1
|
Elliott HR, Bennett CL, Caramaschi D, English S. Negative association between higher maternal pre-pregnancy body mass index and breastfeeding outcomes is not mediated by DNA methylation. Sci Rep 2024; 14:14675. [PMID: 38918574 PMCID: PMC11199553 DOI: 10.1038/s41598-024-65605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/21/2024] [Indexed: 06/27/2024] Open
Abstract
The benefits of breastfeeding for the health and wellbeing of both infants and mothers are well documented, yet global breastfeeding rates are low. One factor associated with low breast feeding is maternal body mass index (BMI), which is used as a measure of obesity. The negative relationship between maternal obesity and breastfeeding is likely caused by a variety of social, psychological, and physiological factors. Maternal obesity may also have a direct biological association with breastfeeding through changes in maternal DNA methylation. Here, we investigate this potential biological association using data from a UK-based cohort study, the Avon Longitudinal Study of Parents and Children (ALSPAC). We find that pre-pregnancy body mass index (BMI) is associated with lower initiation to breastfeed and shorter breastfeeding duration. We conduct epigenome-wide association studies (EWAS) of pre-pregnancy BMI and breastfeeding outcomes, and run candidate-gene analysis of methylation sites associated with BMI identified via previous meta-EWAS. We find that DNA methylation at cg11453712, annotated to PHTP1, is associated with pre-pregnancy BMI. From our results, neither this association nor those at candidate-gene sites are likely to mediate the link between pre-pregnancy BMI and breastfeeding.
Collapse
Affiliation(s)
- Hannah R Elliott
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Chloe L Bennett
- School of Biological Sciences, University of Bristol, Bristol, UK
| | - Doretta Caramaschi
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Faculty of Health and Life Sciences, Department of Psychology, University of Exeter, Exeter, UK
| | - Sinead English
- School of Biological Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
2
|
Adibi JJ, Zhao Y, Koistinen H, Mitchell RT, Barrett ES, Miller R, O'Connor TG, Xun X, Liang HW, Birru R, Smith M, Moog NK. Molecular pathways in placental-fetal development and disruption. Mol Cell Endocrinol 2024; 581:112075. [PMID: 37852527 PMCID: PMC10958409 DOI: 10.1016/j.mce.2023.112075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/11/2023] [Accepted: 09/24/2023] [Indexed: 10/20/2023]
Abstract
The first trimester of pregnancy ranks high in priority when minimizing harmful exposures, given the wide-ranging types of organogenesis occurring between 4- and 12-weeks' gestation. One way to quantify potential harm to the fetus in the first trimester is to measure a corollary effect on the placenta. Placental biomarkers are widely present in maternal circulation, cord blood, and placental tissue biopsied at birth or at the time of pregnancy termination. Here we evaluate ten diverse pathways involving molecules expressed in the first trimester human placenta based on their relevance to normal fetal development and to the hypothesis of placental-fetal endocrine disruption (perturbation in development that results in abnormal endocrine function in the offspring), namely: human chorionic gonadotropin (hCG), thyroid hormone regulation, peroxisome proliferator activated receptor protein gamma (PPARγ), leptin, transforming growth factor beta, epiregulin, growth differentiation factor 15, small nucleolar RNAs, serotonin, and vitamin D. Some of these are well-established as biomarkers of placental-fetal endocrine disruption, while others are not well studied and were selected based on discovery analyses of the placental transcriptome. A literature search on these biomarkers summarizes evidence of placenta-specific production and regulation of each biomarker, and their role in fetal reproductive tract, brain, and other specific domains of fetal development. In this review, we extend the theory of fetal programming to placental-fetal programming.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, University of Pittsburgh School of Public Health, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Yaqi Zhao
- St. Jude's Research Hospital, Memphis, TN, USA
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Rod T Mitchell
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh BioQuarter, Edinburgh, UK
| | - Emily S Barrett
- Environmental and Population Health Bio-Sciences, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - Richard Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Hai-Wei Liang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Rahel Birru
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Megan Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nora K Moog
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
3
|
Gómez-Vilarrubla A, Mas-Parés B, Carreras-Badosa G, Bonmatí-Santané A, Martínez-Calcerrada JM, Niubó-Pallàs M, de Zegher F, Ibáñez L, López-Bermejo A, Bassols J. DNA Methylation Signatures in Paired Placenta and Umbilical Cord Samples: Relationship with Maternal Pregestational Body Mass Index and Offspring Metabolic Outcomes. Biomedicines 2024; 12:301. [PMID: 38397903 PMCID: PMC10886657 DOI: 10.3390/biomedicines12020301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
An epigenomic approach was used to study the impact of maternal pregestational body mass index (BMI) on the placenta and umbilical cord methylomes and their potential effect on the offspring's metabolic phenotype. DNA methylome was assessed in 24 paired placenta and umbilical cord samples. The differentially methylated CpGs associated with maternal pregestational BMI were identified and the metabolic pathways and the potentially related diseases affected by their annotated genes were determined. Two top differentially methylated CpGs were studied in 90 additional samples and the relationship with the offspring's metabolic phenotype was determined. The results showed that maternal pregestational BMI is associated with the methylation of genes involved in endocrine and developmental pathways with potential effects on type 2 diabetes and obesity. The methylation and expression of HADHA and SLC2A8 genes in placenta and umbilical cord were related to several metabolic parameters in the offspring at 6 years (weight SDS, height SDS, BMI SDS, Δ BW-BMI SDS, FM SDS, waist, SBP, TG, HOMA-IR, perirenal fat; all p < 0.05). Our data suggest that epigenetic analysis in placenta and umbilical cord may be useful for identifying individual vulnerability to later metabolic diseases.
Collapse
Affiliation(s)
- Ariadna Gómez-Vilarrubla
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Berta Mas-Parés
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | | | | | - Maria Niubó-Pallàs
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Francis de Zegher
- Department of Development & Regeneration, University of Leuven, 3000 Leuven, Belgium;
| | - Lourdes Ibáñez
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children’s Hospital, 08950 Esplugues de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
- Department of Pediatrics, Dr. Josep Trueta Hospital, 17007 Girona, Spain
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| |
Collapse
|
4
|
Mas-Parés B, Xargay-Torrent S, Gómez-Vilarrubla A, Carreras-Badosa G, Prats-Puig A, De Zegher F, Ibáñez L, Bassols J, López-Bermejo A. Gestational Weight Gain Relates to DNA Methylation in Umbilical Cord, Which, In Turn, Associates with Offspring Obesity-Related Parameters. Nutrients 2023; 15:3175. [PMID: 37513594 PMCID: PMC10386148 DOI: 10.3390/nu15143175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Excessive gestational weight gain (GWG) has a negative impact on offspring's health. Epigenetic modifications mediate these associations by causing changes in gene expression. We studied the association between GWG and DNA methylation in umbilical cord tissue; and determined whether the DNA methylation and the expression of corresponding annotated genes were associated with obesity-related parameters in offspring at 6 years of age. The methylated CpG sites (CpGs) associated with GWG were identified in umbilical cord tissue by genome-wide DNA methylation (n = 24). Twelve top CpGs were validated in a wider sample by pyrosequencing (n = 87), and the expression of their 5 annotated genes (SETD8, TMEM214, SLIT3, RPTOR, and HOXC8) was assessed by RT-PCR. Pyrosequencing results validated the association of SETD8, SLIT3, and RPTOR methylation with GWG and showed that higher levels of SETD8 and RPTOR methylation and lower levels of SLIT3 methylation relate to a higher risk of obesity in the offspring. The association of SETD8 and SLIT3 gene expression with offspring outcomes paralleled the association of methylation levels in opposite directions. Epigenetic changes in the umbilical cord tissue could explain, in part, the relationship between GWG and offspring obesity risk and be early biomarkers for the prevention of overweight and obesity in childhood.
Collapse
Affiliation(s)
- Berta Mas-Parés
- Pediatric Endocrinology Research Group, (Girona Biomedical Research Institute) IDIBGI, 17190 Salt, Spain
| | - Sílvia Xargay-Torrent
- Pediatric Endocrinology Research Group, (Girona Biomedical Research Institute) IDIBGI, 17190 Salt, Spain
| | - Ariadna Gómez-Vilarrubla
- Materno-Fetal Metabolic Research Group, (Girona Biomedical Research Institute) IDIBGI, 17190 Salt, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, (Girona Biomedical Research Institute) IDIBGI, 17190 Salt, Spain
| | - Anna Prats-Puig
- University School of Health and Sport (EUSES), University of Girona, 17190 Salt, Spain
| | - Francis De Zegher
- Department of Development & Regeneration, University of Leuven, 3000 Leuven, Belgium
| | - Lourdes Ibáñez
- Endocrinology Department, Research Institute Sant Joan de Déu, University of Barcelona, 08950 Esplugues, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Judit Bassols
- Materno-Fetal Metabolic Research Group, (Girona Biomedical Research Institute) IDIBGI, 17190 Salt, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, (Girona Biomedical Research Institute) IDIBGI, 17190 Salt, Spain
- Department of Pediatrics, Dr. Josep Trueta Hospital, 17007 Girona, Spain
- Department of Medical Sciences, University of Girona, 17003 Girona, Spain
| |
Collapse
|
5
|
Tehrani JM, Kennedy E, Tung PW, Burt A, Hermetz K, Punshon T, Jackson BP, Hao K, Chen J, Karagas MR, Koestler DC, Lester B, Marsit CJ. Human placental microRNAs dysregulated by cadmium exposure predict neurobehavioral outcomes at birth. Pediatr Res 2023; 93:1410-1418. [PMID: 35906307 PMCID: PMC9884320 DOI: 10.1038/s41390-022-02201-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 06/22/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Prenatal cadmium (Cd) exposure has been implicated in both placental toxicity and adverse neurobehavioral outcomes. Placental microRNAs (miRNAs) may function to developmentally program adverse pregnancy and newborn health outcomes in response to gestational Cd exposure. METHODS In a subset of the Rhode Island Child Health Study (RICHS, n = 115) and the New Hampshire Birth Cohort Study (NHBCS, = 281), we used small RNA sequencing and trace metal analysis to identify Cd-associated expression of placental miRNAs using negative binomial generalized linear models. We predicted mRNAs targeted by Cd-associated miRNAs and relate them to neurobehavioral outcomes at birth through the integration of transcriptomic data and summary scores from the NICU Network Neurobehavioral Scale (NNNS). RESULTS Placental Cd concentrations are significantly associated with the expression level of five placental miRNAs in NHBCS, with similar effect sizes in RICHS. These miRNA target genes overrepresented in nervous system development, and their expression is correlated with NNNS metrics suggestive of atypical neurobehavioral outcomes at birth. CONCLUSIONS Gestational Cd exposure is associated with the expression of placental miRNAs. Predicted targets of these miRNAs are involved in nervous system development and may also regulate placental physiology, allowing their dysregulation to modify developmental programming of early life health outcomes. IMPACT This research aims to address the poor understanding of the molecular mechanisms governing adverse pregnancy and newborn health outcomes in response to Gestational cadmium (Cd) exposure. Our results outline a robust relationship between Cd-associated placental microRNA expression and NICU Network Neurobehavioral Scales (NNNS) at birth indicative of atypical neurobehavior. This study utilized healthy mother-infant cohorts to describe the role of Cd-associated dysregulation of placental microRNAs as a potential mechanism by which adverse neurobehavioral outcomes are developmentally programmed.
Collapse
Affiliation(s)
- Jesse M Tehrani
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Elizabeth Kennedy
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Pei Wen Tung
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Karen Hermetz
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Tracy Punshon
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Brian P Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH, USA
| | - Ke Hao
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Devin C Koestler
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Barry Lester
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, USA
- The Brown Center of the Study of Children at Risk, Brown University, Providence, RI, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
6
|
Tehrani JM, Kennedy EM, Tian FY, Everson TM, Deyssenroth M, Burt A, Hermetz K, Hao K, Chen J, Koestler DC, Marsit CJ. Variation in placental microRNA expression associates with maternal family history of cardiovascular disease. J Dev Orig Health Dis 2023; 14:132-139. [PMID: 35815737 PMCID: PMC9832176 DOI: 10.1017/s2040174422000319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In the United States, cardiovascular disease is the leading cause of death and the rate of maternal mortality remains among the highest of any industrialized nation. Maternal cardiometabolic health throughout gestation and postpartum is representative of placental health and physiology. Both proper placental functionality and placental microRNA expression are essential to successful pregnancy outcomes, and both are highly sensitive to genetic and environmental sources of variation. Placental pathologies, such as preeclampsia, are associated with maternal cardiovascular health but may also contribute to the developmental programming of chronic disease in offspring. However, the role of more subtle alterations to placental function and microRNA expression in this developmental programming remains poorly understood. We performed small RNA sequencing to investigate microRNA in placentae from the Rhode Island Child Health Study (n = 230). MicroRNA counts were modeled on maternal family history of cardiovascular disease using negative binomial generalized linear models. MicroRNAs were considered to be differentially expressed at a false discovery rate (FDR) less than 0.10. Parallel mRNA sequencing data and bioinformatic target prediction software were then used to identify potential mRNA targets of differentially expressed microRNAs. Nine differentially expressed microRNAs were identified (FDR < 0.1). Bioinformatic target prediction revealed 66 potential mRNA targets of these microRNAs, many of which are implicated in TGFβ signaling pathway but also in pathways involving cellular metabolism and immunomodulation. A robust association exists between familial cardiovascular disease and placental microRNA expression which may be implicated in both placental insufficiencies and the developmental programming of chronic disease.
Collapse
Affiliation(s)
- Jesse M. Tehrani
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Elizabeth M. Kennedy
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Fu-Ying Tian
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Todd M. Everson
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Karen Hermetz
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Ke Hao
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Devin C. Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Carmen J. Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
7
|
Xu P, Dong S, Wu L, Bai Y, Bi X, Li Y, Shu C. Maternal and Placental DNA Methylation Changes Associated with the Pathogenesis of Gestational Diabetes Mellitus. Nutrients 2022; 15:nu15010070. [PMID: 36615730 PMCID: PMC9823627 DOI: 10.3390/nu15010070] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is an important metabolic complication of pregnancy, which affects the future health of both the mother and the newborn. The pathogenesis of GDM is not completely clear, but what is clear is that with the development and growth of the placenta, GDM onset and blood glucose is difficult to control, while gestational diabetes patients' blood glucose drops and reaches normal after placenta delivery. This may be associated with placental secretion of insulin-like growth factor, adipokines, tumor necrosis factor-α, cytokines and insulin resistance. Therefore, endocrine secretion of placenta plays a key role in the pathogenesis of GDM. The influence of DNA methylation of these molecules and pathway-related genes on gene expression is also closely related to the pathogenesis of GDM. Here, this review attempts to clarify the pathogenesis of GDM and the related maternal and placental DNA methylation changes and how they affect metabolic pathways.
Collapse
|
8
|
The Complex Roles of Adipokines in Polycystic Ovary Syndrome and Endometriosis. Biomedicines 2022; 10:biomedicines10102503. [PMID: 36289764 PMCID: PMC9598769 DOI: 10.3390/biomedicines10102503] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/30/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) and endometriosis are frequent diseases of the female reproductive tract causing high morbidity as they can significantly affect fertility and quality of life. Adipokines are pleiotropic signaling molecules secreted by white or brown adipose tissues with a central role in energy metabolism. More recently, their involvement in PCOS and endometriosis has been demonstrated. In this review article, we provide an update on the role of adipokines in both diseases and summarize previous findings. We also address the results of multi-omics approaches in adipokine research to examine the role of single nucleotide polymorphisms (SNPs) in genes coding for adipokines and their receptors, the secretome of adipocytes and to identify epigenetic alterations of adipokine genes that might be conferred from mother to child. Finally, we address novel data on the role of brown adipose tissue (BAT), which seems to have notable effects on PCOS. For this review, original research articles on adipokine actions in PCOS and endometriosis are considered, which are listed in the PubMed database.
Collapse
|
9
|
Ortega-Avila JG, García-Muñoz H, Segura Ordoñez A, Salazar Contreras BC. Sexual dimorphism of leptin and adiposity in children between 0 and 10 years: a systematic review and meta-analysis. Biol Sex Differ 2022; 13:47. [PMID: 36064746 PMCID: PMC9446796 DOI: 10.1186/s13293-022-00454-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Differences in adolescents and adults by sex in blood levels of leptin and adiposity have been described; however, it is not yet clear if these differences arise from the prepubertal stage in subjects with a normal-weight. Therefore, we examine whether there are differences by sex in levels of blood leptin and adiposity in children with a normal-weight between 0 and 10 years old. METHODS Search strategy: eligible studies were obtained from three electronic databases (Ovid, Embase and LILACS) and contact with experts. SELECTION CRITERIA healthy children up to 10 years of age with normal-weight according to age. DATA COLLECTION AND ANALYSES data were extracted by four independent reviewers using a predesigned data collection form. For the analysis, we stratified according to age groups (newborns, 0.25-0.5 years, 3-5.9 years, 6-7.9 years, 8-10 years). The statistical analysis was performed in the R program. RESULTS Of the initially identified 13,712 records, 21 were selected in the systematic review and meta-analysis. The sex was associated with the overall effect on blood leptin (pooled MD = 1.72 ng/mL, 95% CI: 1.25-2.19) and body fat percentage (pooled MD = 3.43%, 95% CI: 2.53-4.33), being both higher in girls. This finding was consistent in the majority of age groups. CONCLUSION The results of our meta-analyses support the sexual dimorphism in circulating blood leptin and body fat percentage between girls and boys with normal-weight from prepuberty.
Collapse
Affiliation(s)
- Jose Guillermo Ortega-Avila
- Grupo de Investigación de Ciencias Básicas y Clínicas de la Salud, Departamento de Ciencias Básicas de la Salud, Pontificia Universidad Javeriana, Seccional-Cali, Cali, Colombia
- Grupo de investigación Salud y Movimiento, Facultad de Salud, Universidad Santiago de Cali, Cali, Colombia
| | - Harry García-Muñoz
- Grupo de investigación Salud y Movimiento, Facultad de Salud, Universidad Santiago de Cali, Cali, Colombia
- Grupo de Nutrición, Departamento de Ciencias Fisiológicas, Facultad de Salud, Universidad del Valle, Cali, Colombia
| | - Alejandro Segura Ordoñez
- Grupo de investigación Salud y Movimiento, Facultad de Salud, Universidad Santiago de Cali, Cali, Colombia
- Grupo de Nutrición, Departamento de Ciencias Fisiológicas, Facultad de Salud, Universidad del Valle, Cali, Colombia
| | - Blanca C. Salazar Contreras
- Grupo de investigación Salud y Movimiento, Facultad de Salud, Universidad Santiago de Cali, Cali, Colombia
- Programa de Medicina, Facultad de Salud, Universidad Icesi, Cali, Colombia
| |
Collapse
|
10
|
Marley AR, Ryder JR, Turcotte LM, Spector LG. Maternal obesity and acute lymphoblastic leukemia risk in offspring: A summary of trends, epidemiological evidence, and possible biological mechanisms. Leuk Res 2022; 121:106924. [PMID: 35939888 DOI: 10.1016/j.leukres.2022.106924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/13/2022] [Accepted: 08/01/2022] [Indexed: 10/16/2022]
Abstract
Acute lymphoblastic leukemia, a heterogenous malignancy characterized by uncontrolled proliferation of lymphoid progenitors and generally initiated in utero, is the most common pediatric cancer. Although incidence of ALL has been steadily increasing in recent decades, no clear reason for this trend has been identified. Rising concurrently with ALL incidence, increasing maternal obesity rates may be partially contributing to increasing ALL prevelance. Epidemiological studies, including a recent meta-analysis, have found an association between maternal obesity and leukemogenesis in offspring, although mechanisms underlying this association remain unknown. Therefore, the purpose of this review is to propose possible mechanisms connecting maternal obesity to ALL risk in offspring, including changes to fetal/neonatal epigenetics, altered insulin-like growth factor profiles and insulin resistance, modified adipokine production and secretion, changes to immune cell populations, and impacts on birthweight and childhood obesity/adiposity. We describe how each proposed mechanism is biologically plausible due to their connection with maternal obesity, presence in neonatal and/or fetal tissue, observation in pediatric ALL patients at diagnosis, and association with leukemogenesis, A description of ALL and maternal obesity trends, a summary of epidemiological evidence, a discussion of the pathway from intrauterine environment to subsequent malignancy, and propositions for future directions are also presented.
Collapse
Affiliation(s)
- Andrew R Marley
- Division of Epidemiology and Clinical Research, Department of Pediatrics, University of Minnesota, 420 Delaware St SE MMC 715, Minneapolis, MN 55455, USA.
| | - Justin R Ryder
- Division of Epidemiology and Clinical Research, Department of Pediatrics, University of Minnesota, 420 Delaware St SE MMC 715, Minneapolis, MN 55455, USA; Center for Pediatric Obesity Medicine, Department of Pediatrics, University of Minnesota, 2450 Riverside Ave S AO-102, Minneapolis, MN 55454, USA
| | - Lucie M Turcotte
- Division of Hematology/Oncology, Department of Pediatrics, University of Minnesota, 420 Delaware St SE MMC 484, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, 425 East River Parkway, Minneapolis, MN 55455, USA
| | - Logan G Spector
- Division of Epidemiology and Clinical Research, Department of Pediatrics, University of Minnesota, 420 Delaware St SE MMC 715, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, 425 East River Parkway, Minneapolis, MN 55455, USA
| |
Collapse
|
11
|
Padbury JF, Do BT, Bann CM, Marsit C, Hintz SR, Vohr BR, Lowe J, Newman JE, Granger DA, Payne A, Watterberg K. DNA methylation in former extremely low birth weight newborns: association with cardiovascular and endocrine function. Pediatr Res 2022; 91:1469-1477. [PMID: 33953357 PMCID: PMC8568736 DOI: 10.1038/s41390-021-01531-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/29/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND There is increased risk of cardiovascular, metabolic, and hypertensive disorders in later life in the preterm population. We studied school-age children who had been born extremely premature who had undergone endocrine, cardiovascular, and anthropometric evaluations. METHODS School age measurements of salivary cortisol, adrenal androgens, blood pressure, and anthropometric markers were correlated with DNA methylation of 11-betahydroxysteroid dehydrogenase type 2 (11BHSD2), leptin, and the LINE1 repetitive DNA element. RESULTS We observed a modest correlation between log AUC for salivary cortisol and methylation of leptin in preterm infants and a negative correlation between methylation of region 1 of the glucocorticoid receptor (GR in term-born infants. There was an association between LINE1 methylation and cortisol response to awakening and a negative correlation between LINE1 and systolic blood pressure at 6-7 years. Methylation of the GR promoter region showed a positive association with systolic blood pressure at 6-7 years of age. CONCLUSIONS These results show that extremely preterm birth, followed by complex patterns of endocrine, cardiovascular, and metabolic exposures during early postnatal life, is associated with lasting changes in DNA methylation patterns in genes involved in hypothalamic pituitary adrenal axis function, adrenal hormonal regulation, and cardiometabolic risk. IMPACT Preterm infants have significant environmental and physiological exposures during early life that may have lasting impact on later function. Alterations in hypothalamic pituitary adrenal axis (HPA) function have been associated with these exposures. We examined the associated changes in DNA methylation of important genes involved in HPA function, metabolism, and global DNA methylation. The changes we saw in DNA methylation may help to explain associated cardiovascular, metabolic, and growth disturbance in these children in later life.
Collapse
Affiliation(s)
- James F. Padbury
- Department of Pediatrics, Women & Infants Hospital, Brown University, Providence, RI
| | - Barbara T. Do
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC
| | - Carla M. Bann
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC
| | - Carmen Marsit
- Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA
| | - Susan R. Hintz
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | - Betty R. Vohr
- Department of Pediatrics, Women & Infants Hospital, Brown University, Providence, RI
| | - Jean Lowe
- University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Jamie E. Newman
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC
| | - Douglas A. Granger
- Institute for Interdisciplinary Salivary Bioscience Research, University of California at Irvine, Irvine, CA, Johns Hopkins University School of Medicine, Johns Hopkins University Bloomberg School of Public Health and Johns Hopkins University School of Nursing, Baltimore, MD
| | - Allison Payne
- Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University, Cleveland, OH
| | | | | |
Collapse
|
12
|
Zong X, Wang H, Yang L, Guo Y, Zhao M, Magnussen CG, Xi B. Maternal Pre-pregnancy Body Mass Index Categories and Infant Birth Outcomes: A Population-Based Study of 9 Million Mother–Infant Pairs. Front Nutr 2022; 9:789833. [PMID: 35252291 PMCID: PMC8891137 DOI: 10.3389/fnut.2022.789833] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 01/14/2023] Open
Abstract
Background and Aims Infant adverse birth outcomes have been suggested to contribute to neonatal morbidity and mortality and may cause long-term health consequences. Although evidence suggests maternal prepregnancy body mass index (BMI) categories associate with some birth outcomes, there is no consensus on these associations. We aimed to examine the associations of maternal prepregnancy BMI categories with a wide range of adverse birth outcomes. Methods Data were from a population-based retrospective cohort study of 9,282,486 eligible mother–infant pairs in the U.S. between 2016 and 2018. Maternal prepregnancy BMI was classified as: underweight (<18.5 kg/m2); normal weight (18.5–24.9 kg/m2); overweight (25.0–29.9 kg/m2); obesity grade 1 (30–34.9 kg/m2); obesity grade 2 (35.0–39.9 kg/m2); and obesity grade 3 (≥40 kg/m2). A total of six birth outcomes of the newborn included preterm birth, low birthweight, macrosomia, small for gestational age (SGA), large for gestational age (LGA), and low Apgar score (5-min score <7). Results Maternal prepregnancy overweight and obesity increased the likelihood of infant preterm birth, with odds ratios (ORs) (95% CIs) of 1.04 (1.04–1.05) for overweight, 1.18 (1.17–1.19) for obesity grade 1, 1.31 (1.29–1.32) for obesity grade 2, and 1.47 (1.45–1.48) for obesity grade 3, and also for prepregnancy underweight (OR = 1.32, 95% CI = 1.30–1.34) after adjusting for all potential covariates. Prepregnancy overweight and obesity were associated with higher odds of macrosomia, with ORs (95% CIs) of 1.53 (1.52–1.54) for overweight, 1.92 (1.90–1.93) for obesity grade 1, 2.33 (2.31–2.35) for obesity grade 2, and 2.87 (2.84–2.90) for obesity grade 3. Prepregnancy overweight and obesity was associated with higher odds of LGA, with ORs (95% CIs) of 1.58 (1.57–1.59) for overweight, 2.05 (2.03–2.06) for obesity grade 1, 2.54 (2.52–2.56) for obesity grade 2, and 3.17 (3.14–3.21) for obesity grade 3. Prepregnancy overweight and obesity were also associated with higher odds of low Apgar score, with ORs (95% CIs) of 1.12 (1.11–1.14) for overweight, 1.21 (1.19–1.23) for obesity grade 1, 1.34 (1.31–1.36) for obesity grade 2, and 1.55 (1.51–1.58) for obesity grade 3. Conclusion Our findings suggest maintaining or obtaining a healthy body weight for prepregnancy women could substantially reduce the likelihood of important infant adverse birth outcomes.
Collapse
Affiliation(s)
- Xin'nan Zong
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Growth and Development, Capital Institute of Pediatrics, Beijing, China
| | - Huan Wang
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liu Yang
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yajun Guo
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Min Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Costan G. Magnussen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Bo Xi
| |
Collapse
|
13
|
Vickers MH. Early life nutrition and neuroendocrine programming. Neuropharmacology 2021; 205:108921. [PMID: 34902348 DOI: 10.1016/j.neuropharm.2021.108921] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022]
Abstract
Alterations in the nutritional environment in early life can significantly increase the risk for obesity and a range of development of metabolic disorders in offspring in later life, effects that can be passed onto future generations. This process, termed development programming, provides the framework of the developmental origins of health and disease (DOHaD) paradigm. Early life nutritional compromise including undernutrition, overnutrition or specific macro/micronutrient deficiencies, results in a range of adverse health outcomes in offspring that can be further exacerbated by a poor postnatal nutritional environment. Although the mechanisms underlying programming remain poorly defined, a common feature across the phenotypes displayed in preclinical models is that of altered wiring of neuroendocrine circuits that regulate satiety and energy balance. As such, altered maternal nutritional exposures during critical early periods of developmental plasticity can result in aberrant hardwiring of these circuits with lasting adverse consequences for the offspring. There is also increasing evidence around the role of an altered epigenome and the gut-brain axis in mediating some of the central programming effects observed. Further, although such programming was once considered to result in a permanent change in developmental trajectory, there is evidence, at least from preclinical models, that programming can be reversed via targeted nutritional manipulations during early development. Further work is required at a mechanistic level to allow for identification for early markers of later disease risk, delineation of sex-specific effects and pathways to implementation of strategies aimed at breaking the transgenerational transmission of disease.
Collapse
Affiliation(s)
- M H Vickers
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand.
| |
Collapse
|
14
|
Allbrand M, Eklund D, Cao Y, Nilsson K, Lodefalk M. Gene expression of leptin, leptin receptor isoforms and inflammatory cytokines in placentas of obese women - Associations to birth weight and fetal sex. Placenta 2021; 117:64-71. [PMID: 34773742 DOI: 10.1016/j.placenta.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/26/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Leptin signaling in placentas of obese women may influence fetal growth and may be dependent on fetal sex. The aim of this study was to investigate placental gene expression of leptin, its receptor and inflammatory cytokines in obese mothers in relation to offspring birth weight and sex. METHODS In total, 109 placental tissue samples from severely obese women (body mass index in first trimester ≥35 kg/m2) giving birth vaginally at term to a healthy child were included. Quantitative real-time PCR was used for the analysis of leptin (LEP), its receptor LEPR with two splice variants, interleukin (IL)1B, chemokine (C-X-C motif) ligand 8 (CXCL8), tumour necrosis factor (TNF), IL6, IL10, hypoxia-inducible factor 1-alpha (HIF1A) and insulin receptor (INSR). The subjects were divided into three groups based on LEP expression percentiles (<25th percentile; 25-75th percentile and >75th percentile). RESULTS A reverse U-shaped association between LEP expression and birth weight z-scores was found (R2 = 0.075, p = 0.005). Placental LEPRb expression was downregulated (p = 0.034) in those with highest LEP expression. Female infants had higher birth weight z-scores than males (0.58 (-1.49-2.88) vs 0.21 (-1.50-2.93), p = 0.020) and their placental LEPRb expression was upregulated (p = 0.047). The associations between expression of different genes differed by sex. DISCUSSION A reverse U-shaped relationship between placental LEP expression and offspring birth weight z-scores was found together with sexual dimorphism in LEPRb expression indicating a complex regulation of fetal growth by placental leptin signaling in maternal obesity.
Collapse
Affiliation(s)
- Marianne Allbrand
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Daniel Eklund
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Kerstin Nilsson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Maria Lodefalk
- Department of Paediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden; University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
15
|
Sadashiv, Modi A, Khokhar M, Sharma P, Joshi R, Mishra SS, Bharshankar R, Tiwari S, Singh PK, Bhosale VV, Negi MPS. Leptin DNA Methylation and Its Association with Metabolic Risk Factors in a Northwest Indian Obese Population. J Obes Metab Syndr 2021; 30:304-311. [PMID: 34380782 PMCID: PMC8526297 DOI: 10.7570/jomes20131] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/05/2022] Open
Abstract
Background It is well established that obesity is a major health risk in diabetes and associated diseases. Epigenetic changes, specially DNA methylation, play an important role in regulation of adipokines. The objective of the present study was to evaluate the DNA methylation status at the promoter region of the leptin gene in obese individuals and its association with metabolic risk factors. Methods The study included obese (n=100) and non-obese (n=75) individuals aged 25-45 years, and measured their physical, biochemical parameters (glucose, insulin, and lipid profiles) and leptin, DNA methyltransferase 1 (DNMT1), and DNA methyltransferase 3 beta (DNMT3b) mRNA expressions with real-time reverse transcription-polymerase chain reaction (qRT-PCR). DNA methylation of the leptin gene at the promoter region was analyzed by methyl-specific qPCR . Results The study found that the DNA methylation level at the promoter area of the leptin gene was negatively associated with weight in obese subjects. Furthermore, study findings showed that the DNA methylation level was negatively associated with fasting insulin, glucose, homeostatic model assessment for insulin resistance, and total cholesterol. There was also a higher expression of DNMT1 and DNMT-3b in obese subjects as compared with non-obese subjects. Conclusion The leptin epigenetic profile may be associated with obesity and its associated metabolic risk factors.
Collapse
Affiliation(s)
- Sadashiv
- Department of Biochemistry, All India Institute of Medical Sciences Raebareli, Lucknow, India.,Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Lucknow, India.,Department of Physiology, All India Institute of Medical Sciences Bhopal, Lucknow, India
| | - Anupama Modi
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Lucknow, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Lucknow, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Lucknow, India
| | - Rajnish Joshi
- Deparment of General Medicine, All India Institute of Medical Sciences Bhopal, Lucknow, India
| | | | - Rajay Bharshankar
- Department of Physiology, All India Institute of Medical Sciences Bhopal, Lucknow, India
| | - Sunita Tiwari
- Department of Physiology, King George's Medical University, Lucknow, India
| | - Pankaj Kumar Singh
- Department of Biochemistry, All India Institute of Medical Sciences Bibinagar, Lucknow, India
| | - Vivek Vidyadhar Bhosale
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mahendra Pal Singh Negi
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
16
|
Angulo M, Ramirez-Montaño D, Torres-Canchala L, García X, Lemus R, Aristizabal AM, Floyd-Aristizábal D, Dávalos DM, Diaz-Ordoñez L, Pachajoa H. Methylation Status of GLP2R, LEP and IRS2 in Small for Gestational Age Children with and without Catch-up Growth. J Clin Res Pediatr Endocrinol 2021; 13:136-145. [PMID: 32936762 PMCID: PMC8186343 DOI: 10.4274/jcrpe.galenos.2020.2020.0070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE In small for gestational age (SGA) children, catch-up growth could be influenced by methylation of several genes involved in metabolism. Epigenetics may influence the development of metabolic diseases in adulthood. To compare the methylation of leptin (LEP), glucagon-like peptide-2 receptor (GLP2R), insulin receptor substrate-2 (IRS2) in SGA patients with and without catch-up growth. METHODS Observational prospective study of SGA children. Demographical and clinical variables were collected from clinical records and parents’ questionnaire. Methylation status of LEP, IRS2, and GLP2R promoters was evaluated in DNA extracted from patient and one parent saliva samples. RESULTS Forty-eight SGA patients were included. Twenty-six (54.2%) had catch-up growth phenotype and 22 (45.8%) did not. The median age was 5.2 years [RIC 4.1-6.8] without difference between groups (p=0.306). The catch-up group had increased appetite (42.3% vs 9.1%, p=0.008), family history of dyslipidemia (42.3% vs 27.3%) and diabetes (34.6% vs 22.7%) compared to non-catch-up group. Catch-up patients had significantly larger waist circumference compared to non-catch-up group (median 55 cm [RIC 52-58] versus median 49.5 cm [RIC46-52]; p<0.001). LEP and GLP2R were methylated in all samples. IRS2 was methylated in 60% of SGA patients without difference between groups (p=0.520). CONCLUSION There is no association between IRS2 methylation and catch-up growth among SGA patients. LEP and GLP2R were methylated in all SGA patients. Gene methylation may be implicated in metabolic disease later in life. More studies should be performed to confirm this hypothesis.
Collapse
Affiliation(s)
- Mario Angulo
- Fundación Valle del Lili, Pediatric Endocrinology Service, Colombia, South America
| | - Diana Ramirez-Montaño
- Universidad Icesi, Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras, Colombia, South America,* Address for Correspondence: Universidad Icesi, Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras, Colombia, South America Phone: +57 5552334 ext 4035 E-mail:
| | - Laura Torres-Canchala
- Fundación Valle del Lili, Centro de Investigaciones Clínicas, Colombia, South America
| | - Ximena García
- Universidad Icesi, Facultad de Ciencias de La Salud, Colombia, South America
| | - Rodrigo Lemus
- Universidad Icesi, Facultad de Ciencias de La Salud, Colombia, South America
| | - Ana M. Aristizabal
- Universidad Icesi, Facultad de Ciencias de La Salud, Colombia, South America
| | | | - Diana M. Dávalos
- Universidad Icesi, Facultad de Ciencias de La Salud, Colombia, South America
| | - Lorena Diaz-Ordoñez
- Universidad Icesi, Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras, Colombia, South America
| | - Harry Pachajoa
- Universidad Icesi, Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras, Colombia, South America,Fundación Valle del Lili, Genetics Service, Colombia, South America
| |
Collapse
|
17
|
Werdermann M, Berger I, Scriba LD, Santambrogio A, Schlinkert P, Brendel H, Morawietz H, Schedl A, Peitzsch M, King AJF, Andoniadou CL, Bornstein SR, Steenblock C. Insulin and obesity transform hypothalamic-pituitary-adrenal axis stemness and function in a hyperactive state. Mol Metab 2020; 43:101112. [PMID: 33157254 PMCID: PMC7691554 DOI: 10.1016/j.molmet.2020.101112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/22/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Metabolic diseases are an increasing problem in society with the brain-metabolic axis as a master regulator of the human body for sustaining homeostasis under metabolic stress. However, metabolic inflammation and disease will trigger sustained activation of the hypothalamic-pituitary-adrenal axis. In this study, we investigated the role of metabolic stress on progenitor cells in the hypothalamic-pituitary-adrenal axis. Methods In vitro, we applied insulin and leptin to murine progenitor cells isolated from the pituitary and adrenal cortex and examined the role of these hormones on proliferation and differentiation. In vivo, we investigated two different mouse models of metabolic disease, obesity in leptin-deficient ob/ob mice and obesity achieved via feeding with a high-fat diet. Results Insulin was shown to lead to enhanced proliferation and differentiation of both pituitary and adrenocortical progenitors. No alterations in the progenitors were noted in our chronic metabolic stress models. However, hyperactivation of the hypothalamic-pituitary-adrenal axis was observed and the expression of the appetite-regulating genes Npy and Agrp changed in both the hypothalamus and adrenal. Conclusions It is well-known that chronic stress and stress hormones such as glucocorticoids can induce metabolic changes including obesity and diabetes. In this article, we show for the first time that this might be based on an early sensitization of stem cells of the hypothalamic-pituitary-adrenal axis. Thus, pituitary and adrenal progenitor cells exposed to high levels of insulin are metabolically primed to a hyper-functional state leading to enhanced hormone production. Likewise, obese animals exhibit a hyperactive hypothalamic-pituitary-adrenal axis leading to adrenal hyperplasia. This might explain how stress in early life can increase the risk for developing metabolic syndrome in adulthood. Insulin enhances proliferation and differentiation of adrenocortical and pituitary progenitors. Obesity leads to hyperactivation and priming of the HPA axis. Obesity leads to overexpression of appetite-regulating genes in the hypothalamus. Obesity leads to a decrease in the expression of appetite-regulating genes in the adrenal gland.
Collapse
Affiliation(s)
- Martin Werdermann
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Ilona Berger
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Laura D Scriba
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Alice Santambrogio
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany; Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, SE1 9RT, UK.
| | - Pia Schlinkert
- Department of Pharmacology and Toxicology, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Heike Brendel
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Andreas Schedl
- University of Côte d'Azur, INSERM, CNRS, iBV, Parc Valrose, Nice, 06108, France.
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Aileen J F King
- Department of Diabetes, School of Life Course Sciences, King's College London, Great Maze Pond, London, SE1 9RT, UK.
| | - Cynthia L Andoniadou
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany; Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, SE1 9RT, UK.
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany; Diabetes and Nutritional Sciences Division, King's College London, Guy's Campus, London, SE1 1UL, UK.
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| |
Collapse
|
18
|
Kennedy EM, Hermetz K, Burt A, Everson TM, Deyssenroth M, Hao K, Chen J, Karagas MR, Pei D, Koestler DC, Marsit CJ. Placental microRNA expression associates with birthweight through control of adipokines: results from two independent cohorts. Epigenetics 2020; 16:770-782. [PMID: 33016211 DOI: 10.1080/15592294.2020.1827704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs are non-coding RNAs that regulate gene expression post-transcriptionally. In the placenta, the master regulator of foetal growth and development, microRNAs shape the basic processes of trophoblast biology and specific microRNA have been associated with foetal growth. To comprehensively assess the role of microRNAs in placental function and foetal development, we have performed small RNA sequencing to profile placental microRNAs from two independent mother-infant cohorts: the Rhode Island Child Health Study (n = 225) and the New Hampshire Birth Cohort Study (n = 317). We modelled microRNA counts on infant birthweight percentile (BWP) in each cohort, while accounting for race, sex, parity, and technical factors, using negative binomial generalized linear models. We identified microRNAs that were differentially expressed (DEmiRs) with BWP at false discovery rate (FDR) less than 0.05 in both cohorts. hsa-miR-532-5p (miR-532) was positively associated with BWP in both cohorts. By integrating parallel whole transcriptome and small RNA sequencing in the RICHS cohort, we identified putative targets of miR-532. These targets are enriched for pathways involved in adipogenesis, adipocytokine signalling, energy metabolism, and hypoxia response, and included Leptin, which we further demonstrated to have a decreasing expression with increasing BWP, particularly in male infants. Overall, we have shown a robust and reproducible association of miR-532 with BWP, which could influence BWP through regulation of adipocytokines Leptin and Adiponectin.
Collapse
Affiliation(s)
- Elizabeth M Kennedy
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Karen Hermetz
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Todd M Everson
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ke Hao
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA.,Dartmouth College, Children's Environmental Health and Disease Prevention Research Center at Dartmouth, Lebanon, NH, USA
| | - Dong Pei
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
19
|
Epigenetic signatures associated with maternal body mass index or gestational weight gain: a systematic review. J Dev Orig Health Dis 2020; 12:373-383. [PMID: 32873364 DOI: 10.1017/s2040174420000811] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal body mass index (BMI) and gestational weight gain (GWG) impacts both the mother's and the child's health, and epigenetic modifications have been suggested to mediate some of these effects in offspring. This systematic review aimed to summarize the current literature on associations between maternal BMI and GWG and epigenetic marks. We performed systematic searches in PubMed and EMBASE and manual searches of reference lists. We included 49 studies exploring the association between maternal BMI and/or GWG and DNA methylation or miRNA; 7 performed in maternal tissues, 13 in placental tissue and 38 in different offspring tissues. The most consistent findings were reported for the relationship between maternal BMI, in particular pre-pregnant BMI, and expression of miRNA Let-7d in both maternal blood and placental tissue, methylation of the gene HIF3A in umbilical cord blood and umbilical tissue, and with expression in the miR-210 target gene, BDNF in placental tissue and cord blood. Correspondingly, methylation of BDNF was also found in placental tissue and cord blood. The current evidence suggests that maternal BMI is associated with some epigenetic signatures in the mother, the placenta and her offspring, which could indicate that some of the effects proposed by the Developmental Origins of Health and Disease-hypothesis may be mediated by epigenetic marks. In conclusion, there is a need for large, well-designed studies and meta-analyses that can clarify the relationship between BMI, GWG and epigenetic changes.
Collapse
|
20
|
Precision Nutrition and Childhood Obesity: A Scoping Review. Metabolites 2020; 10:metabo10060235. [PMID: 32521722 PMCID: PMC7345802 DOI: 10.3390/metabo10060235] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/18/2020] [Accepted: 06/02/2020] [Indexed: 01/01/2023] Open
Abstract
Environmental exposures such as nutrition during life stages with high developmental plasticity—in particular, the in utero period, infancy, childhood, and puberty—may have long-lasting influences on risk of chronic diseases, including obesity-related conditions that manifest as early as childhood. Yet, specific mechanisms underlying these relationships remain unclear. Here, we consider the study of ‘omics mechanisms, including nutrigenomics, epigenetics/epigenomics, and metabolomics, within a life course epidemiological framework to accomplish three objectives. First, we carried out a scoping review of population-based literature with a focus on studies that include ‘omics analyses during three sensitive periods during early life: in utero, infancy, and childhood. We elected to conduct a scoping review because the application of multi-‘omics and/or precision nutrition in childhood obesity prevention and treatment is relatively recent, and identifying knowledge gaps can expedite future research. Second, concomitant with the literature review, we discuss the relevance and plausibility of biological mechanisms that may underlie early origins of childhood obesity identified by studies to date. Finally, we identify current research limitations and future opportunities for application of multi-‘omics in precision nutrition/health practice.
Collapse
|
21
|
Kochhar P, Manikandan C, Ravikumar G, Dwarkanath P, Sheela CN, George S, Thomas A, Crasta J, Thomas T, Kurpad AV, Mukhopadhyay A. Placental expression of leptin: fetal sex-independent relation with human placental growth. Eur J Clin Nutr 2020; 74:1603-1612. [PMID: 32382074 DOI: 10.1038/s41430-020-0649-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Leptin (LEP) is a vital placental hormone that is known to affect different aspects of placental function and fetal development. The present study aimed to determine the association of placental LEP transcript abundance with maternal, placental, and newborn parameters. SUBJECTS/METHODS In this retrospective case-control study, placental samples (n = 105) were collected from small (SGA) and appropriate (AGA) for gestational age full-term singleton pregnancies (n = 44 SGA and n = 61 AGA). Placental transcript abundance of LEP was assessed by real-time quantitative PCR after normalization to a reference gene panel. LEP methylation was measured using a quantitative MethyLight assay in a subset of samples (n = 54). RESULTS Placental LEP transcript abundance was negatively and significantly associated with placental weight (β = -3.883, P = 0.015). This association continued to be significant in the SGA group (β = -10.332, P = 0.001), both in female (β = -15.423, P = 0.021) and male births (β = -10.029, P = 0.007). LEP transcript abundance was not associated with LEP methylation levels (Spearman's ρ = 0.148, P = 0.287). CONCLUSION We conclude that placental upregulation of LEP is an integral and fetal sex-independent component of placental growth restriction, which can be potentially targeted through maternal dietary modifications to improve fetoplacental growth.
Collapse
Affiliation(s)
- P Kochhar
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - C Manikandan
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India.,School of Biosciences and Technology; Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| | - G Ravikumar
- Department of Pathology, St John's Medical College Hospital, Bangalore, India
| | - P Dwarkanath
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - C N Sheela
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - S George
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - A Thomas
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - J Crasta
- Department of Pathology, St John's Medical College Hospital, Bangalore, India
| | - T Thomas
- Department of Biostatistics, St. John's Medical College Hospital, Bangalore, India
| | - A V Kurpad
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - A Mukhopadhyay
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India.
| |
Collapse
|
22
|
Echiburú B, Milagro F, Crisosto N, Pérez-Bravo F, Flores C, Arpón A, Salas-Pérez F, Recabarren SE, Sir-Petermann T, Maliqueo M. DNA methylation in promoter regions of genes involved in the reproductive and metabolic function of children born to women with PCOS. Epigenetics 2020; 15:1178-1194. [PMID: 32283997 DOI: 10.1080/15592294.2020.1754674] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Clinical and experimental evidences indicate that epigenetic modifications induced by the prenatal environment are related to metabolic and reproductive derangements in polycystic ovary syndrome (PCOS). Alterations in the leptin and adiponectin systems, androgen signalling and antimüllerian hormone (AMH) levels have been observed in PCOS women and in their offspring. Using a targeted Next-Generation Sequencing (NGS), we studied DNA methylation in promoter regions of the leptin (LEP), leptin receptor (LEPR), adiponectin (ADIPOQ), adiponectin receptor 1 and 2 (ADIPOR1 and ADIPOR2), AMH and androgen receptor (AR) genes in 24 sons and daughters of women with PCOS (12 treated with metformin during pregnancy) and 24 children born to non-PCOS women during early infancy (2-3 months of age). Genomic DNA was extracted from whole blood, bisulphite converted and sequenced by NGS. Girls showed differences between groups in 1 CpG site of LEPR, 2 of LEP, 1 of ADIPOR2 and 2 of AR. Boys showed differences in 5 CpG sites of LEP, 3 of AMH and 9 of AR. Maternal metformin treatment prevented some of these changes in LEP, ADIPOR2 and partially in AR in girls, and in LEP and AMH in boys. Maternal BMI at early pregnancy was inversely correlated with the methylation levels of the ChrX-67544981 site in the whole group of girls (r = -0.530, p = 0.008) and with the global Z-score in all boys (r = -0.539, p = 0.007). These data indicate that the intrauterine PCOS environment predisposes the offspring to acquire certain sex-dependent DNA methylation patterns in the promoter regions of metabolic and reproductive genes.
Collapse
Affiliation(s)
- Bárbara Echiburú
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| | - Fermín Milagro
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra , Pamplona, Spain.,Centro De Investigación Biomédica En Red Fisiopatología De La Obesidad Y Nutrición (Ciberobn), Instituto De Salud Carlos III , Madrid, Spain
| | - Nicolás Crisosto
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile.,Unit of Endocrinology, Clínica Las , Santiago, Chile
| | - Francisco Pérez-Bravo
- Laboratory of Nutritional Genomics, Department of Nutrition, Faculty of Medicine, University of Chile , Santiago, Chile
| | - Cristian Flores
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| | - Ana Arpón
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra , Pamplona, Spain
| | - Francisca Salas-Pérez
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra , Pamplona, Spain
| | - Sergio E Recabarren
- Laboratory of Animal Physiology and Endocrinology, Department of Animal Science, Faculty of Veterinary Sciences, University of Concepcion , Chillán, Chile
| | - Teresa Sir-Petermann
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| | - Manuel Maliqueo
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile , Santiago, Chile
| |
Collapse
|
23
|
Daniels TE, Sadovnikoff AI, Ridout KK, Lesseur C, Marsit CJ, Tyrka AR. Associations of maternal diet and placenta leptin methylation. Mol Cell Endocrinol 2020; 505:110739. [PMID: 32004678 PMCID: PMC7185035 DOI: 10.1016/j.mce.2020.110739] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Maternal diet is an important factor in prenatal development that also has implications for disease risk later in life. The adipokine leptin is a key regulator of energy homeostasis and may be involved in the association between maternal nutrition, maternal obesity, and infant outcomes. DNA methylation of placenta genes may occur in response to exposures and may program subsequent infant development. This study examined maternal diet, placenta leptin gene DNA methylation, and neonatal growth in a sample of healthy neonates and their mothers. METHODS Mothers and their healthy neonates (N = 135) were recruited within 1-2 days following delivery at Women and Infants Hospital in Providence, RI. A structured interview was conducted to assess maternal dietary intake. Maternal pre-pregnancy weight, weight gain during pregnancy, maternal health, medications, and vitamin use were obtained from medical records. Bisulfite pyrosequencing was used to measure methylation of CpG sites in the promoter region of the placenta leptin gene and determine genotype of the leptin single nucleotide polymorphism (SNP) rs2167270, which is known to influence leptin methylation. Bivariate analyses and linear regression models were used to evaluate associations of demographics, diet, and mean leptin methylation. RESULTS Genotype was a significant predictor of placenta leptin DNA methylation (p < .01), and after controlling for this and other relevant maternal and infant covariates, lower levels of leptin methylation were significantly associated with greater intake of carbohydrates (p < .05), in particular added sugars (p < .05) and white/refined carbohydrates (p < .05). Total caloric intake was also associated with placenta leptin methylation (p < .05), however after controlling for relevant covariates, significance diminished to trend-level. There were no significant associations of placenta leptin methylation and intake of protein (p > .05) or fat (p > .05). CONCLUSION These findings underline the importance of intake of carbohydrate consumption for methylation of the placenta leptin gene. Because methylation reduces gene transcription, lower methylation may indicate a placenta response to high caloric intake and carbohydrate food that would result in higher levels of this hormone during fetal development. Further investigation of the developmental ramifications of epigenetic changes to placenta leptin methylation should be pursued.
Collapse
Affiliation(s)
- Teresa E Daniels
- Mood Disorders Research Program and Laboratory for Clinical and Translational Neuroscience, Butler Hospital, Providence, RI, USA; Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, USA.
| | - Alexander I Sadovnikoff
- Mood Disorders Research Program and Laboratory for Clinical and Translational Neuroscience, Butler Hospital, Providence, RI, USA
| | - Kathryn K Ridout
- Department of Psychiatry and Family Medicine, Kaiser Permanente, San Jose, CA, USA
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, USA
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, USA
| | - Audrey R Tyrka
- Mood Disorders Research Program and Laboratory for Clinical and Translational Neuroscience, Butler Hospital, Providence, RI, USA; Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, USA
| |
Collapse
|
24
|
Methylation of the LEP gene promoter in blood at 12 months and BMI at 4 years of age-a population-based cohort study. Int J Obes (Lond) 2020; 44:842-847. [PMID: 32107462 DOI: 10.1038/s41366-020-0553-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
Increasing evidence links epigenetic variation to anthropometric and metabolic measures. Leptin signalling regulates appetite and energy expenditure, and in pregnancy is important for nutrient supply to the foetus. Maternal metabolic health and foetal growth are linked to infant blood leptin gene (LEP) methylation, which has been cross-sectionally associated with adolescent obesity. Despite this, few studies have explored the relationship between infant LEP methylation and childhood anthropometry, or the impact of genetic variation on these relationships. Using a prospective birth cohort, we investigated whether blood LEP promoter methylation at birth and 12 months predicts weight and adiposity at 4-years. Locus-specific methylation data was analysed by partial correlation tests and multivariable linear regression. There was weak evidence of an association of birth LEP methylation with anthropometry measures at 4 years. Methylation at a specific site (cg19594666) at 12 months was inversely associated with 4-year weight (r = -0.11, p = 0.02) and body-mass index (BMI) (r = -0.13, p = 0.007), which persisted following adjustment for weight at birth and at 12 months. Neither association was influenced by genotype. We report the first evidence of an association between LEP methylation in infancy and childhood weight. Replication in additional cohorts is required to determine if this relationship persists.
Collapse
|
25
|
Narapareddy L, Wildman DE, Armstrong DL, Weckle A, Bell AF, Patil CL, Tardif SD, Ross CN, Rutherford JN. Maternal weight affects placental DNA methylation of genes involved in metabolic pathways in the common marmoset monkey (Callithrix jacchus). Am J Primatol 2020; 82:e23101. [PMID: 32020652 PMCID: PMC7154656 DOI: 10.1002/ajp.23101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/09/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022]
Abstract
Accumulating evidence suggests that dysregulation of placental DNA methylation (DNAm) is a mechanism linking maternal weight during pregnancy to metabolic programming outcomes. The common marmoset, Callithrix jaccus, is a platyrrhine primate species that has provided much insight into studies of the primate placenta, maternal condition, and metabolic programming, yet the relationships between maternal weight and placental DNAm are unknown. Here, we report genome‐wide DNAm from term marmoset placentas using reduced representation bisulfite sequencing. We identified 74 genes whose DNAm pattern is associated with maternal weight during gestation. These genes are predominantly involved in energy metabolism and homeostasis, including the regulation of glycolytic and lipid metabolic processes pathways. The placental DNA methylation (DNAm) landscape of the marmoset placenta presents unique differences and similarities with human placental methylation patterns. Maternal weight is associated with placental DNAm in genes that are predominantly involved in energy metabolism and homeostasis. The impact of altered placental DNAm on placental function and development may also contribute to the potential role of placental DNAm in developmental programming in the marmoset monkey.
Collapse
Affiliation(s)
- Laren Narapareddy
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, Georgia
| | - Derek E Wildman
- Genomics Program, College of Public Health, University of South Florida, Tampa, Florida
| | - Don L Armstrong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Amy Weckle
- Illinois Water Resources Center, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Aleeca F Bell
- Department of Women, Children and Family Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
| | - Crystal L Patil
- Department of Women, Children and Family Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
| | - Suzette D Tardif
- Texas Biomedical Research Institute, Southwest National Primate Research Center, San Antonio, Texas
| | - Corinna N Ross
- Program of Biology, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, Texas
| | - Julienne N Rutherford
- Department of Women, Children and Family Health Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
26
|
Stolzenbach F, Valdivia S, Ojeda-Provoste P, Toledo F, Sobrevia L, Kerr B. DNA methylation changes in genes coding for leptin and insulin receptors during metabolic-altered pregnancies. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165465. [DOI: 10.1016/j.bbadis.2019.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/19/2019] [Accepted: 05/02/2019] [Indexed: 01/07/2023]
|
27
|
Mediation Analysis Supports a Causal Relationship between Maternal Hyperglycemia and Placental DNA Methylation Variations at the Leptin Gene Locus and Cord Blood Leptin Levels. Int J Mol Sci 2020; 21:ijms21010329. [PMID: 31947745 PMCID: PMC6982090 DOI: 10.3390/ijms21010329] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/18/2019] [Accepted: 12/31/2019] [Indexed: 12/02/2022] Open
Abstract
Changes in fetal DNA methylation (DNAm) of the leptin (LEP) gene have been associated with exposure to maternal hyperglycemia, but their links with childhood obesity risk are still unclear. We investigated the association between maternal hyperglycemia, placental LEP DNAm (25 5′-C-phosphate-G-3′ (CpG) sites), neonatal leptinemia, and adiposity (i.e., BMI and skinfold thickness (ST) (subscapular (SS) + triceps (TR) skinfold measures, and the ratio of SS:TR) at 3-years-old, in 259 mother–child dyads, from Gen3G birth cohort. We conducted multivariate linear analyses adjusted for gestational age at birth, sex of the child, age at follow-up, and cellular heterogeneity. We assessed the causal role of DNAm in the association between maternal glycemia and childhood outcomes, using mediation analysis. We found three CpGs associated with neonatal leptinemia (p ≤ 0.002). Of these, cg05136031 and cg15758240 were also associated with BMI (β = −2.69, p = 0.05) and fat distribution (β = −0.581, p = 0.05) at 3-years-old, respectively. Maternal glycemia was associated with DNAm at cg15758240 (β = −0.01, p = 0.04) and neonatal leptinemia (β = 0.19, p = 0.004). DNAm levels at cg15758240 mediates 0.8% of the association between maternal glycemia and neonatal leptinemia (p < 0.001). Our results support that DNAm regulation of the leptin pathway in response to maternal glycemia might be involved in programming adiposity in childhood.
Collapse
|
28
|
Interaction of Osteoarthritis and BMI on Leptin Promoter Methylation in Taiwanese Adults. Int J Mol Sci 2019; 21:ijms21010123. [PMID: 31878053 PMCID: PMC6981657 DOI: 10.3390/ijms21010123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/12/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Leptin (LEP) regulates glucose metabolism and energy storage in the body. Osteoarthritis (OA) is associated with the upregulation of serum LEP. LEP promoter methylation is associated with obesity. So far, few studies have explored the association of BMI and OA with LEP methylation. We assessed the interaction between body mass index (BMI) and OA on LEP promoter methylation. Data of 1114 participants comprising 583 men and 558 women, aged 30–70 years were retrieved from the Taiwan Biobank Database (2008–2015). Osteoarthritis was self-reported and cases were those who reported having ever been clinically diagnosed with osteoarthritis. BMI was categorized into underweight, normal weight, overweight, and obesity. The mean LEP promoter methylation level in individuals with osteoarthritis was 0.5509 ± 0.00437 and 0.5375 ± 0.00101 in those without osteoarthritis. The interaction between osteoarthritis and BMI on LEP promoter methylation was significant (p-value = 0.0180). With normal BMI as the reference, the mean LEP promoter methylation level was significantly higher in obese osteoarthritic individuals (β = 0.03696, p-value = 0.0187). However, there was no significant association between BMI and LEP promoter methylation in individuals without osteoarthritis, regardless of BMI. In conclusion, only obesity was significantly associated with LEP promoter methylation (higher levels) specifically in osteoarthritic patients.
Collapse
|
29
|
Genetic variation, intrauterine growth, and adverse pregnancy conditions predict leptin gene DNA methylation in blood at birth and 12 months of age. Int J Obes (Lond) 2019; 44:45-56. [PMID: 31636377 DOI: 10.1038/s41366-019-0472-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Leptin regulates satiety and energy homoeostasis, and plays a key role in placentation in pregnancy. Previous studies have demonstrated regulation of leptin gene (LEP) expression and/or methylation in placenta and cord blood in association with early life exposures, but most have been small and have not considered the influence of genetic variation. Here, we investigated the relationship between maternal factors in pregnancy, infant anthropometry and LEP genetic variation with LEP promoter methylation at birth and 12 months of age. METHODS LEP methylation was measured in cord (n = 877) and 12-month (n = 734) blood in the Barwon Infant Study, a population-based pre-birth cohort. Infant adiposity at birth and 12-months was measured as triceps and subscapular skinfold thickness. Cross-sectional regression tested associations of methylation with pregnancy and anthropometry measures, while longitudinal regression tested if birth anthropometry predicted 12-month LEP methylation levels. RESULTS Male infants had lower LEP methylation in cord blood (-2.07% average methylation, 95% CI (-2.92, -1.22), p < 0.001). Genetic variation strongly influenced DNA methylation at a single CpG site, which was also negatively associated with birth weight (r = -0.10, p = 0.003). Pre-eclampsia was associated with lower cord blood methylation at another CpG site (-6.06%, 95% CI (-10.70, -1.42), p = 0.01). Gestational diabetes was more modestly associated with methylation at two other CpG units. Adiposity at birth was associated with 12-month LEP methylation, modified by rs41457646 genotype. There was no association of LEP methylation with 12-month anthropometric measures. CONCLUSIONS Infant sex, weight, genetic variation, and exposure to pre-eclampsia and gestational diabetes, are associated with LEP methylation in cord blood. Infant adiposity at birth predicts 12-month blood LEP methylation in a genotype-dependent manner. These findings are consistent with genetics and anthropometry driving altered LEP epigenetic profile and expression in infancy. Further work is required to confirm this and to determine the long-term impact of altered LEP methylation on health.
Collapse
|
30
|
Karakosta P, Margetaki K, Fthenou E, Kampouri M, Kyriklaki A, Koutra K, Chalkiadaki G, Roumeliotaki T, Vafeiadi M, Kogevinas M, Mantzoros C, Chatzi L. Cord Leptin is Associated with Neuropsychomotor Development in Childhood. Obesity (Silver Spring) 2019; 27:1693-1702. [PMID: 31479200 PMCID: PMC6756960 DOI: 10.1002/oby.22571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/06/2019] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Leptin is critical for central nervous system development and maturation. This study aimed to evaluate the potential regulatory role of cord leptin in the neuropsychomotor development of children ages 18 months to 6 years. METHODS This study included 424 children from a prospective mother-child cohort (Rhea Study; Crete, Greece) with available cord leptin levels and data on neurodevelopmental outcomes at 18 months (Bayley Scales of Infant and Toddler Development, Third Edition), 4 years (McCarthy Scales of Children's Abilities), and 6 years (Raven's Coloured Progressive Matrices and Trail Making Test). Multivariable linear regression models were used to explore the associations. RESULTS Each 10-ng/mL increase in the cord leptin level was associated with increased scores on the gross motor scale at 18 months (β coefficient: 3.8; 95% CI: 0.0-7.5), with decreased scores in the general cognitive performance (β coefficient: -3.0; 95% CI: -5.5 to -0.4), perceptual performance (β coefficient: -3.4; 95% CI: -6.0 to -9.9), working memory (β coefficient: -3.1; 95% CI: -5.7 to -0.4), executive function (β coefficient -3.1; 95% CI: -5.7 to -0.5), and functions of the posterior cortex (β coefficient: -2.7; 95% CI: -5.2 to -0.1) scales at 4 years, and with a 3.7-unit decrease in the Raven's Coloured Progressive Matrices score at 6 years (β coefficient: -3.7; 95% CI: -6.9 to -0.5). CONCLUSIONS Increased cord leptin levels are associated with enhanced gross motor development at 18 months but decreased cognitive performance in early and middle childhood.
Collapse
Affiliation(s)
- Polyxeni Karakosta
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
- Clinical Microbiology Laboratory, Attikon University
Hospital, National and Kapodistrian University of Athens, Greece
| | - Katerina Margetaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Eleni Fthenou
- Qatar Biobank for Medical Research, Qatar Foundation for
Education, Science and Community, Doha, Doha, Qatar
| | - Mariza Kampouri
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Andriani Kyriklaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Katerina Koutra
- Department of Psychology, School of Social Sciences,
University of Crete, Rethymnon, Greece
| | - Georgia Chalkiadaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Theano Roumeliotaki
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Marina Vafeiadi
- Department of Social Medicine, School of Medicine,
University of Crete, Heraklion, Greece
| | - Manolis Kogevinas
- ISGlobal, Centre for Research in Environmental
Epidemiology (CREAL), Barcelona, Spain
| | - Christos Mantzoros
- Division of Endocrinology, Diabetes, and Metabolism,
Department of Medicine, Beth Israel, Deaconess Medical Center, Harvard Medical
School, Boston, Massachusetts, United States
| | - Lida Chatzi
- Department of Preventive Medicine, University of Southern
California, Los Angeles, CA, United States
| |
Collapse
|
31
|
Wang YH, Xu XX, Sun H, Han Y, Lei ZF, Wang YC, Yan HT, Yang XJ. Cord blood leptin DNA methylation levels are associated with macrosomia during normal pregnancy. Pediatr Res 2019; 86:305-310. [PMID: 31117117 DOI: 10.1038/s41390-019-0435-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND We previously demonstrated an association between placental leptin (LEP) methylation levels and macrosomia without gestational diabetes mellitus (non-GDM). This study further explored the association between LEP methylation in cord blood and non-GDM macrosomia. METHOD We carried out a case-control study of 61 newborns with macrosomia (birth weight ≥4000 g) and 69 newborns with normal birth weight (2500-3999 g). Methylation in the LEP promoter region was mapped by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. RESULTS Average cord blood LEP methylation levels were lower in macrosomia newborns than in control newborns (P < 0.001). Eleven CpG sites were associated with macrosomia. Multivariate logistic regression revealed that low LEP methylation levels [adjusted odds ratio (AOR) = 2.84, 95% confidence interval (CI): 1.72-4.17], high pre-pregnancy body mass index (AOR = 7.44, 95% CI: 1.99-27.75), long gestational age (AOR = 3.18, 95% CI: 1.74-5.79), high cord blood LEP concentration (AOR = 2.25, 95% CI: 1.34-3.77), and male newborn gender (AOR = 3.91, 95% CI: 1.31-11.69) significantly increased the risk of macrosomia. CONCLUSIONS Lower cord blood LEP methylation levels and certain maternal and fetal factors are associated with non-GDM macrosomia.
Collapse
Affiliation(s)
- Yu-Huan Wang
- Department of Obstetrics, Second Affiliated Hospital of Wenzhou Medical University, 325027, Wenzhou, Zhejiang, P.R. China
| | - Xiao-Xi Xu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, University Town, 325035, Wenzhou, Zhejiang, P.R. China
| | - Hao Sun
- Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University, 310000, Hangzhou, P.R. China
| | - Ying Han
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, University Town, 325035, Wenzhou, Zhejiang, P.R. China
| | - Zong-Feng Lei
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, University Town, 325035, Wenzhou, Zhejiang, P.R. China
| | - Yao-Cheng Wang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, University Town, 325035, Wenzhou, Zhejiang, P.R. China
| | - Hong-Tao Yan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, University Town, 325035, Wenzhou, Zhejiang, P.R. China
| | - Xin-Jun Yang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, University Town, 325035, Wenzhou, Zhejiang, P.R. China.
| |
Collapse
|
32
|
Bekkering I, Leeuwerke M, Tanis JC, Schoots MH, Verkaik-Schakel RN, Plösch T, Bilardo CM, Eijsink JJH, Bos AF, Scherjon SA. Differential placental DNA methylation of VEGFA and LEP in small-for-gestational age fetuses with an abnormal cerebroplacental ratio. PLoS One 2019; 14:e0221972. [PMID: 31469872 PMCID: PMC6716778 DOI: 10.1371/journal.pone.0221972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background In Fetal Growth Restriction ‘fetal programming’ may take place via DNA methylation, which has implications for short-term and long-term health outcomes. Small-for-gestational age fetuses are considered fetal growth restricted, characterized by brain-sparing when fetal Doppler hemodynamics are abnormal, expressed as a cerebroplacental ratio (CPR) <1. We aimed to determine whether brain-sparing is associated with altered DNA methylation of selected genes. Methods We compared DNA methylation of six genes in 41 small-for-gestational age placentas with a normal or abnormal CPR. We selected EPO, HIF1A, VEGFA, LEP, PHLDA2, and DHCR24 for their role in angiogenesis, immunomodulation, and placental and fetal growth. DNA methylation was analyzed by pyrosequencing. Results Growth restricted fetuses with an abnormal CPR showed hypermethylation of the VEGFA gene at one CpG (VEGFA-309, p = .001) and an overall hypomethylation of the LEP gene, being significant at two CpGs (LEP-123, p = .049; LEP-51, p = .020). No differences in methylation were observed for the other genes. Conclusions VEGFA and LEP genes are differentially methylated in placentas of small-for-gestational age fetuses with brain-sparing. Hypermethylation of VEGFA-309 in abnormal CPR-placentas could indicate successful compensatory mechanisms. Methylation of LEP-51 is known to suppress LEP expression. Hypomethylation in small-for-gestational age placentas with abnormal CPR may result in hyperleptinemia and predispose to leptin-resistance later in life.
Collapse
Affiliation(s)
- Iris Bekkering
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Neonatology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- * E-mail:
| | - Mariëtte Leeuwerke
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jozien C. Tanis
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Neonatology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Mirthe H. Schoots
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rikst Nynke Verkaik-Schakel
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Caterina M. Bilardo
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jasper J. H. Eijsink
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arend F. Bos
- Department of Neonatology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sicco A. Scherjon
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
33
|
Wróblewski A, Strycharz J, Świderska E, Drewniak K, Drzewoski J, Szemraj J, Kasznicki J, Śliwińska A. Molecular Insight into the Interaction between Epigenetics and Leptin in Metabolic Disorders. Nutrients 2019; 11:nu11081872. [PMID: 31408957 PMCID: PMC6723573 DOI: 10.3390/nu11081872] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022] Open
Abstract
Nowadays, it is well-known that the deregulation of epigenetic machinery is a common biological event leading to the development and progression of metabolic disorders. Moreover, the expression level and actions of leptin, a vast adipocytokine regulating energy metabolism, appear to be strongly associated with epigenetics. Therefore, the aim of this review was to summarize the current knowledge of the epigenetic regulation of leptin as well as the leptin-induced epigenetic modifications in metabolic disorders and associated phenomena. The collected data indicated that the deregulation of leptin expression and secretion that occurs during the course of metabolic diseases is underlain by a variation in the level of promoter methylation, the occurrence of histone modifications, along with miRNA interference. Furthermore, leptin was proven to epigenetically regulate several miRNAs and affect the activity of the histone deacetylases. These epigenetic modifications were observed in obesity, gestational diabetes, metabolic syndrome and concerned various molecular processes like glucose metabolism, insulin sensitivity, liver fibrosis, obesity-related carcinogenesis, adipogenesis or fetal/early postnatal programming. Moreover, the circulating miRNA profiles were associated with the plasma leptin level in metabolic syndrome, and miRNAs were found to be involved in hypothalamic leptin sensitivity. In summary, the evidence suggests that leptin is both a target and a mediator of epigenetic changes that develop in numerous tissues during metabolic disorders.
Collapse
Affiliation(s)
- Adam Wróblewski
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland.
| | - Justyna Strycharz
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Ewa Świderska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Karolina Drewniak
- Student Scientific Society of the Civilization Diseases, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Jacek Kasznicki
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland.
| |
Collapse
|
34
|
Adverse Maternal Metabolic Intrauterine Environment and Placental Epigenetics: Implications for Fetal Metabolic Programming. Curr Environ Health Rep 2019; 5:531-543. [PMID: 30267228 DOI: 10.1007/s40572-018-0217-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW Herein, we summarize existent epidemiological studies relating adverse maternal metabolic environments of maternal obesity and gestational diabetes and placental DNA methylation. RECENT FINDINGS Multiple studies have evaluated associations between intrauterine exposure to gestational diabetes and/or maternal glucose levels and DNA methylation at candidate metabolic genes as well as in epigenome-wide studies. Some of the genomic regions more consistently associated include lipid-related genes (LPL and PPARGC1A), the major histocompatibility complex (MHC), and imprinted genes. Studies solely focused on maternal obesity influences on the placental epigenome are scarce. Understanding the placental mechanisms involved in fetal metabolic programming could lead to discovery of placental biomarkers at birth that predict later-life metabolic risk. Moving forward is important to standardize methods utilized in epigenetics research; consistent methodology can help interpret disparate findings. Larger studies with longitudinal follow-up are needed to address future challenges in fetal programming research.
Collapse
|
35
|
Kochmanski J, Goodrich JM, Peterson KE, Lumeng JC, Dolinoy DC. Neonatal bloodspot DNA methylation patterns are associated with childhood weight status in the Healthy Families Project. Pediatr Res 2019; 85:848-855. [PMID: 30425339 PMCID: PMC6494701 DOI: 10.1038/s41390-018-0227-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 09/19/2018] [Accepted: 10/14/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND This study measured longitudinal DNA methylation dynamics at growth-related genes during childhood, and then tested whether DNA methylation at various stages of childhood was associated with obesity status. METHODS Using neonatal bloodspot (n = 132) and matched childhood blood samples (n = 65), DNA methylation was quantified at a repetitive element (long interspersed nuclear element-1 (LINE-1)), two imprinted genes (IGF2, H19), and four non-imprinted genes (LEP, PPARA, ESR1, SREBF1) related to growth and adiposity. Logistic regression was used to test whether neonatal bloodspot DNA methylation at target genes was associated with log odds of obesity (Y/N) in children recruited from three age groups-12-24 months old (n = 40), 3-5 years of age (n = 40), and 10-12 years of age (n = 52). RESULTS In 3-5 year olds, neonatal bloodspot LINE-1 methylation was negatively associated with obesity (log odds = -0.40, p = 0.04). Across childhood age group in matched blood samples, DNA methylation levels in blood decreased (p < 0.05) at LINE-1, PPARA, ESR1, SREBF1, IGF2, and H19, and increased (p < 0.05) at LEP. CONCLUSIONS Our results suggest that age-related epigenetic changes occur at growth-related genes in the first decade of life, and that gene-specific neonatal bloodspot DNA methylation may be a useful biomarker of obesity likelihood during childhood.
Collapse
Affiliation(s)
- Joseph Kochmanski
- Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jaclyn M Goodrich
- Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Karen E Peterson
- Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Julie C Lumeng
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
- Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
36
|
James P, Sajjadi S, Tomar AS, Saffari A, Fall CHD, Prentice AM, Shrestha S, Issarapu P, Yadav DK, Kaur L, Lillycrop K, Silver M, Chandak GR. Candidate genes linking maternal nutrient exposure to offspring health via DNA methylation: a review of existing evidence in humans with specific focus on one-carbon metabolism. Int J Epidemiol 2018; 47:1910-1937. [PMID: 30137462 PMCID: PMC6280938 DOI: 10.1093/ije/dyy153] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Background Mounting evidence suggests that nutritional exposures during pregnancy influence the fetal epigenome, and that these epigenetic changes can persist postnatally, with implications for disease risk across the life course. Methods We review human intergenerational studies using a three-part search strategy. Search 1 investigates associations between preconceptional or pregnancy nutritional exposures, focusing on one-carbon metabolism, and offspring DNA methylation. Search 2 considers associations between offspring DNA methylation at genes found in the first search and growth-related, cardiometabolic and cognitive outcomes. Search 3 isolates those studies explicitly linking maternal nutritional exposure to offspring phenotype via DNA methylation. Finally, we compile all candidate genes and regions of interest identified in the searches and describe their genomic locations, annotations and coverage on the Illumina Infinium Methylation beadchip arrays. Results We summarize findings from the 34 studies found in the first search, the 31 studies found in the second search and the eight studies found in the third search. We provide details of all regions of interest within 45 genes captured by this review. Conclusions Many studies have investigated imprinted genes as priority loci, but with the adoption of microarray-based platforms other candidate genes and gene classes are now emerging. Despite a wealth of information, the current literature is characterized by heterogeneous exposures and outcomes, and mostly comprise observational associations that are frequently underpowered. The synthesis of current knowledge provided by this review identifies research needs on the pathway to developing possible early life interventions to optimize lifelong health.
Collapse
Affiliation(s)
- Philip James
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, London, UK
| | - Sara Sajjadi
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Ashutosh Singh Tomar
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Ayden Saffari
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, London, UK
| | - Caroline H D Fall
- MRC Life course Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Andrew M Prentice
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, London, UK
| | - Smeeta Shrestha
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore, India
| | - Prachand Issarapu
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Dilip Kumar Yadav
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Lovejeet Kaur
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Karen Lillycrop
- Research Centre for Biological Sciences, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Matt Silver
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, London, UK
| | - Giriraj R Chandak
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
37
|
Tian FY, Rifas-Shiman SL, Cardenas A, Baccarelli AA, DeMeo DL, Litonjua AA, Rich-Edwards JW, Gillman MW, Oken E, Hivert MF. Maternal corticotropin-releasing hormone is associated with LEP DNA methylation at birth and in childhood: an epigenome-wide study in Project Viva. Int J Obes (Lond) 2018; 43:1244-1255. [PMID: 30464231 PMCID: PMC6529291 DOI: 10.1038/s41366-018-0249-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/22/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
Background: Corticotropin-releasing hormone (CRH) plays a central role in regulating the secretion of cortisol which controls a wide range of biological processes. Fetuses overexposed to cortisol have increased risks of disease in later life. DNA methylation may be the underlying association between prenatal cortisol exposure and health effects. We investigated associations between maternal CRH levels and epigenome-wide DNA methylation of cord blood in offsprings and evaluated whether these associations persisted into mid-childhood. Methods: We investigated mother-child pairs enrolled in the prospective Project Viva pre-birth cohort. We measured DNA methylation in 257 umbilical cord blood samples using the HumanMethylation450 Bead Chip. We tested associations of maternal CRH concentration with cord blood cells DNA methylation, adjusting the model for maternal age at enrollment, education, maternal race/ethnicity, pre-pregnancy body mass index, parity, gestational age at delivery, child sex, and cell-type composition in cord blood. We further examined the persistence of associations between maternal CRH levels and DNA methylation in children’s blood cells collected at mid-childhood (N = 239, age: 6.7–10.3 years) additionally adjusting for the children’s age at blood drawn. Results: Maternal CRH levels are associated with DNA methylation variability in cord blood cells at 96 individual CpG sites (False Discovery Rate < 0.05). Among the 96 CpG sites, we identified 3 CpGs located near the LEP gene. Regional analyses confirmed the association between maternal CRH and DNA methylation near LEP. Moreover, higher maternal CRH levels were associated with higher blood-cell DNA methylation of the promoter region of LEP in mid-childhood (P < 0.05, β = 0.64, SE = 0.30). Conclusion: In our cohort, maternal CRH was associated with DNA methylation levels in newborns at multiple loci, notably in the LEP gene promoter. The association between maternal CRH and LEP DNA methylation levels persisted into mid-childhood.
Collapse
Affiliation(s)
- Fu-Ying Tian
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA.,Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Andres Cardenas
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Augusto A Litonjua
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Janet W Rich-Edwards
- Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Matthew W Gillman
- Environmental Influences on Child Health Outcomes (ECHO) Office of the Director, National Institutes of Health, Department of Health and Human Services, Rockville, MD, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA. .,Diabetes Research Center, Massachusetts General Hospital, 50 Staniford Street, Boston, MA, USA. .,Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada. .,Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
38
|
Deyssenroth MA, Gennings C, Liu SH, Peng S, Hao K, Lambertini L, Jackson BP, Karagas MR, Marsit CJ, Chen J. Intrauterine multi-metal exposure is associated with reduced fetal growth through modulation of the placental gene network. ENVIRONMENT INTERNATIONAL 2018; 120:373-381. [PMID: 30125854 PMCID: PMC6288802 DOI: 10.1016/j.envint.2018.08.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 05/20/2023]
Abstract
BACKGROUND Intrauterine metal exposures and aberrations in placental processes are known contributors to being born small for gestational age (SGA). However, studies to date have largely focused on independent effects, failing to account for potential interdependence among these markers. OBJECTIVES We evaluated the inter-relationship between multi-metal indices and placental gene network modules related to SGA status to highlight potential molecular pathways through which in utero multi-metal exposure impacts fetal growth. METHODS Weighted quantile sum (WQS) regression was performed using a panel of 16 trace metals measured in post-partum maternal toe nails collected from the Rhode Island Child Health Study (RICHS, n = 195), and confirmation of the derived SGA-related multi-metal index was conducted using Bayesian kernel machine regression (BKMR). We leveraged existing placental weighted gene coexpression network data to examine associations between the SGA multi-metal index and placental gene expression. Expression of select genes were assessed using RT-PCR in an independent birth cohort, the New Hampshire Birth Cohort Study (NHBCS, n = 237). RESULTS We identified a multi-metal index, predominated by arsenic (As) and cadmium (Cd), that was positively associated with SGA status (Odds ratio = 2.73 [1.04, 7.18]). This index was also associated with the expression of placental gene modules involved in "gene expression" (β = -0.02 [-0.04, -0.01]) and "metabolic hormone secretion" (β = 0.02 [0.00, 0.05]). We validated the association between cadmium exposure and the expression of GRHL1 and INHBA, genes in the "metabolic hormone secretion" module, in NHBCS. CONCLUSION We present a novel approach that integrates the application of advanced bioinformatics and biostatistics methods to delineate potential placental pathways through which trace metal exposures impact fetal growth.
Collapse
Affiliation(s)
- Maya A Deyssenroth
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chris Gennings
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shelley H Liu
- Center for Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 20019, USA
| | - Shouneng Peng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luca Lambertini
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian P Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03755, USA
| | | | - Carmen J Marsit
- Department of Environmental Health, Emory University, Atlanta, GA 30322, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
39
|
Maddock J, Wulaningsih W, Fernandez JC, Ploubidis GB, Goodman A, Bell J, Kuh D, Hardy R. Associations between body size, nutrition and socioeconomic position in early life and the epigenome: A systematic review. PLoS One 2018; 13:e0201672. [PMID: 30096154 PMCID: PMC6086410 DOI: 10.1371/journal.pone.0201672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background Body size, nutrition and socioeconomic position (SEP) in early life have been associated with a wide range of long-term health effects. Epigenetics is one possible mechanism through which these early life exposures can impact later life health. We conducted a systematic review examining the observational evidence for the impact of body size, nutrition and SEP in early life on the epigenome in humans. Methods This systematic review is registered with the PROSPERO database (registration number: CRD42016050193). Three datasets were simultaneously searched using Ovid and the resulting studies were evaluated by at least two independent reviewers. Studies measuring epigenetic markers either at the same time as, or after, the early life exposure and have a measure of body size, nutrition or SEP in early life (up to 12 years), written in English and from a community-dwelling participants were included. Results We identified 90 eligible studies. Seventeen of these papers examined more than one early life exposure of interest. Fifty six papers examined body size, 37 nutrition and 17 SEP. All of the included papers examined DNA methylation (DNAm) as the epigenetic marker. Overall there was no strong evidence for a consistent association between these early life variables in DNAm which may be due to the heterogeneous study designs, data collection methods and statistical analyses. Conclusions Despite these inconclusive results, the hypothesis that the early life environment can impact DNAm, potentially persisting into adult life, was supported by some studies and warrants further investigation. We provide recommendations for future studies.
Collapse
Affiliation(s)
- Jane Maddock
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, United Kingdom
- * E-mail:
| | - Wahyu Wulaningsih
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Juan Castillo Fernandez
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - George B. Ploubidis
- Centre for Longitudinal Studies, UCL Institute of Education, University College London, London, United Kingdom
| | - Alissa Goodman
- Centre for Longitudinal Studies, UCL Institute of Education, University College London, London, United Kingdom
| | - Jordana Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Rebecca Hardy
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
40
|
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: 21] [Impact Index Per Article: 3.0] [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
|
41
|
Schanton M, Maymó JL, Pérez-Pérez A, Sánchez-Margalet V, Varone CL. Involvement of leptin in the molecular physiology of the placenta. Reproduction 2017; 155:R1-R12. [PMID: 29018059 DOI: 10.1530/rep-17-0512] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 12/26/2022]
Abstract
Leptin is a homeostatic regulator in the placenta where it promotes proliferation, protein synthesis and the expression of tolerogenic maternal response molecules such as HLA-G. Leptin also exerts an anti-apoptotic action in placenta controlling the expression of p53 master cell cycle regulator under different stress conditions. On the other hand, leptin is an integrative target of different placental stimuli. The expression of leptin in placenta is regulated by hCG, insulin, steroids, hypoxia and many other growth hormones, suggesting that it might have an important endocrine function in the trophoblastic cells. The leptin expression is induced involving the cAMP/PKA or cAMP/Epac pathways which have profound actions upon human trophoblast function. The activation of PI3K and MAPK pathways also participates in the leptin expression. Estrogens play a central role during pregnancy, particularly 17β-estradiol upregulates the leptin expression in placental cells through genomic and non-genomic actions. The leptin promoter analysis reveals specific elements that are active in placental cells. The transcription factors CREB, AP1, Sp1, NFκB and the coactivator CBP are involved in the placental leptin expression. Moreover, placental leptin promoter is a target of epigenetic marks such as DNA methylation and histone acetylation that regulates not only the leptin expression in placenta during pregnancy but also determines the predisposition of acquiring adult metabolism diseases. Taken together, all these results allow a better understanding of leptin function and regulatory mechanisms of leptin expression in human placental trophoblasts, and support the importance of leptin during pregnancy and in programming adult health.
Collapse
Affiliation(s)
- Malena Schanton
- Departamento de Química BiológicaUniversidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.,Universidad de Buenos AiresCONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Julieta L Maymó
- Departamento de Química BiológicaUniversidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.,Universidad de Buenos AiresCONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Antonio Pérez-Pérez
- Departamento de Bioquímica Médica y Biología MolecularHospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Víctor Sánchez-Margalet
- Departamento de Bioquímica Médica y Biología MolecularHospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Cecilia L Varone
- Departamento de Química BiológicaUniversidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina .,Universidad de Buenos AiresCONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
42
|
Kadakia R, Zheng Y, Zhang Z, Zhang W, Hou L, Josefson JL. Maternal pre-pregnancy BMI downregulates neonatal cord blood LEP methylation. Pediatr Obes 2017; 12 Suppl 1:57-64. [PMID: 27933755 PMCID: PMC5462869 DOI: 10.1111/ijpo.12204] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/02/2016] [Accepted: 10/28/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Neonatal adiposity has many determinants and may be a risk factor for future obesity. Epigenetic regulation of metabolically important genes is a potential contributor. OBJECTIVES The objective of the study is to determine whether methylation changes in the LEP gene in cord blood DNA are impacted by the maternal environment or affect neonatal adiposity measures. METHODS A cross-sectional study of 114 full-term neonates born to healthy mothers with normal glucose tolerance was performed. Cord blood was assayed for leptin and genome-wide DNA methylation profiles via the Illumina 450K platform. Neonatal body composition was measured by air displacement plethysmography. Multivariate linear regression models and semi-partial correlation coefficients were used to analyze associations. False discovery rate was estimated to account for multiple comparisons. RESULTS Maternal pre-pregnancy BMI was associated with decreased methylation at five CpG sites near the LEP transcription start site in an adjusted model (false discovery rate <0.022 for each site). The association between maternal BMI and cord blood leptin approached significance (r = 0.18, p = 0.054). Cord blood leptin was positively correlated with neonatal adiposity measures including birth weight (r = 0.45, p < 0.001), fat mass (r = 0.47, p < 0.001) and percent body fat (r = 0.44, p < 0.001). CONCLUSIONS Maternal pre-pregnancy BMI is strongly associated with decreased cord blood LEP gene methylation and may mediate the well-known association between maternal pre-pregnancy BMI and neonatal adiposity.
Collapse
Affiliation(s)
- Rachel Kadakia
- Division of Endocrinology, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine,Health Sciences Integrated PhD Program, Northwestern University Feinberg School of Medicine
| | - Zhou Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine,Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine,Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine,Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine
| | - Jami L Josefson
- Division of Endocrinology, Ann and Robert H. Lurie Children’s Hospital of Chicago and Department of Pediatrics, Northwestern University Feinberg School of Medicine
| |
Collapse
|
43
|
Houshmand-Oeregaard A, Hansen NS, Hjort L, Kelstrup L, Broholm C, Mathiesen ER, Clausen TD, Damm P, Vaag A. Differential adipokine DNA methylation and gene expression in subcutaneous adipose tissue from adult offspring of women with diabetes in pregnancy. Clin Epigenetics 2017; 9:37. [PMID: 28413567 PMCID: PMC5390345 DOI: 10.1186/s13148-017-0338-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 03/31/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Offspring of women with diabetes in pregnancy are at increased risk of type 2 diabetes mellitus (T2DM), potentially mediated by epigenetic mechanisms. The adipokines leptin, adiponectin, and resistin (genes: LEP, ADIPOQ, RETN) play key roles in the pathophysiology of T2DM. We hypothesized that offspring exposed to maternal diabetes exhibit alterations in epigenetic regulation of subcutaneous adipose tissue (SAT) adipokine transcription. We studied adipokine plasma levels, SAT gene expression, and DNA methylation of LEP, ADIPOQ, and RETN in adult offspring of women with gestational diabetes (O-GDM, N = 82) or type 1 diabetes (O-T1DM, N = 67) in pregnancy, compared to offspring of women from the background population (O-BP, N = 57). RESULTS Compared to O-BP, we found elevated plasma leptin and resistin levels in O-T1DM, decreased gene expression of all adipokines in O-GDM, decreased RETN expression in O-T1DM, and increased LEP and ADIPOQ methylation in O-GDM. In multivariate regression analysis, O-GDM remained associated with increased ADIPOQ methylation and decreased ADIPOQ and RETN gene expression and O-T1DM remained associated with decreased RETN expression after adjustment for potential confounders and mediators. CONCLUSIONS In conclusion, offspring of women with diabetes in pregnancy exhibit increased ADIPOQ DNA methylation and decreased ADIPOQ and RETN gene expression in SAT. However, altered methylation and expression levels were not reflected in plasma protein levels, and the functional implications of these findings remain uncertain.
Collapse
Affiliation(s)
- Azadeh Houshmand-Oeregaard
- Center for Pregnant Women with Diabetes, Department of Obstetrics, Rigshospitalet, Dept. 7821, Blegdamsvej 9, 2100 Copenhagen, Denmark.,Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ninna S Hansen
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Danish Diabetes Academy/Danish PhD School of Molecular Metabolism, Odense, Denmark
| | - Line Hjort
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Danish Diabetes Academy/Danish PhD School of Molecular Metabolism, Odense, Denmark
| | - Louise Kelstrup
- Center for Pregnant Women with Diabetes, Department of Obstetrics, Rigshospitalet, Dept. 7821, Blegdamsvej 9, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christa Broholm
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Elisabeth R Mathiesen
- Center for Pregnant Women with Diabetes, Department of Obstetrics, Rigshospitalet, Dept. 7821, Blegdamsvej 9, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Pregnant Women with Diabetes, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Tine D Clausen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Gynecology and Obstetrics, Nordsjaellands Hospital, University of Copenhagen, Hilleroed, Denmark
| | - Peter Damm
- Center for Pregnant Women with Diabetes, Department of Obstetrics, Rigshospitalet, Dept. 7821, Blegdamsvej 9, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Vaag
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,AstraZeneca, Mölndal, Sweden
| |
Collapse
|
44
|
Declerck K, Remy S, Wohlfahrt-Veje C, Main KM, Van Camp G, Schoeters G, Vanden Berghe W, Andersen HR. Interaction between prenatal pesticide exposure and a common polymorphism in the PON1 gene on DNA methylation in genes associated with cardio-metabolic disease risk-an exploratory study. Clin Epigenetics 2017; 9:35. [PMID: 28396702 PMCID: PMC5382380 DOI: 10.1186/s13148-017-0336-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/30/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Prenatal environmental conditions may influence disease risk in later life. We previously found a gene-environment interaction between the paraoxonase 1 (PON1) Q192R genotype and prenatal pesticide exposure leading to an adverse cardio-metabolic risk profile at school age. However, the molecular mechanisms involved have not yet been resolved. It was hypothesized that epigenetics might be involved. The aim of the present study was therefore to investigate whether DNA methylation patterns in blood cells were related to prenatal pesticide exposure level, PON1 Q192R genotype, and associated metabolic effects observed in the children. METHODS Whole blood DNA methylation patterns in 48 children (6-11 years of age), whose mothers were occupationally unexposed or exposed to pesticides early in pregnancy, were determined by Illumina 450 K methylation arrays. RESULTS A specific methylation profile was observed in prenatally pesticide exposed children carrying the PON1 192R-allele. Differentially methylated genes were enriched in several neuroendocrine signaling pathways including dopamine-DARPP32 feedback (appetite, reward pathways), corticotrophin releasing hormone signaling, nNOS, neuregulin signaling, mTOR signaling, and type II diabetes mellitus signaling. Furthermore, we were able to identify possible candidate genes which mediated the associations between pesticide exposure and increased leptin level, body fat percentage, and difference in BMI Z score between birth and school age. CONCLUSIONS DNA methylation may be an underlying mechanism explaining an adverse cardio-metabolic health profile in children carrying the PON1 192R-allele and prenatally exposed to pesticides.
Collapse
Affiliation(s)
- Ken Declerck
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
| | - Sylvie Remy
- Department of Epidemiology and Social Medicine, Antwerp University, Universiteitsplein 1, Antwerp, Belgium.,Flemish Institute for Technological Research (VITO), Unit Environmental Risk and Health, Boeretang 200, Mol, Belgium
| | - Christine Wohlfahrt-Veje
- Department of Growth and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Greet Schoeters
- Flemish Institute for Technological Research (VITO), Unit Environmental Risk and Health, Boeretang 200, Mol, Belgium.,Department of Biomedical Sciences, Antwerp University, Universiteitsplein 1, Antwerp, Belgium.,Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
| | - Helle R Andersen
- Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
45
|
Saenen ND, Vrijens K, Janssen BG, Roels HA, Neven KY, Vanden Berghe W, Gyselaers W, Vanpoucke C, Lefebvre W, De Boever P, Nawrot TS. Lower Placental Leptin Promoter Methylation in Association with Fine Particulate Matter Air Pollution during Pregnancy and Placental Nitrosative Stress at Birth in the ENVIRONAGE Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:262-268. [PMID: 27623604 PMCID: PMC5289914 DOI: 10.1289/ehp38] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/13/2016] [Accepted: 08/18/2016] [Indexed: 05/02/2023]
Abstract
BACKGROUND Particulate matter with a diameter ≤ 2.5 μm (PM2.5) affects human fetal development during pregnancy. Oxidative stress is a putative mechanism by which PM2.5 may exert its effects. Leptin (LEP) is an energy-regulating hormone involved in fetal growth and development. OBJECTIVES We investigated in placental tissue whether DNA methylation of the LEP promoter is associated with PM2.5 and whether the oxidative/nitrosative stress biomarker 3-nitrotyrosine (3-NTp) is involved. METHODS LEP DNA methylation status of 361 placentas from the ENVIRONAGE birth cohort was assessed using bisulfite-PCR-pyrosequencing. Placental 3-NTp (n = 313) was determined with an ELISA assay. Daily PM2.5 exposure levels were estimated for each mother's residence, accounting for residential mobility during pregnancy, using a spatiotemporal interpolation model. RESULTS After adjustment for a priori chosen covariates, placental LEP methylation was 1.4% lower (95% CI: -2.7, -0.19%) in association with an interquartile range increment (7.5 μg/m3) in second-trimester PM2.5 exposure and 0.43% lower (95% CI: -0.85, -0.02%) in association with a doubling of placental 3-NTp content. CONCLUSIONS LEP methylation status in the placenta was negatively associated with PM2.5 exposure during the second trimester, and with placental 3-NTp, a marker of oxidative/nitrosative stress. Additional research is needed to confirm our findings and to assess whether oxidative/nitrosative stress might contribute to associations between PM2.5 and placental epigenetic events. Potential consequences for health during the neonatal period and later in life warrant further exploration. Citation: Saenen ND, Vrijens K, Janssen BG, Roels HA, Neven KY, Vanden Berghe W, Gyselaers W, Vanpoucke C, Lefebvre W, De Boever P, Nawrot TS. 2017. Lower placental leptin promoter methylation in association with fine particulate matter air pollution during pregnancy and placental nitrosative stress at birth in the ENVIRONAGE cohort. Environ Health Perspect 125:262-268; http://dx.doi.org/10.1289/EHP38.
Collapse
Affiliation(s)
- Nelly D. Saenen
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Karen Vrijens
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Bram G. Janssen
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Harry A. Roels
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Université catholique de Louvain, Brussels, Belgium
| | - Kristof Y. Neven
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Wim Vanden Berghe
- Department of Biomedical Sciences, Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), University of Antwerp, Antwerp, Belgium
| | - Wilfried Gyselaers
- Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department of Obstetrics, East-Limburg Hospital, Genk, Belgium
| | | | | | - Patrick De Boever
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Flemish Institute for Technological Research, Mol, Belgium
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Centre for Environment and Health, Leuven University, Leuven, Belgium
- Address correspondence to T.S. Nawrot, Centre for Environmental Sciences, Agoralaan Building D, 3590 Diepenbeek, Belgium, Telephone: 32-11-268382. E-mail:
| |
Collapse
|
46
|
Pauwels S, Ghosh M, Duca RC, Bekaert B, Freson K, Huybrechts I, A. S. Langie S, Koppen G, Devlieger R, Godderis L. Dietary and supplemental maternal methyl-group donor intake and cord blood DNA methylation. Epigenetics 2017; 12:1-10. [PMID: 27830979 PMCID: PMC5270634 DOI: 10.1080/15592294.2016.1257450] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/19/2016] [Accepted: 10/31/2016] [Indexed: 10/20/2022] Open
Abstract
Maternal nutrition is critically involved in the development and health of the fetus. We evaluated maternal methyl-group donor intake through diet (methionine, betaine, choline, folate) and supplementation (folic acid) before and during pregnancy in relation to global DNA methylation and hydroxymethylation and gene specific (IGF2 DMR, DNMT1, LEP, RXRA) cord blood methylation. A total of 115 mother-infant pairs were enrolled in the MAternal Nutrition and Offspring's Epigenome (MANOE) study. The intake of methyl-group donors was assessed using a food-frequency questionnaire. LC-MS/MS and pyrosequencing were used to measure global and gene specific methylation, respectively. Dietary intake of methyl-groups before and during pregnancy was associated with changes in LEP, DNMT1, and RXRA cord blood methylation. Statistically significant higher cord blood LEP methylation was observed when mothers started folic acid supplementation more than 6 months before conception compared with 3-6 months before conception (34.6 ± 6.3% vs. 30.1 ± 3.6%, P = 0.011, LEP CpG1) or no folic acid used before conception (16.2 ± 4.4% vs. 13.9 ± 3%, P = 0.036 for LEP CpG3 and 24.5 ± 3.5% vs. 22.2 ± 3.5%, P = 0.045 for LEP mean CpG). Taking folic acid supplements during the entire pregnancy resulted in statistically significantly higher cord blood RXRA methylation as compared with stopping supplementation in the second trimester (12.3 ± 1.9% vs. 11.1 ± 2%, P = 0.008 for RXRA mean CpG). To conclude, long-term folic acid use before and during pregnancy was associated with higher LEP and RXRA cord blood methylation, respectively. To date, pregnant women are advised to take a folic acid supplement of 400 µg/day from 4 weeks before until 12 weeks of pregnancy. Our results suggest significant epigenetic modifications when taking a folic acid supplement beyond the current advice.
Collapse
Affiliation(s)
- Sara Pauwels
- KU Leuven - University of Leuven, Department of Public Health and Primary Care, Environment and Health, Leuven, Belgium
- Flemish Institute of Technological Research (VITO), Unit Environmental Risk and Health, Mol, Belgium
| | - Manosij Ghosh
- KU Leuven - University of Leuven, Department of Public Health and Primary Care, Environment and Health, Leuven, Belgium
| | - Radu Corneliu Duca
- KU Leuven - University of Leuven, Department of Public Health and Primary Care, Environment and Health, Leuven, Belgium
| | - Bram Bekaert
- KU Leuven - University of Leuven, Department of Imaging & Pathology, Leuven, Belgium
- KU Leuven - University of Leuven, University Hospitals Leuven, Department of Forensic Medicine, Laboratory of Forensic Genetics and Molecular Archeology, Leuven, Belgium
| | - Kathleen Freson
- KU Leuven - University of Leuven, Center for Molecular and Vascular Biology, Leuven, Belgium
| | - Inge Huybrechts
- International Agency for Research on Cancer, Dietary Exposure Assessment Group, Lyon, France
| | - Sabine A. S. Langie
- Flemish Institute of Technological Research (VITO), Unit Environmental Risk and Health, Mol, Belgium
- Hasselt University, Faculty of Sciences, Diepenbeek, Belgium
| | - Gudrun Koppen
- Flemish Institute of Technological Research (VITO), Unit Environmental Risk and Health, Mol, Belgium
| | - Roland Devlieger
- KU Leuven - University of Leuven, Department of Development and Regeneration, Leuven, Belgium
- University Hospitals of Leuven, Department of Obstetrics and Gynecology, Leuven, Belgium
| | - Lode Godderis
- KU Leuven - University of Leuven, Department of Public Health and Primary Care, Environment and Health, Leuven, Belgium
- IDEWE, External Service for Prevention and Protection at Work, Heverlee, Belgium
| |
Collapse
|
47
|
Abstract
Obesity is a complex, heritable trait influenced by the interplay of genetics, epigenetics, metagenomics and the environment. With the increasing access to high precision diagnostic tools for genetic investigations, numerous genes influencing the phenotype have been identified, especially in early onset severe obesity. This review summarizes the current knowledge on the known genetic causes of obesity and the available therapeutic options. Furthermore, we discuss the role and potential mechanism of epigenetic changes that may be involved as mediators of the environmental influences and that may provide future opportunities for intervention.
Collapse
Affiliation(s)
- Vidhu V Thaker
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY
- Harvard Medical School, Boston, MA
- Division of Endocrinology, Boston Children's Hospital, Boston, MA
| |
Collapse
|
48
|
Thaker VV. GENETIC AND EPIGENETIC CAUSES OF OBESITY. ADOLESCENT MEDICINE: STATE OF THE ART REVIEWS 2017; 28:379-405. [PMID: 30416642 PMCID: PMC6226269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Obesity is a complex, heritable trait influenced by the interplay of genetics, epigenetics, metagenomics and the environment. With the increasing access to high precision diagnostic tools for genetic investigations, numerous genes influencing the phenotype have been identified, especially in early onset severe obesity. This review summarizes the current knowledge on the known genetic causes of obesity and the available therapeutic options. Furthermore, we discuss the role and potential mechanism of epigenetic changes that may be involved as mediators of the environmental influences and that may provide future opportunities for intervention.
Collapse
Affiliation(s)
- Vidhu V Thaker
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY
- Harvard Medical School, Boston, MA
- Division of Endocrinology, Boston Children's Hospital, Boston, MA
| |
Collapse
|
49
|
Gilbert-Diamond D, Emond JA, Baker ER, Korrick SA, Karagas MR. Relation between in Utero Arsenic Exposure and Birth Outcomes in a Cohort of Mothers and Their Newborns from New Hampshire. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1299-307. [PMID: 26955061 PMCID: PMC4977046 DOI: 10.1289/ehp.1510065] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/25/2015] [Accepted: 02/12/2016] [Indexed: 05/15/2023]
Abstract
BACKGROUND Studies suggest that arsenic exposure influences birth outcomes; however, findings are mixed. OBJECTIVE We assessed in utero arsenic exposure in relation to birth outcomes and whether maternal prepregnancy weight and infant sex modified the associations. METHODS Among 706 mother-infant pairs exposed to low levels of arsenic through drinking water and diet, we assessed in utero arsenic exposure using maternal second-trimester urinary arsenic, maternal prepregnancy weight through self-report, and birth outcomes from medical records. RESULTS Median (interquartile range) of total urinary arsenic [tAs; inorganic arsenic (iAs) + monomethylarsonic acid (MMA) + dimethylarsinic acid (DMA)] was 3.4 μg/L (1.7-6.0). In adjusted linear models, each doubling of tAs was associated with a 0.10-cm decrease (95% CI: -0.19, -0.01) in head circumference. Results were similar for MMA and DMA. Ln(tAs) and ln(DMA) were positively associated with birth length in infant males only; among males, each doubling of tAs was associated with a 0.28-cm increase (95% CI: 0.09, 0.46) in birth length (pinteraction = 0.04). Results were similar for DMA. Additionally, arsenic exposure was inversely related to ponderal index, and associations differed by maternal weight. Each ln(tAs) doubling of tAs was associated with a 0.55-kg/m3 lower (95% CI: -0.82, -0.28, p < 0.001) ponderal index for infants of overweight/obese, but not normal-weight, mothers (pinteraction < 0.01). Finally, there was a significant interaction between maternal weight status, infant sex, and arsenic exposure on birth weight (pinteraction = 0.03). In girls born of overweight/obese mothers, each doubling of tAs was associated with a 62.9-g decrease (95% CI: -111.6, -14.2) in birth weight, though the association was null in the other strata. CONCLUSIONS Low-level arsenic exposure may affect fetal growth, and the associations may be modified by maternal weight status and infant sex. CITATION Gilbert-Diamond D, Emond JA, Baker ER, Korrick SA, Karagas MR. 2016. Relation between in utero arsenic exposure and birth outcomes in a cohort of mothers and their newborns from New Hampshire. Environ Health Perspect 124:1299-1307; http://dx.doi.org/10.1289/ehp.1510065.
Collapse
Affiliation(s)
- Diane Gilbert-Diamond
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Children’s Environmental Health and Disease Prevention Center at Dartmouth, Hanover, New Hampshire, USA
| | - Jennifer A. Emond
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Emily R. Baker
- Children’s Environmental Health and Disease Prevention Center at Dartmouth, Hanover, New Hampshire, USA
- Department of Obstetrics and Gynecology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Susan A. Korrick
- Children’s Environmental Health and Disease Prevention Center at Dartmouth, Hanover, New Hampshire, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Children’s Environmental Health and Disease Prevention Center at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
50
|
|