1
|
Uldbjerg CS, Leader J, Minguez-Alarcon L, Chagnon O, Dadd R, Ford J, Fleury E, Williams P, Juul A, Bellinger DC, Calafat AM, Hauser R, Braun JM. Associations of maternal and paternal preconception and maternal pregnancy urinary phthalate biomarker and bisphenol A concentrations with offspring autistic behaviors: The PEACE study. ENVIRONMENTAL RESEARCH 2024; 263:120253. [PMID: 39486680 DOI: 10.1016/j.envres.2024.120253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Environmental chemical exposures in utero may play a role in autism development. While preconception risk factors for autism are increasingly being investigated, little is known about the influence of chemical exposures during the preconception period, particularly for paternal exposures. METHODS In 195 children from the Preconception Environmental exposures And Childhood health Effects (PEACE) cohort born to parents recruited from a fertility clinic in Boston, Massachusetts between 2004 and 2017, we quantified concentrations of 11 phthalate metabolites and bisphenol A (BPA) in urine samples collected from mothers and fathers before conception and mothers throughout pregnancy. When children were 6-15 years old, parents completed the Social Responsiveness Scale (SRS) questionnaire assessing autistic behaviors. We used linear mixed effect models to estimate covariate-adjusted associations of phthalate biomarker and BPA concentrations, separately for maternal preconception (n = 179), paternal preconception (n = 121), and maternal pregnancy (n = 177), with SRS T-scores, based on age and gender, in offspring. We used quantile g-computation models for mixture analyses and evaluated modification by selected dietary factors. RESULTS The mean SRS T-score was 47.7 (±7.4), lower than the normative mean of 50. In adjusted models for individual biomarkers or mixtures, few associations were observed and estimates were generally negative (e.g., lower SRS T-scores) and imprecise. We observed associations of higher mono-isobutyl phthalate (MiBP) concentrations measured in maternal preconception and paternal preconception periods with lower SRS T-scores (βmaternal_precon = -1.6, 95% CI -2.7; -0.4; βpaternal_precon = -2.9, 95% CI -4.6; -1.2) for each loge increase. In a subset of participants with maternal preconception nutrition information, we generally observed stronger inverse associations with higher folate and iron intake, particularly for folate intake and MiBP concentrations. CONCLUSIONS Urinary phthalate biomarker and BPA concentrations during preconception (maternal and paternal) and pregnancy (maternal) were not associated with adverse autistic behaviors in these children. Larger studies are needed to elucidate the observed associations, while considering interactions between maternal nutrition and chemical exposures.
Collapse
Affiliation(s)
- Cecilie Skaarup Uldbjerg
- Department of Growth and Reproduction, Copenhagen University Hospitalet - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMARC), Copenhagen University Hospitalet - Rigshospitalet, Copenhagen, Denmark
| | - Jordana Leader
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lidia Minguez-Alarcon
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Harvard Medical School & Brigham and Women's Hospital, Boston, MA, USA
| | - Olivia Chagnon
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ramace Dadd
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jennifer Ford
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Elvira Fleury
- Department of Epidemiology, Brown University, Providence, RI, USA
| | - Paige Williams
- Departments of Biostatistics and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospitalet - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMARC), Copenhagen University Hospitalet - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David C Bellinger
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Cardiac Neurodevelopment Program, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Psychology, Harvard Medical School, Boston, MA, USA
| | - Antonia M Calafat
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Russ Hauser
- Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, USA.
| |
Collapse
|
2
|
Yano N, Fedulov AV. Targeted DNA Demethylation: Vectors, Effectors and Perspectives. Biomedicines 2023; 11:biomedicines11051334. [PMID: 37239005 DOI: 10.3390/biomedicines11051334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Aberrant DNA hypermethylation at regulatory cis-elements of particular genes is seen in a plethora of pathological conditions including cardiovascular, neurological, immunological, gastrointestinal and renal diseases, as well as in cancer, diabetes and others. Thus, approaches for experimental and therapeutic DNA demethylation have a great potential to demonstrate mechanistic importance, and even causality of epigenetic alterations, and may open novel avenues to epigenetic cures. However, existing methods based on DNA methyltransferase inhibitors that elicit genome-wide demethylation are not suitable for treatment of diseases with specific epimutations and provide a limited experimental value. Therefore, gene-specific epigenetic editing is a critical approach for epigenetic re-activation of silenced genes. Site-specific demethylation can be achieved by utilizing sequence-dependent DNA-binding molecules such as zinc finger protein array (ZFA), transcription activator-like effector (TALE) and clustered regularly interspaced short palindromic repeat-associated dead Cas9 (CRISPR/dCas9). Synthetic proteins, where these DNA-binding domains are fused with the DNA demethylases such as ten-eleven translocation (Tet) and thymine DNA glycosylase (TDG) enzymes, successfully induced or enhanced transcriptional responsiveness at targeted loci. However, a number of challenges, including the dependence on transgenesis for delivery of the fusion constructs, remain issues to be solved. In this review, we detail current and potential approaches to gene-specific DNA demethylation as a novel epigenetic editing-based therapeutic strategy.
Collapse
Affiliation(s)
- Naohiro Yano
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Alexey V Fedulov
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| |
Collapse
|
3
|
Jiang T, Xiao H, Li B, He H, Wang H, Chen L. LOX overexpression programming mediates the osteoclast mechanism of low peak bone mass in female offspring rats caused by pregnant dexamethasone exposure. Cell Commun Signal 2023; 21:84. [PMID: 37095518 PMCID: PMC10124047 DOI: 10.1186/s12964-023-01115-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/25/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Osteoporosis is a degenerative disease characterized by reduced bone mass, with low peak bone mass being the predominant manifestation during development and having an intrauterine origin. Pregnant women at risk of preterm delivery are commonly treated with dexamethasone to promote fetal lung development. However, pregnant dexamethasone exposure (PDE) can lead to reduced peak bone mass and susceptibility to osteoporosis in offspring. In this study, we aimed to investigate the mechanism of PDE-induced low peak bone mass in female offspring from the perspective of altered osteoclast developmental programming. METHODS 0.2 mg/kg.d dexamethasone was injected subcutaneously into rats on gestation days (GDs) 9-20. Some pregnant rats were killed at GD20 to remove fetal rat long bones, the rest were delivered naturally, and some adult offspring rats were given ice water swimming stimulation for two weeks. RESULTS The results showed that the fetal rat osteoclast development was inhibited in the PDE group compared with the control group. In contrast, the adult rat osteoclast function was hyperactivation with reduced peak bone mass. We further found that the promoter region methylation levels of lysyl oxidase (LOX) were decreased, the expression was increased, and the production of reactive oxygen species (ROS) was raised in PDE offspring rat long bone before and after birth. Combined in vivo and in vitro experiments, we confirmed that intrauterine dexamethasone promoted the expression and binding of the glucocorticoid receptor (GR) and estrogen receptor β (ERβ) in osteoclasts and mediated the decrease of LOX methylation level and increase of expression through upregulation of 10-11 translocator protein 3 (Tet3). CONCLUSIONS Taken together, we confirm that dexamethasone causes osteoclast LOX hypomethylation and high expression through the GR/ERβ/Tet3 pathway, leading to elevated ROS production and that this intrauterine epigenetic programming effect can be carried over to postnatal mediating hyperactivation in osteoclast and reduced peak bone mass in adult offspring. This study provides an experimental basis for elucidating the mechanism of osteoclast-mediated intrauterine programming of low peak bone mass in female offspring of PDE and for exploring its early targets for prevention and treatment. Video Abstract.
Collapse
Affiliation(s)
- Tao Jiang
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao Xiao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hangyuan He
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
4
|
Sánchez-Trujillo L, García-Montero C, Fraile-Martinez O, Guijarro LG, Bravo C, De Leon-Luis JA, Saez JV, Bujan J, Alvarez-Mon M, García-Honduvilla N, Saez MA, Ortega MA. Considering the Effects and Maternofoetal Implications of Vascular Disorders and the Umbilical Cord. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1754. [PMID: 36556956 PMCID: PMC9782481 DOI: 10.3390/medicina58121754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
The umbilical cord is a critical anatomical structure connecting the placenta with the foetus, fulfilling multiple functions during pregnancy and hence influencing foetal development, programming and survival. Histologically, the umbilical cord is composed of three blood vessels: two arteries and one vein, integrated in a mucous connective tissue (Wharton's jelly) upholstered by a layer of amniotic coating. Vascular alterations in the umbilical cord or damage in this tissue because of other vascular disorders during pregnancy are worryingly related with detrimental maternofoetal consequences. In the present work, we will describe the main vascular alterations presented in the umbilical cord, both in the arteries (Single umbilical artery, hypoplastic umbilical artery or aneurysms in umbilical arteries) and the vein (Vascular thrombosis, aneurysms or varicose veins in the umbilical vein), together with other possible complications (Velamentous insertion, vasa praevia, hypercoiled or hypocoiled cord, angiomyxoma and haematomas). Likewise, the effect of the main obstetric vascular disorders like hypertensive disorders of pregnancy (specially pre-eclampsia) and chronic venous disease on the umbilical cord will also be summarized herein.
Collapse
Affiliation(s)
- Lara Sánchez-Trujillo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Deparment of Pediatrics, Hospital Universitario Principe de Asturias, 28801 Alcalá de Henares, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Systems Biology, Faculty of Medicine and Health Sciences (Networking Research Center on for Liver and Digestive Diseases (CIBEREHD)), University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Juan A. De Leon-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Jose V. Saez
- Department of Biomedicine and Biotechnology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Príncipe de Asturias, CIBEREHD, 28806 Alcalá de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801 Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| |
Collapse
|
5
|
Feng F, Huang L, Zhou G, Wang J, Zhang R, Li Z, Zhang Y, Ba Y. GPR61 methylation in cord blood: a potential target of prenatal exposure to air pollutants. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:463-472. [PMID: 32478566 DOI: 10.1080/09603123.2020.1773414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/19/2020] [Indexed: 06/11/2023]
Abstract
To explore the impact of air pollutants exposure during pregnancy on infant DNA methylation, we identified correlated methylated genes in maternal and cord blood samples using the Illumina Human Methylation 27 k BeadChip. Quantitative methylation-specific PCR (QMS-PCR) was performed to validate the target gene methylation pattern in 568 participants. Then the association between air pollutants exposure and DNA methylation level in the target gene was investigated. The GPR61 gene with a higher methylation level both in mothers and newborns was identified as the target gene, and we found a positive mother-infant DNA methylation correlation in the promoter region of GPR61. Air pollutants exposure during entire pregnancy was associated with maternal and infant GPR61 DNA methylation. After adjusting confounding variables, maternal air pollutants exposure was still associated with infant GPR61 DNA methylation. In summary, GPR61 methylation in cord blood may be a potential target of prenatal exposure to air pollutants.
Collapse
Affiliation(s)
- Feifei Feng
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Li Huang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Guoyu Zhou
- Department of Environmental Health, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Jia Wang
- Department of Environmental Health, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Ruiqin Zhang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China
| | - Zhiyuan Li
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Yawei Zhang
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Environmental Health Science, Yale School of Public Health, New Haven, CT, USA
| | - Yue Ba
- Department of Environmental Health, Zhengzhou University School of Public Health, Zhengzhou, China
| |
Collapse
|
6
|
DNA Methylation Levels of the TBX5 Gene Promoter Are Associated with Congenital Septal Defects in Mexican Paediatric Patients. BIOLOGY 2022; 11:biology11010096. [PMID: 35053095 PMCID: PMC8773106 DOI: 10.3390/biology11010096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022]
Abstract
The TBX5 gene regulates morphological changes during heart development, and it has been associated with epigenetic abnormalities observed in congenital heart defects (CHD). The aim of this research was to evaluate the association between DNA methylation levels of the TBX5 gene promoter and congenital septal defects. DNA methylation levels of six CpG sites in the TBX5 gene promoter were evaluated using pyrosequencing analysis in 35 patients with congenital septal defects and 48 controls. Average methylation levels were higher in individuals with congenital septal defects than in the controls (p < 0.004). In five CpG sites, we also found higher methylation levels in patients than in the controls (p < 0.05). High methylation levels were associated with congenital septal defects (OR = 3.91; 95% CI = 1.02–14.8; p = 0.045). The analysis of Receiver Operating Characteristic (ROC) showed that the methylation levels of the TBX5 gene could be used as a risk marker for congenital septal defects (AUC = 0.68, 95% CI = 0.56–0.80; p = 0.004). Finally, an analysis of environmental factors indicated that maternal infections increased the risk (OR = 2.90; 95% CI = 1.01–8.33; p = 0.048) of congenital septal defects. Our data suggest that a high DNA methylation of the TBX5 gene could be associated with congenital septal defects.
Collapse
|
7
|
Airborne fine particulate matter induces cognitive and emotional disorders in offspring mice exposed during pregnancy. Sci Bull (Beijing) 2021; 66:578-591. [PMID: 36654428 DOI: 10.1016/j.scib.2020.08.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/04/2020] [Accepted: 08/21/2020] [Indexed: 01/20/2023]
Abstract
Gestational exposure to PM2.5 is associated with adverse postnatal outcomes. PM2.5 can enter alveoli by using intratracheal instillation, even penetrate through lung cells into the blood circulation. Subsequently, they are transferred across the placenta and fetal blood brain barrier, causing the adverse birth outcomes of offspring. This study demonstrated that the gestational exposure resulted in cognitive and emotional disorders in female offspring although the offspring were not exposed to PM2.5. Placental metabolic pathways modulated fetal brain development and played a pivotal role for maternal-placental-fetal interactions in the fetal programming of adult behavioral and mental disorders. Samples of fetus, offspring hippocampus and placenta from the mice exposed to PM2.5 were investigated using a comprehensive approach including mass spectrometry-based lipidomics and three-dimensional imaging. The exposure induced the neuro-degeneration in hippocampus, impairment of placental cytoarchitecture, and reprogramming of lipidome, which might affect the modulation of maternal-fetal cross-talk and result in the behavior disorders of offspring. The variation of spatial distribution of lipids was profoundly affected in dorsal pallium and hippocampal formation regions of fetal brain, offspring hippocampus, as well as labyrinth and junctional zones of placenta. The abundance alteration of lipid markers associated with neurodegenerative diseases was validated in transgenic mouse model with Alzheimer's disease and human cerebrospinal fluid from patients with Parkinson's disease. The finding could help with the selection of more suitable heterogeneous-related substructures targeting PM2.5 exposure and the exploration of PM2.5-induced toxicological effects on neurodegenerative diseases.
Collapse
|
8
|
Intrauterine growth restriction: Clinical consequences on health and disease at adulthood. Reprod Toxicol 2021; 99:168-176. [DOI: 10.1016/j.reprotox.2020.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
|
9
|
Offspring susceptibility to metabolic alterations due to maternal high-fat diet and the impact of inhaled ozone used as a stressor. Sci Rep 2020; 10:16353. [PMID: 33004997 PMCID: PMC7530537 DOI: 10.1038/s41598-020-73361-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
The influence of maternal high-fat diet (HFD) on metabolic response to ozone was examined in Long-Evans rat offspring. F0 females were fed control diet (CD; 10%kcal from fat) or HFD (60%kcal from fat) starting at post-natal day (PND) 30. Rats were bred on PND 72. Dietary regimen was maintained until PND 30 when all offspring were switched to CD. On PND 40, F1 offspring (n = 10/group/sex) were exposed to air or 0.8 ppm ozone for 5 h. Serum samples were collected for global metabolomic analysis (n = 8/group/sex). Offspring from HFD dams had increased body fat and weight relative to CD. Metabolomic analysis revealed significant sex-, diet-, and exposure-related changes. Maternal HFD increased free fatty acids and decreased phospholipids (male > female) in air-exposed rats. Microbiome-associated histidine and tyrosine metabolites were increased in both sexes, while 1,5-anhydroglucitol levels decreased in males indicating susceptibility to insulin resistance. Ozone decreased monohydroxy fatty acids and acyl carnitines and increased pyruvate along with TCA cycle intermediates in females (HFD > CD). Ozone increased various amino acids, polyamines, and metabolites of gut microbiota in HFD female offspring indicating gut microbiome alterations. Collectively, these data suggest that maternal HFD increases offspring susceptibility to metabolic alterations in a sex-specific manner when challenged with environmental stressors.
Collapse
|
10
|
Huang QQ, Tang HHF, Teo SM, Mok D, Ritchie SC, Nath AP, Brozynska M, Salim A, Bakshi A, Holt BJ, Khor CC, Sly PD, Holt PG, Holt KE, Inouye M. Neonatal genetics of gene expression reveal potential origins of autoimmune and allergic disease risk. Nat Commun 2020; 11:3761. [PMID: 32724101 PMCID: PMC7387553 DOI: 10.1038/s41467-020-17477-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic immune-mediated diseases of adulthood often originate in early childhood. To investigate genetic associations between neonatal immunity and disease, we map expression quantitative trait loci (eQTLs) in resting myeloid cells and CD4+ T cells from cord blood samples, as well as in response to lipopolysaccharide (LPS) or phytohemagglutinin (PHA) stimulation, respectively. Cis-eQTLs are largely specific to cell type or stimulation, and 31% and 52% of genes with cis-eQTLs have response eQTLs (reQTLs) in myeloid cells and T cells, respectively. We identified cis regulatory factors acting as mediators of trans effects. There is extensive colocalisation between condition-specific neonatal cis-eQTLs and variants associated with immune-mediated diseases, in particular CTSH had widespread colocalisation across diseases. Mendelian randomisation shows causal neonatal gene expression effects on disease risk for BTN3A2, HLA-C and others. Our study elucidates the genetics of gene expression in neonatal immune cells, and aetiological origins of autoimmune and allergic diseases. Some immune-mediated diseases may originate in early childhood. The authors mapped eQTLs and response eQTLs to various stimuli in neonatal myeloid cells and T cells, and revealed their potential role in immune-mediated diseases using colocalisation and Mendelian randomisation.
Collapse
Affiliation(s)
- Qin Qin Huang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia. .,Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK.
| | - Howard H F Tang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,School of BioSciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Shu Mei Teo
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Danny Mok
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.,British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Artika P Nath
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Marta Brozynska
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, 3010, Australia.,Melbourne School of Population and Global Health, Carlton, VIC, 3053, Australia
| | - Andrew Bakshi
- Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Barbara J Holt
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia
| | - Chiea Chuen Khor
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Peter D Sly
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Kathryn E Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,The London School of Hygiene and Tropical Medicine, London, WC1E 7TH, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia. .,Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK. .,British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK. .,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK. .,The Alan Turing Institute, London, UK. .,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK. .,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Csaba G. Reprogramming of the Immune System by Stress and Faulty Hormonal Imprinting. Clin Ther 2020; 42:983-992. [PMID: 32307123 DOI: 10.1016/j.clinthera.2020.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Hormonal imprinting is taking place perinatally at the first encounter between the developing hormone receptors and their target hormones. However, in this crucial period when the developmental window for physiological imprinting is open, other molecules, such as synthetic hormones and endocrine disruptors can bind to the receptors, leading to faulty imprinting with life-long consequences, especially to the immune system. This review presents the factors of stress and faulty hormonal imprinting that lead to reprogramming of the immune system. METHODS Relevant publications from Pubmed since 1990 were reviewed and synthesized. FINDINGS The developing immune system is rather sensitive to hormonal effects. Faulty hormonal imprinting is able to reprogram the original developmental program present in a given cell, with lifelong consequences, manifested in alteration of hormone binding by receptors, susceptibility to certain (non-infectious) diseases, and triggering of other diseases. As stress mobilizes the hypothalamic-pituitary-adrenal axis if it occurred during gestation or perinatally, it could lead to faulty hormonal imprinting in the immune system, manifested later as allergic and autoimmune diseases or weakness of normal immune defenses. Hormonal imprinting is an epigenetic process and is carried to the offspring without alteration of DNA base sequences. This means that any form of early-life stress alone or in association with hormonal imprinting could be associated with the developmental origin of health and disease (DOHaD). As puberty is also a period of reprogramming, stress or faulty imprinting can change the original (developmental) program, also with life-long consequences. IMPLICATIONS Considering the continuous differentiation of immune cells (from blast-cells) during the whole life, there is a possibility of late-imprinting or stress-activated reprogramming in the immune system at any periods of life, with later pathogenetic consequences.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell, and Immunobiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
12
|
Rogers JM. Smoking and pregnancy: Epigenetics and developmental origins of the metabolic syndrome. Birth Defects Res 2019; 111:1259-1269. [PMID: 31313499 PMCID: PMC6964018 DOI: 10.1002/bdr2.1550] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/19/2019] [Accepted: 06/23/2019] [Indexed: 12/11/2022]
Abstract
Maternal smoking causes lower birth weight, birth defects, and other adverse pregnancy outcomes. Epidemiological evidence over the past four decades has grown stronger and the adverse outcomes attributed to maternal smoking and secondhand smoke exposure have expanded. This review presents findings of latent and persistent metabolic effects in offspring of smoking mothers like those observed in studies of maternal undernutrition during pregnancy. The phenotype of offspring of smoking mothers is like that associated with maternal undernutrition. Born smaller than offspring of nonsmokers, these children have increased risk of being overweight or obese later. Plausible mechanisms include in utero hypoxia, nicotine-induced reductions in uteroplacental blood flow, placental toxicity, or toxic growth restriction from the many toxicants in tobacco smoke. Studies have reported increased risk of insulin resistance, type 2 diabetes and hypertension although the evidence here is weaker than for overweight/obesity. Altered DNA methylation has been consistently documented in smoking mothers' offspring, and these epigenetic alterations are extensive and postnatally durable. A causal link between altered DNA methylation and the phenotypic changes observed in offspring remains to be firmly established, yet the association is strong, and mediation analyses suggest a causal link. Studies examining expression patterns of affected genes during childhood development and associated health outcomes should be instructive in this regard. The adverse effects of exposure to tobacco smoke during pregnancy now clearly include permanent metabolic derangements in offspring that can adversely affect life-long health.
Collapse
Affiliation(s)
- John M Rogers
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina
| |
Collapse
|
13
|
Sharp GC, Lawlor DA. Paternal impact on the life course development of obesity and type 2 diabetes in the offspring. Diabetologia 2019; 62:1802-1810. [PMID: 31451867 PMCID: PMC6731203 DOI: 10.1007/s00125-019-4919-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022]
Abstract
The aetiologies of obesity and type 2 diabetes are incredibly complex, but the potential role of paternal influences remains relatively understudied. A better understanding of paternal influences on offspring risk of obesity and type 2 diabetes could have profound implications for public health, clinical practice and society. In this review, we outline potential biological and social mechanisms through which fathers might exert an impact on the health of their offspring. We also present a systematically compiled overview of the current evidence linking paternal factors to offspring development of obesity and type 2 diabetes throughout the life course. Although evidence is accumulating to support paternal associations with offspring outcomes, more high-quality research is needed to overcome specific methodological challenges and provide stronger causal evidence.
Collapse
Affiliation(s)
- Gemma C Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
- Bristol Dental School, University of Bristol, Bristol, UK.
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and the University of Bristol, Bristol, UK
| |
Collapse
|
14
|
The influence of low dose Bisphenol A on whole genome DNA methylation and chromatin compaction in different human cell lines. Toxicol In Vitro 2019; 58:26-34. [PMID: 30876887 DOI: 10.1016/j.tiv.2019.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 02/23/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022]
|
15
|
Tian FY, Marsit CJ. Environmentally Induced Epigenetic Plasticity in Development: Epigenetic Toxicity and Epigenetic Adaptation. CURR EPIDEMIOL REP 2018; 5:450-460. [PMID: 30984515 PMCID: PMC6456900 DOI: 10.1007/s40471-018-0175-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Epigenetic processes represent important mechanisms underlying developmental plasticity in response to environmental exposures. The current review discusses three classes of environmentally-induced epigenetic changes reflecting two aspects of that plasticity, toxicity effects as well as adaptation in the process of development. RECENT FINDINGS Due to innate resilience, epigenetic changes caused by environmental exposures may not always lead impairments but may allow the organisms to achieve positive developmental outcomes through appropriate adaptation and a buffering response. Thus, some epigenetic adaptive responses to an immediate stimulus or exposure early in life would be expected to have a survival advantage but these same responses may also result in adverse developmental outcomes as they persists into later life stage. Although accumulating literature has identified environmentally induced epigenetic changes and linked them to health outcomes, we currently face challenges in the interpretation of the functional impact of their epigenetic plasticity. SUMMARY Current environmental epigenetic research suggest that epigenetic processes may serve as a mechanism for resilience, and that they can be considered in terms of their impact on toxicity as a negative outcome, but also on adaptation for improved survival or health. This review encourages epigenetic environmental studies to move deeper inside into the functional meaning of epigenetic plasticity in the development.
Collapse
Affiliation(s)
- Fu-Ying Tian
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Carmen J. Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
16
|
Li Y, Deng Y, Deng C, Xie L, Yu L, Liu L, Yuan Y, Liu H, Dai L. Association of long interspersed nucleotide element-1 and interferon regulatory factor 6 methylation changes with nonsyndromic cleft lip with or without cleft palate. Oral Dis 2018; 25:215-222. [PMID: 30153397 DOI: 10.1111/odi.12965] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/09/2018] [Accepted: 07/04/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To examine the possible associations between methylation changes in the promoter regions of long interspersed nucleotide element-1 (LINE-1) and interferon regulatory factor 6 gene (IRF6) and nonsyndromic cleft lip with or without cleft palate (NSCL/P). METHODS A case-control investigation was performed to compare 37 infants affected by NSCL/Ps with 60 babies without cleft malformations. Their genomic DNA samples were obtained, and the LINE-1 and IRF6 methylation levels were measured by using Sequenom MassArray. Unconditional logistic regression was adopted to estimate the odds ratio. RESULTS Infants with NSCL/Ps had a higher methylation level at LINE-1 and IRF6 promoter regions than controls. High levels of LINE-1 (≥64.07%) and IRF6 (≥6.46%) methylation were associated with an increased risk of NSCL/P (LINE-1, OR = 2.63, 95% CI: 1.07-6.57; IRF6, OR = 4.73, 95% CI: 2.10-13.07), and the associations remained to be significant after adjusting for potential confounders. Similar associations were also found for cleft lip only, cleft lip, and palate. CONCLUSION Our study suggested that aberrant methylation of LINE-1 and IRF6 might contribute to the development of NSCL/Ps. Further studies are needed to replicate the findings.
Collapse
Affiliation(s)
- Yanhua Li
- Obstetric and Gynecologic Department, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ying Deng
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Changfei Deng
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liang Xie
- Key Laboratory of Obstetrics & Gynecology and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Yu
- Key Laboratory of Obstetrics & Gynecology and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.,Pediatric Department, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lijun Liu
- Key Laboratory of Obstetrics & Gynecology and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.,Pediatric Department, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yumei Yuan
- Hengyang Women and Children Hospital, Hengyang, China
| | - Hanmin Liu
- Key Laboratory of Obstetrics & Gynecology and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.,Pediatric Department, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Dai
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetrics & Gynecology and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Erickson AC, Sbihi H. Biological embedding, the air we breathe, and carcinogenesis. Lancet Planet Health 2018; 2:e149-e150. [PMID: 29615215 DOI: 10.1016/s2542-5196(18)30053-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 06/08/2023]
Affiliation(s)
- Anders C Erickson
- School of Population and Public Health, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Hind Sbihi
- BC Children's Hospital Research, Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Buck Louis GM, Smarr MM, Patel CJ. The Exposome Research Paradigm: an Opportunity to Understand the Environmental Basis for Human Health and Disease. Curr Environ Health Rep 2018; 4:89-98. [PMID: 28194614 DOI: 10.1007/s40572-017-0126-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW This paper presents an overview of the exposome research paradigm with particular application to understanding human reproduction and development and its implications for health across a lifespan. RECENT FINDINGS The exposome research paradigm has generated considerable discussion about its feasibility and utility for delineating the impact of environmental exposures on human health. Early initiatives are underway, including smaller proof-of-principle studies and larger concerted efforts. Despite the notable challenges underlying the exposome paradigm, analytic techniques are being developed to handle its untargeted approach and correlated and multi-level or hierarchical data structures such initiatives generate, while considering multiple comparisons. The relatively short intervals for critical and sensitive windows of human reproduction and development seem well suited for exposome research and may revolutionize our understanding of later onset diseases. Early initiatives suggest that the exposome paradigm is feasible, but its utility remains to be established with applications to population human health research.
Collapse
Affiliation(s)
- Germaine M Buck Louis
- Office of the Director, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Drive, Room 3148, Rockville, MD, 20852, USA.
| | - Melissa M Smarr
- Office of the Director, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710B Rockledge Drive, Room 3148, Rockville, MD, 20852, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St., Boston, MA, 02115, USA
| |
Collapse
|
19
|
Lin X, Teh AL, Chen L, Lim IY, Tan PF, MacIsaac JL, Morin AM, Yap F, Tan KH, Saw SM, Lee YS, Holbrook JD, Godfrey KM, Meaney MJ, Kobor MS, Chong YS, Gluckman PD, Karnani N. Choice of surrogate tissue influences neonatal EWAS findings. BMC Med 2017; 15:211. [PMID: 29202839 PMCID: PMC5715509 DOI: 10.1186/s12916-017-0970-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/31/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epigenomes are tissue specific and thus the choice of surrogate tissue can play a critical role in interpreting neonatal epigenome-wide association studies (EWAS) and in their extrapolation to target tissue. To develop a better understanding of the link between tissue specificity and neonatal EWAS, and the contributions of genotype and prenatal factors, we compared genome-wide DNA methylation of cord tissue and cord blood, two of the most accessible surrogate tissues at birth. METHODS In 295 neonates, DNA methylation was profiled using Infinium HumanMethylation450 beadchip arrays. Sites of inter-individual variability in DNA methylation were mapped and compared across the two surrogate tissues at birth, i.e., cord tissue and cord blood. To ascertain the similarity to target tissues, DNA methylation profiles of surrogate tissues were compared to 25 primary tissues/cell types mapped under the Epigenome Roadmap project. Tissue-specific influences of genotype on the variable CpGs were also analyzed. Finally, to interrogate the impact of the in utero environment, EWAS on 45 prenatal factors were performed and compared across the surrogate tissues. RESULTS Neonatal EWAS results were tissue specific. In comparison to cord blood, cord tissue showed higher inter-individual variability in the epigenome, with a lower proportion of CpGs influenced by genotype. Both neonatal tissues were good surrogates for target tissues of mesodermal origin. They also showed distinct phenotypic associations, with effect sizes of the overlapping CpGs being in the same order of magnitude. CONCLUSIONS The inter-relationship between genetics, prenatal factors and epigenetics is tissue specific, and requires careful consideration in designing and interpreting future neonatal EWAS. TRIAL REGISTRATION This birth cohort is a prospective observational study, designed to study the developmental origins of health and disease, and was retrospectively registered on 1 July 2010 under the identifier NCT01174875 .
Collapse
Affiliation(s)
- Xinyi Lin
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Duke NUS Medical School, Singapore, 169857, Singapore
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore
| | - Li Chen
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore
| | - Ives Yubin Lim
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore
| | - Pei Fang Tan
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore
| | - Julia L MacIsaac
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Alexander M Morin
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Fabian Yap
- KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Kok Hian Tan
- KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Seang Mei Saw
- Duke NUS Medical School, Singapore, 169857, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, 117597, Singapore.,Singapore Eye Research Institute, Singapore, 169856, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Division of Paediatric Endocrinology and Diabetes, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, 119228, Singapore
| | - Joanna D Holbrook
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,NIHR Biomedical Research Centre, University of Southampton, Southampton, SO16 6YD, UK
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Ludmer Centre for Neuroinformatics and Mental Health, Douglas University Mental Health Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, 1142, New Zealand
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore. .,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
20
|
Rosen-Carole CB, Auinger P, Howard CR, Brownell EA, Lanphear BP. Low-Level Prenatal Toxin Exposures and Breastfeeding Duration: A Prospective Cohort Study. Matern Child Health J 2017; 21:2245-2255. [PMID: 28735496 PMCID: PMC5671900 DOI: 10.1007/s10995-017-2346-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Introduction Maternal exposure to tobacco smoke is associated with shortened breastfeeding duration, but few studies have examined the effects on breastfeeding outcomes of low level exposures to other toxic chemicals. Moreover, it is unclear if passive smoking is associated with duration of breastfeeding. Our objective was therefore to examine the effect of low-level prenatal exposures to common environmental toxins (tobacco smoke, lead, and phthalates) on breastfeeding exclusivity and duration. Methods We conducted an analysis of data from the Health Outcomes and Measures of the Environment (HOME) Study. Serum and urine samples were collected at approximately 16 and 26 weeks gestation and at delivery from 373 women; 302 breastfed their infants. Maternal infant feeding interviews were conducted a maximum of eight times through 30 months postpartum. The main predictor variables for this study were gestational exposures to tobacco smoke (measured by serum cotinine), lead, and phthalates. Passive smoke exposure was defined as cotinine levels of 0.015-3.0 μg/mL. Primary outcomes were duration of any and exclusive breastfeeding. Results Serum cotinine concentrations were negatively associated with the duration of any breastfeeding (29.9 weeks unexposed vs. 24.9 weeks with passive exposure, p = 0.04; and 22.4 weeks with active exposure, p = 0.12; p = 0.03 for linear trend), but not duration of exclusive breastfeeding. Prenatal levels of blood lead and urinary phthalate metabolites were not significantly associated with duration of any or exclusive breastfeeding. Conclusions Passive exposure to tobacco smoke during pregnancy was associated with shortened duration of any breastfeeding.
Collapse
Affiliation(s)
- Casey B Rosen-Carole
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, BOX 777, Rochester, NY, 14642, USA.
| | - Peggy Auinger
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, BOX 777, Rochester, NY, 14642, USA
| | - Cynthia R Howard
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, BOX 777, Rochester, NY, 14642, USA
| | | | | |
Collapse
|
21
|
Ferranti EP, Grossmann R, Starkweather A, Heitkemper M. Biological determinants of health: Genes, microbes, and metabolism exemplars of nursing science. Nurs Outlook 2017; 65:506-514. [PMID: 28576296 PMCID: PMC5657318 DOI: 10.1016/j.outlook.2017.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/13/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Increasingly, nurse scientists are incorporating "omics" measures (e.g., genomics, transcriptomics, proteomics, and metabolomics) in studies of biologic determinants of health and behavior. The role of omics in nursing science can be conceptualized in several ways: (a) as a portfolio of biological measures (biomarkers) to monitor individual risk, (b) as a set of combined data elements that can generate new knowledge based on large and complex patient data sets, (c) as baseline information that promotes health education and potentially personalized interventions, and (d) as a platform to understand how environmental parameters (e.g., diet) interact with the individual's physiology. PURPOSE In this article, we provide exemplars of nursing scientists who use omics to better understand specific health conditions. METHODS We highlight various ongoing nursing research investigations incorporating omics technologies to study chronic pain vulnerability, risk for a pain-related condition, cardiometabolic complications associated with pregnancy, and as biomarkers of response to a dietary intervention. DISCUSSION Omics technologies add an important dimension to nursing science across many foci of investigation. However, there are also challenges and opportunities for nurse scientists who consider using omics in their research. CONCLUSION The integration of omics holds promise for increasing the impact of nursing research and practice on population health outcomes.
Collapse
Affiliation(s)
- Erin P Ferranti
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | | | - Angela Starkweather
- Center for Advancement in Managing Pain and P20 Center for Accelerating Precision Pain Self-Management, University of Connecticut School of Nursing, Storrs, CT
| | - Margaret Heitkemper
- Department of Biobehavioral Nursing and Health Informatics, University of Washington School of Nursing, Seattle, WA.
| |
Collapse
|
22
|
Shaffer RM, Smith MN, Faustman EM. Developing the Regulatory Utility of the Exposome: Mapping Exposures for Risk Assessment through Lifestage Exposome Snapshots (LEnS). ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:085003. [PMID: 28796633 PMCID: PMC5783662 DOI: 10.1289/ehp1250] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/08/2017] [Accepted: 04/04/2017] [Indexed: 05/19/2023]
Abstract
BACKGROUND Exposome-related efforts aim to document the totality of human exposures across the lifecourse. This field has advanced rapidly in recent years but lacks practical application to risk assessment, particularly for children's health. OBJECTIVES Our objective was to apply the exposome to children's health risk assessment by introducing the concept of Lifestage Exposome Snapshots (LEnS). Case studies are presented to illustrate the value of the framework. DISCUSSION The LEnS framework encourages organization of exposome studies based on windows of susceptibility for particular target organ systems. Such analyses will provide information regarding cumulative impacts during specific critical periods of the life course. A logical extension of this framework is that regulatory standards should analyze exposure information by target organ, rather than for a single chemical only or multiple chemicals grouped solely by mechanism of action. CONCLUSIONS The LEnS concept is a practical refinement to the exposome that accounts for total exposures during particular windows of susceptibility in target organ systems. Application of the LEnS framework in risk assessment and regulation will improve protection of children's health by enhancing protection of sensitive developing organ systems that are critical for lifelong health and well-being. https://doi.org/10.1289/EHP1250.
Collapse
Affiliation(s)
- Rachel M Shaffer
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington , Seattle, Washington, USA
- Institute for Risk Analysis and Risk Communication, University of Washington , Seattle, Washington, USA
| | - Marissa N Smith
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington , Seattle, Washington, USA
- Institute for Risk Analysis and Risk Communication, University of Washington , Seattle, Washington, USA
| | - Elaine M Faustman
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington , Seattle, Washington, USA
- Institute for Risk Analysis and Risk Communication, University of Washington , Seattle, Washington, USA
| |
Collapse
|
23
|
Wang Y, Guan H. The Role of DNA Methylation in Lens Development and Cataract Formation. Cell Mol Neurobiol 2017; 37:979-984. [PMID: 27858287 PMCID: PMC11482102 DOI: 10.1007/s10571-016-0447-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/10/2016] [Indexed: 10/20/2022]
Abstract
Epigenetics pertains to heritable alterations in genomic structural modifications without altering genomic DNA sequence. The studies of epigenetic mechanisms include DNA methylation, histone modifications, and microRNAs. DNA methylation may contribute to silencing gene expression which is a major mechanism of epigenetic gene regulation. DNA methylation regulatory mechanisms in lens development and pathogenesis of cataract represent exciting areas of research that have opened new avenues for association with aging and environment. This review addresses our current understanding of the major mechanisms and function of DNA methylation in lens development, age-related cataract, secondary cataract, and complicated cataract. By understanding the role of DNA methylation in the lens disease and development, it is expected to open up a new therapeutic approach to clinical treatment of cataract.
Collapse
Affiliation(s)
- Yong Wang
- Eye Institute, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, China
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, China.
| |
Collapse
|
24
|
Neary JL, Perez SM, Peterson K, Lodge DJ, Carless MA. Comparative analysis of MBD-seq and MeDIP-seq and estimation of gene expression changes in a rodent model of schizophrenia. Genomics 2017; 109:204-213. [PMID: 28365388 PMCID: PMC5526217 DOI: 10.1016/j.ygeno.2017.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/14/2017] [Accepted: 03/26/2017] [Indexed: 12/11/2022]
Abstract
We conducted a comparative study of multiplexed affinity enrichment sequence methodologies (MBD-seq and MeDIP-seq) in a rodent model of schizophrenia, induced by in utero methylazoxymethanol acetate (MAM) exposure. We also examined related gene expression changes using a pooled sample approach. MBD-seq and MeDIP-seq identified 769 and 1771 differentially methylated regions (DMRs) between F2 offspring of MAM-exposed rats and saline control rats, respectively. The assays showed good concordance, with ~56% of MBD-seq-detected DMRs being identified by or proximal to MeDIP-seq DMRs. There was no significant overlap between DMRs and differentially expressed genes, suggesting that DNA methylation regulatory effects may act upon more distal genes, or are too subtle to detect using our approach. Methylation and gene expression gene ontology enrichment analyses identified biological processes important to schizophrenia pathophysiology, including neuron differentiation, prepulse inhibition, amphetamine response, and glutamatergic synaptic transmission regulation, reinforcing the utility of the MAM rodent model for schizophrenia research.
Collapse
Affiliation(s)
- Jennifer L Neary
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227, USA.
| | - Stephanie M Perez
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | - Kara Peterson
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227, USA.
| | - Daniel J Lodge
- Department of Pharmacology, Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | - Melanie A Carless
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227, USA.
| |
Collapse
|
25
|
Kobayashi S, Azumi K, Goudarzi H, Araki A, Miyashita C, Kobayashi S, Itoh S, Sasaki S, Ishizuka M, Nakazawa H, Ikeno T, Kishi R. Effects of prenatal perfluoroalkyl acid exposure on cord blood IGF2/H19 methylation and ponderal index: The Hokkaido Study. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2017; 27:251-259. [PMID: 27553991 DOI: 10.1038/jes.2016.50] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/12/2016] [Indexed: 05/18/2023]
Abstract
Prenatal exposure to perfluoroalkyl acids (PFAAs) influences fetal growth and long-term health. However, whether PFAAs affect offspring DNA methylation patterns to influence health outcomes is yet to be evaluated. Here, we assessed effect of prenatal PFAA exposure on cord blood insulin-like growth factor 2 (IGF2), H19, and long interspersed element 1 (LINE1) methylation and its associations with birth size. Mother-child pairs (N=177) from the Hokkaido Study on Environment and Children's Health were included in the study. Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) levels in maternal serum were measured by liquid chromatography-tandem mass spectrometry. IGF2, H19, and LINE1 methylation in cord blood DNA was determined by pyrosequencing. After full adjustment in multiple linear regression models, IGF2 methylation showed a significant negative association with log-unit increase in PFOA (partial regression coefficient=-0.73; 95% confidence interval: -1.44 to -0.02). Mediation analysis suggested that reduced IGF2 methylation explained ~21% of the observed association between PFOA exposure and reduced ponderal index of the infant at birth. These results indicated that the effects of prenatal PFOA exposure could be mediated through DNA methylation. Further study will be required to determine the potential for long-term adverse health effects of reduced IGF2 methylation induced by PFOA exposure.
Collapse
Affiliation(s)
- Sachiko Kobayashi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Kaoru Azumi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Houman Goudarzi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Atsuko Araki
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Chihiro Miyashita
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sumitaka Kobayashi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sachiko Itoh
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Seiko Sasaki
- Department of Public Health Sciences, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Nakazawa
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Tamiko Ikeno
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Reiko Kishi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
26
|
Marks KJ, Hartman TJ, Taylor EV, Rybak ME, Northstone K, Marcus M. Exposure to phytoestrogens in utero and age at menarche in a contemporary British cohort. ENVIRONMENTAL RESEARCH 2017; 155:287-293. [PMID: 28259093 PMCID: PMC5488334 DOI: 10.1016/j.envres.2017.02.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 06/06/2023]
Abstract
Phytoestrogens are estrogenic compounds that occur naturally in plants. Phytoestrogens can cross the placenta, and animal studies have found associations between in utero exposure to phytoestrogens and markers of early puberty. We investigated the association between in utero exposure to phytoestrogens and early menarche (defined as <11.5 years of age at onset) using data from a nested case-control study within the Avon Longitudinal Study of Parents and Children, a longitudinal study involving families living in the South West of England. Concentrations of six phytoestrogens were measured in maternal urine samples collected during pregnancy. Logistic regression was used to explore associations between tertiles of phytoestrogen concentrations and menarche status, with adjustment for maternal age at menarche, maternal education, pre-pregnancy body mass index (BMI), child birth order, duration of breastfeeding, and gestational age at sample collection. Among 367 mother-daughter dyads, maternal median (interquartile range) creatinine-corrected concentrations (in µg/g creatinine) were: genistein 62.1 (27.1-160.9), daidzein 184.8 (88.8-383.7), equol 4.3 (2.8-9.0), O-desmethylangolensin (O-DMA) 13.0 (4.4-34.5), enterodiol 76.1 (39.1-135.8), and enterolactone 911.7 (448.1-1558.0). In analyses comparing those in the highest tertile relative to those in the lowest tertile of in utero phytoestrogen exposure, higher enterodiol levels were inversely associated with early menarche (odds ratio (OR)=0.47; 95% confidence interval (CI): 0.26-0.83), while higher O-DMA levels were associated with early menarche (OR=1.89; 95% CI: 1.04-3.42). These findings suggest that in utero exposure to phytoestrogens may be associated with earlier age at menarche, though the direction of association differs across phytoestrogens.
Collapse
Affiliation(s)
- Kristin J Marks
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, United States; National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Atlanta, GA 30341, United States.
| | - Terryl J Hartman
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, United States; National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Atlanta, GA 30341, United States
| | - Ethel V Taylor
- National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Atlanta, GA 30341, United States
| | - Michael E Rybak
- National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Atlanta, GA 30341, United States
| | - Kate Northstone
- The National Institute for Health Research Collaboration for Leadership in Applied Health Research and Care West (NIHR CLAHRC West) at University Hospitals Bristol NHS Foundation Trust, 9th Floor, White Friars, Lewins Mead, Bristol BS1 2NT, United Kingdom; School of Social and Community Medicine, University of Bristol, 39 Whatley Road, Bristol BS8 2PS, United Kingdom
| | - Michele Marcus
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, United States
| |
Collapse
|
27
|
Vecoli C, Pulignani S, Andreassi MG. Genetic and Epigenetic Mechanisms Linking Air Pollution and Congenital Heart Disease. J Cardiovasc Dev Dis 2016; 3:jcdd3040032. [PMID: 29367575 PMCID: PMC5715723 DOI: 10.3390/jcdd3040032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/08/2016] [Accepted: 11/26/2016] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies strongly suggest that parental air pollutants exposure during the periconceptional period may play a major role in causing fetal/newborn malformations, including a frequent heterogeneity in the methods applied and a difficulty in estimating the clear effect of environmental toxicants. Moreover, only some couples exposed to toxicants during the pre-conception period give birth to a child with congenital anomalies. The reasons for such phenomena remain elusive but they can be explained by the individual, innate ability to metabolize these contaminants that eventually defines the ultimate dose of a biological active toxicant. In this paper, we reviewed the major evidence regarding the role of parental air pollutant exposure on congenital heart disease (CHD) risk as well as the modulating effect on detoxification systems. Finally, major epigenetic alterations induced by adverse environment contaminants have been revised as possible mechanisms altering a correct heart morphogenesis.
Collapse
Affiliation(s)
- Cecilia Vecoli
- Institute of Clinical Physiology-National Research Council (CNR), Via Moruzzi, 1 56124 Pisa, Italy.
| | - Silvia Pulignani
- Institute of Clinical Physiology-National Research Council (CNR), Via Moruzzi, 1 56124 Pisa, Italy.
| | - Maria Grazia Andreassi
- Institute of Clinical Physiology-National Research Council (CNR), Via Moruzzi, 1 56124 Pisa, Italy.
| |
Collapse
|
28
|
Siddeek B, Lakhdari N, Inoubli L, Paul-Bellon R, Isnard V, Thibault E, Bongain A, Chevallier D, Repetto E, Trabucchi M, Michiels JF, Yzydorczyk C, Simeoni U, Urtizberea M, Mauduit C, Benahmed M. Developmental epigenetic programming of adult germ cell death disease: Polycomb protein EZH2-miR-101 pathway. Epigenomics 2016; 8:1459-1479. [PMID: 27762633 DOI: 10.2217/epi-2016-0061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
AIM The Developmental Origin of Health and Disease refers to the concept that early exposure to toxicants or nutritional imbalances during perinatal life induces changes that enhance the risk of developing noncommunicable diseases in adulthood. Patients/materials & methods: An experimental model with an adult chronic germ cell death phenotype resulting from exposure to a xenoestrogen was used. RESULTS A reciprocal negative feedback loop involving decreased EZH2 protein level and increased miR-101 expression was identified. In vitro and in vivo knockdown of EZH2 induced an apoptotic process in germ cells through increased levels of apoptotic factors (BIM and BAD) and DNA repair alteration via topoisomerase 2B deregulation. The increased miR-101 levels were observed in the animal blood, meaning that miR-101 may be a part of a circulating mark of germ cell death. CONCLUSION miR-101-EZH2 pathway deregulation could represent a novel pathophysiological epigenetic basis for adult germ cell disease with environmental and developmental origins.
Collapse
Affiliation(s)
- Bénazir Siddeek
- Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice F-06204, France.,Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France
| | - Nadjem Lakhdari
- Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice F-06204, France.,Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France
| | - Lilia Inoubli
- Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice F-06204, France.,Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France
| | - Rachel Paul-Bellon
- Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice F-06204, France.,Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France
| | - Véronique Isnard
- Centre Hospitalier Universitaire de Nice, Pôle de Digestif-Obstétrique, Centre de Reproduction, Nice F-06202, France
| | - Emmanuelle Thibault
- Centre Hospitalier Universitaire de Nice, Pôle de Biologie, Centre de Reproduction, Nice F-06202, France
| | - André Bongain
- Centre Hospitalier Universitaire de Nice, Pôle de Digestif-Obstétrique, Centre de Reproduction, Nice F-06202, France
| | - Daniel Chevallier
- Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France.,Centre Hospitalier Universitaire de Nice, Pôle d'Urologie, Service d'Urologie, Nice F-06202, France
| | - Emanuela Repetto
- Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France.,Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10, Nice F-06204, France
| | - Michele Trabucchi
- Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France.,Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10, Nice F-06204, France
| | - Jean-François Michiels
- Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France.,Centre Hospitalier Universitaire de Nice, Pôle de Biologie, Service d'Anatomie et de Cytologie Pathologiques, Nice F-06202, France
| | - Catherine Yzydorczyk
- Division of Paediatrics & DOHaD Laboratory, CHUV & University of Lausanne, CH-1011, Switzerland
| | - Umberto Simeoni
- Division of Paediatrics & DOHaD Laboratory, CHUV & University of Lausanne, CH-1011, Switzerland
| | | | - Claire Mauduit
- Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice F-06204, France.,Université Lyon 1, UFR Médecine Lyon Sud, Lyon F-69921, France.,Hospices Civils de Lyon, Hopital Lyon Sud, Laboratoire d'Anatomie et de Cytologie Pathologiques, Pierre-Bénite F-69495, France
| | - Mohamed Benahmed
- Institut National de la Santé et de la Recherche Médicale, Unité 1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, Nice F-06204, France.,Université de Nice Sophia-Antipolis, Unité de Formation et de Recherche (UFR) Médecine, Nice F-06000, France.,Centre Hospitalier Universitaire de Nice, Département de Recherche Clinique et d'Innovation, Nice F-06001, France
| |
Collapse
|
29
|
Watkins DJ, Milewski S, Domino SE, Meeker JD, Padmanabhan V. Maternal phthalate exposure during early pregnancy and at delivery in relation to gestational age and size at birth: A preliminary analysis. Reprod Toxicol 2016. [PMID: 27352641 DOI: 10.1016/j.reprotox.2016.06.021.maternal] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Epidemiologic studies of in utero phthalate exposure and birth outcomes have had conflicting findings. The objective of this study was to characterize maternal phthalate exposure across pregnancy, examine associations between maternal phthalate levels and infant size and gestational age at birth, and investigate relationships between concurrent bisphenol A (BPA) and phthalate exposure and birth outcomes. Women in the Michigan Mother-Infant Pairs cohort provided urine and blood samples during their first trimester and at delivery. Urinary phthalate metabolites and serum BPA were measured at both time points, and birth weight, length, head circumference, and gestational age were recorded from medical records. Maternal DEHP metabolite concentrations were significantly higher at delivery compared to the first trimester (p<0.05), suggesting increased DEHP exposure late in pregnancy. A number of phthalate metabolites were associated with birth size and gestational age in patterns that varied by sex and timing of exposure, independent of BPA exposure.
Collapse
Affiliation(s)
- Deborah J Watkins
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Samantha Milewski
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Steven E Domino
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - John D Meeker
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Konkel L. Lasting Impact of an Ephemeral Organ: The Role of the Placenta in Fetal Programming. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:A124-9. [PMID: 27479992 PMCID: PMC4937843 DOI: 10.1289/ehp.124-a124] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
|
31
|
Watkins DJ, Milewski S, Domino SE, Meeker JD, Padmanabhan V. Maternal phthalate exposure during early pregnancy and at delivery in relation to gestational age and size at birth: A preliminary analysis. Reprod Toxicol 2016; 65:59-66. [PMID: 27352641 DOI: 10.1016/j.reprotox.2016.06.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/14/2016] [Accepted: 06/24/2016] [Indexed: 11/29/2022]
Abstract
Epidemiologic studies of in utero phthalate exposure and birth outcomes have had conflicting findings. The objective of this study was to characterize maternal phthalate exposure across pregnancy, examine associations between maternal phthalate levels and infant size and gestational age at birth, and investigate relationships between concurrent bisphenol A (BPA) and phthalate exposure and birth outcomes. Women in the Michigan Mother-Infant Pairs cohort provided urine and blood samples during their first trimester and at delivery. Urinary phthalate metabolites and serum BPA were measured at both time points, and birth weight, length, head circumference, and gestational age were recorded from medical records. Maternal DEHP metabolite concentrations were significantly higher at delivery compared to the first trimester (p<0.05), suggesting increased DEHP exposure late in pregnancy. A number of phthalate metabolites were associated with birth size and gestational age in patterns that varied by sex and timing of exposure, independent of BPA exposure.
Collapse
Affiliation(s)
- Deborah J Watkins
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Samantha Milewski
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Steven E Domino
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - John D Meeker
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA.,Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Green BB, Houseman EA, Johnson KC, Guerin DJ, Armstrong DA, Christensen BC, Marsit CJ. Hydroxymethylation is uniquely distributed within term placenta, and is associated with gene expression. FASEB J 2016; 30:2874-84. [PMID: 27118675 DOI: 10.1096/fj.201600310r] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/18/2016] [Indexed: 01/08/2023]
Abstract
The conversion of cytosine to 5-methylcystosine (5mC) is an important regulator of gene expression. 5mC may be enzymatically converted to 5-hydroxymethylcytosine (5hmC), with a potentially distinct regulatory function. We sought to investigate these cytosine modifications and their effect on gene expression by parallel processing of genomic DNA using bisulfite and oxidative bisulfite conversion in conjunction with RNA sequencing. Although values of 5hmC across the placental genome were generally low, we identified ∼21,000 loci with consistently elevated levels of 5-hydroxymethycytosine. Absence of 5hmC was observed in CpG islands and, to a greater extent, in non-CpG island-associated regions. 5hmC was enriched within poised enhancers, and depleted within active enhancers, as defined by H3K27ac and H3K4me1 measurements. 5hmC and 5mC were significantly elevated in transcriptionally silent genes when compared with actively transcribed genes. 5hmC was positively associated with transcription in actively transcribed genes only. Our data suggest that dynamic cytosine regulation, associated with transcription, provides the most complete epigenomic landscape of the human placenta, and will be useful for future studies of the placental epigenome.-Green, B. B., Houseman, E. A., Johnson, K. C., Guerin, D. J., Armstrong, D. A., Christensen, B. C., Marsit, C. J. Hydroxymethylation is uniquely distributed within term placenta, and is associated with gene expression.
Collapse
Affiliation(s)
- Benjamin B Green
- Department of Epidemiology and Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA; and
| | - E Andres Houseman
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Kevin C Johnson
- Department of Epidemiology and Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA; and
| | - Dylan J Guerin
- Department of Epidemiology and Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA; and
| | - David A Armstrong
- Department of Epidemiology and Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA; and
| | - Brock C Christensen
- Department of Epidemiology and Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA; and
| | - Carmen J Marsit
- Department of Epidemiology and Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA; and
| |
Collapse
|
33
|
Relton CL, Hartwig FP, Davey Smith G. From stem cells to the law courts: DNA methylation, the forensic epigenome and the possibility of a biosocial archive. Int J Epidemiol 2015; 44:1083-93. [PMID: 26424516 PMCID: PMC5279868 DOI: 10.1093/ije/dyv198] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The growth in epigenetics continues to attract considerable cross-disciplinary interest, apparently representing an opportunity to move beyond genomics towards the goal of understanding phenotypic variability from molecular through organismal to the societal level. The epigenome may also harbour useful information about life-time exposures (measured or unmeasured) irrespective of their influence on health or disease, creating the potential for a person-specific biosocial archive . Furthermore such data may prove of use in providing identifying information, providing the possibility of a future forensic epigenome . The mechanisms involved in ensuring that environmentally induced epigenetic changes perpetuate across the life course remain unclear. Here we propose a potential role of adult stem cells in maintaining epigenetic states provides a useful basis for formulating such epidemiologically-relevant concepts.
Collapse
Affiliation(s)
- Caroline L Relton
- MRC Integrative Epidemiology Unit, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | | | - George Davey Smith
- MRC Integrative Epidemiology Unit, School of Social & Community Medicine, University of Bristol, Bristol, UK
| |
Collapse
|