1
|
Taschereau A, Doyon M, Arguin M, Allard C, Desgagné V, Cote AM, Massé É, Jacques PÉ, Perron P, Hivert MF, Bouchard L. Cohort profile: the Genetics of Glucose regulation in Gestation and Growth (Gen3G) - a prospective prebirth cohort of mother-child pairs in Sherbrooke, Canada, 3-year and 5-year follow-up visits. BMJ Open 2025; 15:e093434. [PMID: 40122564 PMCID: PMC11931902 DOI: 10.1136/bmjopen-2024-093434] [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: 09/06/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025] Open
Abstract
PURPOSE Initiated in 2010, the Genetics of Glucose regulation in Gestation and Growth (Gen3G) prospective cohort investigates the pathophysiology of impaired glycaemic regulation in pregnancy and evaluates its impact on both the mothers and her offspring health trajectory. Follow-up visits 3 and 5 years after delivery aimed to investigate pregnancy-related risk factors such as maternal obesity and gestational hyperglycaemia in relation to the mother's metabolic health after pregnancy, and with offspring health outcomes such as risk of obesity and neurodevelopmental problems in early childhood. We also investigated molecular mechanisms involved in the fetal programming of these later health outcomes. PARTICIPANTS Of the 1024 women originally recruited in the first trimester of pregnancy, we have targeted the 854 who had complete glucose tolerance test data and the 724 newborns who provided placenta and/or cord blood samples for follow-up recruitment. Of these, 695 mother-child dyads agreed to be contacted for the prospective follow-up visits. 448 and 521 mother-child dyads completed the research visits at 3 and 5 years after delivery respectively. FINDINGS TO DATE At both visits, we collected the mother's and child's medical history, lifestyle (using validated questionnaires), sociodemographic status, anthropometric measurements, mother's blood samples, child's saliva samples and growth charts. At the 5-year-old visit, we additionally collected the mother's and child's urine and stool samples and the child's blood samples; we performed a 75 g oral glucose tolerance test in the mothers and assessed the body composition in children using dual-energy X-ray absorptiometry. Using the Gen3G rich longitudinal data set, we have enhanced the understanding of the pathophysiology and characterisation of the heterogeneity of gestational diabetes mellitus, and we have shown that gestational hyperglycaemia and insulin resistance are associated with offspring epigenetics (DNA methylation) variations in the placenta, cord blood and blood at 5 years of age, as well as with offspring anthropometric, metabolic and neurodevelopmental outcomes in early childhood. FUTURE PLANS We are currently conducting a prospective follow-up of mothers and their children 12 years after delivery to study how prenatal and early-life metabolic factors may programme childhood adiposity and obesogenic dietary behaviours. This follow-up should be completed by the end of 2026.
Collapse
Affiliation(s)
- Amélie Taschereau
- Biochimie et génomique fonctionnelle, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Myriam Doyon
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mélina Arguin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Catherine Allard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Véronique Desgagné
- Biochimie et génomique fonctionnelle, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Anne-Marie Cote
- Department of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Éric Massé
- Biochimie et génomique fonctionnelle, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Patrice Perron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Marie-France Hivert
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
2
|
Liu S, Xu L, Cheng Y, Liu D, Zhang B, Chen X, Zheng M. Methylation of the telomerase gene promoter region in umbilical cord blood of patients with gestational diabetes mellitus is associated with decreased telomerase expression levels and shortened telomere length. Front Endocrinol (Lausanne) 2025; 16:1502329. [PMID: 40134806 PMCID: PMC11932890 DOI: 10.3389/fendo.2025.1502329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Objective This study speculates that gestational diabetes mellitus (GDM) may reduce fetal telomere length (TL),which may be related to modification of methylation in the promoter region of the telomerase (TE) gene promoter region. Methods In this study, umbilical cord blood samples from patients with and without GDM (N = 100 each) were analyzed by prospective case-control. The TL, TE expression levels, and methylation levels of TERT and TERC gene promoter regions in two groups were measured. The significance of the methylation level of each CpG locus employed logistic regression analysis of R software, and the analysis of covariance (ANCOVA) was used to control the influence of confounding factors. Correlation analysis was performed by the Spearman. Results The TL and TE expression levels of the offspring of GDM patients were decreased despite adjusting for PBMI, PWG, and TG. A total of two CpG islands were screened in the promoter region of the TERT gene and three fragments (TERT_2, TERT_3, and TERT_4) containing a total of 70 CpG sites were designed. Additionally, four CpG sites of the TERT gene in the GDM group (TERT_2_40, TERT_2_47, TERT_3_46, and TERT_3_212) showed increased methylation levels compared with the control group (all P < 0.05). In the promoter region of the TERC gene, one CpG island containing 19 CpG loci was screened and designed, and the methylation levels of the two CpG sites were significantly different in TERC_1_67 (0.65 ± 0.21 versus 0.57 ± 0.30; P = 0.040) and TERC_1_120 (0.68 ± 0.23 versus 0.59 ± 0.27; P = 0.014). The methylation levels of TERC gene fragments of GDM patients were significantly higher than those of the control group (0.69 ± 0.06 versus 0.65 ± 0.08, P = 0.001). Conclusion This study revealed that GDM may induce decreased TE expression by increasing the methylation levels of TE genes promoter region, thereby reducing the TL.
Collapse
Affiliation(s)
- Shuhua Liu
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| | - Liping Xu
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
- Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yan Cheng
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
- Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Dehong Liu
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| | - Bin Zhang
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| | - Xianxia Chen
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
- Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Mingming Zheng
- Department of Obstetrics and Gynecology, Hefei Maternal and Child Health Hospital, Hefei, China
- Department of Obstetrics and Gynecology, Anhui Women and Children’s Medical Center, Hefei, China
- Department of Obstetrics and Gynecology, Maternal and Child Medical Center of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Franzago M, Borrelli P, Di Nicola M, Cavallo P, D’Adamo E, Di Tizio L, Gazzolo D, Stuppia L, Vitacolonna E. From Mother to Child: Epigenetic Signatures of Hyperglycemia and Obesity during Pregnancy. Nutrients 2024; 16:3502. [PMID: 39458497 PMCID: PMC11510513 DOI: 10.3390/nu16203502] [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: 09/16/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND In utero exposure to maternal hyperglycemia and obesity can trigger detrimental effects in the newborn through epigenetic programming. We aimed to assess the DNA methylation levels in the promoters of MC4R and LPL genes from maternal blood, placenta, and buccal swab samples collected in children born to mothers with and without obesity and Gestational Diabetes Mellitus (GDM). METHODS A total of 101 Caucasian mother-infant pairs were included in this study. Sociodemographic characteristics, clinical parameters, physical activity, and adherence to the Mediterranean diet were evaluated in the third trimester of pregnancy. Clinical parameters of the newborns were recorded at birth. RESULTS A negative relationship between MC4R DNA methylation on the fetal side of the GDM placenta and birth weight (r = -0.630, p = 0.011) of newborns was found. MC4R DNA methylation level was lower in newborns of GDM women (CpG1: 2.8% ± 3.0%, CpG2: 3.8% ± 3.3%) as compared to those of mothers without GDM (CpG1: 6.9% ± 6.2%, CpG2: 6.8% ± 5.6%; p < 0.001 and p = 0.0033, respectively), and it was negatively correlated with weight (r = -0.229; p = 0.035), head circumference (r = -0.236; p = 0.030), and length (r = -0.240; p = 0.027) at birth. LPL DNA methylation was higher on the fetal side of the placenta in obese patients as compared to normal-weight patients (66.0% ± 14.4% vs. 55.7% ± 15.2%, p = 0.037), and it was associated with maternal total cholesterol (r = 0.770, p = 0.015) and LDL-c (r = 0.783, p = 0.012). CONCLUSIONS These results support the role of maternal MC4R and LPL methylation in fetal programming and in the future metabolic health of children.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| | - Paola Borrelli
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (P.B.); (M.D.N.)
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (P.B.); (M.D.N.)
| | - Pierluigi Cavallo
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
| | - Ebe D’Adamo
- Neonatal Intensive Care Unit, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Luciano Di Tizio
- Department of Obstetrics and Gynaecology, SS. Annunziata Hospital, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Diego Gazzolo
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Neonatal Intensive Care Unit, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
4
|
Li N, Liu HY, Liu SM. Deciphering DNA Methylation in Gestational Diabetes Mellitus: Epigenetic Regulation and Potential Clinical Applications. Int J Mol Sci 2024; 25:9361. [PMID: 39273309 PMCID: PMC11394902 DOI: 10.3390/ijms25179361] [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: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Gestational diabetes mellitus (GDM) represents a prevalent complication during pregnancy, exerting both short-term and long-term impacts on maternal and offspring health. This review offers a comprehensive outline of DNA methylation modifications observed in various maternal and offspring tissues affected by GDM, emphasizing the intricate interplay between DNA methylation dynamics, gene expression, and the pathogenesis of GDM. Furthermore, it explores the influence of environmental pollutants, maternal nutritional supplementation, and prenatal gut microbiota on GDM development through alterations in DNA methylation profiles. Additionally, this review summarizes recent advancements in DNA methylation-based diagnostics and predictive models in early GDM detection and risk assessment for subsequent type 2 diabetes. These insights contribute significantly to our understanding of the epigenetic mechanisms underlying GDM development, thereby enhancing maternal and fetal health outcomes and advocating further efforts in this field.
Collapse
Affiliation(s)
- Nan Li
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Huan-Yu Liu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, 169 Donghu Road, Wuhan 430071, China
| | - Song-Mei Liu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, 169 Donghu Road, Wuhan 430071, China
| |
Collapse
|
5
|
Saucedo R, Ferreira-Hermosillo A, Robledo-Clemente M, Díaz-Velázquez MF, Valencia-Ortega J. Association of DNA Methylation with Infant Birth Weight in Women with Gestational Diabetes. Metabolites 2024; 14:361. [PMID: 39057684 PMCID: PMC11278577 DOI: 10.3390/metabo14070361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Offspring exposed to gestational diabetes mellitus (GDM) exhibit greater adiposity at birth. This early-life phenotype may increase offspring risk of developing obesity, metabolic syndrome, type 2 diabetes, and cardiovascular disease later in life. Infants born to women with GDM have a dysregulation of several hormones, cytokines, and growth factors related to fetal fat mass growth. One of the molecular mechanisms of GDM influencing these factors is epigenetic alterations, such as DNA methylation (DNAm). This review will examine the role of DNAm as a potential biomarker for monitoring fetal growth during pregnancy in women with GDM. This information is relevant since it may provide useful new biomarkers for the diagnosis, prognosis, and treatment of fetal growth and its later-life health consequences.
Collapse
Affiliation(s)
- Renata Saucedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.S.); (A.F.-H.)
| | - Aldo Ferreira-Hermosillo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (R.S.); (A.F.-H.)
| | - Magalhi Robledo-Clemente
- Hospital de Gineco Obstetricia 3, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City 02990, Mexico; (M.R.-C.); (M.F.D.-V.)
| | - Mary Flor Díaz-Velázquez
- Hospital de Gineco Obstetricia 3, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City 02990, Mexico; (M.R.-C.); (M.F.D.-V.)
| | - Jorge Valencia-Ortega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico
| |
Collapse
|
6
|
Yi Y, Wang T, Xu W, Zhang SH. Epigenetic modifications of placenta in women with gestational diabetes mellitus and their offspring. World J Diabetes 2024; 15:378-391. [PMID: 38591094 PMCID: PMC10999040 DOI: 10.4239/wjd.v15.i3.378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/30/2023] [Accepted: 02/06/2024] [Indexed: 03/15/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a pregnancy-related complication characterized by abnormal glucose metabolism in pregnant women and has an important impact on fetal development. As a bridge between the mother and the fetus, the placenta has nutrient transport functions, endocrine functions, etc., and can regulate placental nutrient transport and fetal growth and development according to maternal metabolic status. Only by means of placental transmission can changes in maternal hyperglycemia affect the fetus. There are many reports on the placental pathophysiological changes associated with GDM, the impacts of GDM on the growth and development of offspring, and the prevalence of GDM in offspring after birth. Placental epigenetic changes in GDM are involved in the programming of fetal development and are involved in the pathogenesis of later chronic diseases. This paper summarizes the effects of changes in placental nutrient transport function and hormone secretion levels due to maternal hyperglycemia and hyperinsulinemia on the development of offspring as well as the participation of changes in placental epigenetic modifications due to maternal hyperglycemia in intrauterine fetal programming to promote a comprehensive understanding of the impacts of placental epigenetic modifications on the development of offspring from patients with GDM.
Collapse
Affiliation(s)
- Yan Yi
- Department of Ultrasonography, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Tao Wang
- Clinical Molecular Immunology Center, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Wei Xu
- Department of Ultrasonography, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - San-Hong Zhang
- Department of Pediatric, Xiantao First People’s Hospital, Xiantao 433000, Hubei Province, China
| |
Collapse
|
7
|
Meza-León A, Montoya-Estrada A, Reyes-Muñoz E, Romo-Yáñez J. Diabetes Mellitus and Pregnancy: An Insight into the Effects on the Epigenome. Biomedicines 2024; 12:351. [PMID: 38397953 PMCID: PMC10886464 DOI: 10.3390/biomedicines12020351] [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: 11/16/2023] [Revised: 12/17/2023] [Accepted: 01/08/2024] [Indexed: 02/25/2024] Open
Abstract
Worldwide, diabetes mellitus represents a growing health problem. If it occurs during pregnancy, it can increase the risk of various abnormalities in early and advanced life stages of exposed individuals due to fetal programming occurring in utero. Studies have determined that maternal conditions interfere with the genotypes and phenotypes of offspring. Researchers are now uncovering the mechanisms by which epigenetic alterations caused by diabetes affect the expression of genes and, therefore, the development of various diseases. Among the numerous possible epigenetic changes in this regard, the most studied to date are DNA methylation and hydroxymethylation, as well as histone acetylation and methylation. This review article addresses critical findings in epigenetic studies involving diabetes mellitus, including variations reported in the expression of specific genes and their transgenerational effects.
Collapse
Affiliation(s)
| | | | | | - José Romo-Yáñez
- Coordinación de Endocrinología Ginecológica y Perinatal, Instituto Nacional de Perinatología, Montes Urales 800, Lomas Virreyes, Mexico City 11000, Mexico
| |
Collapse
|
8
|
Lizárraga D, Gómez-Gil B, García-Gasca T, Ávalos-Soriano A, Casarini L, Salazar-Oroz A, García-Gasca A. Gestational diabetes mellitus: genetic factors, epigenetic alterations, and microbial composition. Acta Diabetol 2024; 61:1-17. [PMID: 37660305 DOI: 10.1007/s00592-023-02176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
Gestational diabetes mellitus (GDM) is a common metabolic disorder, usually diagnosed during the third trimester of pregnancy that usually disappears after delivery. In GDM, the excess of glucose, fatty acids, and amino acids results in foetuses large for gestational age. Hyperglycaemia and insulin resistance accelerate the metabolism, raising the oxygen demand, and creating chronic hypoxia and inflammation. Women who experienced GDM and their offspring are at risk of developing type-2 diabetes, obesity, and other metabolic or cardiovascular conditions later in life. Genetic factors may predispose the development of GDM; however, they do not account for all GDM cases; lifestyle and diet also play important roles in GDM development by modulating epigenetic signatures and the body's microbial composition; therefore, this is a condition with a complex, multifactorial aetiology. In this context, we revised published reports describing GDM-associated single-nucleotide polymorphisms (SNPs), DNA methylation and microRNA expression in different tissues (such as placenta, umbilical cord, adipose tissue, and peripheral blood), and microbial composition in the gut, oral cavity, and vagina from pregnant women with GDM, as well as the bacterial composition of the offspring. Altogether, these reports indicate that a number of SNPs are associated to GDM phenotypes and may predispose the development of the disease. However, extrinsic factors (lifestyle, nutrition) modulate, through epigenetic mechanisms, the risk of developing the disease, and some association exists between the microbial composition with GDM in an organ-specific manner. Genes, epigenetic signatures, and microbiota could be transferred to the offspring, increasing the possibility of developing chronic degenerative conditions through postnatal life.
Collapse
Affiliation(s)
- Dennise Lizárraga
- Laboratory of Molecular and Cell Biology, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico
| | - Bruno Gómez-Gil
- Laboratory of Microbial Genomics, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico
| | - Teresa García-Gasca
- Laboratory of Molecular and Cellular Biology, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Avenida de las Ciencias s/n, 76230, Juriquilla, Querétaro, Mexico
| | - Anaguiven Ávalos-Soriano
- Laboratory of Molecular and Cell Biology, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125, Modena, Italy
| | - Azucena Salazar-Oroz
- Maternal-Fetal Department, Instituto Vidalia, Hospital Sharp Mazatlán, Avenida Rafael Buelna y Dr. Jesús Kumate s/n, 82126, Mazatlán, Sinaloa, Mexico
| | - Alejandra García-Gasca
- Laboratory of Molecular and Cell Biology, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico.
| |
Collapse
|
9
|
Waldrop SW, Niemiec S, Wood C, Gyllenhammer LE, Jansson T, Friedman JE, Tryggestad JB, Borengasser SJ, Davidson EJ, Yang IV, Kechris K, Dabelea D, Boyle KE. Cord blood DNA methylation of immune and lipid metabolism genes is associated with maternal triglycerides and child adiposity. Obesity (Silver Spring) 2024; 32:187-199. [PMID: 37869908 PMCID: PMC10872762 DOI: 10.1002/oby.23915] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVE Fetal exposures may impact offspring epigenetic signatures and adiposity. The authors hypothesized that maternal metabolic traits associate with cord blood DNA methylation, which, in turn, associates with child adiposity. METHODS Fasting serum was obtained in 588 pregnant women (27-34 weeks' gestation), and insulin, glucose, high-density lipoprotein cholesterol, triglycerides, and free fatty acids were measured. Cord blood DNA methylation and child adiposity were measured at birth, 4-6 months, and 4-6 years. The association of maternal metabolic traits with DNA methylation (429,246 CpGs) for differentially methylated probes (DMPs) and regions (DMRs) was tested. The association of the first principal component of each DMR with child adiposity was tested, and mediation analysis was performed. RESULTS Maternal triglycerides were associated with the most DMPs and DMRs of all traits tested (261 and 198, respectively, false discovery rate < 0.05). DMRs were near genes involved in immune function and lipid metabolism. Triglyceride-associated CpGs were associated with child adiposity at 4-6 months (32 CpGs) and 4-6 years (2 CpGs). One, near CD226, was observed at both timepoints, mediating 10% and 22% of the relationship between maternal triglycerides and child adiposity at 4-6 months and 4-6 years, respectively. CONCLUSIONS DNA methylation may play a role in the association of maternal triglycerides and child adiposity.
Collapse
Affiliation(s)
- Stephanie W. Waldrop
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Sierra Niemiec
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Cheyret Wood
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Lauren E. Gyllenhammer
- Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jeanie B. Tryggestad
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sarah J. Borengasser
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Elizabeth J. Davidson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Ivana V. Yang
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Kristen E. Boyle
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
| |
Collapse
|
10
|
Taschereau A, Thibeault K, Allard C, Juvinao-Quintero D, Perron P, Lutz SM, Bouchard L, Hivert MF. Maternal glycemia in pregnancy is longitudinally associated with blood DNAm variation at the FSD1L gene from birth to 5 years of age. Clin Epigenetics 2023; 15:107. [PMID: 37386647 PMCID: PMC10308691 DOI: 10.1186/s13148-023-01524-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/23/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND In utero exposure to maternal hyperglycemia has been associated with an increased risk for the development of chronic diseases in later life. These predispositions may be programmed by fetal DNA methylation (DNAm) changes that persist postnatally. However, although some studies have associated fetal exposure to gestational hyperglycemia with DNAm variations at birth, and metabolic phenotypes in childhood, no study has yet examined how maternal hyperglycemia during pregnancy may be associated with offspring DNAm from birth to five years of age. HYPOTHESIS Maternal hyperglycemia is associated with variation in offspring DNAm from birth to 5 years of age. METHODS We estimated maternal hyperglycemia using the area under the curve for glucose (AUCglu) following an oral glucose tolerance test conducted at 24-30 weeks of pregnancy. We quantified DNAm levels in cord blood (n = 440) and peripheral blood at five years of age (n = 293) using the Infinium MethylationEPIC BeadChip (Illumina). Our total sample included 539 unique dyads (mother-child) with 194 dyads having DNAm at both time-points. We first regressed DNAm M-values against the cell types and child age for each time-point separately to account for the difference by time of measurement for these variables. We then used a random intercept model from the linear mixed model (LMM) framework to assess the longitudinal association between maternal AUCglu and the repeated measures of residuals of DNAm. We adjusted for the following covariates as fixed effects in the random intercept model: maternal age, gravidity, smoking status, child sex, maternal body mass index (BMI) (measured at first trimester of pregnancy), and a binary variable for time-point. RESULTS In utero exposure to higher maternal AUCglu was associated with lower offspring blood DNAm levels at cg00967989 located in FSD1L gene (β = - 0.0267, P = 2.13 × 10-8) in adjusted linear regression mixed models. Our study also reports other CpG sites for which DNAm levels were suggestively associated (P < 1.0 × 10-5) with in utero exposure to gestational hyperglycemia. Two of these (cg12140144 and cg07946633) were found in the promotor region of PRDM16 gene (β: - 0.0251, P = 4.37 × 10-07 and β: - 0.0206, P = 2.24 × 10-06, respectively). CONCLUSION Maternal hyperglycemia is associated with offspring DNAm longitudinally assessed from birth to 5 years of age.
Collapse
Affiliation(s)
- Amélie Taschereau
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences (FMHS), Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kathrine Thibeault
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences (FMHS), Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Catherine Allard
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CR-CHUS), Sherbrooke, QC, Canada
| | | | - Patrice Perron
- Centre de Recherche du Centre hospitalier universitaire de Sherbrooke (CR-CHUS), Sherbrooke, QC, Canada
- Department of Medicine, FMHS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sharon M Lutz
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Luigi Bouchard
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences (FMHS), Université de Sherbrooke, Sherbrooke, QC, Canada.
- Department of Medicine, FMHS, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Clinical Department of Laboratory Medicine, Pavillon des Augustines, Centre intégré universitaire de santé et de services sociaux (CIUSSS) du Saguenay-Lac-Saint-Jean - Hôpital de Chicoutimi, 305 rue St-Vallier, Saguenay, QC, G7H 5H6, Canada.
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Evaluation of H19, Mest, Meg3, and Peg3 genes affecting growth and metabolism in umbilical cord blood cells of infants born to mothers with gestational diabetes and healthy mothers in Rafsanjan City, Iran. J Dev Orig Health Dis 2023; 14:182-189. [PMID: 35904097 DOI: 10.1017/s2040174422000393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Hyperglycemia during the first trimester leads to an increased risk of innate malformations as well as death at times close to delivery dates. The methylated genes include those from paternal H19 and PEG3 and those from maternal MEST and MEG3 that are necessary for the growth and regulation of the human fetus and its placenta. The aim of this study was to evaluate and compare the expression of these genes in the cord blood of healthy infants born to mothers with gestational diabetes mellitus (GDM) and healthy mothers.This case-control study was conducted on the cord blood of 40 infants born to mothers with GDM and 35 infants born to healthy mothers. Mothers were identified by measuring oral glucose tolerance in the 24th-26th week of pregnancy. Cord blood was obtained post-delivery, and cord blood mononuclear cells were immediately extracted, using Ficoll solution. Then, RNA extraction and cDNA synthesis were performed, and gene expression of MEG3, PEG3, H19, and MEST was assessed through quantitative real-time PCR.Findings show that the expression levels of MEG3, PEG3, H19, and MEST genes were significantly decreased in mononuclear cord blood cells of infants born to mothers with GDM when compared to those of the healthy control group.These findings reveal that the reduction of imprinted genes in mothers with GDM is most likely due to changes in their methylation by an epigenetic process. Considering the importance of GDM due to its high prevalence and its side effects both for mother and fetus, recognizing their exact mechanisms is of high importance. This has to be studied more widely.
Collapse
|
12
|
Li L, He S, Lin MH, Zhang YP, Kuhl H, Liang XF. Whole-genome resequencing and bisulfite sequencing provide new insights into the feeding habit domestication in mandarin fish ( Siniperca chuatsi). Front Genet 2023; 13:1088081. [PMID: 36712873 PMCID: PMC9878154 DOI: 10.3389/fgene.2022.1088081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
Mandarin fish (Siniperca chuatsi) is one of the most economically important fish in China. However, it has the peculiar feeding habit that it feeds solely on live prey fish since first-feeding, while refuses dead prey fish or artificial diets. After the specific training procedure, partial individuals could accept dead prey fish and artificial diets. The genetic basis of individual difference in artificial diet feeding habit is still unknown. In the present study, the resequencing was performed between 10 individuals which could be domesticated to accept artificial diets and 10 individuals which could not. Through the selective sweep analysis based on heterozygosity (Hp) and population differentiation coefficient (Fst), 57 candidate windows were identified as the putative selected regions for feeding habit domestication of mandarin fish, involved in 149 genes. These genes were related to memory, vision and olfaction function, which could be potential targets of molecular marker assistant breeding of artificial diet feeding trait. Beside of the DNA sequence, we also explored the potential role of DNA methylation in feeding habit domestication in mandarin fish. Whole-genome bisulfite sequencing was performed between the individuals which could be domesticated to accept artificial diets and those could not. 5,976 differentially methylated regions were identified, referring to 3,522 genes, such as the genes involved in cAMP signaling pathway. The DNA methylation changes of these genes might contribute to the adaption of artificial diets in mandarin fish. In conclusion, the putative selected regions and the differentially methylated regions were identified in the whole genome, providing new insights into the feeding habit domestication from live prey fish to artificial diets in mandarin fish. And the involved genes were identified as the candidate genes for molecular breeding of artificial diet utilization in mandarin fish.
Collapse
Affiliation(s)
- Ling Li
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China,Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Shan He
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China,Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Ming-Hui Lin
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China,Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Yan-Peng Zhang
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China,Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Heiner Kuhl
- Department of Ecophysiology and Aquaculture, Leibniz-Institute of Freshwater Ecology and Inland Fisheries, Berlin, Germany,*Correspondence: Xu-Fang Liang, ; Heiner Kuhl,
| | - Xu-Fang Liang
- Chinese Perch Research Center, College of Fisheries, Huazhong Agricultural University, Wuhan, China,Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China,*Correspondence: Xu-Fang Liang, ; Heiner Kuhl,
| |
Collapse
|
13
|
Yang MN, Huang R, Zheng T, Dong Y, Wang WJ, Xu YJ, Mehra V, Zhou GD, Liu X, He H, Fang F, Li F, Fan JG, Zhang J, Ouyang F, Briollais L, Li J, Luo ZC. Genome-wide placental DNA methylations in fetal overgrowth and associations with leptin, adiponectin and fetal growth factors. Clin Epigenetics 2022; 14:192. [PMID: 36585686 PMCID: PMC9801645 DOI: 10.1186/s13148-022-01412-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fetal overgrowth "programs" an elevated risk of type 2 diabetes in adulthood. Epigenetic alterations may be a mechanism in programming the vulnerability. We sought to characterize genome-wide alterations in placental gene methylations in fetal overgrowth and the associations with metabolic health biomarkers including leptin, adiponectin and fetal growth factors. RESULTS Comparing genome-wide placental gene DNA methylations in large-for-gestational-age (LGA, an indicator of fetal overgrowth, n = 30) versus optimal-for-gestational-age (OGA, control, n = 30) infants using the Illumina Infinium Human Methylation-EPIC BeadChip, we identified 543 differential methylation positions (DMPs; 397 hypermethylated, 146 hypomethylated) at false discovery rate < 5% and absolute methylation difference > 0.05 after adjusting for placental cell-type heterogeneity, maternal age, pre-pregnancy BMI and HbA1c levels during pregnancy. Twenty-five DMPs annotated to 20 genes (QSOX1, FCHSD2, LOC101928162, ADGRB3, GCNT1, TAP1, MYO16, NAV1, ATP8A2, LBXCOR1, EN2, INCA1, CAMTA2, SORCS2, SLC4A4, RPA3, UMAD1,USP53, OR2L13 and NR3C2) could explain 80% of the birth weight variations. Pathway analyses did not detect any statistically significant pathways after correcting for multiple tests. We validated a newly discovered differentially (hyper-)methylated gene-visual system homeobox 1 (VSX1) in an independent pyrosequencing study sample (LGA 47, OGA 47). Our data confirmed a hypermethylated gene-cadherin 13 (CDH13) reported in a previous epigenome-wide association study. Adiponectin in cord blood was correlated with its gene methylation in the placenta, while leptin and fetal growth factors (insulin, IGF-1, IGF-2) were not. CONCLUSIONS Fetal overgrowth may be associated with a large number of altered placental gene methylations. Placental VSX1 and CDH13 genes are hypermethylated in fetal overgrowth. Placental ADIPOQ gene methylations and fetal circulating adiponectin levels were correlated, suggesting the contribution of placenta-originated adiponectin to cord blood adiponectin.
Collapse
Affiliation(s)
- Meng-Nan Yang
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China ,grid.17063.330000 0001 2157 2938Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Faculty of Medicine, University of Toronto, L5-240, Murray Street 60, Toronto, ON M5G 1X5 Canada
| | - Rong Huang
- grid.17063.330000 0001 2157 2938Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Faculty of Medicine, University of Toronto, L5-240, Murray Street 60, Toronto, ON M5G 1X5 Canada
| | - Tao Zheng
- grid.16821.3c0000 0004 0368 8293Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Yu Dong
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Wen-Juan Wang
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Ya-Jie Xu
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Vrati Mehra
- grid.17063.330000 0001 2157 2938Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Faculty of Medicine, University of Toronto, L5-240, Murray Street 60, Toronto, ON M5G 1X5 Canada
| | - Guang-Di Zhou
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Xin Liu
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Hua He
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Fang Fang
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Fei Li
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Jian-Gao Fan
- grid.16821.3c0000 0004 0368 8293Center for Fatty Liver, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Jun Zhang
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Fengxiu Ouyang
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China
| | - Laurent Briollais
- grid.17063.330000 0001 2157 2938Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Faculty of Medicine, University of Toronto, L5-240, Murray Street 60, Toronto, ON M5G 1X5 Canada
| | - Jiong Li
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China ,grid.7048.b0000 0001 1956 2722Department of Clinical Medicine-Department of Clinical Epidemiology, Aarhus University, Olof Palmes Allé 43-45, 8200 Aathus, Denmark
| | - Zhong-Cheng Luo
- grid.16821.3c0000 0004 0368 8293Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200092 China ,grid.17063.330000 0001 2157 2938Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Faculty of Medicine, University of Toronto, L5-240, Murray Street 60, Toronto, ON M5G 1X5 Canada
| | | |
Collapse
|
14
|
Gil GP, Ananina G, Maschietto M, Lima SCS, da Silva Costa SM, Baptista LDC, Ito MT, Costa FF, Costa ML, de Melo MB. Epigenetic analysis in placentas from sickle cell disease patients reveals a hypermethylation profile. PLoS One 2022; 17:e0274762. [PMID: 36129958 PMCID: PMC9491616 DOI: 10.1371/journal.pone.0274762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
Pregnancy in Sickle Cell Disease (SCD) women is associated to increased risk of clinical and obstetrical complications. Placentas from SCD pregnancies can present increased abnormal findings, which may lead to placental insufficiency, favoring adverse perinatal outcome. These placental abnormalities are well known and reported, however little is known about the molecular mechanisms, such as epigenetics. Thus, our aim was to evaluate the DNA methylation profile in placentas from women with SCD (HbSS and HbSC genotypes), compared to uncomplicated controls (HbAA). We included in this study 11 pregnant women with HbSS, 11 with HbSC and 21 with HbAA genotypes. Illumina Methylation EPIC BeadChip was used to assess the whole placental DNA methylation. Pyrosequencing was used for array data validation and qRT-PCR was applied for gene expression analysis. Our results showed high frequency of hypermethylated CpGs sites in HbSS and HbSC groups with 73.5% and 76.2% respectively, when compared with the control group. Differentially methylated regions (DMRs) also showed an increased hypermethylation status for the HbSS (89%) and HbSC (86%) groups, when compared with the control group methylation data. DMRs were selected for methylation validation (4 DMRs-HbSS and 3 DMRs the HbSC groups) and after analyses three were validated in the HbSS group, and none in the HbSC group. The gene expression analysis showed differential expression for the PTGFR (-2.97-fold) and GPR56 (3.0-fold) genes in the HbSS group, and for the SPOCK1 (-2.40-fold) and ADCY4 (1.80-fold) genes in the HbSC group. Taken together, these data strongly suggest that SCD (HbSS and HbSC genotypes) can alter placental DNA methylation and lead to gene expression changes. These changes possibly contribute to abnormal placental development and could impact in the clinical course, especially for the fetus, possibly leading to increased risk of abortion, fetal growth restriction (FGR), stillbirth, small for gestational age newborns and prematurity.
Collapse
Affiliation(s)
- Gislene Pereira Gil
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Galina Ananina
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | | | | | - Sueli Matilde da Silva Costa
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Leticia de Carvalho Baptista
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Mirta Tomie Ito
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | | | - Maria Laura Costa
- Department of Obstetrics and Gynecology, University of Campinas, Campinas, São Paulo, Brazil
| | - Mônica Barbosa de Melo
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
15
|
Hjort L, Novakovic B, Cvitic S, Saffery R, Damm P, Desoye G. Placental DNA Methylation in pregnancies complicated by maternal diabetes and/or obesity: State of the Art and research gaps. Epigenetics 2022; 17:2188-2208. [PMID: 35950598 DOI: 10.1080/15592294.2022.2111755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
SUMMARYMaternal diabetes and/or obesity in pregnancy are undoubtedly associated with later disease-risk in the offspring. The placenta, interposed between the mother and the fetus, is a potential mediator of this risk through epigenetic mechanisms, including DNA methylation. In recent years, multiple studies have identified differentially methylated CpG sites in the placental tissue DNA in pregnancies complicated by diabetes and obesity. We reviewed all published original research relevant to this topic and analyzed our findings with the focus of identifying overlaps, contradictions and gaps. Most studies focused on the association of gestational diabetes and/or hyperglycemia in pregnancy and DNA methylation in placental tissue at term. We identified overlaps in results related to specific candidate genes, but also observed a large research gap of pregnancies affected by type 1 diabetes. Other unanswered questions relate to analysis of specific placental cell types and the timing of DNA methylation change in response to diabetes and obesity during pregnancy. Maternal metabolism is altered already in the first trimester involving structural and functional changes in the placenta, but studies into its effects on placental DNA methylation during this period are lacking and urgently needed. Fetal sex is also an important determinant of pregnancy outcome, but only few studies have taken this into account. Collectively, we provide a reference work for researchers working in this large and evolving field. Based on the results of the literature review, we formulate suggestions for future focus of placental DNA methylation studies in pregnancies complicated by diabetes and obesity.
Collapse
Affiliation(s)
- Line Hjort
- Dept. of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Environmental Epigenetics Group, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Boris Novakovic
- Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia.,Dept. of Pediatrics, Melbourne University, Melbourne, VIC, Australia
| | - Silvija Cvitic
- Department of Pediatrics and Adolescent Medicine, Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Medical University of Graz, Austria
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia.,Dept. of Pediatrics, Melbourne University, Melbourne, VIC, Australia
| | - Peter Damm
- Dept. of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark.,Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gernot Desoye
- Dept. of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark.,Dept. of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| |
Collapse
|
16
|
Dietary and Antioxidant Vitamins Limit the DNA Damage Mediated by Oxidative Stress in the Mother-Newborn Binomial. Life (Basel) 2022; 12:life12071012. [PMID: 35888100 PMCID: PMC9323630 DOI: 10.3390/life12071012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 11/16/2022] Open
Abstract
During pregnancy, appropriate nutritional support is necessary for the development of the foetus. Maternal nutrition might protect the foetus from toxic agents such as free radicals due to its antioxidant content. In this study, 90 mothers and their children were recruited. DNA damage mediated by oxidative stress (OS) was determined by the levels of 8-hidroxy-2′-deoxyguanosine (8-OHdG) in the plasma of women and umbilical cord blood. The mothers and newborns were categorised into tertiles according to their 8-OHdG levels for further comparison. No relevant clinical differences were observed in each group. A strong correlation was observed in the mother−newborn binomial for 8-OHdG levels (Rho = 0.694, p < 0.001). In the binomial, a lower level of 8-OHdG was associated with higher consumption of calories, carbohydrates, lipids, and vitamin A (p < 0.05). In addition, the levels of 8-OHdG were only significantly lower in newborns from mothers with a higher consumption of vitamin A and E (p < 0.01). These findings were confirmed by a significant negative correlation between the 8-OHdG levels of newborns and the maternal consumption of vitamins A and E, but not C (Rho = −0.445 (p < 0.001), −0.281 (p = 0.007), and −0.120 (p = 0.257), respectively). Multiple regression analysis showed that the 8-OHdG levels in mothers and newborns inversely correlated with vitamin A (β = −1.26 (p = 0.016) and −2.17 (p < 0.001), respectively) and pregestational body mass index (β = −1.04 (p = 0.007) and −0.977 (p = 0.008), respectively). In conclusion, maternal consumption of vitamins A and E, but not C, might protect newborns from DNA damage mediated by OS.
Collapse
|
17
|
Jeje SO, Adenawoola M, Abosede C. Gestational Nutrition as a Predisposing Factor to Obesity Onset in Offspring: Role for Involvement of Epigenetic Mechanism. Niger J Physiol Sci 2022; 37:1-7. [PMID: 35947841 DOI: 10.54548/njps.v37i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
Maternal lifestyle has been implicated as a predisposing factor in the development of metabolic disorders in adulthood. This lifestyle includes the immediate environment, physical activity and nutrition. Maternal nutrition has direct influence on the developmental programming through biochemical alterations and can lead to modifications in the fetal genome through epigenetic mechanisms. Imbalance in basic micro or macro nutrients due to famine or food deficiency during delicate gestational periods can lead to onset of metabolic syndrome including obesity. A major example is the Dutch famine which led to a serious metabolic disorder in adulthood of affected infants. Notably due to gene variants, individualized responses to nutritional deficiencies are unconventional, therefore intensifying the need to study nutritional genomics during fetal programming. Epigenetic mechanisms can cause hereditary changes without changing the DNA sequence; the major mechanisms include small non-coding RNAs, histone modifications and most stable of all is DNA methylation. The significance association between obesity and DNA methylation is through regulation of genes implicated in lipid and glucose metabolism either directly or indirectly by hypomethylation or hypermethylation. Examples include CPT1A, APOA2, ADRB3 and POMC. Any maternal exposure to malnutrition or overnutrition that can affect genes regulating major metabolic pathways in the fetus, will eventually cause underlying changes that can predispose or cause the onset of metabolic disorder in adulthood. In this review, we examined the interaction between nutrition during gestation and epigenetic programming of metabolic syndrome.
Collapse
|
18
|
Franzago M, Porreca A, D’Ardes M, Di Nicola M, Di Tizio L, Liberati M, Stuppia L, Vitacolonna E. The Obesogenic Environment: Epigenetic Modifications in Placental Melanocortin 4 Receptor Gene Connected to Gestational Diabetes and Smoking. Front Nutr 2022; 9:879526. [PMID: 35571924 PMCID: PMC9100829 DOI: 10.3389/fnut.2022.879526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Background Maternal metabolic insults as well as Gestational Diabetes Mellitus (GDM) influence the fetal health and may affect ‘offspring’s susceptibility to chronic diseases via epigenetic modifications. GDM, the most common metabolic disorder in pregnancy, can be considered the result of complex interactions between genetic and environmental factors. A critical point in this view is the identification of genes which are epigenetically modified under the influence of GDM. The melanocortin 4 receptor (MC4R) gene plays a crucial role in nutritional health by suppressing appetite and participating in energy control regulation. The correlations between pregnant ‘women’s metabolic profiles and placental epigenetic modifications of this gene have been poorly investigated. Objective The aim of this study was to evaluate the effect of GDM and maternal clinical parameters at the third trimester of pregnancy to DNA methylation levels in the placenta at CpG sites of MC4R gene. Design and Methods Socio-demographic and clinical characteristics, Mediterranean diet adherence, smoking habits, and physical activity were assessed at the third trimester of pregnancy of 60 Caucasian pregnant women, of which 33 with GDM. Clinical parameters of the newborns were recorded at birth. MC4R DNA methylation on maternal and fetal sides of the placenta was analyzed using bisulfite pyrosequencing. Results MC4R DNA methylation levels at CpG1 and CpG2 were lower on the fetal side of the placenta in GDM-affected women than in non-GDM-affected recruits (p = 0.033). Moreover, DNA methylation levels on the maternal side at CpG1 were positively related to glucose concentration at 2-h oral glucose tolerance test (OGTT). On the other hand, CpG2 DNA methylation was positively related to both 1-h and 2-h during OGTT. Maternal DNA methylation level at CpG2 was also associated with low density lipoprotein cholesterol (LDL-C) at the third trimester of pregnancy (rho = 0.340, p < 0.05), while CpG1 methylation was negatively related to maternal weight variations at delivery (rho = −0.316, p < 0.05). Significant associations between MC4R DNA methylation on the maternal side and lipid profile at third trimester of pregnancy in women smokers were found. Conclusion Our results suggest that MC4R methylation profile in the placenta is related to maternal metabolic and nutritional conditions, potentially affecting fetal programming and the future metabolic health of the newborn.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
- Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti, Italy
| | - Annamaria Porreca
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Mario D’Ardes
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Luciano Di Tizio
- Department of Obstetrics and Gynaecology, SS. Annunziata Hospital, “G. d’Annunzio” University, Chieti, Italy
| | - Marco Liberati
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti, Italy
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
- Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti, Italy
- *Correspondence: Ester Vitacolonna,
| |
Collapse
|
19
|
Gao M, Cao S, Li N, Liu J, Lyu Y, Li J, Yang X. Risks of overweight in the offspring of women with gestational diabetes at different developmental stages: A meta-analysis with more than half a million offspring. Obes Rev 2022; 23:e13395. [PMID: 34820996 DOI: 10.1111/obr.13395] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022]
Abstract
We conducted a meta-analysis to evaluate the association of maternal gestational diabetes mellitus (GDM) and offspring overweight from birth to adulthood, and to assess the effects of lifestyle interventions in women with GDM on this risk of offspring overweight. We identified literature from PubMed and 12 other electronic databases and retrieved relevant literature published before October 20, 2020. Random-effects model analysis was used to calculate relative risks (RRs) of overweight and weighted mean differences of body mass index among children stratified into different developmental stages. Forty-nine cohort studies (n = 559,377) and four randomized controlled trials (n = 1277) were included. We found that offspring of women with GDM were at an increased risk for overweight with age, from 1.14 (95% confidence interval [CI]: 1.06-1.22) under 5 years, 1.37 (95% CI: 1.31-1.44) at 5 to <10 years, 2.00 (95% CI: 1.79-2.23) at 10 to <18 years, to 2.05 (95% CI: 1.65-2.55) over 18 years of age (p < 0.05 for differences among groups). However, it was not observed that lifestyle interventions for GDM decreased the elevated overweight risk (RR: 0.94, 95% CI: 0.80-1.11, I2 = 0.0%). These findings highlight the need for adopting an active and healthy lifestyle in this high-risk group.
Collapse
Affiliation(s)
- Ming Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Shu Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jinnan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yuanjun Lyu
- Department of Endocrinology, Tianjin Hospital, Tianjin, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.,Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.,Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
20
|
Mora-Janiszewska O, Faryniak-Zuzak A, Darmochwał-Kolarz D. Epigenetic Links between Microbiota and Gestational Diabetes. Int J Mol Sci 2022; 23:1831. [PMID: 35163753 PMCID: PMC8837149 DOI: 10.3390/ijms23031831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 02/01/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is considered a significant and increasing worldwide problem. The growing body of evidence on this topic has allowed us to point out that a hostile intrauterine environment in mothers with GDM via epigenetic mechanisms induces "diabetogenic" and "obesogenic" changes in an offspring's DNA. This sets a vicious intergenerational cycle of metabolic diseases in motion, gradually deteriorating the health of the human population. One of the most important participants of this process seems to be altered microbiota. There is a chance that the identification of specific epigenetic marks may provide a key for future diagnostic, prognostic and therapeutic solutions in the field of personalised medicine. Given the reversibility of most epigenetic changes, there is an opportunity to improve the long-term health of the human population. In this manuscript, we aim to summarise available data on epigenetic changes among women suffering from GDM and their progeny, in association with alterations in the microbiome.
Collapse
|
21
|
Dessì A, Tognazzi C, Bosco A, Pintus R, Fanos V. Metabolomic profiles and microbiota of GDM offspring: The key for future perspective? Front Pediatr 2022; 10:941800. [PMID: 36275053 PMCID: PMC9579340 DOI: 10.3389/fped.2022.941800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Gestational diabetes mellitus (GDM), or any degree of glucose intolerance recognized for the first time during pregnancy, is one of the diseases that most frequently aggravates the course of gestation. Missed or late diagnosis and inadequate treatment are associated with high maternal and fetal morbidity, with possible short- and long-term repercussions. Estimates on the prevalence of GDM are alarming and increasing by about 30% in the last 10-20 years. In addition, there is the negative influence of the SARS-CoV-2 emergency on the glycemic control of pregnant women, making the matter increasingly topical. To date, knowledge on the metabolic maturation of newborns is still incomplete. However, in light of the considerable progress of the theory of "developmental origins of health and disease," the relevant role of the intrauterine environment cannot be overlooked. In fact, due to the high plasticity of the early stages of development, some detrimental metabolic alterations during fetal growth, including maternal hyperglycemia, are associated with a higher incidence of chronic diseases in adult life. In this context, metabolomic analysis which allows to obtain a detailed phenotypic portrait through the dynamic detection of all metabolites in cells, tissues and different biological fluids could be very useful for the early diagnosis and prevention of complications. Indeed, if the diagnostic timing is optimized through the identification of specific metabolites, the detailed understanding of the altered metabolic pathway could also allow better management and more careful monitoring, also from a nutritional profile, of the more fragile children. In this context, a further contribution derives from the analysis of the intestinal microbiota, the main responsible for the fecal metabolome, given its alteration in pregnancies complicated by GDM and the possibility of transmission to offspring. The purpose of this review is to analyze the available data regarding the alterations in the metabolomic profile and microbiota of the offspring of mothers with GDM in order to highlight future prospects for reducing GDM-related complications in children of mothers affected by this disorder.
Collapse
Affiliation(s)
- Angelica Dessì
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Azienda Ospedaliera Universitaria (AOU) Cagliari, University of Cagliari, Cagliari, Italy
| | - Chiara Tognazzi
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Azienda Ospedaliera Universitaria (AOU) Cagliari, University of Cagliari, Cagliari, Italy
| | - Alice Bosco
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Azienda Ospedaliera Universitaria (AOU) Cagliari, University of Cagliari, Cagliari, Italy
| | - Roberta Pintus
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Azienda Ospedaliera Universitaria (AOU) Cagliari, University of Cagliari, Cagliari, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, Azienda Ospedaliera Universitaria (AOU) Cagliari, University of Cagliari, Cagliari, Italy
| |
Collapse
|
22
|
Shashikadze B, Flenkenthaler F, Stöckl JB, Valla L, Renner S, Kemter E, Wolf E, Fröhlich T. Developmental Effects of (Pre-)Gestational Diabetes on Offspring: Systematic Screening Using Omics Approaches. Genes (Basel) 2021; 12:1991. [PMID: 34946940 PMCID: PMC8701487 DOI: 10.3390/genes12121991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/27/2022] Open
Abstract
Worldwide, gestational diabetes affects 2-25% of pregnancies. Due to related disturbances of the maternal metabolism during the periconceptional period and pregnancy, children bear an increased risk for future diseases. It is well known that an aberrant intrauterine environment caused by elevated maternal glucose levels is related to elevated risks for increased birth weights and metabolic disorders in later life, such as obesity or type 2 diabetes. The complexity of disturbances induced by maternal diabetes, with multiple underlying mechanisms, makes early diagnosis or prevention a challenging task. Omics technologies allowing holistic quantification of several classes of molecules from biological fluids, cells, or tissues are powerful tools to systematically investigate the effects of maternal diabetes on the offspring in an unbiased manner. Differentially abundant molecules or distinct molecular profiles may serve as diagnostic biomarkers, which may also support the development of preventive and therapeutic strategies. In this review, we summarize key findings from state-of-the-art Omics studies addressing the impact of maternal diabetes on offspring health.
Collapse
Affiliation(s)
- Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| | - Jan B. Stöckl
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| | - Libera Valla
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eckhard Wolf
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| |
Collapse
|
23
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
24
|
Franzago M, Fraticelli F, Marchioni M, Di Nicola M, Di Sebastiano F, Liberati M, Stuppia L, Vitacolonna E. Fat mass and obesity-associated (FTO) gene epigenetic modifications in gestational diabetes: new insights and possible pathophysiological connections. Acta Diabetol 2021; 58:997-1007. [PMID: 33743080 PMCID: PMC8272710 DOI: 10.1007/s00592-020-01668-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/26/2020] [Indexed: 12/16/2022]
Abstract
AIMS Gestational diabetes mellitus (GDM) can lead to short- and long-term complications for the child. Epigenetic alterations could contribute to explaining the metabolic disturbances associated with foetal programming. Although the role of the FTO gene remains unclear, it affects metabolic phenotypes probably mediated by epigenetic mechanisms. The aim of this study was to assess whether placental DNA epigenetic modifications at FTO promoter-associated cysteine-phosphate-guanine (CpG) sites are correlated with GDM. A secondary aim was to evaluate the association between the placental FTO DNA methylation and the maternal metabolic traits in women with and without GDM. METHODS Socio-demographic characteristics, clinical parameters at the third trimester of pregnancy, Mediterranean diet adherence, and physical activity were assessed in 33 GDM women and 27 controls. Clinical information about the newborns was registered at birth. The FTO rs9939609 (T > A) was genotyped. RESULTS No association between FTO DNA methylation and GDM was found. DNA methylation on the maternal side at the CpG1 was associated with maternal smoking in GDM (p = 0.034), and DNA methylation at the CpG3 was correlated with smoking or former smoking in controls (p = 0.023). A higher level of TGs was correlated with higher foetal placental DNA methylation at the CpG2 (p = 0.036) in GDM. An inverse association between HDL-C and maternal placental DNA methylation at the CpG3 in controls (p = 0.045) was found. An association between FTO rs9939609 and neonatal birthweight (p = 0.033) was detected. CONCLUSIONS In the awareness that the obesity pathophysiology is complex, the study adds a piece to this intricate mosaic.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, "G. D'Annunzio" University, Chieti-Pescara, Via dei Vestini, 66100, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Federica Fraticelli
- Department of Medicine and Aging, School of Medicine and Health Sciences, "G. D'Annunzio" University, Chieti-Pescara, Via dei Vestini, 66100, Chieti, Italy
| | - Michele Marchioni
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, "G.D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, "G.D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Francesca Di Sebastiano
- Department of Obstetric and Gynaecology, SS. Annunziata Hospital, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Marco Liberati
- Department of Medicine and Aging, School of Medicine and Health Sciences, "G. D'Annunzio" University, Chieti-Pescara, Via dei Vestini, 66100, Chieti, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, "G. D'Annunzio" University, Chieti-Pescara, Via dei Vestini, 66100, Chieti, Italy.
- Center for Advanced Studies and Technology (CAST), "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
25
|
Chen C, Jiang Y, Yan T, Chen Y, Yang M, Lv M, Xi F, Lu J, Zhao B, Luo Q. Placental maternally expressed gene 3 differentially methylated region methylation profile is associated with maternal glucose concentration and newborn birthweight. J Diabetes Investig 2021; 12:1074-1082. [PMID: 33090678 PMCID: PMC8169366 DOI: 10.1111/jdi.13432] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS/INTRODUCTION Emerging evidence shows that epigenetic modifications occurring during fetal development in response to intrauterine exposures could be one of the mechanisms involved in the early determinants of adult metabolic disorders. This study aimed to investigate whether the placental maternally expressed gene 3 (MEG3) deoxyribonucleic acid (DNA) methylation profile is associated with maternal gestational diabetes mellitus status and newborn birthweight. MATERIALS AND METHODS Samples for measurement were collected from 23 women with gestational diabetes mellitus and 23 healthy controls. MEG3 gene expression and DNA methylation levels were assessed using quantitative real-time polymerase chain reaction and MethylTargetTM, respectively. Pearson correlation analyses were used to examine associations between placental DNA methylation levels and clinical variables of interest. The associated results were adjusted by multivariate linear regression for maternal age, body mass index, height, gestational age and newborn sex as confounders. RESULTS We found that the DNA methylation levels in the MEG3 differentially methylated region were significantly different between the gestational diabetes mellitus and control groups on the maternal side of the placenta (40.64 ± 2.15 vs 38.33 ± 2.92; P = 0.004). Furthermore, the mean MEG3 DNA methylation levels were correlated positively with maternal fasting glucose concentrations (R = 0.603, P < 0.001) and newborn birthweight (R = 0.568, P < 0.001). CONCLUSIONS The placental DNA methylation status in the MEG3 differentially methylated region was correlated with maternal glucose concentrations and newborn birthweight. These epigenetic adaptations might contribute to late-onset obesity, underlining the adverse intrauterine environment.
Collapse
Affiliation(s)
- Cheng Chen
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Ying Jiang
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Ting Yan
- Jinhua Municipal Central HospitalJinhuaChina
| | - Yuan Chen
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Mengmeng Yang
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Min Lv
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Fangfang Xi
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Juefei Lu
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Baihui Zhao
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiong Luo
- Department of ObstetricsWomen’s HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
26
|
Lizárraga D, García-Gasca A. The Placenta as a Target of Epigenetic Alterations in Women with Gestational Diabetes Mellitus and Potential Implications for the Offspring. EPIGENOMES 2021; 5:epigenomes5020013. [PMID: 34968300 PMCID: PMC8594713 DOI: 10.3390/epigenomes5020013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a pregnancy complication first detected in the second or third trimester in women that did not show evident glucose intolerance or diabetes before gestation. In 2019, the International Diabetes Federation reported that 15.8% of live births were affected by hyperglycemia during pregnancy, of which 83.6% were due to gestational diabetes mellitus, 8.5% were due to diabetes first detected in pregnancy, and 7.9% were due to diabetes detected before pregnancy. GDM increases the susceptibility to developing chronic diseases for both the mother and the baby later in life. Under GDM conditions, the intrauterine environment becomes hyperglycemic, while also showing high concentrations of fatty acids and proinflammatory cytokines, producing morphological, structural, and molecular modifications in the placenta, affecting its function; these alterations may predispose the baby to disease in adult life. Molecular alterations include epigenetic mechanisms such as DNA and RNA methylation, chromatin remodeling, histone modifications, and expression of noncoding RNAs (ncRNAs). The placenta is a unique organ that originates only in pregnancy, and its main function is communication between the mother and the fetus, ensuring healthy development. Thus, this review provides up-to-date information regarding two of the best-documented (epigenetic) mechanisms (DNA methylation and miRNA expression) altered in the human placenta under GDM conditions, as well as potential implications for the offspring.
Collapse
|
27
|
Priyanka PP, Yenugu S. Coiled-Coil Domain-Containing (CCDC) Proteins: Functional Roles in General and Male Reproductive Physiology. Reprod Sci 2021; 28:2725-2734. [PMID: 33942254 DOI: 10.1007/s43032-021-00595-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/22/2021] [Indexed: 01/10/2023]
Abstract
The coiled-coil domain-containing (CCDC) proteins have been implicated in a variety of physiological and pathological processes. Their functional roles vary from their interaction with molecular components of signaling pathways to determining the physiological functions at the cellular and organ level. Thus, they govern important functions like gametogenesis, embryonic development, hematopoiesis, angiogenesis, and ciliary development. Further, they are implicated in the pathogenesis of a large number of cancers. Polymorphisms in CCDC genes are associated with the risk of lifetime diseases. Because of their role in many biological processes, they have been extensively studied. This review concisely presents the functional role of CCDC proteins that have been studied in the last decade. Studies on CCDC proteins continue to be an active area of investigation because of their indispensable functions. However, there is ample opportunity to further understand the involvement of CCDC proteins in many more functions. It is anticipated that basing on the available literature, the functional role of CCDC proteins will be explored much further.
Collapse
Affiliation(s)
| | - Suresh Yenugu
- Department of Animal Biology, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
28
|
Zhu W, Shen Y, Liu J, Fei X, Zhang Z, Li M, Chen X, Xu J, Zhu Q, Zhou W, Zhang M, Liu S, Du J. Epigenetic alternations of microRNAs and DNA methylation contribute to gestational diabetes mellitus. J Cell Mol Med 2020; 24:13899-13912. [PMID: 33085184 PMCID: PMC7753873 DOI: 10.1111/jcmm.15984] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/29/2020] [Indexed: 01/01/2023] Open
Abstract
This study aimed to identify epigenetic alternations of microRNAs and DNA methylation for gestational diabetes mellitus (GDM) diagnosis and treatment using in silico approach. Data of mRNA and miRNA expression microarray (GSE103552 and GSE104297) and DNA methylation data set (GSE106099) were obtained from the GEO database. Differentially expressed genes (DEGs), differentially expressed miRNAs (DEMs) and differentially methylated genes (DMGs) were obtained by limma package. Functional and enrichment analyses were performed with the DAVID database. The protein‐protein interaction (PPI) network was constructed by STRING and visualized in Cytoscape. Simultaneously, a connectivity map (CMap) analysis was performed to screen potential therapeutic agents for GDM. In GDM, 184 low miRNA‐targeting up‐regulated genes and 234 high miRNA‐targeting down‐regulated genes as well as 364 hypomethylation–high‐expressed genes and 541 hypermethylation–low‐expressed genes were obtained. They were mainly enriched in terms of axon guidance, purine metabolism, focal adhesion and proteasome, respectively. In addition, 115 genes (67 up‐regulated and 48 down‐regulated) were regulated by both aberrant alternations of miRNAs and DNA methylation. Ten chemicals were identified as putative therapeutic agents for GDM and four hub genes (IGF1R, ATG7, DICER1 and RANBP2) were found in PPI and may be associated with GDM. Overall, this study identified a series of differentially expressed genes that are associated with epigenetic alternations of miRNA and DNA methylation in GDM. Ten chemicals and four hub genes may be further explored as potential drugs and targets for GDM diagnosis and treatment, respectively.
Collapse
Affiliation(s)
- Weiqiang Zhu
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Shandong University, Jinan, China.,NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Yupei Shen
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Junwei Liu
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Xiaoping Fei
- The First people's Hospital of Kunshan, Kunshan, China
| | - Zhaofeng Zhang
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Min Li
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Xiaohong Chen
- Department of Obstetrics and Gynecology, Shanghai Pudong New Area Health Care Hospital For Women & Children, Shanghai, China
| | - Jianhua Xu
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Qianxi Zhu
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Weijin Zhou
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Shandong University, Jinan, China
| | | | - Jing Du
- NHC Key Lab. of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research), Pharmacy School, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Gagné-Ouellet V, Breton E, Thibeault K, Fortin CA, Desgagné V, Girard Tremblay É, Cardenas A, Guérin R, Perron P, Hivert MF, Bouchard L. Placental Epigenome-Wide Association Study Identified Loci Associated with Childhood Adiposity at 3 Years of Age. Int J Mol Sci 2020; 21:ijms21197201. [PMID: 33003475 PMCID: PMC7582906 DOI: 10.3390/ijms21197201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/22/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to identify placental DNA methylation (DNAm) variations associated with adiposity at 3 years of age. We quantified placental DNAm using the Infinium MethylationEPIC BeadChips. We assessed associations between DNAm at single-CpGs and skinfold thickness using robust linear regression models adjusted for gestational age, child's sex, age at follow-up and cellular heterogeneity. We sought replication of DNAm association with child adiposity in an independent cohort. We quantified placental mRNA levels for annotated gene using qRT-PCR and tested for correlation with DNAm. Lower DNAm at cg22593959 and cg22436429 was associated with higher adiposity (β = -1.18, q = 0.002 and β = -0.82, q = 0.04). The cg22593959 is located in an intergenic region (chr7q31.3), whereas cg22436429 is within the TFAP2E gene (1p34.3). DNAm at cg22593959 and cg22436429 was correlated with mRNA levels at FAM3C (rs = -0.279, p = 0.005) and TFAP2E (rs = 0.216, p = 0.03). In an independent cohort, the association between placental DNAm at cg22593959 and childhood adiposity was of similar strength and direction (β = -3.8 ± 4.1, p = 0.36), yet non-significant. Four genomic regions were also associated with skinfold thickness within FMN1, MAGI2, SKAP2 and BMPR1B genes. We identified placental epigenetic variations associated with adiposity at 3 years of age suggesting that childhood fat accretion patterns might be established during fetal life.
Collapse
Affiliation(s)
- Valérie Gagné-Ouellet
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Edith Breton
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Kathrine Thibeault
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Carol-Ann Fortin
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Véronique Desgagné
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean—Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 5H6, Canada
| | - Élise Girard Tremblay
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA;
| | - Renée Guérin
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean—Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 5H6, Canada
| | - Patrice Perron
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (P.P.); (M.-F.H.)
- Centre de Recherche du CHUS, Sherbrooke, QC J1H 5N4, Canada
| | - Marie-France Hivert
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (P.P.); (M.-F.H.)
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Luigi Bouchard
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean—Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 5H6, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
30
|
Geurtsen ML, Jaddoe VWV, Gaillard R, Felix JF. Associations of maternal early-pregnancy blood glucose and insulin concentrations with DNA methylation in newborns. Clin Epigenetics 2020; 12:134. [PMID: 32894192 PMCID: PMC7487846 DOI: 10.1186/s13148-020-00924-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 08/25/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Intrauterine exposure to a disturbed maternal glucose metabolism is associated with adverse offspring outcomes. DNA methylation is a potential mechanism underlying these associations. We examined whether maternal early-pregnancy glucose and insulin concentrations are associated with newborn DNA methylation. In a population-based prospective cohort study among 935 pregnant women, maternal plasma concentrations of non-fasting glucose and insulin were measured at a median of 13.1 weeks of gestation (95% range 9.4-17.4). DNA methylation was measured using the Infinium HumanMethylation450 BeadChip (Ilumina). We analyzed associations of maternal early-pregnancy glucose and insulin concentrations with single-CpG DNA methylation using robust linear regression models. Differentially methylated regions were analyzed using the dmrff package in R. We stratified the analyses on normal weight versus overweight or obese women. We also performed a look-up of CpGs and differently methylated regions from previous studies to be associated with maternal gestational diabetes, hyperglycemia or hyperinsulinemia, or with type 2 diabetes in adults. RESULTS Maternal early-pregnancy glucose and insulin concentrations were not associated with DNA methylation at single CpGs nor with differentially methylated regions in the total group. In analyses stratified on maternal BMI, maternal early-pregnancy glucose concentrations were associated with DNA methylation at one CpG (cg03617420, XKR6) among normal weight women and at another (cg12081946, IL17D) among overweight or obese women. No stratum-specific associations were found for maternal early-pregnancy insulin concentrations. The two CpGs were not associated with birth weight or childhood glycemic measures (p values > 0.1). Maternal early-pregnancy insulin concentrations were associated with one CpG known to be related to adult type 2 diabetes. Enrichment among nominally significant findings in our maternal early-pregnancy glucose concentrations was found for CpGs identified in a previous study on adult type 2 diabetes. CONCLUSIONS Maternal early-pregnancy glucose concentrations, but not insulin concentrations, were associated with DNA methylation at one CpG each in the subgroups of normal weight and of overweight or obese women. No associations were present in the full group. The role of these CpGs in mechanisms underlying offspring health outcomes needs further study. Future studies should replicate our results in larger samples with early-pregnancy information on maternal fasting glucose metabolism.
Collapse
Affiliation(s)
- Madelon L Geurtsen
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Romy Gaillard
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands.
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
31
|
Kheirkhah Rahimabad P, Arshad SH, Holloway JW, Mukherjee N, Hedman A, Gruzieva O, Andolf E, Kere J, Pershagen G, Almqvist C, Jiang Y, Chen S, Karmaus W. Association of Maternal DNA Methylation and Offspring Birthweight. Reprod Sci 2020; 28:218-227. [PMID: 32754889 DOI: 10.1007/s43032-020-00281-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/27/2020] [Indexed: 04/01/2025]
Abstract
This study aims to evaluate the association of maternal DNA methylation (DNAm) during pregnancy and offspring birthweight. One hundred twenty-two newborn-mother dyads from the Isle of Wight (IOW) cohort were studied to identify differentially methylated cytosine-phosphate-guanine sites (CpGs) in maternal blood associated with offspring birthweight. Peripheral blood samples were drawn from mothers at 22-38 weeks of pregnancy for epigenome-wide DNAm assessment using the Illumina Infinium HumanMethylation450K array. Candidate CpGs were identified using a course of 100 repetitions of a training and testing process with robust regressions. CpGs were considered informative if they showed statistical significance in at least 80% of training and testing samples. Linear mixed models adjusting for covariates were applied to further assess the selected CpGs. The Swedish Born Into Life cohort was used to replicate our findings (n = 33). Eight candidate CpGs corresponding to the genes LMF1, KIF9, KLHL18, DAB1, VAX2, CD207, SCT, SCYL2, DEPDC4, NECAP1, and SFRS3 in mothers were identified as statistically significantly associated with their children's birthweight in the IOW cohort and confirmed by linear mixed models after adjusting for covariates. Of these, in the replication cohort, three CpGs (cg01816814, cg23153661, and cg17722033 with p values = 0.06, 0.175, and 0.166, respectively) associated with four genes (LMF1, VAX2, CD207, and NECAP1) were marginally significant. Biological pathway analyses of three of the genes revealed cellular processes such as endocytosis (possibly sustaining an adequate maternal-fetal interface) and metabolic processes such as regulation of lipoprotein lipase activity (involved in providing substrates for the developing fetus). Our results contribute to an epigenetic understanding of maternal involvement in offspring birthweight. Measuring DNAm levels of maternal CpGs may in the future serve as a diagnostic tool recognizing mothers at risk for pregnancies ending with altered birthweights.
Collapse
Affiliation(s)
- Parnian Kheirkhah Rahimabad
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA.
| | - Syed Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,The David Hide Asthma and Allergy Research Centre, Isle of Wight, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, England, UK
| | - John W Holloway
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, England, UK.,Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Nandini Mukherjee
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Anna Hedman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Olena Gruzieva
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Ellika Andolf
- Department of Clinical Sciences, Danderyd Hospital, Stockholm, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden.,Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Goran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Unit of Pediatric Allergy and Pulmonology at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Su Chen
- Department of Mathematical Sciences, University of Memphis, Memphis, TN, USA
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| |
Collapse
|
32
|
Lima RS, Assis Silva Gomes J, Moreira PR. An overview about DNA methylation in childhood obesity: Characteristics of the studies and main findings. J Cell Biochem 2020; 121:3042-3057. [DOI: 10.1002/jcb.29544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Rafael Silva Lima
- Laboratory of Cell‐Cell Interactions, Department of Morphology, Institute of Biological SciencesFederal University of Minas Gerais Minas Gerais Brazil
| | - Juliana Assis Silva Gomes
- Laboratory of Cell‐Cell Interactions, Department of Morphology, Institute of Biological SciencesFederal University of Minas Gerais Minas Gerais Brazil
| | - Paula Rocha Moreira
- Laboratory of Cell‐Cell Interactions, Department of Morphology, Institute of Biological SciencesFederal University of Minas Gerais Minas Gerais Brazil
| |
Collapse
|
33
|
Franzago M, Santurbano D, Vitacolonna E, Stuppia L. Genes and Diet in the Prevention of Chronic Diseases in Future Generations. Int J Mol Sci 2020; 21:ijms21072633. [PMID: 32290086 PMCID: PMC7178197 DOI: 10.3390/ijms21072633] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 03/30/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Nutrition is a modifiable key factor that is able to interact with both the genome and epigenome to influence human health and fertility. In particular, specific genetic variants can influence the response to dietary components and nutrient requirements, and conversely, the diet itself is able to modulate gene expression. In this context and the era of precision medicine, nutrigenetic and nutrigenomic studies offer significant opportunities to improve the prevention of metabolic disturbances, such as Type 2 diabetes, gestational diabetes, hypertension, and cardiovascular diseases, even with transgenerational effects. The present review takes into account the interactions between diet, genes and human health, and provides an overview of the role of nutrigenetics, nutrigenomics and epigenetics in the prevention of non-communicable diseases. Moreover, we focus our attention on the mechanism of intergenerational or transgenerational transmission of the susceptibility to metabolic disturbances, and underline that the reversibility of epigenetic modifications through dietary intervention could counteract perturbations induced by lifestyle and environmental factors.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
| | | | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Correspondence:
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
34
|
Abstract
The placenta is exposed to metabolic derangements in the maternal and fetal circulation. The effects of the early placental "exposome" determine further trajectories. Overstimulation of the fetal pancreas in early gestation results in fetal hyperinsulinemia, augmenting glucose transfer with adverse effects on the fetus. The manifold placental changes at the end of pregnancy can be regarded as adaptive responses to protect the fetus from diabetes and obesity. The causal role of the placenta, if any, in mediating long-term effects on offspring development is an important area of current and future research.
Collapse
Affiliation(s)
- Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, Graz 8036, Austria.
| | - Mila Cervar-Zivkovic
- Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, Graz 8036, Austria
| |
Collapse
|
35
|
Zhu Z, Cao F, Li X. Epigenetic Programming and Fetal Metabolic Programming. Front Endocrinol (Lausanne) 2019; 10:764. [PMID: 31849831 PMCID: PMC6901800 DOI: 10.3389/fendo.2019.00764] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/21/2019] [Indexed: 12/30/2022] Open
Abstract
Fetal metabolic programming caused by the adverse intrauterine environment can induce metabolic syndrome in adult offspring. Adverse intrauterine environment introduces fetal long-term relatively irreversible changes in organs and metabolism, and thus causes fetal metabolic programming leading metabolic syndrome in adult offspring. Fetal metabolic programming of obesity and insulin resistance plays a key role in this process. The mechanism of fetal metabolic programming is still not very clear. It is suggested that epigenetic programming, also induced by the adverse intrauterine environment, is a critical underlying mechanism of fetal metabolic programming. Fetal epigenetic programming affects gene expression changes and cellular function through epigenetic modifications without DNA nucleotide sequence changes. Epigenetic modifications can be relatively stably retained and transmitted through mitosis and generations, and thereby induce the development of metabolic syndrome in adult offspring. This manuscript provides an overview of the critical role of epigenetic programming in fetal metabolic programming.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Children's Hospital of Soochow University, Suzhou, China
- Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, China
| | - Fang Cao
- Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, China
| | - Xiaozhong Li
- Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
36
|
Hjort L, Novakovic B, Grunnet LG, Maple-Brown L, Damm P, Desoye G, Saffery R. Diabetes in pregnancy and epigenetic mechanisms-how the first 9 months from conception might affect the child's epigenome and later risk of disease. Lancet Diabetes Endocrinol 2019; 7:796-806. [PMID: 31128973 DOI: 10.1016/s2213-8587(19)30078-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/11/2019] [Accepted: 02/21/2019] [Indexed: 12/23/2022]
Abstract
Diabetes in pregnancy is not only associated with increased risk of pregnancy complications and subsequent maternal metabolic disease, but also increases the risk of long-term metabolic disease in the offspring. At the interface between genetic and environmental factors, epigenetic variation established in utero represents a plausible link between the in utero environment and later disease susceptibility. The identification of an epigenetic fingerprint of diabetes in pregnancy linked to the metabolic health of the offspring might provide novel biomarkers for the identification of offspring most at risk, before the onset of metabolic dysfunction, for targeted monitoring and intervention. In this Personal View, we (1) highlight the scale of the problem of diabetes in pregnancy, (2) summarise evidence for the variation in offspring epigenetic profiles following exposure to diabetes in utero, and (3) outline potential future approaches to further understand the mechanisms by which exposure to maternal metabolic dysfunction in pregnancy is transmitted through generations.
Collapse
Affiliation(s)
- Line Hjort
- Department of Endocrinology, Diabetes and Bone-metabolic Research Unit, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Center for Pregnant Women with Diabetes, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; The Danish Diabetes Academy, Odense, Denmark.
| | - Boris Novakovic
- Cancer and Disease Epigenetics, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Pediatrics, Melbourne University, Melbourne, VIC, Australia
| | - Louise G Grunnet
- Department of Endocrinology, Diabetes and Bone-metabolic Research Unit, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; The Danish Diabetes Academy, Odense, Denmark
| | - Louise Maple-Brown
- Wellbeing and Preventable Chronic Diseases Division, Menzies School of Health Research, Darwin, NT, Australia; Endocrinology Department, Royal Darwin Hospital, Darwin, NT, Australia
| | - Peter Damm
- Center for Pregnant Women with Diabetes, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Richard Saffery
- Cancer and Disease Epigenetics, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Pediatrics, Melbourne University, Melbourne, VIC, Australia
| |
Collapse
|
37
|
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: 27] [Impact Index Per Article: 4.5] [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
|
38
|
Franzago M, Fraticelli F, Stuppia L, Vitacolonna E. Nutrigenetics, epigenetics and gestational diabetes: consequences in mother and child. Epigenetics 2019; 14:215-235. [PMID: 30865571 PMCID: PMC6557546 DOI: 10.1080/15592294.2019.1582277] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gestational Diabetes Mellitus (GDM) is the most common metabolic condition during pregnancy and may result in short- and long-term complications for both mother and offspring. The complexity of phenotypic outcomes seems influenced by genetic susceptibility, nutrient-gene interactions and lifestyle interacting with clinical factors. There is strong evidence that not only the adverse genetic background but also the epigenetic modifications in response to nutritional and environmental factors could influence the maternal hyperglycemia in pregnancy and the foetal metabolic programming. In this view, the correlation between epigenetic modifications and their transgenerational effects represents a very interesting field of study. The present review gives insight into the role of gene variants and their interactions with nutrients in GDM. In addition, we provide an overview of the epigenetic changes and their role in the maternal-foetal transmission of chronic diseases. Overall, the knowledge of epigenetic modifications induced by an adverse intrauterine and perinatal environment could shed light on the potential pathophysiological mechanisms of long-term disease development in the offspring and provide useful tools for their prevention.
Collapse
Affiliation(s)
- Marica Franzago
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy.,b Molecular Genetics, Unit , CeSI-Met , Chieti , Italy
| | - Federica Fraticelli
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| | - Liborio Stuppia
- b Molecular Genetics, Unit , CeSI-Met , Chieti , Italy.,c Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| | - Ester Vitacolonna
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| |
Collapse
|
39
|
Kerr B, Leiva A, Farías M, Contreras-Duarte S, Toledo F, Stolzenbach F, Silva L, Sobrevia L. Foetoplacental epigenetic changes associated with maternal metabolic dysfunction. Placenta 2018; 69:146-152. [PMID: 29699712 DOI: 10.1016/j.placenta.2018.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/08/2018] [Accepted: 04/09/2018] [Indexed: 02/08/2023]
Abstract
Metabolic-related diseases are attributed to a sedentary lifestyle and eating habits, and there is now an increased awareness regarding pregnancy as a preponderant window in the programming of adulthood health and disease. The developing foetus is susceptible to the maternal environment; hence, any unfavourable condition will result in foetal physiological adaptations that could have a permanent impact on its health. Some of these alterations are maintained via epigenetic modifications capable of modifying gene expression in metabolism-related genes. Children born to mothers with dyslipidaemia, pregestational or gestational obesity, and gestational diabetes mellitus, have a predisposition to develop metabolic alterations during adulthood. CpG methylation-associated alterations to the expression of several genes in the human placenta play a crucial role in the mother-to-foetus transfer of nutrients and macromolecules. Identification of epigenetic modifications in metabolism-related tissues of offspring from metabolic-altered pregnancies is essential to obtain insights into foetal programming controlling newborn, childhood, and adult metabolism. This review points out the importance of the foetal milieu in the programming and development of human disease and provides evidence of this being the underlying mechanism for the development of adulthood metabolic disorders in maternal dyslipidaemia, pregestational or gestational obesity, and gestational diabetes mellitus.
Collapse
Affiliation(s)
- Bredford Kerr
- Laboratory of Biology, Centro de Estudios Científicos (CECs), Valdivia 5110466, Chile.
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Marcelo Farías
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Susana Contreras-Duarte
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad Del Bío-Bío, Chillán 3780000, Chile
| | - Francisca Stolzenbach
- Laboratory of Biology, Centro de Estudios Científicos (CECs), Valdivia 5110466, Chile; Faculty of Science, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Luis Silva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen (UMCG), Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia.
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW This review outlines recent advances in placental lipid transport in relation to maternal metabolic status and pregnancy outcome. A particular focus of this review will be on the way these findings may influence our understanding of placental transfer of the essential fatty acid docosahexaenoic acid (DHA) which is crucial for fetal neurodevelopment and of lipid transfer as a predisposing factor for childhood obesity. RECENT FINDINGS Placental metabolism may determine the quantity and composition of fatty acids delivered to the fetus. Maternal factors, such as obesity, appear to regulate placental lipid metabolism and may influence fatty acids delivery to the fetus. Although the role of placental metabolism is now recognized, new evidence also suggests important roles for nontraditional fatty acid transporters such as Mfsd2a which facilitates transfer of DHA. SUMMARY Placental lipid metabolism is likely to be a determinant of placental transfer of fatty acids to the fetus. Maternal conditions, such as obesity, have now been shown to regulate placental lipid metabolism and thus may influence fatty acid transfer and fetal development. However, it is not yet clear how regulation of placental lipid metabolism affects fatty acid delivery to the fetus and its long-term health.
Collapse
Affiliation(s)
- Rohan M Lewis
- Faculty of Medicine, University of Southampton, Southampton General Hospital, England, UK
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz
| | | |
Collapse
|