201
|
Lunde ER, Washburn SE, Golding MC, Bake S, Miranda RC, Ramadoss J. Alcohol-Induced Developmental Origins of Adult-Onset Diseases. Alcohol Clin Exp Res 2016; 40:1403-14. [PMID: 27254466 PMCID: PMC5067080 DOI: 10.1111/acer.13114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/25/2016] [Indexed: 12/16/2022]
Abstract
Fetal alcohol exposure may impair growth, development, and function of multiple organ systems and is encompassed by the term fetal alcohol spectrum disorders (FASD). Research has so far focused on the mechanisms, prevention, and diagnosis of FASD, while the risk for adult-onset chronic diseases in individuals exposed to alcohol in utero is not well explored. David Barker's hypothesis on Developmental Origins of Health and Disease (DOHaD) suggests that insults to the milieu of the developing fetus program it for adult development of chronic diseases. In the 25 years since the introduction of this hypothesis, epidemiological and animal model studies have made significant advancements in identifying in utero developmental origins of chronic adult-onset diseases affecting cardiovascular, endocrine, musculoskeletal, and psychobehavioral systems. Teratogen exposure is an established programming agent for adult diseases, and recent studies suggest that prenatal alcohol exposure correlates with adult onset of neurobehavioral deficits, cardiovascular disease, endocrine dysfunction, and nutrient homeostasis instability, warranting additional investigation of alcohol-induced DOHaD, as well as patient follow-up well into adulthood for affected individuals. In utero epigenetic alterations during critical periods of methylation are a key potential mechanism for programming and susceptibility of adult-onset chronic diseases, with imprinted genes affecting metabolism being critical targets. Additional studies in epidemiology, phenotypic characterization in response to timing, dose, and duration of exposure, as well as elucidation of mechanisms underlying FASD-DOHaD inter relation, are thus needed to clinically define chronic disease associated with prenatal alcohol exposure. These studies are critical to establish interventional strategies that decrease incidence of these adult-onset diseases and promote healthier aging among individuals affected with FASD.
Collapse
Affiliation(s)
- Emilie R. Lunde
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shannon E. Washburn
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Michael C. Golding
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shameena Bake
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA
| | - Rajesh C. Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA
| | - Jayanth Ramadoss
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
202
|
Derraik JGB, Lundgren M, Cutfield WS, Ahlsson F. Body Mass Index, Overweight, and Obesity in Swedish Women Born Post-term. Paediatr Perinat Epidemiol 2016; 30:320-4. [PMID: 27016177 DOI: 10.1111/ppe.12292] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND There is increasing evidence that post-term birth (≥42 weeks of gestation) is associated with adverse long-term outcomes. We assessed whether women born post-term displayed increased risk of overweight and obesity in adulthood. METHODS Data were collected at first antenatal visit (~10-12 weeks of gestation) on singleton Swedish women aged ≥18 years in 1991-2009 (mean age 26.1 years), who were born post-term (n = 27 153) or at term (37-41 weeks of gestation; n = 184 245). Study outcomes were evaluated for continuous associations with gestational age. Stratified analyses were carried out comparing women born post-term or at term. Analyses were also run with a 2-week buffer between groups to account for possible errors in gestational age estimation, comparing women born very post-term (≥43 weeks of gestation; n = 5761) to those born within a narrower term window (38-40 weeks of gestation; n = 130 110). RESULTS Increasing gestational age was associated with greater adult weight and body mass index (BMI). Stratified analyses showed that women born post-term were 0.5 kg heavier and had BMI 0.2 kg/m(2) greater than those born at term. Differences were more marked between women born very post-term (≥43 weeks) vs. a narrower term group (38-40 weeks): 1.0 kg and 0.3 kg/m(2) . The adjusted relative risks of overweight/obesity and obesity in women born very post-term were 1.13 and 1.12 times higher, respectively, than in those born at term. CONCLUSIONS Post-term birth is associated with greater BMI and increased risk of overweight and obesity in adulthood, particularly among women born ≥43 weeks of gestation.
Collapse
Affiliation(s)
- José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Maria Lundgren
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Fredrik Ahlsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
203
|
Kautzky-Willer A, Harreiter J, Pacini G. Sex and Gender Differences in Risk, Pathophysiology and Complications of Type 2 Diabetes Mellitus. Endocr Rev 2016; 37:278-316. [PMID: 27159875 PMCID: PMC4890267 DOI: 10.1210/er.2015-1137] [Citation(s) in RCA: 1195] [Impact Index Per Article: 132.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The steep rise of type 2 diabetes mellitus (T2DM) and associated complications go along with mounting evidence of clinically important sex and gender differences. T2DM is more frequently diagnosed at lower age and body mass index in men; however, the most prominent risk factor, which is obesity, is more common in women. Generally, large sex-ratio differences across countries are observed. Diversities in biology, culture, lifestyle, environment, and socioeconomic status impact differences between males and females in predisposition, development, and clinical presentation. Genetic effects and epigenetic mechanisms, nutritional factors and sedentary lifestyle affect risk and complications differently in both sexes. Furthermore, sex hormones have a great impact on energy metabolism, body composition, vascular function, and inflammatory responses. Thus, endocrine imbalances relate to unfavorable cardiometabolic traits, observable in women with androgen excess or men with hypogonadism. Both biological and psychosocial factors are responsible for sex and gender differences in diabetes risk and outcome. Overall, psychosocial stress appears to have greater impact on women rather than on men. In addition, women have greater increases of cardiovascular risk, myocardial infarction, and stroke mortality than men, compared with nondiabetic subjects. However, when dialysis therapy is initiated, mortality is comparable in both males and females. Diabetes appears to attenuate the protective effect of the female sex in the development of cardiac diseases and nephropathy. Endocrine and behavioral factors are involved in gender inequalities and affect the outcome. More research regarding sex-dimorphic pathophysiological mechanisms of T2DM and its complications could contribute to more personalized diabetes care in the future and would thus promote more awareness in terms of sex- and gender-specific risk factors.
Collapse
Affiliation(s)
- Alexandra Kautzky-Willer
- Gender Medicine Unit (A.K.-W., J.H.), Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; and Metabolic Unit (G.P.), Institute of Neuroscience, National Research Council, 35127 Padua, Italy
| | - Jürgen Harreiter
- Gender Medicine Unit (A.K.-W., J.H.), Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; and Metabolic Unit (G.P.), Institute of Neuroscience, National Research Council, 35127 Padua, Italy
| | - Giovanni Pacini
- Gender Medicine Unit (A.K.-W., J.H.), Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; and Metabolic Unit (G.P.), Institute of Neuroscience, National Research Council, 35127 Padua, Italy
| |
Collapse
|
204
|
Boone-Heinonen J, Markwardt S, Fortmann SP, Thornburg KL. Overcoming birth weight: can physical activity mitigate birth weight-related differences in adiposity? Pediatr Obes 2016; 11:166-73. [PMID: 25988588 PMCID: PMC4653090 DOI: 10.1111/ijpo.12040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Individuals born at low or high birth weight (BW) have elevated adiposity. The extent to which physical activity can mitigate this risk is unknown. OBJECTIVES The aim of this study was to determine if associations between BW and adiposity vary by self-reported moderate-to-vigorous physical activity (MVPA) among adolescents. METHODS We used data on adolescents in the National Health and Nutrition Examination Survey (1999-2006; 12-15 years; n = 4064). Using gender-stratified linear regression, we modelled body mass index (BMI) and waist circumference (WC) z-scores as a function of low, normal and high BW, MVPA (weekly Metabolic Equivalent of Task hours) and MVPA*BW cross-product terms, adjusting for sociodemographics, diet and, in WC models, BMI. RESULTS Among girls with low MVPA, those born with high BW had greater BMI than normal BW; this difference diminished with greater MVPA (coefficient [95% confidence interval]: low MVPA: 0.72 [0.29, 1.14]; high MVPA: -0.04 [-0.48, 0.39]; P for interaction = 0.05). Among boys, MVPA did not modify the associations between BW and BMI. WC was unrelated to BW, regardless of MVPA. CONCLUSIONS Findings suggest that effects of high BW in total adiposity can be more easily modified with MVPA in adolescent girls than in boys.
Collapse
Affiliation(s)
- Janne Boone-Heinonen
- Department of Public Health and Preventive Medicine, Oregon Health & Science University; Portland, OR, USA
| | - Sheila Markwardt
- Department of Public Health and Preventive Medicine, Oregon Health & Science University; Portland, OR, USA
| | | | - Kent L. Thornburg
- Bob and Charlee Moore Institute for Nutrition and Wellness, Oregon Health & Science University; Portland, OR, USA
| |
Collapse
|
205
|
Lee A, Mathilda Chiu YH, Rosa MJ, Jara C, Wright RO, Coull BA, Wright RJ. Prenatal and postnatal stress and asthma in children: Temporal- and sex-specific associations. J Allergy Clin Immunol 2016; 138:740-747.e3. [PMID: 26953156 DOI: 10.1016/j.jaci.2016.01.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/22/2015] [Accepted: 01/07/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND Temporal- and sex-specific effects of perinatal stress have not been examined for childhood asthma. OBJECTIVES We examined associations between prenatal and/or postnatal stress and children's asthma (n = 765) and effect modification by sex in a prospective cohort study. METHODS Maternal negative life events were ascertained prenatally and postpartum. Negative life event scores were categorized as 0, 1 to 2, 3 to 4, or 5 or greater to assess exposure-response relationships. We examined effects of prenatal and postnatal stress on children's asthma by age 6 years, modeling each as independent predictors, mutually adjusting for prenatal and postnatal stress, and finally considering interactions between prenatal and postnatal stress. Effect modification by sex was examined in stratified analyses and by fitting interaction terms. RESULTS When considering stress in each period independently, among boys, a dose-response relationship was evident for each level increase on the ordinal scale prenatally (odds ratio [OR], 1.38; 95% CI, 1.06-1.79; P value for trend = .03) and postnatally (OR, 1.53; 95% CI, 1.16-2.01; P value for trend = .001); among girls, only the postnatal trend was significant (OR, 1.60; 95% CI, 1.14-2.22; P value for trend = .005). Higher stress in both the prenatal and postnatal periods was associated with increased odds of receiving a diagnosis of asthma in girls (OR, 1.37; 95% CI, 0.98-1.91; Pinteraction = .07) but not boys (OR, 1.08; 95% CI, 0.82-1.42; Pinteraction = .61). CONCLUSIONS Although boys were more vulnerable to stress during the prenatal period, girls were more affected by postnatal stress and cumulative stress across both periods in relation to asthma. Understanding sex and temporal differences in response to early-life stress might provide unique insight into the cause and natural history of asthma.
Collapse
Affiliation(s)
- Alison Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yueh-Hsiu Mathilda Chiu
- Department of Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Maria José Rosa
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Calvin Jara
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert O Wright
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY; Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Brent A Coull
- Department of Biostatistics, Harvard School of Public Health, Boston, Mass; Department of Environmental Health, Harvard School of Public Health, Boston, Mass
| | - Rosalind J Wright
- Department of Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, NY; Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
206
|
Junien C, Panchenko P, Fneich S, Pirola L, Chriett S, Amarger V, Kaeffer B, Parnet P, Torrisani J, Bolaños Jimenez F, Jammes H, Gabory A. [Epigenetics in transgenerational responses to environmental impacts: from facts and gaps]. Med Sci (Paris) 2016; 32:35-44. [PMID: 26850605 DOI: 10.1051/medsci/20163201007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The existence of non-genetic and non-cultural mechanisms that transfer information on the memory of parental exposures to various environments, determining the reactivity of the following generations to their environments during their life, are of growing interest. Yet fundamental questions remain about the nature, the roles and relative importance of epigenetic marks and processes, non-coding RNAs, or other mechanisms, and their persistence over generations. A model incorporating the various transmission systems, their cross-talks and windows of susceptibility to the environment as a function of sex/gender of parent and offspring, has yet to be built.
Collapse
Affiliation(s)
- Claudine Junien
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | - Polina Panchenko
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | - Sara Fneich
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | | | | | - Valérie Amarger
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Bertrand Kaeffer
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Patricia Parnet
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Jérome Torrisani
- Inserm UMR1037, Centre de recherches en cancérologie de Toulouse, Université de Toulouse III Paul Sabatier, F-31037 Toulouse, France
| | - Francisco Bolaños Jimenez
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Hélène Jammes
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | - Anne Gabory
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| |
Collapse
|
207
|
Peugnet P, Robles M, Wimel L, Tarrade A, Chavatte-Palmer P. Management of the pregnant mare and long-term consequences on the offspring. Theriogenology 2016; 86:99-109. [PMID: 26954944 DOI: 10.1016/j.theriogenology.2016.01.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/22/2016] [Accepted: 01/29/2016] [Indexed: 01/21/2023]
Abstract
The study of early developmental conditioning of health and disease in adulthood is particularly relevant in the horse, which is bred mainly to perform in demanding sport challenges. On the basis of this concept, the management of the broodmare could be considered an effective means to produce animals with the desired features. Knowledge on the Developmental Origins of Health and Disease in the equine species remains relatively scarce, with some experimental studies and one single epidemiologic study. Data highlight the determinant role of the maternal environment for postnatal body conformation, immune response, energy homeostasis, osteoarticular status and thyroidal, adrenocortical, and cardiovascular functions of the foal. Most research, however, focuses on the first months/years after birth. Long-term effects on the adult horse phenotype have not been investigated so far.
Collapse
Affiliation(s)
- Pauline Peugnet
- UMR 1198 Developmental Biology and Reproduction, French Institute for Agricultural Research (INRA), Jouy-en-Josas, France.
| | - Morgane Robles
- UMR 1198 Developmental Biology and Reproduction, French Institute for Agricultural Research (INRA), Jouy-en-Josas, France
| | - Laurence Wimel
- Experimental farm, French Horse and Riding Institute (IFCE), Chamberet, France
| | - Anne Tarrade
- UMR 1198 Developmental Biology and Reproduction, French Institute for Agricultural Research (INRA), Jouy-en-Josas, France
| | - Pascale Chavatte-Palmer
- UMR 1198 Developmental Biology and Reproduction, French Institute for Agricultural Research (INRA), Jouy-en-Josas, France
| |
Collapse
|
208
|
Vilahur N, Vahter M, Broberg K. The Epigenetic Effects of Prenatal Cadmium Exposure. Curr Environ Health Rep 2016; 2:195-203. [PMID: 25960943 PMCID: PMC4417128 DOI: 10.1007/s40572-015-0049-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prenatal exposure to the highly toxic and common pollutant cadmium has been associated with adverse effects on child health and development. However, the underlying biological mechanisms of cadmium toxicity remain partially unsolved. Epigenetic disruption due to early cadmium exposure has gained attention as a plausible mode of action, since epigenetic signatures respond to environmental stimuli and the fetus undergoes drastic epigenomic rearrangements during embryogenesis. In the current review, we provide a critical examination of the literature addressing prenatal cadmium exposure and epigenetic effects in human, animal, and in vitro studies. We conducted a PubMed search and obtained eight recent studies addressing this topic, focusing almost exclusively on DNA methylation. These studies provide evidence that cadmium alters epigenetic signatures in the DNA of the placenta and of the newborns, and some studies indicated marked sexual differences for cadmium-related DNA methylation changes. Associations between early cadmium exposure and DNA methylation might reflect interference with de novo DNA methyltransferases. More studies, especially those including environmentally relevant doses, are needed to confirm the toxicoepigenomic effects of prenatal cadmium exposure and how that relates to the observed health effects of cadmium in childhood and later life.
Collapse
Affiliation(s)
- Nadia Vilahur
- Institute of Environmental Medicine, Unit of Metals and Health, Karolinska Institutet, Nobels väg 13, Box 210, SE-171 77 Stockholm, Sweden
| | - Marie Vahter
- Institute of Environmental Medicine, Unit of Metals and Health, Karolinska Institutet, Nobels väg 13, Box 210, SE-171 77 Stockholm, Sweden
| | - Karin Broberg
- Institute of Environmental Medicine, Unit of Metals and Health, Karolinska Institutet, Nobels väg 13, Box 210, SE-171 77 Stockholm, Sweden
| |
Collapse
|
209
|
Gopalakrishnan K, Mishra JS, Chinnathambi V, Vincent KL, Patrikeev I, Motamedi M, Saade GR, Hankins GD, Sathishkumar K. Elevated Testosterone Reduces Uterine Blood Flow, Spiral Artery Elongation, and Placental Oxygenation in Pregnant Rats. Hypertension 2016; 67:630-9. [PMID: 26781277 DOI: 10.1161/hypertensionaha.115.06946] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
Elevated maternal testosterone levels are shown to cause fetal growth restriction, eventually culminating in sex-specific adult-onset hypertension that is more pronounced in males than in females. In this study, we tested whether uteroplacental and fetoplacental disturbances underlie fetal growth restriction and if these changes vary in male and female placentas. Pregnant Sprague-Dawley rats were injected with vehicle (n=16) or testosterone propionate (0.5 mg/kg per day from gestation day 15-19; n=16). On gestation day 20, we quantified uterine artery blood flow using microultrasound, visualized placental arterial network using x-ray microcomputed tomography, determined fetoplacental hypoxia using pimonidazole and hypoxia-inducible factor-1α, and used Affymetrix array to determine changes in placental expression of genes involved in vascular development. Plasma testosterone levels increased 2-fold in testosterone-injected rats. Placental and fetal weights were lower in rats with elevated testosterone. Uterine artery blood flow was lower, and resistance index was higher in the testosterone group. Radial and spiral artery diameter and length, the number of fetoplacental arterial branches, and umbilical artery diameter were reduced in the testosterone group. In addition, markers of hypoxia in the placentas and fetuses were elevated in the testosterone group. The magnitude of changes in placental vasculature and hypoxia was greater in males than in females and was associated with sex-specific alteration of unique sets of genes involved in angiogenesis and blood vessel morphogenesis. The results demonstrate that elevated testosterone during gestation induces a decrease in uterine arterial blood flow and fetal sex-related uteroplacental vascular changes, which may set the stage for subsequent sex differences in adult-onset diseases.
Collapse
Affiliation(s)
- Kathirvel Gopalakrishnan
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Jay S Mishra
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Vijayakumar Chinnathambi
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Kathleen L Vincent
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Igor Patrikeev
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Massoud Motamedi
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - George R Saade
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Gary D Hankins
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston
| | - Kunju Sathishkumar
- From the Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology (K.G., J.S.M., V.C., K.L.V., G.R.S., G.D.H., K.S.) and Center for Biomedical Engineering (K.L.V., I.P., M.M.), University of Texas Medical Branch, Galveston.
| |
Collapse
|
210
|
Rubin LP. Maternal and pediatric health and disease: integrating biopsychosocial models and epigenetics. Pediatr Res 2016; 79:127-35. [PMID: 26484619 DOI: 10.1038/pr.2015.203] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 08/13/2015] [Indexed: 11/09/2022]
Abstract
The concepts of allostasis (stability through adaptation) and accumulated life stress (McEwen's allostatic load) aim to understand childhood and adult outcomes. Chronic malnutrition, changes in social condition, and adverse early-life experiences may program phenotypes and contribute to long-lasting disease risk. However, integration of life course approaches, social and economic contexts, and comparison among different biopsychosocial models has not generally been explored. This review critically examines the literature and evaluates recent insights into how environmental stress can alter lifelong hypothalamic-pituitary-adrenal axis and immune system responsiveness and induce metabolic and neurodevelopmental maladaptation. Models of biopsychosocial stress overlap but may consider different conditions. Concepts include allostasis, which incorporates hormonal responses to predictable environmental changes, and Geronimus's "weathering," which aims to explain how socially structured, repeated stress can accumulate and increase disease vulnerability. Weathering emphasizes roles of internalized/interpersonal racism in outcomes disparities. For Mexican immigrants and Mexican Americans, the "acculturation" framework has proven especially useful to explore disparities, including preterm birth and neuropsychiatric risks in childhood. Complexities of stress assessments and recent research into epigenetic mechanisms mediating effects of physical, nutritional, psychological, and social stress are reviewed.
Collapse
Affiliation(s)
- Lewis P Rubin
- Department of Pediatrics, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, Texas
| |
Collapse
|
211
|
Bronson SL, Bale TL. The Placenta as a Mediator of Stress Effects on Neurodevelopmental Reprogramming. Neuropsychopharmacology 2016; 41:207-18. [PMID: 26250599 PMCID: PMC4677129 DOI: 10.1038/npp.2015.231] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/10/2015] [Accepted: 07/30/2015] [Indexed: 02/07/2023]
Abstract
Adversity experienced during gestation is a predictor of lifetime neuropsychiatric disease susceptibility. Specifically, maternal stress during pregnancy predisposes offspring to sex-biased neurodevelopmental disorders, including schizophrenia, attention deficit/hyperactivity disorder, and autism spectrum disorders. Animal models have demonstrated disease-relevant endophenotypes in prenatally stressed offspring and have provided unique insight into potential programmatic mechanisms. The placenta has a critical role in the deleterious and sex-specific effects of maternal stress and other fetal exposures on the developing brain. Stress-induced perturbations of the maternal milieu are conveyed to the embryo via the placenta, the maternal-fetal intermediary responsible for maintaining intrauterine homeostasis. Disruption of vital placental functions can have a significant impact on fetal development, including the brain, outcomes that are largely sex-specific. Here we review the novel involvement of the placenta in the transmission of the maternal adverse environment and effects on the developing brain.
Collapse
Affiliation(s)
- Stefanie L Bronson
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Tracy L Bale
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| |
Collapse
|
212
|
Quarini C, Pearson RM, Stein A, Ramchandani PG, Lewis G, Evans J. Are female children more vulnerable to the long-term effects of maternal depression during pregnancy? J Affect Disord 2016; 189:329-35. [PMID: 26469300 PMCID: PMC4650986 DOI: 10.1016/j.jad.2015.09.039] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 08/24/2015] [Accepted: 09/21/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Female fetuses are more vulnerable to high levels of maternal glucocorticoids. We examined whether exposure to prenatal maternal depression, a condition associated with high glucocorticoids, carries greater risk for depression at 12 and 18 years in girls. METHODS Our sample comprised 7959 mothers and children from the Avon Longitudinal Study of Parents and Children following imputation for missing data. Maternal depression was assessed pre-and post-natally, and offspring depression at ages 12 and 18. We used logistic regression models to examine the relationship between exposure to prenatal and postnatal depression and offspring depression at 18 and 12 and interactions with gender. RESULTS There was an interaction between prenatal depression and gender (P=0.027) and between postnatal depression and gender (P=0.027) for offspring depression at 18. Following adjustment in pre-natally depressed mothers, the odds ratio for offspring depression at 18 was 1.55 (95% c.i. 1.03-2.34) for girls and 0.54 (0.23-1.26) for boys. In post-natally depressed mothers, the odds ratio for offspring depression at 18 was 1.15 (0.70-1.89) in girls and 3.13 (1.52-6.45) in boys. However there was no evidence for interaction between prenatal or postnatal depression and gender (P=0.559 and 0.780 respectively) for offspring depression at 12. LIMITATIONS As expected with this large cohort spanning over 18 years, there was loss-to-follow-up. CONCLUSIONS This is the first evidence in humans that increased vulnerability of female fetuses to maternal stress responses during pregnancy persists into adolescence. One explanation for gender differences emerging later is more depressive symptomatology is attributed to heritable risk at 12, whereas biological processes involved in brain development at 18 may be influenced by foetal programming. If replicated, this study has potential to help understand intergenerational transmission of depression, a leading cause of morbidity worldwide.
Collapse
Affiliation(s)
| | - Rebecca M Pearson
- School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Alan Stein
- Department of Psychiatry, University of Oxford, Warneford Lane, Oxford OX3 7JX, UK
| | - Paul G Ramchandani
- Centre for Mental Health, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Glyn Lewis
- UCL Division of Psychiatry, University College London, Charles Bell house, 67-73 Riding House Street, London W1W 7EJ, UK
| | - Jonathan Evans
- School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| |
Collapse
|
213
|
Kroener L, Wang ET, Pisarska MD. Predisposing Factors to Abnormal First Trimester Placentation and the Impact on Fetal Outcomes. Semin Reprod Med 2015; 34:27-35. [PMID: 26696276 DOI: 10.1055/s-0035-1570029] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Normal placentation during the first trimester sets the stage for the rest of pregnancy and involves a finely orchestrated cellular and molecular interplay of maternal and fetal tissues. The resulting intrauterine environment plays an important role in fetal programming and the future health of the fetus, and is impacted by multiple genetic and epigenetic factors. Abnormalities in placentation and spiral artery invasion can lead to ischemia, placental disease, and adverse obstetrical outcomes including preeclampsia, intrauterine growth restriction, and placental abruption. Although first trimester placentation is affected by multiple factors, preconception environmental influences such as mode of conception, including assisted reproductive technologies which result in fertilization in vitro and intrauterine influences due to sex differences, are emerging as potential significant factors impacting first trimester placentation.
Collapse
Affiliation(s)
- Lindsay Kroener
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Erica T Wang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Margareta D Pisarska
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
214
|
Mandò C, Calabrese S, Mazzocco MI, Novielli C, Anelli GM, Antonazzo P, Cetin I. Sex specific adaptations in placental biometry of overweight and obese women. Placenta 2015; 38:1-7. [PMID: 26907375 DOI: 10.1016/j.placenta.2015.12.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/30/2015] [Accepted: 12/14/2015] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Placental biometry at birth has been shown to predict chronic disease in later life. We hypothesized that maternal overweight/obesity, a state of low-grade inflammation and risk factor for adverse pregnancy outcome, could negatively influence placental development and that differences would be sex-specific. METHODS 696 women (537 normal-weight, NW; 112 overweight, OW; 47 obese, OB) with singleton uncomplicated pregnancies were prospectively enrolled at term delivery. Gestational age, maternal (age, height, pre-pregnancy BMI, gestational weight gain -GWG, hemoglobin, hematocrit and glycemia), fetal (weight, length, ponderal index, cranial circumference) and placental (weight, diameters) data were collected. Placental area, thickness and efficiency (fetal/placental weight ratio, F/P) were calculated. RESULTS GWG was within standard recommendations in OB, while OW exceeded it. Placental weight was significantly higher in OW versus NW, but not in OB, leading to significantly higher placental thickness and lower F/P in this group. In the total population, a significant interaction effect between maternal BMI and fetal sex on placental weight and efficiency was found. Indeed, differences in placental parameters were present only in female offspring. DISCUSSION In our population of OW and OB uncomplicated pregnancies only OW women, presenting GWG over standard recommendations, had thicker and less efficient placentas. We also reported different placental adaptation depending on fetal sex, with significant changes only in female fetuses. This may be part of a female-specific strategy aiming to ensure survival if another adverse event occurs. Customized counseling according to maternal BMI and fetal sex should be evaluated in clinical care.
Collapse
Affiliation(s)
- Chiara Mandò
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy.
| | - Stefania Calabrese
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy
| | - Martina Ilaria Mazzocco
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy
| | - Chiara Novielli
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy
| | - Gaia Maria Anelli
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy
| | - Patrizio Antonazzo
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy
| | - Irene Cetin
- Department of Mother and Child, Hospital L. Sacco, Department of Biomedical and Clinical Sciences L. Sacco, and Center for Fetal Research Giorgio Pardi, Università degli studi di Milano, Italy
| |
Collapse
|
215
|
Milagro FI, Riezu-Boj JI, Martinez JA. Epigenetic Determinants of Weight Management: Methylation Signatures. Curr Nutr Rep 2015. [DOI: 10.1007/s13668-015-0140-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
216
|
Gray C, Vickers MH, Dyson RM, Reynolds CM, Berry MJ. Magnesium sulfate has sex-specific, dose-dependent vasodilator effects on preterm placental vessels. Biol Sex Differ 2015; 6:22. [PMID: 26543552 PMCID: PMC4634574 DOI: 10.1186/s13293-015-0040-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/28/2015] [Indexed: 12/26/2022] Open
Abstract
Background Women at risk of preterm delivery receive magnesium sulfate (MgSO4) in the pre-delivery phase to reduce their child’s risk of neurodevelopmental complications associated with preterm birth. However, the mechanisms underpinning its placental vascular role remain uncertain. Methods The aim of this study was to examine MgSO4 action on vascular tone in male and female human placental vessels from term and preterm deliveries. Vessels were obtained from placental biopsy following birth at term (37–41 weeks) or preterm gestation (<36 weeks of gestation). The vessels were mounted on a pressure myograph, pre-constricted with synthetic endoperoxide prostaglandin PGH2 (U46619) (0.1–100 μmol/l), and percentage of relaxation was calculated following incubation with bradykinin. Experiments were carried out in the presence of MgSO4 (0.2 mmol/l), NΨ-nitro-L-arginine methyl ester (L-NAME) (0.1 mmol/l), indomethacin (10 μmol/l), Ca2+-activated K+ channel blocker TRAM-34 (1 μM) and apamin (3 μM) to assess mechanisms of vascular function. Vascular [calcium ions (Ca2+)] was analysed using a colorimetric calcium assay. Results Vasodilation in vessels from preterm males was significantly blunted in the presence of MgSO4 when compared to preterm female and term male and female vessels. Overall, MgSO4 was observed to differentially modulate placental vascular tone and vascular calcium concentrations in a sex-specific manner. Conclusions As MgSO4 regulates human placental blood flow via specific pathways, foetal sex-specific MgSO4 treatment regimes may be necessary. In an era of increasing awareness of individualised medicine, sex-specific effects may be of importance when developing strategies to optimise care in high-risk patients.
Collapse
Affiliation(s)
- Clint Gray
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand ; Centre for Translational Physiology, University of Otago, Wellington, New Zealand ; Gravida: National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Gravida: National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Rebecca M Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand ; Centre for Translational Physiology, University of Otago, Wellington, New Zealand ; Department of Paediatrics, Graduate School of Medicine and IHMRI, University of Wollongong, Wollongong, NSW Australia
| | - Clare M Reynolds
- Gravida: National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand ; Centre for Translational Physiology, University of Otago, Wellington, New Zealand ; Capital and Coast District Health Board, Wellington, New Zealand
| |
Collapse
|
217
|
Panchenko PE, Lemaire M, Fneich S, Voisin S, Jouin M, Junien C, Gabory A. [Epigenetics and Nutrition: maternal nutrition impacts on placental development and health of offspring]. Biol Aujourdhui 2015; 209:175-87. [PMID: 26514387 DOI: 10.1051/jbio/2015021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 12/22/2022]
Abstract
The environment, defined broadly by all that is external to the individual, conditions the phenotype during development, particularly the susceptibility to develop non-communicable diseases. This notion, called Developmental Origins of Health and Disease (DOHaD), is based on numerous epidemiological studies as well as animal models. Thus, parental nutrition and obesity can predispose the offspring to develop metabolic and cardiovascular diseases in adulthood. The known underlying mechanisms include an altered development of tissues that adapt to maternal metabolic condition, and a placental dysfunction, which in turn impacts fetal growth and development. Epigenetic mechanisms modulate gene expression without affecting the DNA sequence itself. The main epigenetic marks are DNA methylation and histone post-translational modifications. These marks are erased and set-up during gametogenesis and development in order to ensure cellular identity. Therefore, they can lead to a memorisation of early environment and induce long-term alteration of cell and tissue functions, which will condition the susceptibility to non-communicable diseases. The placenta is a programming agent of adult disease. The environment, such as smoking or psychosocial stress, is able to modify epigenetic processes in placenta, such as small RNA expression and DNA methylation. We showed that placenta is sensitive to maternal obesity and maternal nutrition, in terms of histology, transcription and epigenetic marks. A clear sexual dimorphism is remarkable in the placental response to maternal environment. In adulthood, the phenotype is also different between males and females. Epigenetic mechanisms could underlie this differential response of males and females to the same environment. The DOHaD can no longer be ignored in Biology of Reproduction. The prevention of non-communicable diseases must take this new paradigm into account. Research will allow a better comprehension of the mechanisms of this early conditioning and the marked sexual dimorphism it is associated to.
Collapse
Affiliation(s)
- Polina E Panchenko
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France - Université Pierre et Marie Curie, Paris VI, 75005 Paris, France
| | - Marion Lemaire
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| | - Sara Fneich
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| | - Sarah Voisin
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France - Université Pierre et Marie Curie, Paris VI, 75005 Paris, France
| | - Mélanie Jouin
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| | - Claudine Junien
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| | - Anne Gabory
- INRA, UMR1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| |
Collapse
|
218
|
Abstract
The placenta is an ephemeral but critical organ for the survival of all eutherian mammals and marsupials. It is the primary messenger system between the mother and fetus, where communicational signals, nutrients, waste, gases, and extrinsic factors are exchanged. Although the placenta may buffer the fetus from various environmental insults, placental dysfunction might also contribute to detrimental developmental origins of adult health and disease effects. The placenta of one sex over the other might possess greater ability to respond and buffer against environmental insults. Given the potential role of the placenta in effecting the lifetime health of the offspring, it is not surprising that there has been a resurging interest in this organ, including the Human Placental Project launched by the National Institutes of Child Health and Human Development. In this review, we will compare embryological development of the laboratory mouse and human chorioallantoic placentae. Next, evidence that various species, including humans, exhibit normal sex-dependent structural and functional placental differences will be examined followed by how in utero environmental changes (nutritional state, stress, and exposure to environmental chemicals) might interact with fetal sex to affect this organ. Recent data also suggest that paternal state impacts placental function in a sex-dependent manner. The research to date linking placental maladaptive responses and later developmental origins of adult health and disease effects will be explored. Finally, we will focus on how sex chromosomes and epimutations may contribute to sex-dependent differences in placental function, the unanswered questions, and future directions that warrant further consideration.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Departments of Bond Life Sciences Center, Biomedical Sciences, Genetics Area Program, and Research Faculty Member for the Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
219
|
Grandjean P, Barouki R, Bellinger DC, Casteleyn L, Chadwick LH, Cordier S, Etzel RA, Gray KA, Ha EH, Junien C, Karagas M, Kawamoto T, Paige Lawrence B, Perera FP, Prins GS, Puga A, Rosenfeld CS, Sherr DH, Sly PD, Suk W, Sun Q, Toppari J, van den Hazel P, Walker CL, Heindel JJ. Life-Long Implications of Developmental Exposure to Environmental Stressors: New Perspectives. Endocrinology 2015; 156:3408-15. [PMID: 26241067 PMCID: PMC4588822 DOI: 10.1210/en.2015-1350] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) paradigm is one of the most rapidly expanding areas of biomedical research. Environmental stressors that can impact on DOHaD encompass a variety of environmental and occupational hazards as well as deficiency and oversupply of nutrients and energy. They can disrupt early developmental processes and lead to increased susceptibility to disease/dysfunctions later in life. Presentations at the fourth Conference on Prenatal Programming and Toxicity in Boston, in October 2014, provided important insights and led to new recommendations for research and public health action. The conference highlighted vulnerable exposure windows that can occur as early as the preconception period and epigenetics as a major mechanism than can lead to disadvantageous "reprogramming" of the genome, thereby potentially resulting in transgenerational effects. Stem cells can also be targets of environmental stressors, thus paving another way for effects that may last a lifetime. Current testing paradigms do not allow proper characterization of risk factors and their interactions. Thus, relevant exposure levels and combinations for testing must be identified from human exposure situations and outcome assessments. Testing of potential underpinning mechanisms and biomarker development require laboratory animal models and in vitro approaches. Only few large-scale birth cohorts exist, and collaboration between birth cohorts on a global scale should be facilitated. DOHaD-based research has a crucial role in establishing factors leading to detrimental outcomes and developing early preventative/remediation strategies to combat these risks.
Collapse
Affiliation(s)
- Philippe Grandjean
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Robert Barouki
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - David C Bellinger
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Ludwine Casteleyn
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Lisa H Chadwick
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Sylvaine Cordier
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Ruth A Etzel
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Kimberly A Gray
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Eun-Hee Ha
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Claudine Junien
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Margaret Karagas
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Toshihiro Kawamoto
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - B Paige Lawrence
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Frederica P Perera
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Gail S Prins
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Alvaro Puga
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Cheryl S Rosenfeld
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - David H Sherr
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Peter D Sly
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - William Suk
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Qi Sun
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Jorma Toppari
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Peter van den Hazel
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Cheryl L Walker
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| | - Jerrold J Heindel
- Departments of Environmental Health (P.G., D.C.B.) and Nutrition (Q.S.), Harvard T.H. Chan School of Public Health; Children's Hospital (D.C.B.), Harvard Medical School; and Channing Division of Network Medicine (Q.S.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Environmental Medicine (P.G.), Institute of Public Health, University of Southern Denmark, 5000 Odense, Denmark; Institut National de la Santé et de la Recherche Médicale unit 1124 (R.B.), Université Paris Descartes, Hôpital Necker enfants malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; University of Leuven (L.C.), Center for Human Genetics, 3000 Leuven, Belgium; Division of Extramural Research and Training (L.H.C., K.A.G., W.S., J.J.H.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709; Institut National de la Santé et de la Recherche Médicale, Unit 1085 (S.C.), University Rennes I, F-35000 Rennes, France; Joseph J. Zilber School of Public Health (R.A.E.), University of Wisconsin, Milwaukee, Wisconsin 53201; Department of Preventive Medicine (E.-H.H.), School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea; Institut National de la Recherche Agronomique (C.J.), MR Unité Mixte de Recherche 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France; Université Versailles-St-Quentin (C.J.), 78000 Versailles, France; Children's Environmental Health and Disease Prevention Research Center and Department of Epidemiology, Geisel School of Medicine at Dartmouth (M.K.), Hanover, New Hampshire 03766; Department of Environmental Health (T.K.), University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan; Department of Environmental Medicine (B.P.L.), University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Department of Environmental Health Sciences and Columbia Center for Children's Environmental Heal
| |
Collapse
|
220
|
Rosenfeld CS. Microbiome Disturbances and Autism Spectrum Disorders. Drug Metab Dispos 2015; 43:1557-71. [PMID: 25852213 DOI: 10.1124/dmd.115.063826] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 04/06/2015] [Indexed: 12/18/2022] Open
Abstract
Autism spectrum disorders (ASDs) are considered a heterogenous set of neurobehavioral diseases, with the rates of diagnosis dramatically increasing in the past few decades. As genetics alone does not explain the underlying cause in many cases, attention has turned to environmental factors as potential etiological agents. Gastrointestinal disorders are a common comorbidity in ASD patients. It was thus hypothesized that a gut-brain link may account for some autistic cases. With the characterization of the human microbiome, this concept has been expanded to include the microbiota-gut-brain axis. There are mounting reports in animal models and human epidemiologic studies linking disruptive alterations in the gut microbiota or dysbiosis and ASD symptomology. In this review, we will explore the current evidence that gut dysbiosis in animal models and ASD patients correlates with disease risk and severity. The studies to date have surveyed how gut microbiome changes may affect these neurobehavioral disorders. However, we harbor other microbiomes in the body that might impact brain function. We will consider microbial colonies residing in the oral cavity, vagina, and the most recently discovered one in the placenta. Based on the premise that gut microbiota alterations may be causative agents in ASD, several therapeutic options have been tested, such as diet modulations, prebiotics, probiotics, synbiotics, postbiotics, antibiotics, fecal transplantation, and activated charcoal. The potential benefits of these therapies will be considered. Finally, the possible mechanisms by which changes in the gut bacterial communities may result in ASD and related neurobehavioral disorders will be examined.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Bond Life Sciences Center, Thompson Center for Autism and Neurobehavioral Disorders, Genetics Area Program, and Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
221
|
Matheson H, Veerbeek JHW, Charnock-Jones DS, Burton GJ, Yung HW. Morphological and molecular changes in the murine placenta exposed to normobaric hypoxia throughout pregnancy. J Physiol 2015; 594:1371-88. [PMID: 26278110 PMCID: PMC4771784 DOI: 10.1113/jp271073] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/04/2015] [Indexed: 11/22/2022] Open
Abstract
Key points Exposure of pregnant mice to chronic hypoxia at 13% O2 induces fetal growth restriction but increases placental weight. Sex dimorphism induces differential responses in placental weight to hypoxia. The male placenta is heavier than the female and is associated with less severe fetal growth restriction. Increases in maternal arterial/venous blood spaces and higher protein kinase B (Akt)‐mechanistic target of rapamycin growth signalling could contribute to the heavier hypoxic placenta. Placental endoplasmic reticulum stress is elevated equally in both sexes in response to hypoxia. In comparison, oxidative stress is only apparent in female placentas. Chronic hypoxia induces down‐regulation of placental mitochondrial electron transport chain complexes protein subunits but does not cause intracellular energy depletion.
Abstract Chronic hypoxia is a common complication of pregnancy, arising through malperfusion of the placenta or pregnancy at high altitude. The present study investigated the effects of hypoxia on the growth of the placenta, which is the organ that interfaces between the mother and her fetus. Mice were housed in an hypoxic environment for the whole of gestation. An atmosphere of 13% oxygen induced fetal growth restriction (1182 ± 9 mg, n = 90 vs. 1044 ± 11 mg, n = 62, P < 0.05) but enhanced placental weight (907 ± 11 mg, n = 90 vs. 998 ± 15 mg, n = 62,P < 0.05). Stereological analyses revealed an increase in the volume of maternal blood spaces in the placenta, consistent with increased flow. At the molecular level, we observed activation of the protein kinase B (Akt)‐mechanistic target of rapamycin growth and proliferation pathway. Chronic hypoxia also triggered mild endoplasmic reticulum stress, a conserved homeostatic response that mediates translational arrest through phosphorylation of eukaryotic initiation factor 2 subunit α. Surprisingly, although subunits of the mitochondrial electron transport chain complexes were reduced at the protein level, there was no evidence of intracellular energy depletion. Finally, we demonstrated sex‐specific placental responses to chronic hypoxia. Placentas from male fetuses were heavier (1082 ± 2 mg, n = 30 vs. 928 ± 2 mg, n = 34, P < 0.05) and less susceptible to hypoxia‐induced oxidative stress than those from females. Their capacity to adapt may explain why male fetuses were significantly less growth restricted at embryonic day 18.5 than their female counterparts. These findings are consistent with the concept that male fetuses are more aggressive with respect to their nutrient demands, which may place them at greater risk of adverse outcomes under limiting conditions. Exposure of pregnant mice to chronic hypoxia at 13% O2 induces fetal growth restriction but increases placental weight. Sex dimorphism induces differential responses in placental weight to hypoxia. The male placenta is heavier than the female and is associated with less severe fetal growth restriction. Increases in maternal arterial/venous blood spaces and higher protein kinase B (Akt)‐mechanistic target of rapamycin growth signalling could contribute to the heavier hypoxic placenta. Placental endoplasmic reticulum stress is elevated equally in both sexes in response to hypoxia. In comparison, oxidative stress is only apparent in female placentas. Chronic hypoxia induces down‐regulation of placental mitochondrial electron transport chain complexes protein subunits but does not cause intracellular energy depletion.
Collapse
Affiliation(s)
- Hannah Matheson
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jan H W Veerbeek
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Birth Center and the Division of Woman and Baby, University Medical Center Utrecht, Utrecht, The Netherlands
| | - D Stephen Charnock-Jones
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital, Cambridge, UK.,National Institute for Health Research, Cambridge Comprehensive Biomedical Research Centre, Cambridge, UK
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hong Wa Yung
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
222
|
DeBenedictis B, Guan H, Yang K. Prenatal Exposure to Bisphenol A Disrupts Mouse Fetal Liver Maturation in a Sex-Specific Manner. J Cell Biochem 2015; 117:344-50. [PMID: 26146954 DOI: 10.1002/jcb.25276] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/30/2015] [Indexed: 12/31/2022]
Abstract
Bisphenol A (BPA) is one of the most prevalent endocrine disrupting chemicals in the environment. Developmental exposure to BPA is known to be associated with liver dysfunction and diseases, such as hepatic steatosis, liver tumors, metabolic syndrome, and altered hepatic gene expression, and DNA methylation profiles. However, the effects of BPA on rodent liver development are unknown. The present study was undertaken to address this important question using the mouse as an experimental model. Pregnant mice were exposed to BPA via diet from embryonic day 7.5 (E7.5) to E18.5. At E18.5, fetal livers were collected, and analyzed for changes in the expression of key hepatocyte maturation markers. We found the following significant alterations in BPA-exposed female but not male fetal livers: (a) levels of the mature hepatocyte markers, albumin and glycogen synthase proteins, were decreased (-65% and -40%, respectively); (b) levels of the immature hepatocyte marker, α-fetoprotein, were increased (+43%); (c) the level of C/EBP-α protein, the master transcription factor essential for hepatocyte maturation, was down-regulated (-50%); and (d) the level of PCNA protein (marker of proliferation) was elevated (+40%), while that of caspase-3 protein and activity (markers of apoptosis) was reduced (-40% and -55%, respectively), suggestive of a perturbed balance between cell proliferation and apoptosis in BPA-exposed female fetuses. Taken together, these findings demonstrate that prenatal exposure to BPA disrupts the mouse fetal liver maturation in a sex-specific manner, and suggest a fetal origin for BPA-induced hepatic dysfunction and diseases.
Collapse
Affiliation(s)
- Bianca DeBenedictis
- Departments of Obstetrics and Gynaecology, Western University, London, Ontario, Canada, N6C 2V5.,Departments of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6C 2V5.,Children's Health Research Institute, Western University, London, Ontario, Canada, N6C 2V5
| | - Haiyan Guan
- Departments of Obstetrics and Gynaecology, Western University, London, Ontario, Canada, N6C 2V5.,Departments of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6C 2V5.,Children's Health Research Institute, Western University, London, Ontario, Canada, N6C 2V5
| | - Kaiping Yang
- Departments of Obstetrics and Gynaecology, Western University, London, Ontario, Canada, N6C 2V5.,Departments of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6C 2V5.,Children's Health Research Institute, Western University, London, Ontario, Canada, N6C 2V5
| |
Collapse
|
223
|
Mohanty AF, Thompson ML, Burbacher TM, Siscovick DS, Williams MA, Enquobahrie DA. Periconceptional Seafood Intake and Fetal Growth. Paediatr Perinat Epidemiol 2015; 29:376-87. [PMID: 26147526 PMCID: PMC4536156 DOI: 10.1111/ppe.12205] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Previous reports of associations of maternal seafood intake with fetal growth were inconsistent. Further, little is known whether associations differ across seafood subtypes or fetal growth indices. METHODS Among 3141 participants of the Omega study, a pregnancy cohort study, we investigated associations of periconceptional shell, lean, and fatty fish intake with fetal growth indices. We categorised food frequency questionnaire reported seafood intake into frequencies of: <0.2 servings/month, 0.2 servings/month -<0.5 servings/week, 0.5-1 servings/week, and >1 servings/week. We abstracted birthweight, birth length, and head circumference from medical records. Using generalised linear models with a log link, the Poisson family, and robust standard errors, we estimated relative risks and 95% confidence intervals (CI) for low birthweight (LBW, <2500 g) and linear regression models to estimate mean differences for continuous fetal growth indices across seafood intake categories. RESULTS Medians (interquartile range) of shell, lean, and fatty fish intake were 0.3 (0-0.9), 0.5 (0-1.0), and 0.5 (0.1-1.0) servings/week, respectively. Lean fish intake of >1 servings/week (vs. <0.2 servings/month) was associated with a 2.2-fold higher risk of LBW (95% CI 1.2, 4.1). Shellfish intake of >1 servings/week (vs. <0.2 servings/month) was associated with a 0.6 kg/m(3) higher mean ponderal index (95% CI 0.0, 1.2 kg/m(3) ). There was no evidence for associations of total seafood or seafood subtype intake with other fetal growth indices. CONCLUSIONS Higher intakes of lean fish and shellfish were associated with a higher risk of LBW and higher mean ponderal index, respectively. Findings highlight the importance of considerations of seafood subtype in similar investigations.
Collapse
Affiliation(s)
- April F Mohanty
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA
| | | | - Thomas M Burbacher
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - David S Siscovick
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, Department of Medicine, University of Washington, Seattle, WA
| | | | - Daniel A Enquobahrie
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, Center for Perinatal Studies, Swedish Medical Center, Seattle, WA
| |
Collapse
|
224
|
Derraik JGB, Ahlsson F, Lundgren M, Jonsson B, Cutfield WS. First-borns have greater BMI and are more likely to be overweight or obese: a study of sibling pairs among 26,812 Swedish women. J Epidemiol Community Health 2015; 70:78-81. [PMID: 26311896 DOI: 10.1136/jech-2014-205368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 07/27/2015] [Indexed: 11/04/2022]
Abstract
BACKGROUND A number of large studies have shown phenotypic differences between first-borns and later-borns among adult men. In this study, we aimed to assess whether birth order was associated with height and BMI in a large cohort of Swedish women. METHODS Information was obtained from antenatal clinic records from the Swedish National Birth Register over 20 years (1991-2009). Maternal anthropometric data early in pregnancy (at approximately 10-12 weeks of gestation) were analysed on 13,406 pairs of sisters who were either first-born or second-born (n=26,812). RESULTS Early in pregnancy, first-born women were of BMI that was 0.57 kg/m(2) (2.4%) greater than their second-born sisters (p<0.0001). In addition, first-borns had greater odds of being overweight (OR 1.29; p<0.0001) or obese (OR 1.40; p<0.0001) than second-borns. First-borns were also negligibly taller (+1.2 mm) than their second-born sisters. Of note, there was a considerable increase in BMI over the 18-year period covered by this study, with an increment of 0.11 kg/m(2) per year (p<0.0001). CONCLUSIONS Our study corroborates other large studies on men, and the steady reduction in family size may contribute to the observed increase in adult BMI worldwide.
Collapse
Affiliation(s)
- José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Fredrik Ahlsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Maria Lundgren
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Björn Jonsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
225
|
Sex differences in the early life correlates of natural antibody concentrations. J Dev Orig Health Dis 2015; 6:501-11. [PMID: 26279187 DOI: 10.1017/s2040174415001373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Innate-like B1a lymphocytes arise from long-lived progenitors produced exclusively by fetal stem cells. Any insults coinciding with this early lymphopoietic wave could have a permanent impact on the B1a population and its unique protein products, the natural antibodies (NAb). We investigated early life nutritional influences on NAb concentrations of pre-adolescent children (n=290) in rural Nepal for whom we had extensive information on exposures from pregnancy and early infancy. Infant size and growth were strongly associated with NAb concentrations at 9-13 years of age among males (e.g., for neonatal weight: βBOYS=0.43; P<0.001), but not females (e.g., for neonatal weight: βGIRLS=-0.16; P=0.26). In females, season of birth was associated with NAb concentrations, with marked reductions among girls born during the pre-monsoon (March-May; βGIRLS=-0.39; P=0.01) and pre-harvest (September-November; βGIRLS=-0.35; P=0.03) seasons. Our findings suggest that nutritional or other environmental influences on immune development may vary by sex, with potential consequences for immune function during infancy and long-term risk of immune-mediated disease.
Collapse
|
226
|
de Vries GJ, Forger NG. Sex differences in the brain: a whole body perspective. Biol Sex Differ 2015; 6:15. [PMID: 26279833 PMCID: PMC4536872 DOI: 10.1186/s13293-015-0032-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/28/2015] [Indexed: 12/29/2022] Open
Abstract
Most writing on sexual differentiation of the mammalian brain (including our own) considers just two organs: the gonads and the brain. This perspective, which leaves out all other body parts, misleads us in several ways. First, there is accumulating evidence that all organs are sexually differentiated, and that sex differences in peripheral organs affect the brain. We demonstrate this by reviewing examples involving sex differences in muscles, adipose tissue, the liver, immune system, gut, kidneys, bladder, and placenta that affect the nervous system and behavior. The second consequence of ignoring other organs when considering neural sex differences is that we are likely to miss the fact that some brain sex differences develop to compensate for differences in the internal environment (i.e., because male and female brains operate in different bodies, sex differences are required to make output/function more similar in the two sexes). We also consider evidence that sex differences in sensory systems cause male and female brains to perceive different information about the world; the two sexes are also perceived by the world differently and therefore exposed to differences in experience via treatment by others. Although the topic of sex differences in the brain is often seen as much more emotionally charged than studies of sex differences in other organs, the dichotomy is largely false. By putting the brain firmly back in the body, sex differences in the brain are predictable and can be more completely understood.
Collapse
Affiliation(s)
- Geert J. de Vries
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA 30302-5030 USA
| | - Nancy G. Forger
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA 30302-5030 USA
| |
Collapse
|
227
|
Induction of autophagy through the activating transcription factor 4 (ATF4)-dependent amino acid response pathway in maternal skeletal muscle may function as the molecular memory in response to gestational protein restriction to alert offspring to maternal nutrition. Br J Nutr 2015. [DOI: 10.1017/s0007114515002172] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of the present study was to investigate the mechanistic basis of protein deficiency during pregnancy in mother that is transduced to offspring. To this end, timed-pregnant Sprague–Dawley rats were fed either a control (20 % of energy from protein) or low-protein (LP, 8 % of energy from protein) diet during gestation. Tissues were collected after delivery from rat dams, and skeletal muscle was collected at postnatal day 38 from the offspring. Quantitative RT-PCR and Western blot analyses were performed to determine mRNA and protein levels. Histological analysis was performed to evaluate myofibre size. LP dams gained significantly less weight during pregnancy, developed muscle atrophy, and had significantly lower circulating threonine and histidine levels than control dams. The mRNA expression of the well-known amino acid response (AAR) pathway-related target genes was increased only in the skeletal muscle of LP dams, as well as the protein expression levels of activating transcription factor 4 (ATF4) and phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α). The mRNA expression of autophagy-related genes was significantly increased in the skeletal muscle of LP dams. Moreover, the mRNA expression of genes involved in both AAR and autophagy pathways remained elevated and was memorised in the muscle of LP offspring that consumed a post-weaning control diet. Additionally, the LP diet increased an autophagy marker, microtubule-associated proteins 1A/1B light chain 3B (LC3B) protein expression in the skeletal muscle of rat dams, consistent with the initiation of autophagy. The LP diet further increased ATF4 binding at the predicted regions of AAR and autophagy pathway-related genes. Increased binding of ATF4 unveils the crucial role of ATF4 in the activation of autophagy in response to protein restriction. Our data suggest that molecular changes in maternal muscle are memorised in the offspring long after gestational protein restriction, reinforcing the role of maternal signalling in programming offspring health.
Collapse
|
228
|
Affiliation(s)
- Joshua B Rubin
- Departments of Pediatrics and Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
229
|
Paquette AG, Marsit CJ. The developmental basis of epigenetic regulation of HTR2A and psychiatric outcomes. J Cell Biochem 2015; 115:2065-72. [PMID: 25043477 DOI: 10.1002/jcb.24883] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 07/09/2014] [Indexed: 12/21/2022]
Abstract
The serotonin receptor 5-HT2A (encoded by HTR2A) is an important regulator of fetal brain development and adult cognitive function. Environmental signals that induce epigenetic changes of serotonin response genes, including HTR2A, have been implicated in adverse mental health outcomes. The objective of this perspective article is to address the medical implications of HTR2A epigenetic regulation, which has been associated with both infant neurobehavioral outcomes and adult mental health. Ongoing research has identified a region of the HTR2A promoter that has been associated with a number of medical outcomes in adults and infants, including bipolar disorder, schizophrenia, chronic fatigue syndrome, borderline personality disorder, suicidality, and neurobehavioral outcomes. Epigenetic regulation of HTR2A has been studied in several different types of tissues, including the placenta. The placenta is an important source of serotonin during fetal neurodevelopment, and placental epigenetic variation of HTR2A has been associated with infant neurobehavioral outcomes, which may represent the basis of adult mental health disorders. Further analysis is needed to identify intrinsic and extrinsic factors that modulate HTR2A methylation, and the mechanism by which this epigenetic variation influences fetal growth and leads to altered brain development, manifesting in psychiatric disorders.
Collapse
Affiliation(s)
- Alison G Paquette
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | | |
Collapse
|
230
|
Filis P, Nagrath N, Fraser M, Hay DC, Iredale JP, O'Shaughnessy P, Fowler PA. Maternal Smoking Dysregulates Protein Expression in Second Trimester Human Fetal Livers in a Sex-Specific Manner. J Clin Endocrinol Metab 2015; 100:E861-70. [PMID: 25803269 PMCID: PMC4533306 DOI: 10.1210/jc.2014-3941] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CONTEXT Maternal smoking during pregnancy has adverse effects on the offspring (eg, increased likelihood of metabolic syndrome and infertility), which may involve alterations in fetal liver function. OBJECTIVE Our aim was to analyze, for the first time, the human fetal liver proteome to identify pathways affected by maternal smoking. DESIGN Fetal liver proteins extracted from elective second trimester pregnancy terminations (12-16 weeks of gestation) were divided in four balanced groups based on sex and maternal smoking. SETTING AND PARTICIPANTS Livers were collected from 24 morphologically normal fetuses undergoing termination for nonmedical reasons and analyzed at the Universities of Aberdeen and Glasgow. MAIN OUTCOME MEASURES Protein extracts were resolved by 2D-PAGE and analyzed with SameSpots software. Ingenuity pathway analysis was used to investigate likely roles of dysregulated proteins identified by tandem liquid chromatography/mass spectroscopy. RESULTS Significant expression differences between one or more groups (fetal sex and/or maternal smoking) were found in 22 protein spots. Maternal smoking affected proteins with roles in post-translational protein processing and secretion (ERP29, PDIA3), stress responses and detoxification (HSP90AA1, HSBP1, ALDH7A1, CAT), and homeostasis (FTL1, ECHS1, GLUD1, AFP, SDHA). Although proteins involved in necrosis and cancer development were affected in both sexes, pathways affecting cellular homeostasis, inflammation, proliferation, and apoptosis were affected in males and pathways affecting glucose metabolism were affected in females. CONCLUSIONS The fetal liver exhibits marked sex differences at the protein level, and these are disturbed by maternal smoking. The foundations for smoke-induced post-natal diseases are likely to be due to sex-specific effects on diverse pathways.
Collapse
|
231
|
Reynolds CM, Vickers MH, Harrison CJ, Segovia SA, Gray C. Maternal high fat and/or salt consumption induces sex-specific inflammatory and nutrient transport in the rat placenta. Physiol Rep 2015; 3:3/5/e12399. [PMID: 25991721 PMCID: PMC4463828 DOI: 10.14814/phy2.12399] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Maternal high fat and salt consumption are associated with developmental programming of disease in adult offspring. Inadequacies in placental nutrient transport may explain these ‘programmed effects’. Diet-induced inflammation may have detrimental effects on placental function leading to alteration of key nutrient transporters. We examined the effects of maternal high fat and/or salt diets on markers of placental nutrient transport and inflammation. Sprague–Dawley rats were assigned to (1) control (CD; 1% Salt 10% kcal from fat); (2) high salt (SD; 4% salt, 10% kcal from fat); (3) high fat (HF; 1% Salt 45% kcal from fat) or (4) high fat high salt (HFSD; 4% salt, 45% kcal from fat) 21 days prior to and throughout gestation. At embryonic day 18, dams were killed by isoflurane anesthesia followed by decapitation; placenta/fetuses were weighed, sexed, and collected for molecular analysis. Maternal SD, HF, and HFSD consumption decreased weight of placenta derived from male offspring; however, weight of placenta derived from female offspring was only reduced with maternal HF diet. This was associated with increased expression of LPL, SNAT2, GLUT1, and GLUT4 in placenta derived from male offspring suggesting increased fetal exposure to free fatty acids and glucose. Maternal SD, HF, and HFSD diet consumption increased expression of proinflammatory mediators IL-1β, TNFα, and CD68 in male placenta. Our results suggest that a proinflammatory placental profile results in detrimental alterations in nutrient transport which may contribute to the developmental origins of cardio-metabolic disturbances in offspring throughout life.
Collapse
Affiliation(s)
- Clare M Reynolds
- Liggins Institute and Gravida, National Centre for Growth and Development University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Liggins Institute and Gravida, National Centre for Growth and Development University of Auckland, Auckland, New Zealand
| | - Claudia J Harrison
- Liggins Institute and Gravida, National Centre for Growth and Development University of Auckland, Auckland, New Zealand
| | - Stephanie A Segovia
- Liggins Institute and Gravida, National Centre for Growth and Development University of Auckland, Auckland, New Zealand
| | - Clint Gray
- Liggins Institute and Gravida, National Centre for Growth and Development University of Auckland, Auckland, New Zealand
| |
Collapse
|
232
|
Daraki V, Georgiou V, Papavasiliou S, Chalkiadaki G, Karahaliou M, Koinaki S, Sarri K, Vassilaki M, Kogevinas M, Chatzi L. Metabolic profile in early pregnancy is associated with offspring adiposity at 4 years of age: the Rhea pregnancy cohort Crete, Greece. PLoS One 2015; 10:e0126327. [PMID: 25970502 PMCID: PMC4430416 DOI: 10.1371/journal.pone.0126327] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/19/2015] [Indexed: 12/14/2022] Open
Abstract
Context Maternal pre-pregnancy obesity may increase the risk of childhood obesity but it is unknown whether other metabolic factors in early pregnancy such as lipid profile and hypertension are associated with offspring cardiometabolic traits. Objective Our objective was to investigate whether fasting lipid, glucose, and insulin levels during early pregnancy and maternal pre-pregnancy weight status, are associated with offspring adiposity measures, lipid levels and blood pressure at preschool age. Design and Methods The study included 618 mother-child pairs of the pregnancy cohort “Rhea” study in Crete, Greece. Pregnant women were recruited at the first prenatal visit (mean: 12weeks, SD: 0.7). A subset of 348 women provided fasting serum samples for glucose and lipid measurements. Outcomes measures were body mass index, abdominal circumference, sum of skinfold thickness, and blood pressure measurements at 4 years of age. A subsample of 525 children provided non-fasting blood samples for lipid measurements. Results Pre-pregnancy overweight/obesity was associated with greater risk of offspring overweight/obesity (RR: 1.83, 95%CI: 1.19, 2.81), central adiposity (RR: 1.97, 95%CI: 1.11, 3.49), and greater fat mass by 5.10mm (95%CI: 2.49, 7.71) at 4 years of age. These associations were more pronounced in girls. An increase of 40mg/dl in fasting serum cholesterol levels in early pregnancy was associated with greater skinfold thickness by 3.30mm (95%CI: 1.41, 5.20) at 4 years of age after adjusting for pre-pregnancy BMI and several other confounders. An increase of 10mmHg in diastolic blood pressure in early pregnancy was associated with increased risk of offspring overweight/obesity (RR: 1.22, 95%CI: 1.03, 1.45), and greater skinfold thickness by 1.71mm (95% CI: 0.57, 2.86) at 4 years of age. Conclusions Metabolic dysregulation in early pregnancy may increase the risk of obesity at preschool age.
Collapse
Affiliation(s)
- Vasiliki Daraki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece; Clinic of Endocrinology, University Hospital of Crete, Heraklion, Greece
| | - Vaggelis Georgiou
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | | | - Georgia Chalkiadaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Marianna Karahaliou
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Stella Koinaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Katerina Sarri
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Maria Vassilaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Manolis Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; IMIM (Hospital del Mar Research Institute), Barcelona, Spain; CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain; National School of Public Health, Athens, Greece
| | - Leda Chatzi
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
233
|
Mohanty AF, Farin FM, Bammler TK, MacDonald JW, Afsharinejad Z, Burbacher TM, Siscovick DS, Williams MA, Enquobahrie DA. Infant sex-specific placental cadmium and DNA methylation associations. ENVIRONMENTAL RESEARCH 2015; 138:74-81. [PMID: 25701811 PMCID: PMC4385453 DOI: 10.1016/j.envres.2015.02.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/26/2015] [Accepted: 02/03/2015] [Indexed: 05/18/2023]
Abstract
BACKGROUND Recent evidence suggests that maternal cadmium (Cd) burden and fetal growth associations may vary by fetal sex. However, mechanisms contributing to these differences are unknown. OBJECTIVES Among 24 maternal-infant pairs, we investigated infant sex-specific associations between placental Cd and placental genome-wide DNA methylation. METHODS We used ANOVA models to examine sex-stratified associations of placental Cd (dichotomized into high/low Cd using sex-specific Cd median cutoffs) with DNA methylation at each cytosine-phosphate-guanine site or region. Statistical significance was defined using a false discovery rate cutoff (<0.10). RESULTS Medians of placental Cd among females and males were 5 and 2 ng/g, respectively. Among females, three sites (near ADP-ribosylation factor-like 9 (ARL9), siah E3 ubiquitin protein ligase family member 3 (SIAH3), and heparin sulfate (glucosamine) 3-O-sulfotransferase 4 (HS3ST4) and one region on chromosome 7 (including carnitine O-octanoyltransferase (CROT) and TP5S target 1 (TP53TG1)) were hypomethylated in high Cd placentas. Among males, high placental Cd was associated with methylation of three sites, two (hypomethylated) near MDS1 and EVI1 complex locus (MECOM) and one (hypermethylated) near spalt-like transcription factor 1 (SALL1), and two regions (both hypomethylated, one on chromosome 3 including MECOM and another on chromosome 8 including rho guanine nucleotide exchange factor (GEF) 10 (ARHGEF10). Differentially methylated sites were at or close to transcription start sites of genes involved in cell damage response (SIAH3, HS3ST4, TP53TG1) in females and cell differentiation, angiogenesis and organ development (MECOM, SALL1) in males. CONCLUSIONS Our preliminary study supports infant sex-specific placental Cd-DNA methylation associations, possibly accounting for previously reported differences in Cd-fetal growth associations across fetal sex. Larger studies are needed to replicate and extend these findings. Such investigations may further our understanding of epigenetic mechanisms underlying maternal Cd burden with suboptimal fetal growth associations.
Collapse
Affiliation(s)
- April F Mohanty
- Cardiovascular Health Research Unit, University of Washington, 1730 Minor Ave, Seattle, WA 98101, USA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA.
| | - Fred M Farin
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt Way N.E., Suite #100, Seattle, WA 98105, USA.
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt Way N.E., Suite #100, Seattle, WA 98105, USA.
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt Way N.E., Suite #100, Seattle, WA 98105, USA.
| | - Zahra Afsharinejad
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt Way N.E., Suite #100, Seattle, WA 98105, USA.
| | - Thomas M Burbacher
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Box: 357234, 1705 N.E. Pacific Street, Seattle, WA 98195, USA.
| | - David S Siscovick
- Cardiovascular Health Research Unit, University of Washington, 1730 Minor Ave, Seattle, WA 98101, USA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Michelle A Williams
- Department of Epidemiology, Harvard School of Public Health, Kresge Building, 9th Floor, 677 Huntington Ave., Boston, MA 02115, USA.
| | - Daniel A Enquobahrie
- Cardiovascular Health Research Unit, University of Washington, 1730 Minor Ave, Seattle, WA 98101, USA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA; Center for Perinatal Studies, Swedish Medical Center, 1124 Columbia Street, Suite 750, Seattle, WA 98104, USA.
| |
Collapse
|
234
|
Adverse childhood experiences and physiological wear-and-tear in midlife: Findings from the 1958 British birth cohort. Proc Natl Acad Sci U S A 2015; 112:E738-46. [PMID: 25646470 DOI: 10.1073/pnas.1417325112] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Allostatic load (AL) is a measure of overall physiological wear-and-tear over the life course, which could partially be the consequence of early life exposures. AL could allow a better understanding of the potential biological pathways playing a role in the construction of the social gradient in adult health. To explore the biological embedding hypothesis, we examined whether adverse childhood experiences (ACEs) are associated with elevated AL in midlife. We used imputed data on 3,782 women and 3,753 men of the National Child Development Study in Britain followed up seven times. ACEs were measured using prospective data collected at ages 7, 11, and 16. AL was operationalized using data from the biomedical survey collected at age 44 on 14 parameters representing four biological systems. We examined the role of adult health behaviors, body mass index (BMI), and socioeconomic status as potential mediators using a path analysis. ACEs were associated with higher AL for both men and women after adjustment for early life factors and childhood pathologies. The path analysis showed that the association between ACEs and AL was largely explained by early adult factors at age 23 and 33. For men, the total mediated effect was 59% (for two or more ACEs) via health behaviors, education level, and wealth. For women, the mediated effect represented 76% (for two or more ACEs) via smoking, BMI, education level, and wealth. Our results indicate that early psychosocial stress has an indirect lasting impact on physiological wear-and-tear via health behaviors, BMI, and socioeconomic factors in adulthood.
Collapse
|
235
|
Roy S, Dhobale M, Dangat K, Mehendale S, Lalwani S, Joshi S. Differential oxidative stress levels in mothers with preeclampsia delivering male and female babies. J Matern Fetal Neonatal Med 2015; 28:1973-80. [DOI: 10.3109/14767058.2014.974537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
236
|
Tarrade A, Panchenko P, Junien C, Gabory A. Placental contribution to nutritional programming of health and diseases: epigenetics and sexual dimorphism. J Exp Biol 2015; 218:50-8. [DOI: 10.1242/jeb.110320] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The recent and rapid worldwide increase in non-communicable diseases challenges the assumption that genetic factors are the primary contributors to such diseases. A new concept of the ‘developmental origins of health and disease’ (DOHaD) is at stake and therefore requires a paradigm shift. Maternal obesity and malnutrition predispose offspring to develop metabolic syndrome, a vicious cycle leading to transmission to subsequent generation(s), with differences in response and susceptibility according to the sex of the individual. The placenta is a programming agent of adult health and disease. Adaptations of placental phenotype in response to maternal diet and metabolic status alter fetal nutrient supply. This implies important epigenetic changes that are, however, still poorly documented in DOHaD studies, particularly concerning overnutrition. The aim of this review is to discuss the emerging knowledge on the relationships between the effect of maternal nutrition or metabolic status on placental function and the risk of diseases later in life, with a specific focus on epigenetic mechanisms and sexual dimorphism. Explaining the sex-specific causal variables and how males versus females respond and adapt to environmental perturbations should help physicians and patients to anticipate disease susceptibility.
Collapse
Affiliation(s)
- Anne Tarrade
- INRA, UMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| | - Polina Panchenko
- INRA, UMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| | - Claudine Junien
- INRA, UMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
- UVSQ, Université Versailles-Saint-Quentin-en-Yvelines, France
| | - Anne Gabory
- INRA, UMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| |
Collapse
|
237
|
Gohir W, Ratcliffe EM, Sloboda DM. Of the bugs that shape us: maternal obesity, the gut microbiome, and long-term disease risk. Pediatr Res 2015; 77:196-204. [PMID: 25314580 DOI: 10.1038/pr.2014.169] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/15/2014] [Indexed: 01/06/2023]
Abstract
Chronic disease risk is inextricably linked to our early-life environment, where maternal, fetal, and childhood factors predict disease risk later in life. Currently, maternal obesity is a key predictor of childhood obesity and metabolic complications in adulthood. Although the mechanisms are unclear, new and emerging evidence points to our microbiome, where the bacterial composition of the gut modulates the weight gain and altered metabolism that drives obesity. Over the course of pregnancy, maternal bacterial load increases, and gut bacterial diversity changes and is influenced by pre-pregnancy- and pregnancy-related obesity. Alterations in the bacterial composition of the mother have been shown to affect the development and function of the gastrointestinal tract of her offspring. How these microbial shifts influence the maternal-fetal-infant relationship is a topic of hot debate. This paper will review the evidence linking nutrition, maternal obesity, the maternal gut microbiome, and fetal gut development, bringing together clinical observations in humans and experimental data from targeted animal models.
Collapse
Affiliation(s)
- Wajiha Gohir
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | | | - Deborah M Sloboda
- 1] Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada [2] Department of Pediatrics, McMaster University, Hamilton, Canada [3] Department of Obstetrics and Gynecology, McMaster University, Hamilton, Canada
| |
Collapse
|
238
|
Kurek M, Żądzińska E, Sitek A, Borowska-Strugińska B, Rosset I, Lorkiewicz W. Prenatal factors associated with the neonatal line thickness in human deciduous incisors. HOMO-JOURNAL OF COMPARATIVE HUMAN BIOLOGY 2014; 66:251-63. [PMID: 25618810 DOI: 10.1016/j.jchb.2014.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 11/30/2014] [Indexed: 01/10/2023]
Abstract
The neonatal line (NNL) is used to distinguish developmental events observed in enamel which occurred before and after birth. However, there are few studies reporting relationship between the characteristics of the NNL and factors affecting prenatal conditions. The aim of the study was to determine prenatal factors that may influence the NNL thickness in human deciduous teeth. The material consisted of longitudinal ground sections of 60 modern human deciduous incisors obtained from full-term healthy children with reported birth histories and prenatal factors. All teeth were sectioned in the labio-lingual plane using diamond blade (Buechler IsoMet 1000). Final specimens were observed using scanning electron microscopy at magnifications 320×. For each tooth, linear measurements of the NNL thickness were taken on its labial surface at the three levels from the cemento-enamel junction. The difference in the neonatal line thickness between tooth types and between males and females was statistically significant. A multiple regression analyses confirmed influence of two variables on the NNL thickness standardised on tooth type and the children's sex (z-score values). These variables are the taking of an antispasmodic medicine by the mother during pregnancy and the season of the child's birth. These two variables together explain nearly 17% of the variability of the NNL. Children of mothers taking a spasmolytic medicine during pregnancy were characterised by a thinner NNL compared with children whose mothers did not take such medication. Children born in summer and spring had a thinner NNL than children born in winter. These results indicate that the prenatal environment significantly contributes to the thickness of the NNL influencing the pace of reaching the post-delivery homeostasis by the newborn's organism.
Collapse
Affiliation(s)
- M Kurek
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237 Łódź, Poland.
| | - E Żądzińska
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237 Łódź, Poland
| | - A Sitek
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237 Łódź, Poland
| | - B Borowska-Strugińska
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237 Łódź, Poland
| | - I Rosset
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237 Łódź, Poland
| | - W Lorkiewicz
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237 Łódź, Poland
| |
Collapse
|
239
|
Franconi F, Rosano G, Campesi I. Need for gender-specific pre-analytical testing: the dark side of the moon in laboratory testing. Int J Cardiol 2014; 179:514-35. [PMID: 25465806 DOI: 10.1016/j.ijcard.2014.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 01/16/2023]
Abstract
Many international organisations encourage studies in a sex-gender perspective. However, research with a gender perspective presents a high degree of complexity, and the inclusion of sex-gender variable in experiments presents many methodological questions, the majority of which are still neglected. Overcoming these issues is fundamental to avoid erroneous results. Here, pre-analytical aspects of the research, such as study design, choice of utilised specimens, sample collection and processing, animal models of diseases, and the observer's role, are discussed. Artefacts in this stage of research could affect the predictive value of all analyses. Furthermore, the standardisation of research subjects according to their lifestyles and, if female, to their life phase and menses or oestrous cycle, is urgent to harmonise research worldwide. A sex-gender-specific attention to pre-analytical aspects could produce a decrease in the time for translation from the bench to bedside. Furthermore, sex-gender-specific pre-clinical pharmacological testing will enable adequate assessment of pharmacokinetic and pharmacodynamic actions of drugs and will enable, where appropriate, an adequate gender-specific clinical development plan. Therefore, sex-gender-specific pre-clinical research will increase the gender equity of care and will produce more evidence-based medicine.
Collapse
Affiliation(s)
- Flavia Franconi
- Department of Biomedical Sciences, University of Sassari, National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari, Italy; Vicepresident of Basilicata Region.
| | - Giuseppe Rosano
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, United Kingdom
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems, Osilo, Sassari, Italy
| |
Collapse
|
240
|
Davis EP, Pfaff D. Sexually dimorphic responses to early adversity: implications for affective problems and autism spectrum disorder. Psychoneuroendocrinology 2014; 49:11-25. [PMID: 25038479 PMCID: PMC4165713 DOI: 10.1016/j.psyneuen.2014.06.014] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 05/28/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
During gestation, development proceeds at a pace that is unmatched by any other stage of the life cycle. For these reasons the human fetus is particularly susceptible not only to organizing influences, but also to pathogenic disorganizing influences. Growing evidence suggests that exposure to prenatal adversity leads to neurological changes that underlie lifetime risks for mental illness. Beginning early in gestation, males and females show differential developmental trajectories and responses to stress. It is likely that sex-dependent organization of neural circuits during the fetal period influences differential vulnerability to mental health problems. We consider in this review evidence that sexually dimorphic responses to early life stress are linked to two developmental disorders: affective problems (greater female prevalence) and autism spectrum disorder (greater male prevalence). Recent prospective studies illustrating the neurodevelopmental consequences of fetal exposure to stress and stress hormones for males and females are considered here. Plausible biological mechanisms including the role of the sexually differentiated placenta are discussed.
Collapse
Affiliation(s)
- Elysia Poggi Davis
- Neurodevelopmental Research Program, Department of Psychology, University of Denver, Denver, CO, USA; Department of Psychiatry and Human Behavior, University of California Irvine, Orange, CA 92868, USA.
| | - Donald Pfaff
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY 10021, USA
| |
Collapse
|
241
|
Song HJ, Paek YJ, Choi MK, Lee HJ. Gender differences in the relationship between risk of hypertension and fruit intake. Prev Med 2014; 67:154-9. [PMID: 25045835 DOI: 10.1016/j.ypmed.2014.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/27/2014] [Accepted: 07/11/2014] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To investigate the relationship between hypertension and fruit intake in an Asian population. METHOD This study was based on the data from 2007, 2008 and 2009 Korea National Health and Nutrition Examination Survey. In the final analysis, a total of 9791 subjects (men=3819, women=5972) were included. Daily energy and nutrient intakes were assessed using 24-h recall. The odds ratios (ORs) for hypertension were assessed by using logistic regression and multivariable models. RESULTS A total of 10.6% of individuals were classified as having hypertension. Compared with the lowest quintile of fruit intake, the fifth quintile showed the lowest likelihood of hypertension (OR 0.73; 95% confidence interval [CI], 0.61-0.88) after adjusting for age and gender. For women, the likelihood of hypertension in the 2nd, 3rd, 4th and 5th quintiles of fruit intake decreased to 0.67 (95% CI, 0.34-1.30), 0.76 (0.56-1.05), 0.90 (0.67-1.22) and 0.54 (0.38-0.77), respectively, after adjusting for confounding factors (P value for trend=0.0011). An inverse association of fruit intake and hypertension was shown only in non-obese women. For men and obese women, there was no relationship between fruit intake and hypertension. CONCLUSION Dietary fruit recommendation for hypertension should be taken into account together with ethnic background, gender as well as the presence of obesity in individuals.
Collapse
Affiliation(s)
- Hong Ji Song
- Department of Family Medicine, Hallym University Sacred Heart Hospital, College of Medicine, Hallym University, Anyang-si, South Korea
| | - Yu Jin Paek
- Department of Family Medicine, Hallym University Sacred Heart Hospital, College of Medicine, Hallym University, Anyang-si, South Korea
| | - Min Kyu Choi
- Department of Family Medicine, Kangnam Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, South Korea
| | - Hae-Jeung Lee
- Department of Food & Nutrition, Eulji University, Seongnam-si, South Korea.
| |
Collapse
|
242
|
Paquette AG, Lester BM, Koestler DC, Lesseur C, Armstrong DA, Marsit CJ. Placental FKBP5 genetic and epigenetic variation is associated with infant neurobehavioral outcomes in the RICHS cohort. PLoS One 2014; 9:e104913. [PMID: 25115650 PMCID: PMC4130612 DOI: 10.1371/journal.pone.0104913] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/15/2014] [Indexed: 12/21/2022] Open
Abstract
Adverse maternal environments can lead to increased fetal exposure to maternal cortisol, which can cause infant neurobehavioral deficits. The placenta regulates fetal cortisol exposure and response, and placental DNA methylation can influence this function. FK506 binding protein (FKBP5) is a negative regulator of cortisol response, FKBP5 methylation has been linked to brain morphology and mental disorder risk, and genetic variation of FKBP5 was associated with post-traumatic stress disorder in adults. We hypothesized that placental FKBP5 methylation and genetic variation contribute to gene expression control, and are associated with infant neurodevelopmental outcomes assessed using the Neonatal Intensive Care Unit (NICU) Network Neurobehavioral Scales (NNNS). In 509 infants enrolled in the Rhode Island Child Health Study, placental FKBP5 methylation was measured at intron 7 using quantitative bisulfite pyrosequencing. Placental FKBP5 mRNA was measured in a subset of 61 infants by quantitative PCR, and the SNP rs1360780 was genotyped using a quantitative allelic discrimination assay. Relationships between methylation, expression and NNNS scores were examined using linear models adjusted for confounding variables, then logistic models were created to determine the influence of methylation on membership in high risk groups of infants. FKBP5 methylation was negatively associated with expression (P = 0.08, r = -0.22); infants with the TT genotype had higher expression than individuals with CC and CT genotypes (P = 0.06), and those with CC genotype displayed a negative relationship between methylation and expression (P = 0.06, r = -0.43). Infants in the highest quartile of FKBP5 methylation had increased risk of NNNS high arousal compared to infants in the lowest quartile (OR 2.22, CI 1.07-4.61). TT genotype infants had increased odds of high NNNS stress abstinence (OR 1.98, CI 0.92-4.26). Placental FKBP5 methylation reduces expression in a genotype specific fashion, and genetic variation supersedes this effect. These genetic and epigenetic differences in expression may alter the placenta's ability to modulate cortisol response and exposure, leading to altered neurobehavioral outcomes.
Collapse
Affiliation(s)
- Alison G. Paquette
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Barry M. Lester
- Department of Pediatrics, Center for the Study of Children at Risk, Women and Infants Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Devin C. Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Corina Lesseur
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - David A. Armstrong
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Carmen J. Marsit
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Department of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
243
|
Samsam M, Ahangari R, Naser SA. Pathophysiology of autism spectrum disorders: Revisiting gastrointestinal involvement and immune imbalance. World J Gastroenterol 2014; 20:9942-9951. [PMID: 25110424 PMCID: PMC4123375 DOI: 10.3748/wjg.v20.i29.9942] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 05/22/2014] [Accepted: 06/13/2014] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) comprise a group of neurodevelopmental abnormalities that begin in early childhood and are characterized by impairment of social communication and behavioral problems including restricted interests and repetitive behaviors. Several genes have been implicated in the pathogenesis of ASD, most of them are involved in neuronal synaptogenesis. A number of environmental factors and associated conditions such as gastrointestinal (GI) abnormalities and immune imbalance have been linked to the pathophysiology of ASD. According to the March 2012 report released by United States Centers for Disease Control and Prevention, the prevalence of ASD has sharply increased during the recent years and one out of 88 children suffers now from ASD symptoms. Although there is a strong genetic base for the disease, several associated factors could have a direct link to the pathogenesis of ASD or act as modifiers of the genes thus aggravating the initial problem. Many children suffering from ASD have GI problems such as abdominal pain, chronic diarrhea, constipation, vomiting, gastroesophageal reflux, and intestinal infections. A number of studies focusing on the intestinal mucosa, its permeability, abnormal gut development, leaky gut, and other GI problem raised many questions but studies were somehow inconclusive and an expert panel of American Academy of Pediatrics has strongly recommended further investigation in these areas. GI tract has a direct connection with the immune system and an imbalanced immune response is usually seen in ASD children. Maternal infection or autoimmune diseases have been suspected. Activation of the immune system during early development may have deleterious effect on various organs including the nervous system. In this review we revisited briefly the GI and immune system abnormalities and neuropeptide imbalance and their role in the pathophysiology of ASD and discussed some future research directions.
Collapse
|
244
|
King V, Norman JE, Seckl JR, Drake AJ. Post-weaning diet determines metabolic risk in mice exposed to overnutrition in early life. Reprod Biol Endocrinol 2014; 12:73. [PMID: 25082159 PMCID: PMC4120004 DOI: 10.1186/1477-7827-12-73] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/26/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Maternal overnutrition during pregnancy is associated with an increased risk of obesity and cardiometabolic disease in the offspring; a phenomenon attributed to 'developmental programming'. The post-weaning development of obesity may associate with exacerbation of the programmed metabolic phenotype. In mice, we have previously shown that exposure to maternal overnutrition causes increased weight gain in offspring before weaning, but exerts no persistent effects on weight or glucose tolerance in adulthood. In order to determine whether post-weaning exposure to a cafeteria diet might lead to an exacerbation of programmed effects, offspring born and raised by mothers on control (CON) or cafeteria (DIO) diets were transferred onto either CON or DIO diets at weaning. FINDINGS Post-weaning DIO caused the development of obesity, with hyperglycaemia and hyperinsulinaemia in males; and obesity with hyperinsulinaemia in females and with increased cholesterol levels in both sexes. Exposure to maternal overnutrition during pregnancy and lactation caused only subtle additional effects on offspring phenotype. CONCLUSIONS These results suggest that post-weaning exposure to a high-fat high-sugar diet has a more profound effect on offspring weight gain and glucose tolerance than exposure to maternal overnutrition. These data emphasise the importance of optimising early life nutrition in offspring of both obese and lean mothers.
Collapse
Affiliation(s)
- Vicky King
- MRC/University of Edinburgh Centre for Reproductive Health, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jane E Norman
- MRC/University of Edinburgh Centre for Reproductive Health, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jonathan R Seckl
- Endocrinology Unit, University/BHF Centre for Cardiovascular Science, University of Edinburgh, QMRI, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Amanda J Drake
- Endocrinology Unit, University/BHF Centre for Cardiovascular Science, University of Edinburgh, QMRI, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
245
|
Pembrey M, Saffery R, Bygren LO. Human transgenerational responses to early-life experience: potential impact on development, health and biomedical research. J Med Genet 2014; 51:563-72. [PMID: 25062846 PMCID: PMC4157403 DOI: 10.1136/jmedgenet-2014-102577] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mammalian experiments provide clear evidence of male line transgenerational effects on health and development from paternal or ancestral early-life exposures such as diet or stress. The few human observational studies to date suggest (male line) transgenerational effects exist that cannot easily be attributed to cultural and/or genetic inheritance. Here we summarise relevant studies, drawing attention to exposure sensitive periods in early life and sex differences in transmission and offspring outcomes. Thus, variation, or changes, in the parental/ancestral environment may influence phenotypic variation for better or worse in the next generation(s), and so contribute to common, non-communicable disease risk including sex differences. We argue that life-course epidemiology should be reframed to include exposures from previous generations, keeping an open mind as to the mechanisms that transmit this information to offspring. Finally, we discuss animal experiments, including the role of epigenetic inheritance and non-coding RNAs, in terms of what lessons can be learnt for designing and interpreting human studies. This review was developed initially as a position paper by the multidisciplinary Network in Epigenetic Epidemiology to encourage transgenerational research in human cohorts.
Collapse
Affiliation(s)
- Marcus Pembrey
- School of Social & Community Medicine, University of Bristol, Bristol, UK UCL Institute of Child Health, London, UK
| | - Richard Saffery
- Murdoch Childrens Research Institute, Parkville, Australia Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Lars Olov Bygren
- Department of Biosciences and Rehabilitation, Karolinska Institutet, Huddinge, Sweden Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
246
|
Kautzky-Willer A. Gendermedizin. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2014; 57:1022-30. [DOI: 10.1007/s00103-014-2011-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
247
|
Bronson SL, Bale TL. Prenatal stress-induced increases in placental inflammation and offspring hyperactivity are male-specific and ameliorated by maternal antiinflammatory treatment. Endocrinology 2014; 155:2635-46. [PMID: 24797632 PMCID: PMC4060181 DOI: 10.1210/en.2014-1040] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adverse experiences during gestation such as maternal stress and infection are known risk factors for neurodevelopmental disorders, including schizophrenia, autism, and attention deficit/hyperactivity disorder. The mechanisms by which these distinct exposures may confer similar psychiatric vulnerability remain unclear, although likely involve pathways common to both stress and immune responses at the maternal-fetal interface. We hypothesized that maternal stress-induced activation of immune pathways within the placenta, the sex-specific maternal-fetal intermediary, may contribute to prenatal stress programming effects on the offspring. Therefore, we assessed for markers indicative of stress-induced placental inflammation, and examined the ability of maternal nonsteroidal antiinflammatory drug (NSAID) treatment to ameliorate placental effects and thereby rescue the stress-dysregulation phenotype observed in our established mouse model of early prenatal stress (EPS). As expected, placental gene expression analyses revealed increased levels of immune response genes, including the proinflammatory cytokines IL-6 and IL-1β, specifically in male placentas. NSAID treatment partially ameliorated these EPS effects. Similarly, in adult offspring, males displayed stress-induced locomotor hyperactivity, a hallmark of dopaminergic dysregulation, which was ameliorated by maternal NSAID treatment. Fitting with these outcomes and supportive of dopamine pathway involvement, expression of dopamine D1 and D2 receptors was altered by EPS in males. These studies support an important interaction between maternal stress and a proinflammatory state in the long-term programming effects of maternal stress.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/immunology
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Blotting, Western
- Brain/drug effects
- Brain/immunology
- Brain/metabolism
- Female
- Gene Expression/drug effects
- Gene Expression/immunology
- Humans
- Interleukin-1beta/genetics
- Interleukin-1beta/immunology
- Interleukin-6/genetics
- Interleukin-6/immunology
- Male
- Maze Learning/drug effects
- Mice
- Mice, Inbred C57BL
- Motor Activity/drug effects
- Motor Activity/immunology
- Placenta/drug effects
- Placenta/immunology
- Placenta/metabolism
- Pregnancy
- Prenatal Exposure Delayed Effects/immunology
- Prenatal Exposure Delayed Effects/psychology
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/immunology
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Sex Factors
- Stress, Psychological/immunology
- Stress, Psychological/psychology
- Tyrosine 3-Monooxygenase/immunology
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Stefanie L Bronson
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104
| | | |
Collapse
|
248
|
Maternal pravastatin prevents altered fetal brain development in a preeclamptic CD-1 mouse model. PLoS One 2014; 9:e100873. [PMID: 24963809 PMCID: PMC4071009 DOI: 10.1371/journal.pone.0100873] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/30/2014] [Indexed: 12/14/2022] Open
Abstract
Objective Using an animal model, we have previously shown that preeclampsia results in long-term adverse neuromotor outcomes in the offspring, and this phenotype was prevented by antenatal treatment with pravastatin. This study aims to localize the altered neuromotor programming in this animal model and to evaluate the role of pravastatin in its prevention. Materials and Methods For the preeclampsia model, pregnant CD-1 mice were randomly allocated to injection of adenovirus carrying sFlt-1 or its control virus carrying mFc into the tail vein. Thereafter they received pravastatin (sFlt-1-pra “experimental group”) or water (sFlt-1 “positive control”) until weaning. The mFc group (“negative control”) received water. Offspring at 6 months of age were sacrificed, and whole brains underwent magnetic resonance imaging (MRI). MRIs were performed using an 11.7 Tesla vertical bore MRI scanner. T2 weighted images were acquired to evaluate the volumes of 28 regions of interest, including areas involved in adaptation and motor, spatial and sensory function. Cytochemistry and cell quantification was performed using neuron-specific Nissl stain. One-way ANOVA with multiple comparison testing was used for statistical analysis. Results Compared with control offspring, male sFlt-1 offspring have decreased volumes in the fimbria, periaquaductal gray, stria medullaris, and ventricles and increased volumes in the lateral globus pallidus and neocortex; however, female sFlt-1 offspring showed increased volumes in the ventricles, stria medullaris, and fasciculus retroflexus and decreased volumes in the inferior colliculus, thalamus, and lateral globus pallidus. Neuronal quantification via Nissl staining exhibited decreased cell counts in sFlt-1 offspring neocortex, more pronounced in males. Prenatal pravastatin treatment prevented these changes. Conclusion Preeclampsia alters brain development in sex-specific patterns, and prenatal pravastatin therapy prevents altered neuroanatomic programming in this animal model.
Collapse
|
249
|
Feuer SK, Liu X, Donjacour A, Lin W, Simbulan RK, Giritharan G, Piane LD, Kolahi K, Ameri K, Maltepe E, Rinaudo PF. Use of a mouse in vitro fertilization model to understand the developmental origins of health and disease hypothesis. Endocrinology 2014; 155:1956-69. [PMID: 24684304 PMCID: PMC3990843 DOI: 10.1210/en.2013-2081] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The Developmental Origins of Health and Disease hypothesis holds that alterations to homeostasis during critical periods of development can predispose individuals to adult-onset chronic diseases such as diabetes and metabolic syndrome. It remains controversial whether preimplantation embryo manipulation, clinically used to treat patients with infertility, disturbs homeostasis and affects long-term growth and metabolism. To address this controversy, we have assessed the effects of in vitro fertilization (IVF) on postnatal physiology in mice. We demonstrate that IVF and embryo culture, even under conditions considered optimal for mouse embryo culture, alter postnatal growth trajectory, fat accumulation, and glucose metabolism in adult mice. Unbiased metabolic profiling in serum and microarray analysis of pancreatic islets and insulin sensitive tissues (liver, skeletal muscle, and adipose tissue) revealed broad changes in metabolic homeostasis, characterized by systemic oxidative stress and mitochondrial dysfunction. Adopting a candidate approach, we identify thioredoxin-interacting protein (TXNIP), a key molecule involved in integrating cellular nutritional and oxidative states with metabolic response, as a marker for preimplantation stress and demonstrate tissue-specific epigenetic and transcriptional TXNIP misregulation in selected adult tissues. Importantly, dysregulation of TXNIP expression is associated with enrichment for H4 acetylation at the Txnip promoter that persists from the blastocyst stage through adulthood in adipose tissue. Our data support the vulnerability of preimplantation embryos to environmental disturbance and demonstrate that conception by IVF can reprogram metabolic homeostasis through metabolic, transcriptional, and epigenetic mechanisms with lasting effects for adult growth and fitness. This study has wide clinical relevance and underscores the importance of continued follow-up of IVF-conceived offspring.
Collapse
Affiliation(s)
- Sky K Feuer
- Department of Obstetrics, Gynecology and Reproductive Sciences (S.K.F., X.L., A.D., W.L., R.K.S., G.G., L.D.P., K.K., P.F.R.), and Department of Pediatrics (K.A., E.M.), University of California San Francisco, San Francisco, California 94143; Nevada Center for Reproductive Medicine (G.G.), Reno, Nevada 89511; Obstetric and Gynecology Department (L.D.P.), University of Turin, Turin, Italy; and Oregon Health & Science University (K.K.), Portland, Oregon 97239
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Lesseur C, Paquette AG, Marsit CJ. Epigenetic Regulation of Infant Neurobehavioral Outcomes. MEDICAL EPIGENETICS 2014; 2:71-79. [PMID: 25089125 PMCID: PMC4116357 DOI: 10.1159/000361026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During fetal development and early-infancy, environmental signals can induce epigenetic changes that alter neurobehavioral development and later-life mental health. Several neurodevelopmental genetic diseases influence epigenetic regulatory genes and genomic imprinting. Recently, brain epigenetic marks have been involved in idiopathic neurodevelopmental disorders including autism spectrum disorders (ASD). The placenta is an important regulator of the intrauterine environment that links maternal and fetal nervous systems. Placental epigenetic signatures have been associated with neurodevelopment of healthy newborns quantified through the NICU Network Neurobehavioral Scales (NNNS). Associations have been observed for DNA methylation of genes involved in cortisol (NR3C1, HSD11B), serotonin (HTR2A), and metabolic (LEP) pathways. Dysregulation of imprinted genes and microRNAs has also been associated with neurobehavior assessed by NNNS. Further analysis is needed to characterize the mechanisms by which the epigenome influences neurodevelopment, and the connection between this dysregulation and mental health disorders. In the future, epigenetic marks could serve as functional biomarkers of mental health and cognitive function.
Collapse
Affiliation(s)
- Corina Lesseur
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA
| | - Alison G. Paquette
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA
| | - Carmen J. Marsit
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA
- Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, 1 Medical Center Drive, 7927 Rubin Building, Lebanon, NH 03756, USA
| |
Collapse
|