soderborg, T. K., Clark, S. E., Mulligan, C. E., Janssen, R. C., Babcock, L., Ir, D., Lemas, D. J., Johnson, L. K., Weir, T., Lenz, L. L., Frank, D. N., Hernandez, T. L., Kuhn, K. A., D’Alessandro, A., Barbour, L. A., Kasmi, K. C. El, & Friedman, J. E., The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nature Communications, 9 (2018). https://doi.org/10.1038/s41467-018-06929-0.Google Scholar
Castaner, O., Goday, A., Park, Y.-M., Lee, S.-H., Magkos, F., Shiow, S.-A. T. E., & Schröder, H., The gut microbiome profile in obesity: A systematic review. International Journal of Endocrinology, 2018 (2018). https://doi.org/10.1155/2018/4095789.Google Scholar
Barrea, L., Muscogiuri, G., Annunziata, G., Laudisio, D., Pugliese, G., Salzano, C., Colao, A., & Savastano, S., From gut microbiota dysfunction to obesity: Could short-chain fatty acids stop this dangerous course? Hormones, 18 (2019) 245–250. https://doi.org/10.1007/s42000-019-00100-0.Google Scholar
Kim, K. N., Yao, Y., & Ju, S. Y., Short chain fatty acids and fecal microbiota abundance in humans with obesity: A systematic review and meta-analysis. Nutrients, 11 (2019). https://doi.org/10.3390/nu11102512.Google Scholar
Saad, M. J. A., Santos, A., & Prada, P. O., Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology, 31 (2016) 283–293. https://doi.org/10.1152/physiol.00041.2015.Google Scholar
Differding, M. K., Benjamin-Neelon, S. E., Hoyo, C., Østbye, T., & Mueller, N. T., Timing of complementary feeding is associated with gut microbiota diversity and composition and short chain fatty acid concentrations over the first year of life. BMC Microbiology, 20 (2020) 56. https://doi.org/10.1186/s12866-020-01723-9.Google Scholar
Tsukuda, N., K. Yahagi, T. Hara, Y. Watanabe, H. Matsumoto, H. Mori, K. Higashi, H. Tsuji, S. Matsumoto, K. Kurokawa, & T. Matsuki, Key bacterial taxa and metabolic pathways affecting gut short-chain fatty acid profiles in early life. ISME J, 15 (2021) 2574–2590. https://doi.org/10.1038/s41396-021-00937-7.Google Scholar
Bridgman, S. L., Azad, M. B., Field, C. J., Haqq, A. M., Becker, A. B., Mandhane, P. J., Subbarao, P., Turvey, S. E., Sears, M. R., Scott, J. A., Wishart, D. S., & Kozyrskyj, A. L., Fecal short-chain fatty acid variations by breastfeeding status in infants at 4 months: Differences in relative versus absolute concentrations. Frontiers in Nutrition, 4 (2017). https://doi.org/10.3389/fnut.2017.00011.Google Scholar
Tun, H. M., Bridgman, S. L., Chari, R., Field, C. J., Guttman, D. S., Becker, A. B., Mandhane, P. J., Turvey, S. E., Subbarao, P., Sears, M. R., Scott, J. A., & Kozyrskyj, A. L., Roles of birth mode and infant gut microbiota in intergenerational transmission of overweight and obesity from mother to offspring. JAMA Pediatrics, 172 (2018) 368–377. https://doi.org/10.1001/jamapediatrics.2017.5535.Google Scholar
van der Vossen, E. W. J., M. C. de Goffau, E. Levin, & M. Nieuwdorp, Recent insights into the role of microbiome in the pathogenesis of obesity. Therapeutic Advances in Gastroenterology, 15 (2022). https://doi.org/10.1177/17562848221115320.Google Scholar
Stanislawski, M. A., Dabelea, D., Wagner, B. D., Iszatt, N., Dahl, C., Sontag, M. K., Knight, R., Lozupone, C. A., & Eggesbø, M., Gut microbiota in the first 2 years of life and the association with body mass index at age 12 in a Norwegian Birth Cohort. mBio, 9 (2018). https://doi.org/10.1128/mBio.01751-18.Google Scholar
Netea, M. G., Joosten, L. A. B. B., Latz, E., Mills, K. H. G. G., Natoli, G., Stunnenberg, H. G., O’Neill, L. A. J., Xavier, R. J., ONeill, L. A. J., & Xavier, R. J., Trained immunity: A program of innate immune memory in health and disease (American Association for the Advancement of Science, 2016). https://doi.org/10.1126/science.aaf1098.Google Scholar
Safari, Z. & Gérard, P., The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cellular and Molecular Life Sciences, 76 (2019) 1541–1558. https://doi.org/10.1007/s00018-019-03011-w.Google Scholar
Cani, P. D., Amar, J., Iglesias, M. A., Poggi, M., Knauf, C., Bastelica, D., Neyrinck, A. M., Fava, F., Tuohy, K. M., Chabo, C., Waget, A. Lie, Delmé, E., Cousin, B. Atrice, Sulpice, T., Chamontin, B., Ferriè, J., Tanti, J.-F., Gibson, G. R., Casteilla, L., Delzenne, N. M., Alessi, M. C., & Burcelin, R. My, Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes, 56 (2007) 1761–1772. https://doi.org/10.2337/db06-1491.Google Scholar
Vatanen, T., Kostic, A. D., D’Hennezel, E., Siljander, H., Franzosa, E. A., Yassour, M., Kolde, R., Vlamakis, H., Arthur, T. D., Hämäläinen, A. M., Peet, A., Tillmann, V., Uibo, R., Mokurov, S., Dorshakova, N., Ilonen, J., Virtanen, S. M., Szabo, S. J., Porter, J. A., Lähdesmäki, H., Huttenhower, C., Gevers, D., Cullen, T. W., Knip, M., & Xavier, R. J., Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell, 165 (2016) 842–853. https://doi.org/10.1016/j.cell.2016.04.007.Google Scholar
Wampach, L., Heintz-Buschart, A., Fritz, J. V., Ramiro-Garcia, J., Habier, J., Herold, M., Narayanasamy, S., Kaysen, A., Hogan, A. H., Bindl, L., Bottu, J., Halder, R., Sjöqvist, C., May, P., Andersson, A. F., de Beaufort, C., & Wilmes, P., Birth mode is associated with earliest strain-conferred gut microbiome functions and immunostimulatory potential. Nature Communications, 9 (2018) 1–14. https://doi.org/10.1038/s41467-018-07631-x.Google Scholar
Kameyama, K. & Itoh, K., Intestinal colonization by a lachnospiraceae bacterium contributes to the development of diabetes in obese mice. Microbes and Environments, 29 (2014) 427–430. https://doi.org/10.1264/jsme2.ME14054.Google Scholar
Loomba, R. & Sanyal, A. J., The global NAFLD epidemic. Nature Reviews Gastroenterology and Hepatology, 10 (2013) 686–690. https://doi.org/10.1038/nrgastro.2013.171.Google Scholar
Goyal, N. P. & Schwimmer, J. B., The progression and natural history of pediatric nonalcoholic fatty liver disease. Clinics in Liver Disease, 20 (2016) 325–338. https://doi.org/10.1016/j.cld.2015.10.003.Google Scholar
Ibrahim, S. H., Jonas, M. M., Taylor, S. A., Sanchez, L. H. Gutierrez, Wolf, J. L., & Sundaram, S. S., Liver diseases in the perinatal period: Interactions between mother and infant. Hepatology, 71 (2020) 1474–1485. https://doi.org/10.1002/hep.31109.Google Scholar
Doycheva, I., Issa, D., Watt, K. D., Lopez, R., Rifai, G., & Alkhouri, N., Nonalcoholic steatohepatitis is the most rapidly increasing indication for liver transplantation in young adults in the United States. Journal of Clinical Gastroenterology, 52 (2018) 339–346. https://doi.org/10.1097/MCG.0000000000000925.Google Scholar
Jayakumar, S. & Loomba, R., Review article: Emerging role of the gut microbiome in the progression of nonalcoholic fatty liver disease and potential therapeutic implications. Alimentary Pharmacology and Therapeutics, 50 (2019) 144–158. https://doi.org/10.1111/apt.15314.Google Scholar
Leatham-Jensen, M. P., Frimodt-Møller, J., Adediran, J., Mokszycki, M. E., Banner, M. E., Caughron, J. E., Krogfelt, K. A., Conway, T., & Cohen, P. S., The streptomycin-treated mouse intestine selects Escherichia coli envZ missense mutants that interact with dense and diverse intestinal microbiota. Infection and Immunity, 80 (2012) 1716–1727. https://doi.org/10.1128/IAI.06193-11.Google Scholar
Koch, M. A., Reiner, G. L., Lugo, K. A., Seher, T. D., Ludington, W. B., & Barton, G. M., Maternal IgG and IgA antibodies Dampen Mucosal T Helper cell responses in early life. Cell, 165 (2016) 827–841. https://doi.org/10.1016/j.cell.2016.04.055.Google Scholar
McCoy, K. D. & Thomson, C. A., The impact of maternal microbes and microbial colonization in early life on hematopoiesis. The Journal of Immunology, 200 (2018) 2519–2526. https://doi.org/10.4049/jimmunol.1701776.Google Scholar
Dogra, S. K., C. K. Chung, D. Wang, O. Sakwinska, S. C. Mottaz, & N. Sprenger, Nurturing the early life gut microbiome and immune maturation for long term health. Microorganisms, 9 (2021) 2110. https://doi.org/10.3390/MICROORGANISMS9102110.Google Scholar

References

Barker, DJ, Winter, PD, Osmond, C, Margetts, B, Simmonds, SJ. Weight in infancy and death from ischaemic heart disease. Lancet. 1989;2(8663), 577–580.Google Scholar
Fleming, TP, Watkins, AJ, Velazquez, MA, et al. Origins of lifetime health around the time of conception: causes and consequences. Lancet. 2018;391(10132), 1842–1852.Google Scholar
Eckersley-Maslin, MA, Alda-Catalinas, C, Reik, W. Dynamics of the epigenetic landscape during the maternal-to-zygotic transition. Nat Rev Mol Cell Biol. 2018;19(7), 436–450.Google Scholar
Velazquez, MA, Fleming, TP, Watkins, AJ. Periconceptional environment and the developmental origins of disease. J Endocrinol. 2019;242(1), T33T49.Google Scholar
Crawford, GE, Ledger, WL. In vitro fertilisation/intracytoplasmic sperm injection beyond 2020. BJOG. 2019;126(2), 237–243.Google Scholar
Roseboom, TJ. Developmental plasticity and its relevance to assisted human reproduction. Hum Reprod. 2018;33(4), 546–552.Google Scholar
Hann, M, Roberts, SA, D’Souza, SW, Clayton, P, Macklon, N, Brison, DR. The growth of assisted reproductive treatment-conceived children from birth to 5 years: a national cohort study. BMC Med. 2018;16(1), 224.Google Scholar
Turkgeldi, E, Yagmur, H, Seyhan, A, Urman, B, Ata, B. Short and long term outcomes of children conceived with assisted reproductive technology. Eur J Obstet Gynecol Reprod Biol. 2016;207, 129–136.Google Scholar
Hargreave, M, Jensen, A, Hansen, MK, et al. Association between fertility treatment and cancer risk in children. JAMA. 2019;322(22), 2203–2210.Google Scholar
Sakka, SD, Loutradis, D, Kanaka-Gantenbein, C, et al. Absence of insulin resistance and low-grade inflammation despite early metabolic syndrome manifestations in children born after in vitro fertilization. Fertil Steril. 2010;94(5), 1693–1699.Google Scholar
Cavoretto, P, Candiani, M, Giorgione, V, et al. Risk of spontaneous preterm birth in singleton pregnancies conceived after IVF/ICSI treatment: meta-analysis of cohort studies. Ultrasound Obstet Gynecol. 2018;51(1), 43–53.Google Scholar
Hart, R, Norman, RJ. The longer-term health outcomes for children born as a result of IVF treatment: Part I – General health outcomes. Hum Reprod Update. 2013;19(3), 232–243.Google Scholar
Hart, R, Norman, RJ. The longer-term health outcomes for children born as a result of IVF treatment. Part II – Mental health and development outcomes. Hum Reprod Update. 2013;19(3), 244–250.Google Scholar
Kuiper, DB, Seggers, J, Schendelaar, P, et al. Asthma and asthma medication use among 4-year-old offspring of subfertile couples – association with IVF? Reprod Biomed Online. 2015;31(5), 711–714.Google Scholar
Ludwig, AK, Katalinic, A, Thyen, U, Sutcliffe, AG, Diedrich, K, Ludwig, M. Physical health at 5.5 years of age of term-born singletons after intracytoplasmic sperm injection: results of a prospective, controlled, single-blinded study. Fertil Steril. 2009;91(1), 115–124.Google Scholar
Guo, XY, Liu, XM, Jin, L, et al. Cardiovascular and metabolic profiles of offspring conceived by assisted reproductive technologies: a systematic review and meta-analysis. Fertil Steril. 2017;107(3), 622–631 e625.Google Scholar
Schendelaar, P, Van den Heuvel, ER, Heineman, MJ, et al. Increased time to pregnancy is associated with less optimal neurological condition in 4-year-old singletons, in vitro fertilization itself is not. Hum Reprod. 2014;29(12), 2773–2786.Google Scholar
Vanky, E, Engen Hanem, LG, Abbott, DH. Children born to women with polycystic ovary syndrome-short- and long-term impacts on health and development. Fertil Steril. 2019;111(6), 1065–1075.Google Scholar
Simard, M, Laprise, C, Girard, SL. Impact of paternal age at conception on human health. Clin Chem. 2019;65(1), 146–152.Google Scholar
Seggers, J, Haadsma, ML, Bastide-van Gemert, S, et al. Blood pressure and anthropometrics of 4-y-old children born after preimplantation genetic screening: follow-up of a unique, moderately sized, randomized controlled trial. Pediatr Res. 2013;74(5), 606–614.Google Scholar
Berntsen, S, Soderstrom-Anttila, V, Wennerholm, UB, et al. The health of children conceived by ART: ‘the chicken or the egg?’. Hum Reprod Update. 2019;25(2), 137–158.Google Scholar
Mintjens, S, Menting, MD, Gemke, R, et al. The effects of intrauterine insemination and single embryo transfer or modified natural cycle in vitro fertilization on offspring’s health-Follow-up of a randomized clinical trial. Eur J Obstet Gynecol Reprod Biol. 2019;242, 131–138.Google Scholar
Lazaraviciute, G, Kauser, M, Bhattacharya, S, Haggarty, P, Bhattacharya, S. A systematic review and meta-analysis of DNA methylation levels and imprinting disorders in children conceived by IVF/ICSI compared with children conceived spontaneously. Hum Reprod Update. 2014;20(6), 840–852.Google Scholar
White, CR, Denomme, MM, Tekpetey, FR, Feyles, V, Power, SG, Mann, MR. High frequency of imprinted methylation errors in human preimplantation embryos. Sci Rep. 2015;5, 17311.Google Scholar
Song, S, Ghosh, J, Mainigi, M, et al. DNA methylation differences between in vitro- and in vivo-conceived children are associated with ART procedures rather than infertility. Clin Epigenetics. 2015;7(7), 41.Google Scholar
Williams, CL, Teeling, JL, Perry, VH, Fleming, TP. Mouse maternal systemic inflammation at the zygote stage causes blunted cytokine responsiveness in lipopolysaccharide-challenged adult offspring. BMC Biol. 2011;9, 49.Google Scholar
Novakovic, B, Lewis, S, Halliday, J, et al. Assisted reproductive technologies are associated with limited epigenetic variation at birth that largely resolves by adulthood. Nat Commun. 2019;10(1), 3922.Google Scholar
Dumoulin, JC, Land, JA, Van Montfoort, AP, et al. Effect of in vitro culture of human embryos on birthweight of newborns. Hum Reprod. 2010;25(3), 605–612.Google Scholar
Nelissen, EC, Van Montfoort, AP, Smits, LJ, et al. IVF culture medium affects human intrauterine growth as early as the second trimester of pregnancy. Hum Reprod. 2013;28(8), 2067–2074.Google Scholar
Kleijkers, SH, Mantikou, E, Slappendel, E, et al. Influence of embryo culture medium (G5 and HTF) on pregnancy and perinatal outcome after IVF: a multicenter RCT. Hum Reprod. 2016;31(10), 2219–2230.Google Scholar
Zandstra, H, Smits, LJM, van Kuijk, SMJ, et al. No effect of IVF culture medium on cognitive development of 9-year-old children. Hum Reprod Open. 2018;2018(4), hoy018.Google Scholar
Zandstra, H, Van Montfoort, AP, Dumoulin, JC. Does the type of culture medium used influence birthweight of children born after IVF? Hum Reprod. 2015;30(3), 530–542.Google Scholar
Duranthon, V, Chavatte-Palmer, P. Long term effects of ART: What do animals tell us? Mol Reprod Dev. 2018;85(4), 348–368.Google Scholar
Ealy, AD, Wooldridge, LK, McCoski, SR. BOARD INVITED REVIEW: post-transfer consequences of in vitro-produced embryos in cattle. J Anim Sci. 2019;97(6), 2555–2568.Google Scholar
Glujovsky, D, Farquhar, C, Quinteiro Retamar, AM, Alvarez Sedo, CR, Blake, D. Cleavage stage versus blastocyst stage embryo transfer in assisted reproductive technology. Cochrane Database Syst Rev. 2016; doi: 10.1002/14651858.CD002118.pub5(6), CD002118.Google Scholar
Maheshwari, A, Pandey, S, Amalraj Raja, E, Shetty, A, Hamilton, M, Bhattacharya, S. Is frozen embryo transfer better for mothers and babies? Can cumulative meta-analysis provide a definitive answer? Hum Reprod Update. 2018;24(1), 35–58.Google Scholar
Castillo, CM, Horne, G, Fitzgerald, CT, Johnstone, ED, Brison, DR, Roberts, SA. The impact of IVF on birthweight from 1991 to 2015: a cross-sectional study. Hum Reprod. 2019;34(5), 920–931.Google Scholar
Pelkonen, S, Gissler, M, Koivurova, S, et al. Physical health of singleton children born after frozen embryo transfer using slow freezing: a 3-year follow-up study. Hum Reprod. 2015;30(10), 2411–2418.Google Scholar
Kimber, SJ, Sneddon, SF, Bloor, DJ, et al. Expression of genes involved in early cell fate decisions in human embryos and their regulation by growth factors. Reproduction. 2008;135(5), 635–647.Google Scholar
Nassan, FL, Chavarro, JE, Tanrikut, C. Diet and men’s fertility: does diet affect sperm quality? Fertil Steril. 2018;110(4), 570–577.Google Scholar
Watkins, AJ, Sirovica, S, Stokes, B, Isaacs, M, Addison, O, Martin, RA. Paternal low protein diet programs preimplantation embryo gene expression, fetal growth and skeletal development in mice. Biochimica et biophysica acta. 2017;1863(6), 1371–1381.Google Scholar
Lambrot, R, Xu, C, Saint-Phar, S, et al. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat Commun. 2013;4, 2889; doi: 10.1038/ncomms3889.Google Scholar
Morgan, HL, Paganopoulou, P, Akhtar, S, et al. Paternal diet impairs F1 and F2 offspring vascular function through sperm and seminal plasma specific mechanisms in mice. J Physiol. 2019; doi: 10.1113/JP278270.Google Scholar
McPherson, NO, Fullston, T, Kang, WX, et al. Paternal under-nutrition programs metabolic syndrome in offspring which can be reversed by antioxidant/vitamin food fortification in fathers. Sci Rep. 2016;6, 27010.Google Scholar
Pang, TYC, Short, AK, Bredy, TW, Hannan, AJ. Transgenerational paternal transmission of acquired traits: stress-induced modification of the sperm regulatory transcriptome and offspring phenotypes. Curr Opin Behav Sci. 2017;14, 140–147.Google Scholar
Campbell, JM, Lane, M, Owens, JA, Bakos, HW. Paternal obesity negatively affects male fertility and assisted reproduction outcomes: a systematic review and meta-analysis. Reprod Biomed Online. 2015;31(5), 593–604.Google Scholar
Hammoud, SS, Nix, DA, Hammoud, AO, Gibson, M, Cairns, BR, Carrell, DT. Genome-wide analysis identifies changes in histone retention and epigenetic modifications at developmental and imprinted gene loci in the sperm of infertile men. Hum Reprod. 2011;26(9), 2558–2569.Google Scholar
Aston, KI, Uren, PJ, Jenkins, TG, et al. Aberrant sperm DNA methylation predicts male fertility status and embryo quality. Fertil Steril. 2015;104(6), 1388–1397 e1381–1385.Google Scholar
Benchaib, M, Braun, V, Ressnikof, D, et al. Influence of global sperm DNA methylation on IVF results. Hum Reprod. 2005;20(3), 768–773.Google Scholar
Colaco, S, Sakkas, D. Paternal factors contributing to embryo quality. J Assist Reprod Genet. 2018; doi: 10.1007/s10815-018-1304-4.Google Scholar
Chen, Q, Yan, M, Cao, Z, et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351(6271), 397–400.Google Scholar
Morgan, HL, Watkins, AJ. The influence of seminal plasma on offspring development and health. Semin Cell Dev Biol. 2020;97, 131–137.Google Scholar
Robillard, PY, Hulsey, TC, Perianin, J, Janky, E, Miri, EH, Papiernik, E. Association of pregnancy-induced hypertension with duration of sexual cohabitation before conception. Lancet. 1994;344(8928), 973–975.Google Scholar
Bromfield, JJ, Schjenken, JE, Chin, PY, Care, AS, Jasper, MJ, Robertson, SA. Maternal tract factors contribute to paternal seminal fluid impact on metabolic phenotype in offspring. Proc Natl Acad Sci U S A. 2014;111(6), 2200–2205.Google Scholar
Watkins, AJ, Dias, I, Tsuro, H, et al. Paternal diet programs offspring health through sperm- and seminal plasma-specific pathways in mice. Proc Natl Acad Sci U S A. 2018;115(40), 10064–10069.Google Scholar
Lane, M, Robker, RL, Robertson, SA. Parenting from before conception. Science. 2014;345(6198), 756–760.Google Scholar
Lane, M, Gardner, DK. Understanding cellular disruptions during early embryo development that perturb viability and fetal development. Reprod Fertil Dev. 2005;17(3), 371–378.Google Scholar
Feil, D, Henshaw, RC, Lane, M. Day 4 embryo selection is equal to day 5 using a new embryo scoring system validated in single embryo transfers. Hum Reprod. 2008;23(7), 1505–1510.Google Scholar
Mitchell, M, Schulz, SL, Armstrong, DT, Lane, M. Metabolic and mitochondrial dysfunction in early mouse embryos following maternal dietary protein intervention. Biol Reprod. 2009;80(4), 622–630.Google Scholar
McPherson, NO, Shehadeh, H, Fullston, T, Zander-Fox, DL, Lane, M. Dietary micronutrient supplementation for 12 days in obese male mice restores sperm oxidative stress. Nutrients. 2019;11(9): 2196; doi: 10.3390/nu11092196.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×