1
|
Cortese R, Cataldo K, Hummel J, Smith G, Ortega M, Richey M, Winn H, Gozal D, Goodman JR. Fetal growth disorders detection during first trimester gestation through comprehensive maternal circulating DNA profiling. Hum Mol Genet 2025:ddaf055. [PMID: 40263746 DOI: 10.1093/hmg/ddaf055] [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: 12/23/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Early diagnosis, close follow-up and timely delivery constitute the main elements for appropriate detection and management of Fetal Growth Disorders (FGD). We hypothesized that fetoplacental FGD-associated alterations can be detected in circulating DNA (cirDNA) samples isolated from maternal blood, as early as the first gestational trimester. METHODS Plasma cirDNA was isolated from samples prospectively collected during first trimester gestation (n = 56). Small, Large and Appropriate for Gestational Age (SGA n = 11, LGA n = 18, and AGA n = 29, respectively) status was determined at birth according to weight and gestational age. cirDNA amount, fragmentation, mitochondrial/nuclear ratio and cirDNA methylation profiles were quantified using qPCR-based assays. Machine learning approaches were applied to build a molecular signature for prediction of LGA and SGA. Prediction accuracy was assessed by Receiver-Operating Curve (ROC) analysis, and Positive and Negative Predictive values (PPV and NPV, respectively) were calculated. RESULTS Total concentration of plasma cirDNA, cirDNA fragmentation and ratio of mitochondrial/nuclear cirDNA were increased in SGA and LGA compared to AGA pregnancies. Out of the 10 selected loci, we detected 5 genes (HSD2, RASSF1, CYP19A1, IL10, and LEP) showing significant differential methylation differences (p < 0.05) across the SGA, AGA and LGA samples at first trimester. cirDNA marker signature discriminated between FGD and AGA pregnancies with high accuracy (AUC > 0.95), achieving 88.8% PPV and 85.7% NPV. CONCLUSIONS Maternal blood cirDNA profiles accurately detects early gestation FGD. The proposed novel marker panel hold great potential for implementation of low invasive approaches for reliable prediction of FGDs, enabling a disruptive path toward precision medicine in FGD.
Collapse
Affiliation(s)
- Rene Cortese
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Department of Pediatrics, School of Medicine, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Institute for Data Science and Informatics, University of Missouri, Columbia, 22 Heinkel Building, MO 65211 United States
| | - Kylie Cataldo
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Department of Pediatrics, School of Medicine, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Justin Hummel
- Institute for Data Science and Informatics, University of Missouri, Columbia, 22 Heinkel Building, MO 65211 United States
| | - Gracie Smith
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Madison Ortega
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Madison Richey
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Hung Winn
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
- Saint Luke's Hospital Maternal Fetal Medicine Specialists, Kansas City, MO, 64111, United States
| | - David Gozal
- Department of Pediatrics, School of Medicine, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| | - Jean Ricci Goodman
- Department of Gynecology, Obstetrics and Women's Health, University of Missouri, 1 Hospital Dr. Columbia, MO 65212, United States
| |
Collapse
|
2
|
Łuszczki E, Wyszyńska J, Dymek A, Drożdż D, González-Ramos L, Hartgring I, García-Carbonell N, Mazur A, Erdine S, Parnarauskienė J, Alvarez-Pitti J. The Effect of Maternal Diet and Lifestyle on the Risk of Childhood Obesity. Metabolites 2024; 14:655. [PMID: 39728436 DOI: 10.3390/metabo14120655] [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: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Childhood obesity is a global health problem that affects at least 41 million children under the age of five. Increased BMI in children is associated with serious long-term health consequences, such as type 2 diabetes, cardiovascular disease, and psychological problems, including depression and low self-esteem. Although the etiology of obesity is complex, research suggests that the diet and lifestyle of pregnant women play a key role in shaping metabolic and epigenetic changes that can increase the risk of obesity in their children. Excessive gestational weight gain, unhealthy dietary patterns (including the Western diet), and pregnancy complications (such as gestational diabetes) are some of the modifiable factors that contribute to childhood obesity. The purpose of this narrative review is to summarize the most important and recent information on the impact of the diet and lifestyle of pregnant women on the risk of childhood obesity. Methods: This article is a narrative review that aims to summarize the available literature on the impact of pregnant women's diet and lifestyle on the risk of obesity in their offspring, with a focus on metabolic and epigenetic mechanisms. Results/Conclusions: Current evidence suggests that a pregnant woman's lifestyle and diet can significantly contribute to lowering the risk of obesity in their offspring. However, further high-quality research is needed to understand better the metabolic and epigenetic relationships concerning maternal factors that predispose offspring to obesity.
Collapse
Affiliation(s)
- Edyta Łuszczki
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Justyna Wyszyńska
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Agnieszka Dymek
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Pediatric Institute, Jagiellonian University Medical College, 31-007 Krakow, Poland
| | - Laura González-Ramos
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Isa Hartgring
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Nuria García-Carbonell
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
| | - Artur Mazur
- Institute of Medical Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Serap Erdine
- Cerrahpasa Faculty of Medicine, Department of Cardiology, Istanbul University-Cerrahpasa, 34320 Istanbul, Turkey
| | - Justė Parnarauskienė
- Pediatric Department, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Julio Alvarez-Pitti
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3
|
Ducros L, Lavoie-Rochon AS, Pichaud N, Lamarre SG. Metabolic rate and mitochondrial physiology adjustments in Arctic char (Salvelinus alpinus) during cyclic hypoxia. J Exp Biol 2024; 227:jeb247834. [PMID: 39319396 DOI: 10.1242/jeb.247834] [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: 04/08/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Diel fluctuations of oxygen levels characterize cyclic hypoxia and pose a significant challenge to wild fish populations. Although recent research has been conducted on the effects of hypoxia and reoxygenation, mechanisms by which fish acclimatize to cyclic hypoxia remain unclear, especially in hypoxia-sensitive species. We hypothesized that acclimation to cyclic hypoxia requires a downregulation of aerobic metabolic rate and an upregulation of mitochondrial respiratory capacities to mitigate constraints on aerobic metabolism and the elevated risk of oxidative stress upon reoxygenation. We exposed Arctic char (Salvelinus alpinus) to 10 days of cyclic hypoxia and measured their metabolic rate and mitochondrial physiology to determine how they cope with fluctuating oxygen concentrations. We measured oxygen consumption as a proxy of metabolic rate and observed that Arctic char defend their standard metabolic rate but decrease their routine metabolic rate during hypoxic phases, presumably through the repression of spontaneous swimming activities. At the mitochondrial level, acute cyclic hypoxia increases oxygen consumption without ADP (CI-LEAK) in the liver and heart. Respiration in the presence of ADP (OXPHOS) temporarily increases in the liver and decreases in the heart. Cytochrome c oxidase oxygen affinity also increases at day 3 in the liver. However, no change occurs in the brain, which is likely primarily preserved through preferential perfusion (albeit not measured in this study). Finally, in vivo measurements of reactive oxygen species revealed the absence of an oxidative burst in mitochondria in the cyclic hypoxia group. Our study shows that Arctic char acclimatize to cyclic hypoxia through organ-specific mitochondrial adjustments.
Collapse
Affiliation(s)
- Loïck Ducros
- Département de Biologie, Université de Moncton, Moncton, NB, Canada, E1A 3E9
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB, Canada, E1A 3E9
| | - A S Lavoie-Rochon
- Département de Biologie, Université de Moncton, Moncton, NB, Canada, E1A 3E9
| | - N Pichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB, Canada, E1A 3E9
| | - S G Lamarre
- Département de Biologie, Université de Moncton, Moncton, NB, Canada, E1A 3E9
| |
Collapse
|
4
|
Libretti A, Savasta F, Nicosia A, Corsini C, De Pedrini A, Leo L, Laganà AS, Troìa L, Dellino M, Tinelli R, Sorrentino F, Remorgida V. Exploring the Father's Role in Determining Neonatal Birth Weight: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1661. [PMID: 39459447 PMCID: PMC11509116 DOI: 10.3390/medicina60101661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
Birth weight, which exhibits variability across different populations, is influenced by a mix of genetic, environmental, and dietary factors originating from both the mother and father. Maternal characteristics, including age, socioeconomic status, prior pregnancies, weight, height, and weight increase throughout pregnancy, have a substantial influence on fetal growth and the health of the infant. On the other hand, the influence of paternal characteristics on the weight of newborns is still not fully comprehended in a consistent manner. Birth weight is an important factor that can help predict various maternal complications, such as the probability of having a C-section, experiencing postpartum hemorrhage or infections. It can also indicate future health challenges like asthma, cognitive impairment, and chronic diseases such as hypertension and diabetes. Nineteen publications were found through a thorough search of the Medline, PubMed, and Scopus databases, which provide insights into how paternal variables contribute to variations in birth weight. Significantly, the age of the father was found to be associated with higher chances of preterm birth and having a smaller size for gestational age in premature infants, while full-term children were more likely to have a larger size for gestational age. In addition, there is a constant correlation between the height of the father and the birth weight of the child. Taller dads are more likely to have babies with a higher birth weight and a lower likelihood of being small for gestational age (SGA). Although there were some discrepancies in the data about the weight and BMI of fathers, it was found that the height of fathers played a significant role in determining the size of the fetus and the weight of the newborn. While there may be differences in the conducted studies, these findings provide valuable insights into the complex connection between parental characteristics and fetal development. This data can be utilized to enhance clinical treatment strategies and enhance our comprehension of outcomes for neonates. Further homogeneous investigations are required to conclusively validate and build upon these findings.
Collapse
Affiliation(s)
- Alessandro Libretti
- Department of Gynaecology and Obstetrics, University Hospital Maggiore della Carità, 28100 Novara, Italy; (F.S.); (A.N.); (A.D.P.); (L.T.); (V.R.)
- School of Gynaecology and Obstetrics, University of Eastern Piedmont, 28100 Novara, Italy
| | - Federica Savasta
- Department of Gynaecology and Obstetrics, University Hospital Maggiore della Carità, 28100 Novara, Italy; (F.S.); (A.N.); (A.D.P.); (L.T.); (V.R.)
- School of Gynaecology and Obstetrics, University of Eastern Piedmont, 28100 Novara, Italy
| | - Anthony Nicosia
- Department of Gynaecology and Obstetrics, University Hospital Maggiore della Carità, 28100 Novara, Italy; (F.S.); (A.N.); (A.D.P.); (L.T.); (V.R.)
- School of Gynaecology and Obstetrics, University of Eastern Piedmont, 28100 Novara, Italy
| | - Christian Corsini
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
- Department of Urology, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alberto De Pedrini
- Department of Gynaecology and Obstetrics, University Hospital Maggiore della Carità, 28100 Novara, Italy; (F.S.); (A.N.); (A.D.P.); (L.T.); (V.R.)
| | - Livio Leo
- Department of Gynecology and Obstetrics, Hopital Beauregard, AUSL Valleè d’Aoste, 11100 Aosta, Italy;
| | - Antonio Simone Laganà
- Unit of Obstetrics and Gynecology, “Paolo Giaccone” Hospital, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Libera Troìa
- Department of Gynaecology and Obstetrics, University Hospital Maggiore della Carità, 28100 Novara, Italy; (F.S.); (A.N.); (A.D.P.); (L.T.); (V.R.)
- School of Gynaecology and Obstetrics, University of Eastern Piedmont, 28100 Novara, Italy
| | - Miriam Dellino
- Department of Interdisciplinary Medicine (DIM), Unit of Obstetrics and Gynecology, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Raffaele Tinelli
- Department of Obstetrics and Gynecology, “Valle d’Itria” Hospital, Martina Franca, 74015 Taranto, Italy;
| | - Felice Sorrentino
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynecology, University of Foggia, 71122 Foggia, Italy;
| | - Valentino Remorgida
- Department of Gynaecology and Obstetrics, University Hospital Maggiore della Carità, 28100 Novara, Italy; (F.S.); (A.N.); (A.D.P.); (L.T.); (V.R.)
- School of Gynaecology and Obstetrics, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
5
|
Kafida M, Karela M, Giakountis A. RNA-Independent Regulatory Functions of lncRNA in Complex Disease. Cancers (Basel) 2024; 16:2728. [PMID: 39123456 PMCID: PMC11311644 DOI: 10.3390/cancers16152728] [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: 07/06/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
During the metagenomics era, high-throughput sequencing efforts both in mice and humans indicate that non-coding RNAs (ncRNAs) constitute a significant fraction of the transcribed genome. During the past decades, the regulatory role of these non-coding transcripts along with their interactions with other molecules have been extensively characterized. However, the study of long non-coding RNAs (lncRNAs), an ncRNA regulatory class with transcript lengths that exceed 200 nucleotides, revealed that certain non-coding transcripts are transcriptional "by-products", while their loci exert their downstream regulatory functions through RNA-independent mechanisms. Such mechanisms include, but are not limited to, chromatin interactions and complex promoter-enhancer competition schemes that involve the underlying ncRNA locus with or without its nascent transcription, mediating significant or even exclusive roles in the regulation of downstream target genes in mammals. Interestingly, such RNA-independent mechanisms often drive pathological manifestations, including oncogenesis. In this review, we summarize selective examples of lncRNAs that regulate target genes independently of their produced transcripts.
Collapse
Affiliation(s)
| | | | - Antonis Giakountis
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, Mezourlo, 41500 Larissa, Greece
| |
Collapse
|
6
|
Teng X, He H, Yu H, Zhang X, Xing J, Shen J, Li C, Wang M, Shao L, Wang Z, Yang H, Zhang Y, Wu Q. LncRNAs in the Dlk1-Dio3 Domain Are Essential for Mid-Embryonic Heart Development. Int J Mol Sci 2024; 25:8184. [PMID: 39125754 PMCID: PMC11311489 DOI: 10.3390/ijms25158184] [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: 06/14/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
The Dlk1-Dio3 domain is important for normal embryonic growth and development. The heart is the earliest developing and functioning organ of the embryo. In this study, we constructed a transcriptional termination model by inserting termination sequences and clarified that the lack of long non-coding RNA (lncRNA) expression in the Dlk1-Dio3 domain caused the death of maternal insertion mutant (MKI) and homozygous mutant (HOMO) mice starting from E13.5. Parental insertion mutants (PKI) can be born and grow normally. Macroscopically, dying MKI and HOMO embryos showed phenomena such as embryonic edema and reduced heart rate. Hematoxylin and eosin (H.E.) staining showed thinning of the myocardium in MKI and HOMO embryos. In situ hybridization (IHC) and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) showed downregulation of lncGtl2, Rian, and Mirg expression in MKI and HOMO hearts. The results of single-cell RNA sequencing (scRNA-Seq) analysis indicated that the lack of lncRNA expression in the Dlk1-Dio3 domain led to reduced proliferation of epicardial cells and may be an important cause of cardiac dysplasia. In conclusion, this study demonstrates that Dlk1-Dio3 domain lncRNAs play an integral role in ventricular development.
Collapse
Affiliation(s)
- Xiangqi Teng
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Hongjuan He
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Haoran Yu
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Ximeijia Zhang
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Jie Xing
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Jiwei Shen
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Chenghao Li
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Mengyun Wang
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Lan Shao
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Ziwen Wang
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Haopeng Yang
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Yan Zhang
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
| | - Qiong Wu
- Faculty of Life Sciences and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China; (X.T.); (H.H.); (H.Y.); (X.Z.); (J.X.); (J.S.); (C.L.); (M.W.); (L.S.); (Z.W.); (H.Y.); (Y.Z.)
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
7
|
Ormindean CM, Ciortea R, Bucuri CE, Măluțan AM, Iuhas CI, Porumb CG, Ormindean V, Roman MP, Nati ID, Suciu V, Mihu D. Obesity, a Single Pathology Influencing Both Mother and Child-A Retrospective Analysis in Hospital Settings. J Pers Med 2024; 14:683. [PMID: 39063937 PMCID: PMC11277859 DOI: 10.3390/jpm14070683] [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: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity, characterized by an excess of adipose tissue, has become a significant global health issue. The prevalence of obesity has increased markedly in recent decades worldwide, with a sharp rise also observed in developing countries, particularly in urban areas. Addressing obesity during pregnancy is crucial for several reasons and presents challenges for specialists in obstetrics and gynecology. OBJECTIVES The aim of the present study was to investigate the correlation between obesity and its implications for childbirth. MATERIALS AND METHODS We conducted a retrospective study involving 1513 patients, grouped into normal-weight, overweight, and obese categories using corrected BMI values. We performed comparative analyses to explore the association between BMI and various outcomes: the method of delivery, the Apgar score at birth, the incidence of fetal distress, fetal birth weight, the presence of pregnancy-associated pathologies, and the occurrence of postpartum hemorrhage. Descriptive statistical analysis was utilized to characterize the demographic and clinical features of the patients and newborns. RESULTS By examining variables such as the occurrence of fetal distress during labor, the Apgar score at delivery, and the mode of delivery, we identified an association between increasing BMI and complications during labor and delivery. The results indicate that a higher BMI is linked with increased complications and variations in the mode of delivery. CONCLUSIONS Obesity is the most common health issue among women of reproductive age and requires long-term care. It can contribute to numerous pregnancy-associated pathologies and affect both mother and child during labor and delivery. Obesity is associated with lower Apgar scores, the increased incidence of fetal distress, and a higher rate of cesarean section deliveries. Although the absolute risk of serious complications for mother, fetus, and newborn is low among women with obesity, adopting healthy eating and exercise behaviors prior to pregnancy, ideally, or as early in pregnancy as possible, can help minimize excessive weight gain during pregnancy.
Collapse
Affiliation(s)
| | - Razvan Ciortea
- 2nd Department of Obstetrics and Gynaecology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (C.M.O.); (C.E.B.); (V.O.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Zhang X, He H, Yu H, Teng X, Wang Z, Li C, Li J, Yang H, Shen J, Wu T, Zhang F, Zhang Y, Wu Q. Maternal RNA transcription in Dlk1-Dio3 domain is critical for proper development of the mouse placental vasculature. Commun Biol 2024; 7:363. [PMID: 38521877 PMCID: PMC10960817 DOI: 10.1038/s42003-024-06038-3] [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: 07/20/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
The placenta is a unique organ for ensuring normal embryonic growth in the uterine. Here, we found that maternal RNA transcription in Dlk1-Dio3 imprinted domain is essential for placentation. PolyA signals were inserted into Gtl2 to establish a mouse model to prevent the expression of maternal RNAs in the domain. The maternal allele knock-in (MKI) and homozygous (HOMO) placentas showed an expanded junctional zone, reduced labyrinth and poor vasculature impacting both fetal and maternal blood spaces. The MKI and HOMO models displayed dysregulated gene expression in the Dlk1-Dio3 domain. In situ hybridization detected Dlk1, Gtl2, Rtl1, miR-127 and Rian dysregulated in the labyrinth vasculature. MKI and HOMO induced Dlk1 to lose imprinting, and DNA methylation changes of IG-DMR and Gtl2-DMR, leading to abnormal gene expression, while the above changes didn't occur in paternal allele knock-in placentas. These findings demonstrate that maternal RNAs in the Dlk1-Dio3 domain are involved in placental vasculature, regulating gene expression, imprinting status and DNA methylation.
Collapse
Affiliation(s)
- Ximeijia Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Hongjuan He
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Haoran Yu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Xiangqi Teng
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Ziwen Wang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Chenghao Li
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Jiahang Li
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Haopeng Yang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Jiwei Shen
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Tong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Fengwei Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Yan Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China.
| |
Collapse
|
9
|
Aryee SND, Owusu-Adjei D, Osei-Amponsah R, Skinner BM, Amuzu-Aweh EN, Ahunu B, Enright A, Sargent CA. Population Structure and Genomic Characterisation of the Ashanti Dwarf Pig of Ghana. Animals (Basel) 2024; 14:792. [PMID: 38473177 DOI: 10.3390/ani14050792] [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: 01/31/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
There is still limited information on the genomic structure and genetic diversity of African pigs. Genetic diversity studies can contribute significantly to the genetic improvement and conservation of African pigs. This study presents a genetic diversity analysis and population structure of pig breeds in Ghana, with a focus on the Ashanti Dwarf pig (ADP), an indigenous pig breed of Ghana. A total of 167 pigs sampled in Ghana and populations consisting of Ashanti Dwarf pigs (n = 106), exotics (mostly European pigs) (n = 11), crosses (between indigenous and exotic breeds) (n = 44), and unknown breeds (nondescript) (n = 6) were genotyped using Porcine SNP60K BeadChip. Moderate heterozygosity levels, ranging from 0.28 for Ashanti Dwarf pigs to 0.31 for exotic pigs (mostly European pigs), were observed. Principal component analysis of the pig populations within Ghana resulted in two distinct clusters of pigs: (i) Northern and (ii) Southern regional clusters. The PCA based on breed also resulted in four clusters: (i) ADPs; (ii) exotics (iii) crossbreeds between ADP and exotics; (iv) unknown breed types. The PCA demonstrated that the clustering was influenced by genetics, geographical location, production systems, and practices. ADMIXTURE-based analysis also showed that the populations within Ghana are admixed. FST analysis revealed SNPs associated with QTLs for traits such as disease resilience and growth among ADP populations within the different regional and ecological zones of Ghana.
Collapse
Affiliation(s)
- Sethlina Naa Dodua Aryee
- Department of Pathology, University of Cambridge, Cambridge CB2 1TN, UK
- Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Dennis Owusu-Adjei
- Department of Animal Science, University of Ghana, Accra P.O. Box LG43, Ghana
| | | | | | | | - Benjamin Ahunu
- Department of Animal Science, University of Ghana, Accra P.O. Box LG43, Ghana
| | - Anton Enright
- Department of Pathology, University of Cambridge, Cambridge CB2 1TN, UK
| | | |
Collapse
|
10
|
Eggermann T. Human Reproduction and Disturbed Genomic Imprinting. Genes (Basel) 2024; 15:163. [PMID: 38397153 PMCID: PMC10888310 DOI: 10.3390/genes15020163] [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: 01/02/2024] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Genomic imprinting is a specific mode of gene regulation which particularly accounts for the factors involved in development. Its disturbance affects the fetus, the course of pregnancy and even the health of the mother. In children, aberrant imprinting signatures are associated with imprinting disorders (ImpDis). These alterations also affect the function of the placenta, which has consequences for the course of the pregnancy. The molecular causes of ImpDis comprise changes at the DNA level and methylation disturbances (imprinting defects/ImpDefs), and there is an increasing number of reports of both pathogenic fetal and maternal DNA variants causing ImpDefs. These ImpDefs can be inherited, but prediction of the pregnancy complications caused is difficult, as they can cause miscarriages, aneuploidies, health issues for the mother and ImpDis in the child. Due to the complexity of imprinting regulation, each pregnancy or patient with suspected altered genomic imprinting requires a specific workup to identify the precise molecular cause and also careful clinical documentation. This review will cover the current knowledge on the molecular causes of aberrant imprinting signatures and illustrate the need to identify this basis as the prerequisite for personalized genetic and reproductive counselling of families.
Collapse
Affiliation(s)
- Thomas Eggermann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH University Aachen, Pauwelsstr. 3, D-52074 Aachen, Germany
| |
Collapse
|
11
|
Chen LN, Shou ZX, Jin X. Interaction Between Genetic Susceptibility and COVID-19 Pathogenesis in Pediatric Multisystem Inflammatory Disorders: The Role of Immune Responses. Viral Immunol 2024; 37:1-11. [PMID: 38271561 DOI: 10.1089/vim.2023.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
Numerous studies have highlighted the emergence of coronavirus disease (COVID-19) symptoms reminiscent of Kawasaki disease in children, including fever, heightened multisystem inflammation, and multiorgan involvement, posing a life-threatening complication. Consequently, extensive research endeavors in pediatric have aimed to elucidate the intricate relationship between COVID-19 infection and the immune system. COVID-19 profoundly impacts immune cells, culminating in a cytokine storm that particularly inflicts damage on the pulmonary system. The gravity and vulnerability to COVID-19 are closely intertwined with the vigor of the immune response. In this context, the human leukocyte antigen (HLA) molecule assumes pivotal significance in shaping immune responses. Genetic scrutiny of HLA has unveiled the presence of at least one deleterious allele in children afflicted with multisystem inflammatory syndrome in children (MIS-C). Furthermore, research has demonstrated that COVID-19 exploits the angiotensin-converting enzyme 2 (ACE-2) receptor, transmembrane serine protease type 2, and various other genes to gain entry into host cells, with individuals harboring ACE-2 polymorphisms being at higher risk. Pediatric studies have employed diverse genetic methodologies, such as genome-wide association studies (GWAS) and whole exome sequencing, to scrutinize target genes. These investigations have pinpointed two specific genomic loci linked to the severity and susceptibility of COVID-19, with the HLA locus emerging as a notable risk factor. In this comprehensive review article, we endeavor to assess the available evidence and consolidate data, offering insights into current clinical practices and delineating avenues for future research. Our objective is to advance early diagnosis, stabilization, and appropriate management strategies to mitigate genetic susceptibility's impact on the incidence of COVID-19 in pediatric patients with multisystem inflammation.
Collapse
Affiliation(s)
- Li-Na Chen
- Department of Pediatric, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Zhang-Xuan Shou
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue Jin
- Department of Pharmacy, Center for Clinical Pharmacy, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
12
|
Guo Z, Liu Y, Zhan S, Cao J, Wang L, Guo J, Li L, Zhang H, Zhong T. Expression patterns and DNA methylation profile of GTL2 gene in goats. Anim Biotechnol 2023; 34:3617-3625. [PMID: 36911908 DOI: 10.1080/10495398.2023.2184698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Gene trap locus 2 (GTL2), a long non-coding paternal imprinting gene, participates in various biological processes, including cell proliferation, differentiation, and apoptosis, by regulating the transcription of target mRNA, which is tightly related to the growth of the organic and maintenance of function. In this study, DNA methylation patterns of CpG islands (CGI) of GTL2 were explored, and its expression level was quantified in six tissues, rumen epithelium cells, and skeletal muscle cells in goats. GTL2 expression levels were measured by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), and the methylation model was confirmed by bisulfite-sequencing PCR (BSP). CGI methylation of GTL2 indicated a moderate methylation (ranging from 81.42 to 86.83%) in the brain, heart, liver, kidney, lung, and longissimus dorsi. GTL2 is most highly expressed in brain tissues, but there is no significant difference in the other five tissues. In addition, in the rumen epithelium cell proliferation, GTL2 expression was highest at 60 h, followed by 72 h, and almost unchanged at 12-48 h. In the skeletal muscle cell differentiation, GTL2 expression was highest at 0 and 24 h, significantly decreasing at 72 and 128 h. Pearson correlation analysis did not indicate a clear relationship between methylation and GTL2 expression levels, suggesting that other regulatory factors may modulate GTL2 expression. This study will provide a better understanding of the expression regulation mechanism of genes in the delta-like homolog 1 gene (DLK1)-GTL2 domain.
Collapse
Affiliation(s)
- Ziwei Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yue Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
13
|
Beiraghdar M, Beiraghdar M, Khosravi S. The methylation status of GATA3 potentially predicts the outcomes of assisted reproductive technologies. HUM FERTIL 2023; 26:1279-1285. [PMID: 36625441 DOI: 10.1080/14647273.2023.2164871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/30/2022] [Indexed: 01/11/2023]
Abstract
Evaluation of methylation status of genes in sperm samples has been suggested for diagnosis of male infertility as well as prognosis of assisted reproductive technologies (ART) outcomes. In this study, we compared the methylation pattern of the GATA3 gene in infertile and fertile men as well as in infertile men with positive and negative ART outcome based on clinical pregnancy. Ejaculates were obtained from 42 infertile men with a negative ART outcome (group 1), 30 infertile men with a positive ART outcome (group 2), and 21 fertile men (control). Then, samples were subjected to genomic DNA isolation and subsequent TUNEL assay and methylation-specific PCR. The number of infertile men with at least one methylated allele of GATA3 was significantly higher compared to the control group (p = 0.022). Also, the number of patients with at least one methylated allele was significantly higher in group 1 compared to group 2 (p = 0.013). Moreover, the TUNEL assay revealed that the amount of sperm DNA fragmentation is higher in group 1 compared to group 2 (p = 0.008). The findings of our study demonstrated that the degree of GATA3 methylation can potentially differentiate between infertile and fertile men and more importantly can potentially predict the outcome of ART.
Collapse
Affiliation(s)
- Mina Beiraghdar
- Department of Biology, Faculty of Basic Science, Islamic Azad University of Center Tehran Branch, Tehran, Iran
| | - Mozhdeh Beiraghdar
- Department of pathology, specialist of anatomical and clinical pathology, University of Isfahan, Isfahan, Iran
| | - Sharifeh Khosravi
- Department of Genetics and Molecular Biology, Isfahan University of Medical Science, Isfahan, Iran
- Genetic Lab in Majesty of Maryam Infertility Center, Martyr Beheshti Hospital, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
14
|
John RM, Higgs MJ, Isles AR. Imprinted genes and the manipulation of parenting in mammals. Nat Rev Genet 2023; 24:783-796. [PMID: 37714957 DOI: 10.1038/s41576-023-00644-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 09/17/2023]
Abstract
Genomic imprinting refers to the parent-of-origin expression of genes, which originates from epigenetic events in the mammalian germ line. The evolution of imprinting may reflect a conflict over resource allocation early in life, with silencing of paternal genes in offspring soliciting increased maternal provision and silencing of maternal genes limiting demands on the mother. Parental caregiving has been identified as an area of potential conflict, with several imprinted genes serendipitously found to directly influence the quality of maternal care. Recent systems biology approaches, based on single-cell RNA sequencing data, support a more deliberate relationship, which is reinforced by the finding that imprinted genes expressed in the offspring influence the quality of maternal caregiving. These bidirectional, reiterative relationships between parents and their offspring are critical both for short-term survival and for lifelong wellbeing, with clear implications for human health.
Collapse
|
15
|
Lopez-Tello J, Yong HEJ, Sandovici I, Dowsett GKC, Christoforou ER, Salazar-Petres E, Boyland R, Napso T, Yeo GSH, Lam BYH, Constancia M, Sferruzzi-Perri AN. Fetal manipulation of maternal metabolism is a critical function of the imprinted Igf2 gene. Cell Metab 2023; 35:1195-1208.e6. [PMID: 37437545 DOI: 10.1016/j.cmet.2023.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/12/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023]
Abstract
Maternal-offspring interactions in mammals involve both cooperation and conflict. The fetus has evolved ways to manipulate maternal physiology to enhance placental nutrient transfer, but the mechanisms involved remain unclear. The imprinted Igf2 gene is highly expressed in murine placental endocrine cells. Here, we show that Igf2 deletion in these cells impairs placental endocrine signaling to the mother, without affecting placental morphology. Igf2 controls placental hormone production, including prolactins, and is crucial to establish pregnancy-related insulin resistance and to partition nutrients to the fetus. Consequently, fetuses lacking placental endocrine Igf2 are growth restricted and hypoglycemic. Mechanistically, Igf2 controls protein synthesis and cellular energy homeostasis, actions dependent on the placental endocrine cell type. Igf2 loss also has additional long-lasting effects on offspring metabolism in adulthood. Our study provides compelling evidence for an intrinsic fetal manipulation system operating in placenta that modifies maternal metabolism and fetal resource allocation, with long-term consequences for offspring metabolic health.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Hannah E J Yong
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A(∗)STAR), 30 Medical Drive, Singapore 117609, Singapore
| | - Ionel Sandovici
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0SW, UK; Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Georgina K C Dowsett
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Efthimia R Christoforou
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Esteban Salazar-Petres
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Rebecca Boyland
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Royal Devon and Exeter Hospital NHS Trust, Barrack Rd, Exeter EX2 5DW, UK
| | - Tina Napso
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Giles S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Brian Y H Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Miguel Constancia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0SW, UK; Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science and, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
16
|
Shi Q, Qi K. Developmental origins of health and disease: Impact of paternal nutrition and lifestyle. Pediatr Investig 2023; 7:111-131. [PMID: 37324600 PMCID: PMC10262906 DOI: 10.1002/ped4.12367] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/29/2023] [Indexed: 09/20/2023] Open
Abstract
Most epidemiological and experimental studies have focused on maternal influences on offspring's health. The impact of maternal undernutrition, overnutrition, hypoxia, and stress is linked to adverse offspring outcomes across a range of systems including cardiometabolic, respiratory, endocrine, and reproduction among others. During the past decade, it has become evident that paternal environmental factors are also linked to the development of diseases in offspring. In this article, we aim to outline the current understanding of the impact of male health and environmental exposure on offspring development, health, and disease and explore the mechanisms underlying the paternal programming of offspring health. The available evidence suggests that poor paternal pre-conceptional nutrition and lifestyle, and advanced age can increase the risk of negative outcomes in offspring, via both direct (genetic/epigenetic) and indirect (maternal uterine environment) effects. Beginning at preconception, and during utero and the early life after birth, cells acquire an epigenetic memory of the early exposure which can be influential across the entire lifespan and program a child's health. Potentially not only mothers but also fathers should be advised that maintaining a healthy diet and lifestyle is important to improve offspring health as well as the parental health status. However, the evidence is mostly based on animal studies, and well-designed human studies are urgently needed to verify findings from animal data.
Collapse
Affiliation(s)
- Qiaoyu Shi
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Kemin Qi
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| |
Collapse
|
17
|
Stoccoro A, Nicolì V, Coppedè F, Grossi E, Fedrizzi G, Menotta S, Lorenzoni F, Caretto M, Carmignani A, Pistolesi S, Burgio E, Fanos V, Migliore L. Prenatal Environmental Stressors and DNA Methylation Levels in Placenta and Peripheral Tissues of Mothers and Neonates Evaluated by Applying Artificial Neural Networks. Genes (Basel) 2023; 14:836. [PMID: 37107594 PMCID: PMC10138241 DOI: 10.3390/genes14040836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Exposure to environmental stressors during pregnancy plays an important role in influencing subsequent susceptibility to certain chronic diseases through the modulation of epigenetic mechanisms, including DNA methylation. Our aim was to explore the connections between environmental exposures during gestation with DNA methylation of placental cells, maternal and neonatal buccal cells by applying artificial neural networks (ANNs). A total of 28 mother-infant pairs were enrolled. Data on gestational exposure to adverse environmental factors and on mother health status were collected through the administration of a questionnaire. DNA methylation analyses at both gene-specific and global level were analyzed in placentas, maternal and neonatal buccal cells. In the placenta, the concentrations of various metals and dioxins were also analyzed. Analysis of ANNs revealed that suboptimal birth weight is associated with placental H19 methylation, maternal stress during pregnancy with methylation levels of NR3C1 and BDNF in placentas and mother's buccal DNA, respectively, and exposure to air pollutants with maternal MGMT methylation. Associations were also observed between placental concentrations of lead, chromium, cadmium and mercury with methylation levels of OXTR in placentas, HSD11B2 in maternal buccal cells and placentas, MECP2 in neonatal buccal cells, and MTHFR in maternal buccal cells. Furthermore, dioxin concentrations were associated with placental RELN, neonatal HSD11B2 and maternal H19 gene methylation levels. Current results suggest that exposure of pregnant women to environmental stressors during pregnancy could induce aberrant methylation levels in genes linked to several pathways important for embryogenesis in both the placenta, potentially affecting foetal development, and in the peripheral tissues of mothers and infants, potentially providing peripheral biomarkers of environmental exposure.
Collapse
Affiliation(s)
- Andrea Stoccoro
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Fabio Coppedè
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Enzo Grossi
- Autism Research Unit, Villa Santa Maria Foundation, 22038 Tavernerio, Italy
| | - Giorgio Fedrizzi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Chemical Department, Via P. Fiorini 5, 40127 Bologna, Italy
| | - Simonetta Menotta
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Chemical Department, Via P. Fiorini 5, 40127 Bologna, Italy
| | - Francesca Lorenzoni
- Division of Neonatology and NICU, Department of Clinical and Experimental Medicine, 56126 Pisa, Italy
| | - Marta Caretto
- Obstetrics and Gynecology Unit 1, Department of Experimental and Clinical Medicine, University of Pisa, 56126 Pisa, Italy
| | - Arianna Carmignani
- Obstetrics and Gynecology Unit 2, Pisa University Hospital, 56126 Pisa, Italy
| | - Sabina Pistolesi
- First Division of Pathology, Department of Laboratory Medicine, Pisa University Hospital, 56126 Pisa, Italy
| | - Ernesto Burgio
- European Cancer and Environment Research Institute (ECERI), 1000 Brussels, Belgium
| | - Vassilios Fanos
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, 09124 Cagliari, Italy
| | - Lucia Migliore
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
- Department of Laboratory Medicine, Pisa University Hospital, 56126 Pisa, Italy
| |
Collapse
|
18
|
Ihirwe RG, Martel J, Rahimi S, Trasler J. Protective and sex-specific effects of moderate dose folic acid supplementation on the placenta following assisted reproduction in mice. FASEB J 2023; 37:e22677. [PMID: 36515682 PMCID: PMC10108070 DOI: 10.1096/fj.202201428r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Epigenetic defects induced by assisted reproductive technologies (ART) have been suggested as a potential mechanism contributing to suboptimal placentation. Here, we hypothesize that ART perturbs DNA methylation (DNAme) and gene expression during early placenta development, leading to abnormal placental phenotypes observed at term. Since folic acid (FA) plays a crucial role in epigenetic regulation, we propose that FA supplementation can rescue ART-induced placental defects. Female mice were placed on a control diet (CD), a moderate 4-fold (FAS4) or high dose 10-fold (FAS10) FA-supplemented diet prior to ART and compared to a natural mating group. ART resulted in 41 and 28 differentially expressed genes (DEGs) in E10.5 female and male placentas, respectively. Many DEGs were implicated in early placenta development and associated with DNAme changes; a number clustered at known imprinting control regions (ICR). In females, FAS4 partially corrected alterations in gene expression while FAS10 showed evidence of male-biased adverse effects. DNAme and gene expression for five genes involved in early placentation (Phlda2, EphB2, Igf2, Peg3, L3mbtl1) were followed up in placentas from normal as well as delayed and abnormal embryos. Phlda2 and Igf2 expression levels were lowest after ART in placentas of female delayed embryos. Moreover, ART concomitantly reduced DNAme at the Kcnq1ot1 ICR which regulates Phlda2 expression; FAS4 partially improved DNAme in a sex-specific manner. In conclusion, ART-associated placental DNAme and transcriptome alterations observed at mid-gestation are sex-specific; they may help explain adverse placental phenotypes detected at term and are partially corrected by maternal moderate dose FA supplementation.
Collapse
Affiliation(s)
- Rita Gloria Ihirwe
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Josée Martel
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Sophia Rahimi
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jacquetta Trasler
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada.,Department of Pediatrics, McGill University, Montreal, Quebec, Canada.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
19
|
Shi Q, Liu X, Fan X, Wang R, Qi K. Paternal dietary ratio of n-6: n-3 polyunsaturated fatty acids programs offspring leptin expression and gene imprinting in mice. Front Nutr 2022; 9:1043876. [PMID: 36618698 PMCID: PMC9816484 DOI: 10.3389/fnut.2022.1043876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
Background This study determined the effects of the paternal dietary ratio of n-6: n-3 polyunsaturated fatty acids (PUFAs) on leptin expression in the offspring and associated gene imprinting in a mouse model. Methods Three- to four-week-old male C57BL/6J mice (F0) were fed an n-3 PUFA-deficient (n-3 D) diet, a diet with normal n-3 PUFA content (n-3 N; n-6: n-3 = 4.3:1), or a diet with a high n-3 PUFA content (n-3 H; n-6: n-3 = 1.5:1) for 8 weeks. Two subsequent generations were generated by mating F0 and F1 male mice with 10-week-old virgin female C57 BL/6J mice, to produce F1 and F2 offspring. Results Compared to the paternal n-3 D diet, paternal n-3 N and n-3 H diets reduced adipose mRNA expression of leptin (Lep) and its plasma concentrations in juvenile F1 male and female offspring, and adult F1 male and F2 female offspring, with upregulated Lep receptor mRNA expression in the hypothalamus. Meanwhile, paternal n-3 N and n-3 H diets altered the expression of the imprinted genes H19, Igf2, Igf2r, Plagl1, Cdkn1c, Kcnq1ot1, Peg3, and Grb10 in the adipose tissue of juvenile and adult F1 males, with almost no effects on F1 females, while more effects were observed in the adult F2 females than F2 males. Principal component analysis verified that Plagl1, Cdkn1c, and Kcnq1ot1 contributed the most to variation in adipose tissue expression in all offspring. Some of these genes (Plagl1, Cdkn1c, Kcnq1ot1, Peg3, and Grb10) were altered by the paternal n-3 N and n-3 H diets in the F1 and F2 generation testes as well. Furthermore, adipose Lep expression was positively correlated with expressions of H19, Igf2r, Plagl1, and Kcnq1ot1 in juvenile F1 males and females, negatively correlated with the Kcnq1ot1 expression in adult F1 males, and positively correlated with the Plagl1 expression in adult F2 females. Conclusion These data imply that paternal Plagl1, Cdkn1c, and Kcnq1ot1 might be part of the pathways involved in offspring leptin programming. Therefore, a lower ratio of n-6: n-3 PUFAs, with higher intake of n-3 PUFAs in paternal pre-conception, may help maintain the offspring's optimal leptin pattern in a sex-specific manner through multiple generations, and thereby, be beneficial for the offspring's long-term health.
Collapse
|
20
|
Zhang L, Chen H, Cui C, Liang L, Ge H, Meng L, Zhang C. Effects of oocyte vitrification on gene expression in the liver and kidney tissues of adult offspring. J Assist Reprod Genet 2022; 39:2635-2646. [PMID: 36223009 PMCID: PMC9723011 DOI: 10.1007/s10815-022-02611-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/31/2022] [Indexed: 12/14/2022] Open
Abstract
Oocyte vitrification is an important assisted reproductive technology (ART) that preserves the fertility of unmarried patients with malignant tumors, and promotes the development of the oocyte donation program. In recent years, the effects of ART, including the vitrification of oocytes and embryos on the health of offspring, have attracted much attention; however, it is difficult to conduct long-term follow-up and biochemical evaluation in humans. In this study, we detected the effect of oocyte vitrification on gene expression in the organs of adult mice offspring by RNA sequencing for the first time. Our results showed that only a small amount of gene expression was significantly affected. Seven genes (Tpm3, Hspe1-rs1, Ntrk2, Cyp4a31, Asic5, Cyp4a14, Retsat) were abnormally expressed in the liver, and ten genes (Lbp, Hspe1-rs1, Prxl2b, Pfn3, Gm9008, Bglap3, Col8a1, Hmgcr, Ero1lb, Ifi44l) were abnormal in the kidney. Several genes were related to metabolism and disease occurrence in the liver or kidney. Besides, we paid special attention to the expression of known imprinted genes and DNA methylation-related genes in adult organs, which are susceptible to oocyte cryopreservation in the preimplantation stage. As a result, some of these transcripts were detected in adult organs, but they were not affected by oocyte vitrification. In conclusion, we first report that oocyte vitrification did not significantly change the global gene expression in offspring organs; nonetheless, it can still influence the transcription of a few functional genes. The potential adverse effects caused by oocyte vitrification need attention and further study.
Collapse
Affiliation(s)
- Lei Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Huanhuan Chen
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Chenchen Cui
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Linlin Liang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Hengtao Ge
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Li Meng
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China.
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China.
| | - Cuilian Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China.
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China.
| |
Collapse
|
21
|
Shi C, Yan Z, Zhang Y, Qin L, Wu W, Gao C, Gao L, Liu J, Cui Y. Effects of putrescine on the quality and epigenetic modification of mouse oocytes during in vitro maturation. Reprod Fertil Dev 2022; 34:957-970. [PMID: 36031717 DOI: 10.1071/rd22064] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
CONTEXT Low ovarian putrescine levels and decreased peak values following luteinising hormone peaks are related to poor oocyte quantity and quality in ageing women. AIMS To investigate the effects of putrescine supplementation in in vitro maturation (IVM) medium on oocyte quality and epigenetic modification. METHODS Germinal vesicle oocytes retrieved from the ovaries of 8-week-old and 9-month-old mice were divided into four groups (the young, young+difluoromethylornithine (DFMO), ageing and ageing+putrescine groups) and cultured in IVM medium with or without 1mM putrescine or DFMO for 16h. The first polar body extrusion (PBE), cleavage and embryonic development were evaluated. Spindles, chromosomes, mitochondria and reactive oxygen species (ROS) were measured. The expression levels of SIRT1, H3K9ac, H3K9me2, H3K9me3, and 5mC levels were evaluated. Sirt1 and imprinted genes were detected. RESULTS The PBE was higher in the ageing+putrescine group than in the ageing group. Putrescine increased the total and inner cell mass cell numbers of blastocysts in ageing oocytes. Putrescine decreased aberrant spindles and chromosome aneuploidy, increased the mitochondrial membrane potential and decreased ROS levels. Putrescine increased SIRT1 expression and attenuated the upregulation of H3K9ac levels in ageing oocytes. Putrescine did not affect 5mC, H3K9me2 or H3K9me3 levels or imprinted gene expression. CONCLUSIONS Putrescine supplementation during IVM improved the maturation and quality of ageing oocytes and promoted embryonic development by decreasing ROS generation, maintaining mitochondrial and spindle function and correcting aberrant epigenetic modification. IMPLICATIONS Putrescine shows application potential for human-assisted reproduction, especially for IVM of oocytes from ageing women.
Collapse
Affiliation(s)
- Chennan Shi
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Zhengjie Yan
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yuexin Zhang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Lianju Qin
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Chao Gao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Li Gao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| |
Collapse
|
22
|
Vidal AC, Moylan CA, Wilder J, Grant DJ, Murphy SK, Hoyo C. Racial disparities in liver cancer: Evidence for a role of environmental contaminants and the epigenome. Front Oncol 2022; 12:959852. [PMID: 36072796 PMCID: PMC9441658 DOI: 10.3389/fonc.2022.959852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/21/2022] [Indexed: 01/09/2023] Open
Abstract
Liver cancer incidence has tripled since the early 1980s, making this disease one of the fastest rising types of cancer and the third leading cause of cancer-related deaths worldwide. In the US, incidence varies by geographic location and race, with the highest incidence in the southwestern and southeastern states and among racial minorities such as Hispanic and Black individuals. Prognosis is also poorer among these populations. The observed ethnic disparities do not fully reflect differences in the prevalence of risk factors, e.g., for cirrhosis that may progress to liver cancer or from genetic predisposition. Likely substantial contributors to risk are environmental factors, including chemical and non-chemical stressors; yet, the paucity of mechanistic insights impedes prevention efforts. Here, we review the current literature and evaluate challenges to reducing liver cancer disparities. We also discuss the hypothesis that epigenetic mediators may provide biomarkers for early detection to support interventions that reduce disparities.
Collapse
Affiliation(s)
- Adriana C. Vidal
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| | - Cynthia A. Moylan
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Duke University, Durham, NC, United States
| | - Julius Wilder
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Duke University, Durham, NC, United States
| | - Delores J. Grant
- Department of Biomedical and Biological Sciences, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, United States
| | - Susan K. Murphy
- Department of Obstetrics and Gynecology, Division of Research, School of Medicine, Duke University, Durham, NC, United States
| | - Cathrine Hoyo
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
23
|
Zubcevic J, Watkins J, Lin C, Bautista B, Hatch HM, Tevosian SG, Hayward LF. Nicotine Exposure during Rodent Pregnancy Alters the Composition of Maternal Gut Microbiota and Abundance of Maternal and Amniotic Short Chain Fatty Acids. Metabolites 2022; 12:metabo12080735. [PMID: 36005607 PMCID: PMC9414314 DOI: 10.3390/metabo12080735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/28/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Tobacco smoking is the leading cause of preventable death. Numerous reports link smoking in pregnancy with serious adverse outcomes, such as miscarriage, stillbirth, prematurity, low birth weight, perinatal morbidity, and infant mortality. Corollaries of consuming nicotine in pregnancy, separate from smoking, are less explored, and the mechanisms of nicotine action on maternal–fetal communication are poorly understood. This study examined alterations in the maternal gut microbiome in response to nicotine exposure during pregnancy. We report that changes in the maternal gut microbiota milieu are an important intermediary that may mediate the prenatal nicotine exposure effects, affect gene expression, and alter fetal exposure to circulating short-chain fatty acids (SCFAs) and leptin during in utero development.
Collapse
Affiliation(s)
- Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
- Correspondence: (J.Z.); (S.G.T.)
| | - Jacqueline Watkins
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Cindy Lin
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Byrell Bautista
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Heather M. Hatch
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Sergei G. Tevosian
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
- Correspondence: (J.Z.); (S.G.T.)
| | - Linda F. Hayward
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
24
|
Zuccarello D, Sorrentino U, Brasson V, Marin L, Piccolo C, Capalbo A, Andrisani A, Cassina M. Epigenetics of pregnancy: looking beyond the DNA code. J Assist Reprod Genet 2022; 39:801-816. [PMID: 35301622 PMCID: PMC9050975 DOI: 10.1007/s10815-022-02451-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/01/2022] [Indexed: 12/19/2022] Open
Abstract
Epigenetics is the branch of genetics that studies the different mechanisms that influence gene expression without direct modification of the DNA sequence. An ever-increasing amount of evidence suggests that such regulatory processes may play a pivotal role both in the initiation of pregnancy and in the later processes of embryonic and fetal development, thus determining long-term effects even in adult life. In this narrative review, we summarize the current knowledge on the role of epigenetics in pregnancy, from its most studied and well-known mechanisms to the new frontiers of epigenetic regulation, such as the role of ncRNAs and the effects of the gestational environment on fetal brain development. Epigenetic mechanisms in pregnancy are a dynamic phenomenon that responds both to maternal-fetal and environmental factors, which can influence and modify the embryo-fetal development during the various gestational phases. Therefore, we also recapitulate the effects of the most notable environmental factors that can affect pregnancy and prenatal development, such as maternal nutrition, stress hormones, microbiome, and teratogens, focusing on their ability to cause epigenetic modifications in the gestational environment and ultimately in the fetus. Despite the promising advancements in the knowledge of epigenetics in pregnancy, more experience and data on this topic are still needed. A better understanding of epigenetic regulation in pregnancy could in fact prove valuable towards a better management of both physiological pregnancies and assisted reproduction treatments, other than allowing to better comprehend the origin of multifactorial pathological conditions such as neurodevelopmental disorders.
Collapse
Affiliation(s)
- Daniela Zuccarello
- Clinical Genetics Unit, Department of Women's and Children's Health, University Hospital of Padova, Padua, Italy.
| | - Ugo Sorrentino
- Clinical Genetics Unit, Department of Women's and Children's Health, University Hospital of Padova, Padua, Italy
| | - Valeria Brasson
- Clinical Genetics Unit, Department of Women's and Children's Health, University Hospital of Padova, Padua, Italy
| | - Loris Marin
- Gynaecological Clinic, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Chiara Piccolo
- Clinical Genetics Unit, Department of Women's and Children's Health, University Hospital of Padova, Padua, Italy
| | | | - Alessandra Andrisani
- Gynaecological Clinic, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Matteo Cassina
- Clinical Genetics Unit, Department of Women's and Children's Health, University Hospital of Padova, Padua, Italy
| |
Collapse
|
25
|
Gutherz OR, Deyssenroth M, Li Q, Hao K, Jacobson JL, Chen J, Jacobson SW, Carter RC. Potential roles of imprinted genes in the teratogenic effects of alcohol on the placenta, somatic growth, and the developing brain. Exp Neurol 2021; 347:113919. [PMID: 34752786 DOI: 10.1016/j.expneurol.2021.113919] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
Despite several decades of research and prevention efforts, fetal alcohol spectrum disorders (FASD) remain the most common preventable cause of neurodevelopmental disabilities worldwide. Animal and human studies have implicated fetal alcohol-induced alterations in epigenetic programming as a chief mechanism in FASD. Several studies have demonstrated fetal alcohol-related alterations in methylation and expression of imprinted genes in placental, brain, and embryonic tissue. Imprinted genes are epigenetically regulated in a parent-of-origin-specific manner, in which only the maternal or paternal allele is expressed, and the other allele is silenced. The chief functions of imprinted genes are in placental development, somatic growth, and neurobehavior-three domains characteristically affected in FASD. In this review, we summarize the growing body of literature characterizing prenatal alcohol-related alterations in imprinted gene methylation and/or expression and discuss potential mechanistic roles for these alterations in the teratogenic effects of prenatal alcohol exposure. Future research is needed to examine potential physiologic mechanisms by which alterations in imprinted genes disrupt development in FASD, which may, in turn, elucidate novel targets for intervention. Furthermore, mechanistic alterations in imprinted gene expression and/or methylation in FASD may inform screening assays that identify individuals with FASD neurobehavioral deficits who may benefit from early interventions.
Collapse
Affiliation(s)
- Olivia R Gutherz
- Institute of Human Nutrition, Columbia University Medical Center, United States of America
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, United States of America
| | - Qian Li
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, United States of America
| | - Ke Hao
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, United States of America
| | - Joseph L Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, United States of America; Department of Human Biology, University of Cape Town Faculty of Health Sciences, South Africa
| | - Jia Chen
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, United States of America
| | - Sandra W Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, United States of America; Department of Human Biology, University of Cape Town Faculty of Health Sciences, South Africa
| | - R Colin Carter
- Institute of Human Nutrition, Columbia University Medical Center, United States of America; Departments of Emergency Medicine and Pediatrics, Columbia University Medical Center, United States of America.
| |
Collapse
|
26
|
Cimino I, Rimmington D, Tung YCL, Lawler K, Larraufie P, Kay RG, Virtue S, Lam BYH, Fagnocchi L, Ma MKL, Saudek V, Zvetkova I, Vidal-Puig A, Yeo GSH, Farooqi IS, Pospisilik JA, Gribble FM, Reimann F, O'Rahilly S, Coll AP. Murine neuronatin deficiency is associated with a hypervariable food intake and bimodal obesity. Sci Rep 2021; 11:17571. [PMID: 34475432 PMCID: PMC8413370 DOI: 10.1038/s41598-021-96278-8] [Citation(s) in RCA: 4] [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: 05/15/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Neuronatin (Nnat) has previously been reported to be part of a network of imprinted genes downstream of the chromatin regulator Trim28. Disruption of Trim28 or of members of this network, including neuronatin, results in an unusual phenotype of a bimodal body weight. To better characterise this variability, we examined the key contributors to energy balance in Nnat+/-p mice that carry a paternal null allele and do not express Nnat. Consistent with our previous studies, Nnat deficient mice on chow diet displayed a bimodal body weight phenotype with more than 30% of Nnat+/-p mice developing obesity. In response to both a 45% high fat diet and exposure to thermoneutrality (30 °C) Nnat deficient mice maintained the hypervariable body weight phenotype. Within a calorimetry system, food intake in Nnat+/-p mice was hypervariable, with some mice consuming more than twice the intake seen in wild type littermates. A hyperphagic response was also seen in Nnat+/-p mice in a second, non-home cage environment. An expected correlation between body weight and energy expenditure was seen, but corrections for the effects of positive energy balance and body weight greatly diminished the effect of neuronatin deficiency on energy expenditure. Male and female Nnat+/-p mice displayed subtle distinctions in the degree of variance body weight phenotype and food intake and further sexual dimorphism was reflected in different patterns of hypothalamic gene expression in Nnat+/-p mice. Loss of the imprinted gene Nnat is associated with a highly variable food intake, with the impact of this phenotype varying between genetically identical individuals.
Collapse
Affiliation(s)
- Irene Cimino
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Debra Rimmington
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Y C Loraine Tung
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust‑MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Pierre Larraufie
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005, Paris, France
| | - Richard G Kay
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Samuel Virtue
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Marcella K L Ma
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Vladimir Saudek
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Ilona Zvetkova
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust‑MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - J Andrew Pospisilik
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Fiona M Gribble
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Frank Reimann
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK
| | - Anthony P Coll
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0SL, UK.
| |
Collapse
|
27
|
Juliusdottir T, Steinthorsdottir V, Stefansdottir L, Sveinbjornsson G, Ivarsdottir EV, Thorolfsdottir RB, Sigurdsson JK, Tragante V, Hjorleifsson KE, Helgadottir A, Frigge ML, Thorgeirsson G, Benediktsson R, Sigurdsson EL, Arnar DO, Steingrimsdottir T, Jonsdottir I, Holm H, Gudbjartsson DF, Thorleifsson G, Thorsteinsdottir U, Stefansson K. Distinction between the effects of parental and fetal genomes on fetal growth. Nat Genet 2021; 53:1135-1142. [PMID: 34282336 DOI: 10.1038/s41588-021-00896-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/11/2021] [Indexed: 01/12/2023]
Abstract
Birth weight is a common measure of fetal growth that is associated with a range of health outcomes. It is directly affected by the fetal genome and indirectly by the maternal genome. We performed genome-wide association studies on birth weight in the genomes of the child and parents and further analyzed birth length and ponderal index, yielding a total of 243 fetal growth variants. We clustered those variants based on the effects of transmitted and nontransmitted alleles on birth weight. Out of 141 clustered variants, 22 were consistent with parent-of-origin-specific effects. We further used haplotype-specific polygenic risk scores to directly test the relationship between adult traits and birth weight. Our results indicate that the maternal genome contributes to increased birth weight through blood-glucose-raising alleles while blood-pressure-raising alleles reduce birth weight largely through the fetal genome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kristjan E Hjorleifsson
- deCODE genetics/Amgen, Reykjavik, Iceland.,Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
| | | | | | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Rafn Benediktsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - David O Arnar
- deCODE genetics/Amgen, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Thora Steingrimsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Obstetrics and Gynecology, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Immunology, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavik, Iceland. .,Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
28
|
Abstract
Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.
Collapse
Affiliation(s)
- Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, and Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Theresa L Powell
- Department of Pediatrics and Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Daniel B Hardy
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
29
|
Cullen SM, Hassan N, Smith-Raska M. Effects of non-inherited ancestral genotypes on offspring phenotypes. Biol Reprod 2021; 105:747-760. [PMID: 34159361 DOI: 10.1093/biolre/ioab120] [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: 02/03/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
It is well established that environmental exposures can modify the profile of heritable factors in an individual's germ cells, ultimately affecting the inheritance of phenotypes in descendants. Similar to exposures, an ancestor's genotype can also affect the inheritance of phenotypes across generations, sometimes in offspring who do not inherit the genetic aberration. This can occur via a variety of prenatal, in utero, or postnatal mechanisms. In this review, we discuss the evidence for this process in mammals, with a focus on examples that are potentially mediated through the germline, while also considering alternate routes of inheritance. Non-inherited ancestral genotypes may influence descendant's disease risk to a much greater extent than currently appreciated, and focused evaluation of this phenomenon may reveal novel mechanisms of inheritance.
Collapse
Affiliation(s)
- Sean M Cullen
- Division of Newborn Medicine, Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1252D, New York, NY 10021
| | - Nora Hassan
- Division of Newborn Medicine, Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1252D, New York, NY 10021
| | - Matthew Smith-Raska
- Division of Newborn Medicine, Department of Pediatrics, Weill Cornell Medicine, 413 East 69th Street, Room 1252D, New York, NY 10021
| |
Collapse
|
30
|
Migliore L, Nicolì V, Stoccoro A. Gender Specific Differences in Disease Susceptibility: The Role of Epigenetics. Biomedicines 2021; 9:652. [PMID: 34200989 PMCID: PMC8228628 DOI: 10.3390/biomedicines9060652] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 01/08/2023] Open
Abstract
Many complex traits or diseases, such as infectious and autoimmune diseases, cancer, xenobiotics exposure, neurodevelopmental and neurodegenerative diseases, as well as the outcome of vaccination, show a differential susceptibility between males and females. In general, the female immune system responds more efficiently to pathogens. However, this can lead to over-reactive immune responses, which may explain the higher presence of autoimmune diseases in women, but also potentially the more adverse effects of vaccination in females compared with in males. Many clinical and epidemiological studies reported, for the SARS-CoV-2 infection, a gender-biased differential response; however, the majority of reports dealt with a comparable morbidity, with males, however, showing higher COVID-19 adverse outcomes. Although gender differences in immune responses have been studied predominantly within the context of sex hormone effects, some other mechanisms have been invoked: cellular mosaicism, skewed X chromosome inactivation, genes escaping X chromosome inactivation, and miRNAs encoded on the X chromosome. The hormonal hypothesis as well as other mechanisms will be examined and discussed in the light of the most recent epigenetic findings in the field, as the concept that epigenetics is the unifying mechanism in explaining gender-specific differences is increasingly emerging.
Collapse
Affiliation(s)
- Lucia Migliore
- Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, 56126 Pisa, Italy; (V.N.); (A.S.)
- Department of Laboratory Medicine, Azienda Ospedaliero Universitaria Pisana, 56124 Pisa, Italy
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, 56126 Pisa, Italy; (V.N.); (A.S.)
| | - Andrea Stoccoro
- Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, 56126 Pisa, Italy; (V.N.); (A.S.)
| |
Collapse
|
31
|
Cannarella R, Crafa A, Condorelli RA, Mongioì LM, La Vignera S, Calogero AE. Relevance of sperm imprinted gene methylation on assisted reproductive technique outcomes and pregnancy loss: a systematic review. Syst Biol Reprod Med 2021; 67:251-259. [PMID: 34080930 DOI: 10.1080/19396368.2021.1909667] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recent evidence suggests that gamete-imprinted genes play a role in embryo and placenta development and growth. This systematic review aimed to evaluate whether altered methylation of sperm-imprinted genes associates with sperm DNA fragmentation (SDF), pregnancy loss rate and assisted reproductive technique (ART) outcome. To accomplish this, Pubmed, MEDLINE, Cochrane, Academic One Files, Google Scholar, and Scopus databases were used for search strategy from each database inception until December 2020. Specific keywords were used. Studies satisfying the PECOS (Population, Exposure, Comparison/Comparator, Outcomes, Study design) model were retrieved. Ten studies could be included in the qualitative analysis. A significant association was reported between increased SDF rate and aberrant methylation of H19/IGF2 and KCNQ1 genes by two studies. A significantly lower H19 methylation was found in patients with idiopathic recurrent pregnancy loss (RPL) and in infertile patients compared to fertile men. Methylation of GLT2, PEG1/MEST, and ZAC/PLACL1 were similar in patients with RPL and controls. The ART outcome was similar in patients with aberrant and normal methylation of H19, SNRPN, KCNQ1OT1, PEG1/MEST, LIT1, PEG3, NESPAS, and GLT2. By contrast, a study showed an association between altered GLT2 methylation and more inferior ART results. If further confirmed by well-sized studies, these data might be helpful to identify possible epigenetic predictors of ART outcome. Particularly, aberrant methylation of H19/IGF2 and KCNQ1 genes might represent interesting targets that deserve further investigation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Laura M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
32
|
Dini P, Kalbfleisch T, Uribe-Salazar JM, Carossino M, Ali HES, Loux SC, Esteller-Vico A, Norris JK, Anand L, Scoggin KE, Rodriguez Lopez CM, Breen J, Bailey E, Daels P, Ball BA. Parental bias in expression and interaction of genes in the equine placenta. Proc Natl Acad Sci U S A 2021; 118:e2006474118. [PMID: 33853939 PMCID: PMC8072238 DOI: 10.1073/pnas.2006474118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Most autosomal genes in the placenta show a biallelic expression pattern. However, some genes exhibit allele-specific transcription depending on the parental origin of the chromosomes on which the copy of the gene resides. Parentally expressed genes are involved in the reciprocal interaction between maternal and paternal genes, coordinating the allocation of resources between fetus and mother. One of the main challenges of studying parental-specific allelic expression (allele-specific expression [ASE]) in the placenta is the maternal cellular remnant at the fetomaternal interface. Horses (Equus caballus) have an epitheliochorial placenta in which both the endometrial epithelium and the epithelium of the chorionic villi are juxtaposed with minimal extension into the uterine mucosa, yet there is no information available on the allelic gene expression of equine chorioallantois (CA). In the current study, we present a dataset of 1,336 genes showing ASE in the equine CA (https://pouya-dini.github.io/equine-gene-db/) along with a workflow for analyzing ASE genes. We further identified 254 potentially imprinted genes among the parentally expressed genes in the equine CA and evaluated the expression pattern of these genes throughout gestation. Our gene ontology analysis implies that maternally expressed genes tend to decrease the length of gestation, while paternally expressed genes extend the length of gestation. This study provides fundamental information regarding parental gene expression during equine pregnancy, a species with a negligible amount of maternal cellular remnant in its placenta. This information will provide the basis for a better understanding of the role of parental gene expression in the placenta during gestation.
Collapse
Affiliation(s)
- Pouya Dini
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke 9820, Belgium
| | - Theodore Kalbfleisch
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202
| | - José M Uribe-Salazar
- Department of Biochemistry and Molecular Medicine, Genome Center, Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA 95616
| | - Mariano Carossino
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
| | - Hossam El-Sheikh Ali
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
- Theriogenology Department, Faculty of Veterinary Medicine, University of Mansoura, 35516, Egypt
| | - Shavahn C Loux
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
| | - Alejandro Esteller-Vico
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
| | - Jamie K Norris
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
| | - Lakshay Anand
- Environmental Epigenetics and Genetics Group, Department of Horticulture, University of Kentucky, Lexington, KY 40546
| | - Kirsten E Scoggin
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
| | - Carlos M Rodriguez Lopez
- Environmental Epigenetics and Genetics Group, Department of Horticulture, University of Kentucky, Lexington, KY 40546
| | - James Breen
- South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Ernest Bailey
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503
| | - Peter Daels
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke 9820, Belgium
| | - Barry A Ball
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40503;
| |
Collapse
|
33
|
Maternal Obesity and Risk of Low Birth Weight, Fetal Growth Restriction, and Macrosomia: Multiple Analyses. Nutrients 2021; 13:nu13041213. [PMID: 33916963 PMCID: PMC8067544 DOI: 10.3390/nu13041213] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 11/17/2022] Open
Abstract
The associations between maternal pre-pregnancy obesity and low birth weight (LBW, <2500 g) remain inconclusive. Therefore, birth weight in a Polish prospective cohort of 912 mothers was investigated depending on the pre-pregnancy body mass index (BMI). The whole cohort and the subgroup of gestational weight gain (GWG) in the range of the Institute of Medicine (IOM) recommendations, as well as 'healthy' women (who did not develop diabetes or hypertension in this pregnancy) were investigated. Adjusted odds ratios (AOR) of the newborn outcomes (with 95% confidence intervals, CI) for obesity (BMI ≥ 30 kg/m2) vs. normal BMI (18.5-24.9 kg/m2) were calculated using multiple logistic regression. Risk profiles (in the Lowess method) were presented for BMI values (kg/m2) and threshold BMI values were calculated. (1) In the cohort, LBW affected 6.6% of pregnancies, fetal growth restriction (FGR) 2.3%, and macrosomia 10.6%. (2) The adjusted risk of macrosomia was more than three-fold higher for obesity compared to normal BMI in the whole cohort (AOR = 3.21 (1.69-6.1), p < 0.001) and the result was maintained in the subgroups. A 17-fold higher adjusted LBW risk for obesity was found (AOR = 17.42 (1.5-202.6), p = 0.022), but only in the normal GWG subgroup. The FGR risk profile was U-shaped: in the entire cohort, the risk was more than three times higher for obesity (AOR = 3.12 (1.02-9.54), p = 0.045) and underweight (AOR = 3.84 (1.13-13.0), p = 0.031). (3) The risk profiles showed that the highest BMI values were found to be associated with a higher risk of these three newborn outcomes and the threshold BMI was 23.7 kg/m2 for macrosomia, 26.2 kg/m2 for LBW, and 31.8 kg/m2 for FGR. These results confirm the multidirectional effects of obesity on fetal growth (low birth weight, fetal growth restriction, and macrosomia). The results for LBW were heavily masked by the effects of abnormal gestational weight gain.
Collapse
|
34
|
Caniçais C, Vasconcelos S, Ramalho C, Marques CJ, Dória S. Deregulation of imprinted genes expression and epigenetic regulators in placental tissue from intrauterine growth restriction. J Assist Reprod Genet 2021; 38:791-801. [PMID: 33389447 PMCID: PMC8079450 DOI: 10.1007/s10815-020-02047-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/21/2020] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Intrauterine growth restriction (IUGR) is a fetal growth complication that can be caused by ineffective nutrient transfer from the mother to the fetus via the placenta. Abnormal placental development and function have been correlated with abnormal expression of imprinted genes, which are regulated by epigenetic modifications at imprinting control regions (ICRs). In this study, we analyzed the expression of imprinted genes known to be involved in fetal growth and epigenetic regulators involved in DNA methylation, as well as DNA methylation at the KvDMR1 imprinting control region and global levels of DNA hydroxymethylation, in IUGR cases. METHODS Expression levels of imprinted genes and epigenetic regulators were analyzed in term placental samples from 21 IUGR cases and 9 non-IUGR (control) samples, by RT-qPCR. Additionally, KvDMR1 methylation was analyzed by bisulfite sequencing and combined bisulfite restriction analysis (COBRA) techniques. Moreover, global DNA methylation and hydroxymethylation levels were also measured. RESULTS We observed increased expression of PHLDA2, CDKN1C, and PEG10 imprinted genes and of DNMT1, DNMT3A, DNMT3B, and TET3 epigenetic regulators in IUGR placentas. No differences in methylation levels at the KvDMR1 were observed between the IUGR and control groups; similarly, no differences in global DNA methylation and hydromethylation were detected. CONCLUSION Our study shows that deregulation of epigenetic mechanisms, namely increased expression of imprinted genes and epigenetic regulators, might be associated with IUGR etiology. Therefore, this study adds knowledge to the molecular mechanisms underlying IUGR, which may contribute to novel prediction tools and future therapeutic options for the management of IUGR pregnancies.
Collapse
Affiliation(s)
- Carla Caniçais
- Department of Genetics, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Sara Vasconcelos
- Department of Genetics, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Carla Ramalho
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hospital São João, Porto, Portugal
| | - C Joana Marques
- Department of Genetics, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
| | - Sofia Dória
- Department of Genetics, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
| |
Collapse
|
35
|
Scagliotti V, Esse R, Willis TL, Howard M, Carrus I, Lodge E, Andoniadou CL, Charalambous M. Dynamic Expression of Imprinted Genes in the Developing and Postnatal Pituitary Gland. Genes (Basel) 2021; 12:genes12040509. [PMID: 33808370 PMCID: PMC8066104 DOI: 10.3390/genes12040509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/19/2022] Open
Abstract
In mammals, imprinted genes regulate many critical endocrine processes such as growth, the onset of puberty and maternal reproductive behaviour. Human imprinting disorders (IDs) are caused by genetic and epigenetic mechanisms that alter the expression dosage of imprinted genes. Due to improvements in diagnosis, increasing numbers of patients with IDs are now identified and monitored across their lifetimes. Seminal work has revealed that IDs have a strong endocrine component, yet the contribution of imprinted gene products in the development and function of the hypothalamo-pituitary axis are not well defined. Postnatal endocrine processes are dependent upon the production of hormones from the pituitary gland. While the actions of a few imprinted genes in pituitary development and function have been described, to date there has been no attempt to link the expression of these genes as a class to the formation and function of this essential organ. This is important because IDs show considerable overlap, and imprinted genes are known to define a transcriptional network related to organ growth. This knowledge deficit is partly due to technical difficulties in obtaining useful transcriptomic data from the pituitary gland, namely, its small size during development and cellular complexity in maturity. Here we utilise high-sensitivity RNA sequencing at the embryonic stages, and single-cell RNA sequencing data to describe the imprinted transcriptome of the pituitary gland. In concert, we provide a comprehensive literature review of the current knowledge of the role of imprinted genes in pituitary hormonal pathways and how these relate to IDs. We present new data that implicate imprinted gene networks in the development of the gland and in the stem cell compartment. Furthermore, we suggest novel roles for individual imprinted genes in the aetiology of IDs. Finally, we describe the dynamic regulation of imprinted genes in the pituitary gland of the pregnant mother, with implications for the regulation of maternal metabolic adaptations to pregnancy.
Collapse
Affiliation(s)
- Valeria Scagliotti
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London SE19RT, UK; (V.S.); (R.C.F.E.); (I.C.)
| | - Ruben Esse
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London SE19RT, UK; (V.S.); (R.C.F.E.); (I.C.)
| | - Thea L. Willis
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE19RT, UK; (T.L.W.); (E.L.); (C.L.A.)
| | - Mark Howard
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, London SE19RT, UK;
| | - Isabella Carrus
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London SE19RT, UK; (V.S.); (R.C.F.E.); (I.C.)
| | - Emily Lodge
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE19RT, UK; (T.L.W.); (E.L.); (C.L.A.)
| | - Cynthia L. Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE19RT, UK; (T.L.W.); (E.L.); (C.L.A.)
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Marika Charalambous
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London SE19RT, UK; (V.S.); (R.C.F.E.); (I.C.)
- Correspondence:
| |
Collapse
|
36
|
Roque-Jiménez JA, Rosa-Velázquez M, Pinos-Rodríguez JM, Vicente-Martínez JG, Mendoza-Cervantes G, Flores-Primo A, Lee-Rangel HA, Relling AE. Role of Long Chain Fatty Acids in Developmental Programming in Ruminants. Animals (Basel) 2021; 11:ani11030762. [PMID: 33801880 PMCID: PMC8001802 DOI: 10.3390/ani11030762] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The objective of the current review is to provide a broad perspective on developmental program aspects of dietary n-3 FA supplementation in ruminants during pre-conception, conception, pregnancy, early life, including its effects on production, lipid metabolism, and health of the offspring. Offspring growth and metabolism could change depending on the FA profile and the stage of gestation when the dam is supplemented. Despite this extended review we are highlighting areas that we consider that there is a lack of information. Abstract Nutrition plays a critical role in developmental programs. These effects can be during gametogenesis, gestation, or early life. Omega-3 polyunsaturated fatty acids (PUFA) are essential for normal physiological functioning and for the health of humans and all domestic species. Recent studies have demonstrated the importance of n-3 PUFA in ruminant diets during gestation and its effects on pre-and postnatal offspring growth and health indices. In addition, different types of fatty acids have different metabolic functions, which affects the developmental program differently depending on when they are supplemented. This review provides a broad perspective of the effect of fatty acid supplementation on the developmental program in ruminants, highlighting the areas of a developmental program that are better known and the areas that more research may be needed.
Collapse
Affiliation(s)
- José Alejandro Roque-Jiménez
- Facultad de Agronomía y Veterinaria, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78321, Mexico; (J.A.R.-J.); (H.A.L.-R.)
| | - Milca Rosa-Velázquez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91710, Mexico; (M.R.-V.); (J.M.P.-R.); (J.G.V.-M.); (A.F.-P.)
| | - Juan Manuel Pinos-Rodríguez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91710, Mexico; (M.R.-V.); (J.M.P.-R.); (J.G.V.-M.); (A.F.-P.)
| | - Jorge Genaro Vicente-Martínez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91710, Mexico; (M.R.-V.); (J.M.P.-R.); (J.G.V.-M.); (A.F.-P.)
| | | | - Argel Flores-Primo
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91710, Mexico; (M.R.-V.); (J.M.P.-R.); (J.G.V.-M.); (A.F.-P.)
| | - Héctor Aarón Lee-Rangel
- Facultad de Agronomía y Veterinaria, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78321, Mexico; (J.A.R.-J.); (H.A.L.-R.)
| | - Alejandro E. Relling
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
- Correspondence: ; Tel.: +1-330-263-3900
| |
Collapse
|
37
|
Salimi M, Shirazi A, Norouzian M, Jafari A, Edalatkhah H, Mehravar M, Majidi M, Mehrazar MM. H19/Igf2 Expression and Methylation of Histone 3 in Mice Chimeric Blastocysts. Rep Biochem Mol Biol 2021; 9:357-365. [PMID: 33649730 DOI: 10.29252/rbmb.9.3.357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Background Currently, the efficient production of chimeric mice and their survival are still challenging. Recent researches have indicated that preimplantation embryo culture media and manipulation lead to abnormal methylation of histone in the H19/Igf2 promotor region and consequently alter their gene expression pattern. This investigation was designed to evaluate the relationship between the methylation state of histone H3 and H19/Igf2 expression in mice chimeric blastocysts. Methods Mouse 129/Sv embryonic stem cells (mESCs) expressing the green fluorescent protein (mESCs-GFP) were injected into the perivitelline space of 2.5 days post-coitis (dpc) embryos (C57BL/6) using a micromanipulator. H3K4 and H3K9 methylation, and H19 and Igf2 expression was measured by immunocytochemistry and q-PCR, respectively, in blastocysts. Results Histone H3 trimethylation in H3K4 and H3K9 in chimeric blastocysts was significantly less and greater, respectively (p< 0.05), than in controls. H19 expression was significantly less (p< 0.05), while Igf2 expression was less, but not significantly so, in chimeric than in control blastocysts. Conclusion Our results showed, that the alteration ofH3K4me3 and H3K9me3 methylation, change H19/Igf2 expression in chimeric blastocysts.
Collapse
Affiliation(s)
- Maryam Salimi
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.,Department of Gametes and Cloning, Research Institute of Animal Embryo Technology, Shahrekord University, Shahrekord, Iran
| | - Mohsen Norouzian
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ameneh Jafari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haleh Edalatkhah
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Maryam Mehravar
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Majidi
- Food and Drug Laboratory Research Center, Food and Drug Organization, MOH & ME, Tehran, Iran
| | - Mohammad Mahdi Mehrazar
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
38
|
|
39
|
Maternal Melatonin Deficiency Leads to Endocrine Pathologies in Children in Early Ontogenesis. Int J Mol Sci 2021; 22:ijms22042058. [PMID: 33669686 PMCID: PMC7922827 DOI: 10.3390/ijms22042058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
The review summarizes the results of experimental and clinical studies aimed at elucidating the causes and pathophysiological mechanisms of the development of endocrine pathology in children. The modern data on the role of epigenetic influences in the early ontogenesis of unfavorable factors that violate the patterns of the formation of regulatory mechanisms during periods of critical development of fetal organs and systems and contribute to the delayed development of pathological conditions are considered. The mechanisms of the participation of melatonin in the regulation of metabolic processes and the key role of maternal melatonin in the formation of the circadian system of regulation in the fetus and in the protection of the genetic program of its morphofunctional development during pregnancy complications are presented. Melatonin, by controlling DNA methylation and histone modification, prevents changes in gene expression that are directly related to the programming of endocrine pathology in offspring. Deficiency and absence of the circadian rhythm of maternal melatonin underlies violations of the genetic program for the development of hormonal and metabolic regulatory mechanisms of the functional systems of the child, which determines the programming and implementation of endocrine pathology in early ontogenesis, contributing to its development in later life. The significance of this factor in the pathophysiological mechanisms of endocrine disorders determines a new approach to risk assessment and timely prevention of offspring diseases even at the stage of family planning.
Collapse
|
40
|
Chang S, Wang Y, Xin Y, Wang S, Luo Y, Wang L, Zhang H, Li J. DNA methylation abnormalities of imprinted genes in congenital heart disease: a pilot study. BMC Med Genomics 2021; 14:4. [PMID: 33407475 PMCID: PMC7789576 DOI: 10.1186/s12920-020-00848-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is resulted from the interaction of genetic aberration and environmental factors. Imprinted genes, which are regulated by epigenetic modifications, are essential for the normal embryonic development. However, the role of imprinted genes in the etiology of CHD remains unclear. METHODS After the samples were treated with bisulfate salt, imprinted genes methylation were measured by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. T test and One-way ANOVA were performed to evaluate the differences among groups. Odds ratios (ORs) were performed to evaluate the incidence risk of CHD in relation to methylation levels. RESULTS We investigated the alterations of imprinted gene germline differential methylation regions (gDMRs) methylation in patients with CHD. Eighteen imprinted genes that are known to affect early embryonic development were selected and the methylation modification genes were detected by massarray in 27 CHD children and 28 healthy children. Altered gDMR methylation level of 8 imprinted genes was found, including 2 imprinted genes with hypermethylation of GRB10 and MEST and 6 genes with hypomethylation of PEG10, NAP1L5, INPP5F, PLAGL1, NESP and MEG3. Stratified analysis showed that the methylation degree of imprinted genes was different in different types of CHD. Risk analysis showed that 6 imprinted genes, except MEST and NAP1L5, within a specific methylation level range were the risk factors for CHD CONCLUSION: Altered methylation of imprinted genes is associated with CHD and varies in different types of CHD. Further experiments are warranted to identify the methylation characteristics of imprinted genes in different types of CHD and clarify the etiologies of imprinted genes in CHD.
Collapse
Affiliation(s)
- Shaoyan Chang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yubo Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yu Xin
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shuangxing Wang
- Department of Cardiac Surgery, Children's Hospital Affiliated to Capital Institute of Pediatrics, No. 2 Yabao Road, Chao Yang District, Beijing, 100020, China
| | - Yi Luo
- Department of Cardiac Surgery, Children's Hospital Affiliated to Capital Institute of Pediatrics, No. 2 Yabao Road, Chao Yang District, Beijing, 100020, China
| | - Li Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hui Zhang
- Department of Cardiac Surgery, Children's Hospital Affiliated to Capital Institute of Pediatrics, No. 2 Yabao Road, Chao Yang District, Beijing, 100020, China.
| | - Jia Li
- Clinical Physiology Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou City, 510000, Guangdong Province, China.
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510000, Guangdong Province, China.
| |
Collapse
|
41
|
Zhao H, Tang J, Zhu Q, He H, Li S, Jin L, Zhang X, Zhu L, Guo J, Zhang D, Luo Q, Chen G. Associations of prenatal heavy metals exposure with placental characteristics and birth weight in Hangzhou Birth Cohort: Multi-pollutant models based on elastic net regression. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 742:140613. [PMID: 32640390 DOI: 10.1016/j.scitotenv.2020.140613] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The human placenta is vulnerable to environmental pollutants, but the associations between exposure to multiple, correlated metals and placental characteristics have not been studied. METHODS The current study population was derived from the Hangzhou Birth Cohort Study. Whole blood and urine samples were collected from mothers during 20-28 gestational week. The concentrations of 11 metals in blood and 11 metals in urine were evaluated by inductively coupled plasma mass spectrometry. The data on placental characteristics and birth weight were retrieved from medical records. The elastic net (ENET) model combined with unpenalized regression model was employed to estimate the relationship between levels of metals and placental characteristics (placental weight, chorionic disc area, chorionic disc eccentricity, placental thickness, placental-fetal birth weight ratio) and birth weight. Mediation analysis was performed to explore the mediated effect of placenta on the association of prenatal metals exposure with birth weight. RESULTS Among 512 participants with urine metal levels, the ENET model retained Cadmium (Cd) and Selenium (Se) for placental weight. Further unpenalized regression model including Cd and Se simultaneously showed that one-unite increased natural-logarithm (ln)-transformed urine creatinine corrected (CC) Cd levels was associated with reductions in placental weight of -7.2 g (95% confidence interval (CI): -14.0, -0.4). Among 483 participants with blood metal levels, similarly, blood Cd levels were negatively associated with placental weight (β = -7.5, 95% CI: -17.0, 1.9). Furthermore, mediation analysis demonstrated that urine CC-Cd level was associated with a 21.3 g decrease (95% CI: -42.0, -2.5, p = 0.024) in birth weight through a reduction in placental weight, while blood Cd levels presented a negative association at borderline significance. CONCLUSION Our findings suggest a mediation effect of the placenta in the relationship between prenatal Cd exposure and lower birth weight. Additional studies with repeated assessment of exposure and more placental parameters are warranted to confirm this relationship.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Tang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Qinheng Zhu
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongsen He
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Shuai Li
- Department of Clinical Laboratory, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanfei Jin
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Zhang
- Jiashan County Center for Disease Control and Prevention, Jiaxing, China
| | - Longtao Zhu
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Guo
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiong Luo
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guangdi Chen
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Bioelectromagnetics Laboratory, Institute of Environmental Health, Zhejiang University School of Public Health, Hangzhou, China.
| |
Collapse
|
42
|
Bowman CE, Arany Z, Wolfgang MJ. Regulation of maternal-fetal metabolic communication. Cell Mol Life Sci 2020; 78:1455-1486. [PMID: 33084944 DOI: 10.1007/s00018-020-03674-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
Pregnancy may be the most nutritionally sensitive stage in the life cycle, and improved metabolic health during gestation and early postnatal life can reduce the risk of chronic disease in adulthood. Successful pregnancy requires coordinated metabolic, hormonal, and immunological communication. In this review, maternal-fetal metabolic communication is defined as the bidirectional communication of nutritional status and metabolic demand by various modes including circulating metabolites, endocrine molecules, and other secreted factors. Emphasis is placed on metabolites as a means of maternal-fetal communication by synthesizing findings from studies in humans, non-human primates, domestic animals, rabbits, and rodents. In this review, fetal, placental, and maternal metabolic adaptations are discussed in turn. (1) Fetal macronutrient needs are summarized in terms of the physiological adaptations in place to ensure their proper allocation. (2) Placental metabolite transport and maternal physiological adaptations during gestation, including changes in energy budget, are also discussed. (3) Maternal nutrient limitation and metabolic disorders of pregnancy serve as case studies of the dynamic nature of maternal-fetal metabolic communication. The review concludes with a summary of recent research efforts to identify metabolites, endocrine molecules, and other secreted factors that mediate this communication, with particular emphasis on serum/plasma metabolomics in humans, non-human primates, and rodents. A better understanding of maternal-fetal metabolic communication in health and disease may reveal novel biomarkers and therapeutic targets for metabolic disorders of pregnancy.
Collapse
Affiliation(s)
- Caitlyn E Bowman
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zoltan Arany
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
43
|
Jiménez KM, Morel A, Parada-Niño L, Alejandra González-Rodriguez M, Flórez S, Bolívar-Salazar D, Becerra-Bayona S, Aguirre-García A, Gómez-Murcia T, Fernanda Castillo L, Carlosama C, Ardila J, Vaiman D, Serrano N, Laissue P. Identifying new potential genetic biomarkers for HELLP syndrome using massive parallel sequencing. Pregnancy Hypertens 2020; 22:181-190. [PMID: 33059327 DOI: 10.1016/j.preghy.2020.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 07/20/2020] [Accepted: 09/05/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Preeclampsia (PE) is a frequently occurring multisystemic disease affecting ~5% of pregnancies. PE patients may develop HELLP syndrome (haemolysis, elevated liver enzymes, and low platelet), a mother and foetus life-threatening condition. Research into HELLP's genetic origin has been relatively unsuccessful, mainly because normal placental function and blood pressure regulation involve the fine-regulation of hundreds of genes. OBJECTIVE To identify new genes and mutations constituting potential biomarkers for HELLP syndrome. STUDY DESIGN The present case-control study involved whole-exome sequencing of 79 unrelated HELLP women. Candidate variants were screened in a control population constituted by 176 individuals. Stringent bioinformatics filters were used for selecting potentially etiological sequence variants in a subset of 487 genes. We used robust in silico mutation modelling for predicting the potential effect on protein structure. RESULTS We identified numerous sequence variants in genes related to angiogenesis/coagulation/blood pressure regulation, cell differentiation/communication/adhesion, cell cycle and transcriptional gene regulation, extracellular matrix biology, lipid metabolism and immunological response. Five sequence variants generated premature stop codons in genes playing an essential role in placental physiology (STOX1, PDGFD, IGF2, MMP1 and DNAH11). Six variants (ERAP1- p.Ile915Thr, ERAP2- p.Leu837Ser, COMT-p.His192Gln, CSAD-p.Pro418Ser, CDH1- p.Ala298Thr and CCR2-p.Met249Lys) led to destabilisation of protein structure as they had significant energy and residue interaction-related changes. We identified at least two mutations in 57% of patients, arguing in favour of a polygenic origin for the HELLP syndrome. CONCLUSION Our results provide novel evidence regarding PE/HELLP's genetic origin, leading to new biomarkers, having potential clinical usefulness, being proposed.
Collapse
Affiliation(s)
- Karen Marcela Jiménez
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Adrien Morel
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Laura Parada-Niño
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - María Alejandra González-Rodriguez
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Stephanie Flórez
- Hospital Universitario Mayor Méderi, Universidad del Rosario, Bogotá, Colombia
| | - David Bolívar-Salazar
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | | | - Angel Aguirre-García
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Hospital Universitario Mayor Méderi, Universidad del Rosario, Bogotá, Colombia
| | - Tatiana Gómez-Murcia
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Hospital Universitario Mayor Méderi, Universidad del Rosario, Bogotá, Colombia
| | - Luisa Fernanda Castillo
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Carolina Carlosama
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Javier Ardila
- Hospital Universitario Mayor Méderi, Universidad del Rosario, Bogotá, Colombia
| | - Daniel Vaiman
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014 Paris, France
| | - Norma Serrano
- Research Centre, Fundación Cardiovascular de Colombia (FCV), Bucaramanga, Colombia
| | - Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014 Paris, France; Orphan Diseases Group, Biopas Laboratoires, Bogotá, Colombia.
| |
Collapse
|
44
|
Biwer C, Kawam B, Chapelle V, Silvestre F. The Role of Stochasticity in the Origin of Epigenetic Variation in Animal Populations. Integr Comp Biol 2020; 60:1544-1557. [PMID: 32470118 DOI: 10.1093/icb/icaa047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epigenetic mechanisms such as DNA methylation modulate gene expression in a complex fashion are consequently recognized as among the most important contributors to phenotypic variation in natural populations of plants, animals, and microorganisms. Interactions between genetics and epigenetics are multifaceted and epigenetic variation stands at the crossroad between genetic and environmental variance, which make these mechanisms prominent in the processes of adaptive evolution. DNA methylation patterns depend on the genotype and can be reshaped by environmental conditions, while transgenerational epigenetic inheritance has been reported in various species. On the other hand, DNA methylation can influence the genetic mutation rate and directly affect the evolutionary potential of a population. The origin of epigenetic variance can be attributed to genetic, environmental, or stochastic factors. Generally less investigated than the first two components, variation lacking any predictable order is nevertheless present in natural populations and stochastic epigenetic variation, also referred to spontaneous epimutations, can sustain phenotypic diversity. Here, potential sources of such stochastic epigenetic variability in animals are explored, with a focus on DNA methylation. To this day, quantifying the importance of stochasticity in epigenetic variability remains a challenge. However, comparisons between the mutation and the epimutation rates showed a high level of the latter, suggesting a significant role of spontaneous epimutations in adaptation. The implications of stochastic epigenetic variability are multifold: by affecting development and subsequently phenotype, random changes in epigenetic marks may provide additional phenotypic diversity, which can help natural populations when facing fluctuating environments. In isogenic lineages and asexually reproducing organisms, poor or absent genetic diversity can hence be tolerated. Further implication of stochastic epigenetic variability in adaptation is found in bottlenecked invasive species populations and populations using a bet-hedging strategy.
Collapse
Affiliation(s)
| | | | | | - F Silvestre
- Institute of Earth, Life and Environment (ILEE), University of Namur, 61 rue de Bruxelles, Namur, 5000, Belgium
| |
Collapse
|
45
|
Ivanov DO, Evsyukova II, Mazzoccoli G, Anderson G, Polyakova VO, Kvetnoy IM, Carbone A, Nasyrov RA. The Role of Prenatal Melatonin in the Regulation of Childhood Obesity. BIOLOGY 2020; 9:biology9040072. [PMID: 32260529 PMCID: PMC7235795 DOI: 10.3390/biology9040072] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/23/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
There is a growing awareness that pregnancy can set the foundations for an array of diverse medical conditions in the offspring, including obesity. A wide assortment of factors, including genetic, epigenetic, lifestyle, and diet can influence foetal outcomes. This article reviews the role of melatonin in the prenatal modulation of offspring obesity. A growing number of studies show that many prenatal risk factors for poor foetal metabolic outcomes, including gestational diabetes and night-shift work, are associated with a decrease in pineal gland-derived melatonin and associated alterations in the circadian rhythm. An important aspect of circadian melatonin’s effects is mediated via the circadian gene, BMAL1, including in the regulation of mitochondrial metabolism and the mitochondrial melatoninergic pathway. Alterations in the regulation of mitochondrial metabolic shifts between glycolysis and oxidative phosphorylation in immune and glia cells seem crucial to a host of human medical conditions, including in the development of obesity and the association of obesity with the risk of other medical conditions. The gut microbiome is another important hub in the pathoetiology and pathophysiology of many medical conditions, with negative consequences mediated by a decrease in the short-chain fatty acid, butyrate. The effects of butyrate are partly mediated via an increase in the melatoninergic pathway, indicating interactions of the gut microbiome with melatonin. Some of the effects of melatonin seem mediated via the alpha 7 nicotinic receptor, whilst both melatonin and butyrate may regulate obesity through the opioidergic system. Oxytocin, a recently recognized inhibitor of obesity, may also be acting via the opioidergic system. The early developmental regulation of these processes and factors by melatonin are crucial to the development of obesity and many diverse comorbidities.
Collapse
Affiliation(s)
- Dmitry O. Ivanov
- Saint-Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia; (D.O.I.); (V.O.P.); (R.A.N.)
| | - Inna I. Evsyukova
- Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia;
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
- Correspondence: ; Tel.: +039-0882-410255
| | | | - Victoria O. Polyakova
- Saint-Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia; (D.O.I.); (V.O.P.); (R.A.N.)
| | - Igor M. Kvetnoy
- Saint-Petersburg State University, University Embankment 7/9, 199034 St. Petersburg, Russia;
| | - Annalucia Carbone
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Ruslan A. Nasyrov
- Saint-Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia; (D.O.I.); (V.O.P.); (R.A.N.)
| |
Collapse
|
46
|
Yan Z, Li Q, Zhang L, Kang B, Fan W, Deng T, Zhu J, Wang Y. The growth and development conditions in mouse offspring derived from ovarian tissue cryopreservation and orthotopic transplantation. J Assist Reprod Genet 2020; 37:923-932. [PMID: 32221789 DOI: 10.1007/s10815-020-01734-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/02/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To investigate the potential development or metabolic risk in offspring derived from mice with transplanted frozen-thawed ovarian tissue. METHODS Mice ovaries were intervened by vitrification (group V) and slow-freezing (group S) cryopreservation and orthotopic transplantation. Orthotopic transplantation of fresh ovarian (group F) and natural mating (group C) served as control groups. The fertility restoration and health conditions of generations were assessed by offspring counts, anti-fatigue and motor ability, and organ morphology. The methylation rate and expression level of imprinted genes (IGF2R, H19, SNRPN, and PLAGL1) were used to predict the potential risk of development in transplanted generations. RESULTS Both the percentage of normal morphological follicles in different developmental periods and the litter size of receipt mice were comparable in all three transplanted groups. There was no significant difference in offspring mice's birth defects, body weight gain, anti-fatigue ability, or exercise capacity among the four groups. The methylation rate of IGF2R, H19, and PLAGL1 showed a significant variation in cryopreservation groups as compared with control groups, as well as a difference in gene expression. The SNRPN appeared to be stable in methylation status. There were no differences in mRNA expression in all groups. CONCLUSIONS The different ovarian tissue cryopreservation methods did not influence either maternal fertility function or offspring growth. However, these technologies could affect the methylation rate and expression level of some development-related imprinting genes in the offspring, which may lead to some indeterminacy risk.
Collapse
Affiliation(s)
- Zhe Yan
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Li
- Department of Thoracic Oncology, West China hospital, Sichuan University, Chengdu, 610041, China
| | - Long Zhang
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, 610041, China.,Key laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China
| | - Beijia Kang
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, 610041, China.,Key laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China
| | - Wei Fan
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, 610041, China.,Key laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China
| | - Tang Deng
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, 610041, China.,Key laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China
| | - Jiang Zhu
- Department of Thoracic Oncology, West China hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Wang
- Reproductive Medical Center of West China 2nd University Hospital, Sichuan University, Ren Min Nan Lu, Chengdu, 610041, China. .,Key laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China.
| |
Collapse
|
47
|
Laissue P, Vaiman D. Exploring the Molecular Aetiology of Preeclampsia by Massive Parallel Sequencing of DNA. Curr Hypertens Rep 2020; 22:31. [PMID: 32172383 DOI: 10.1007/s11906-020-01039-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This manuscript aims to review (for the first time) studies describing NGS sequencing of preeclampsia (PE) women's DNA. RECENT FINDINGS Describing markers for the early detection of PE is an essential task because, although associated molecular dysfunction begins early on during pregnancy, the disease's clinical signs usually appear late in pregnancy. Although several biochemical biomarkers have been proposed, their use in clinical environments is still limited, thereby encouraging research into PE's genetic origin. Hundreds of genes involved in numerous implantation- and placentation-related biological processes may be coherent candidates for PE aetiology. Next-generation sequencing (NGS) offers new technical possibilities for PE studying, as it enables large genomic regions to be analysed at affordable cost. This technique has facilitated the description of genes contributing to the molecular origin of a significant amount of monogenic and complex diseases. Regarding PE, NGS of DNA has been used in familial and isolated cases, thereby enabling new genes potentially related to the phenotype to be proposed. For a better understanding of NGS, technical aspects, applications and limitations are presented initially. Thereafter, NGS studies of DNA in familial and non-familial cases are described, including pitfalls and positive findings. The information given here should enable scientists and clinicians to analyse and design new studies permitting the identification of novel clinically useful molecular PE markers.
Collapse
Affiliation(s)
- Paul Laissue
- Biopas Laboratoires, Biopas Group, Bogotá, Colombia. .,Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France. .,CIGGUR Genetics Group, School of Medicine and Health Sciences, El Rosario University, Bogotá, Colombia.
| | - Daniel Vaiman
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France
| |
Collapse
|
48
|
Cannarella R, Condorelli RA, Mongioì LM, La Vignera S, Calogero AE. Molecular Biology of Spermatogenesis: Novel Targets of Apparently Idiopathic Male Infertility. Int J Mol Sci 2020; 21:E1728. [PMID: 32138324 PMCID: PMC7084762 DOI: 10.3390/ijms21051728] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
Male infertility affects half of infertile couples and, currently, a relevant percentage of cases of male infertility is considered as idiopathic. Although the male contribution to human fertilization has traditionally been restricted to sperm DNA, current evidence suggest that a relevant number of sperm transcripts and proteins are involved in acrosome reactions, sperm‒oocyte fusion and, once released into the oocyte, embryo growth and development. The aim of this review is to provide updated and comprehensive insight into the molecular biology of spermatogenesis, including evidence on spermatogenetic failure and underlining the role of the sperm-carried molecular factors involved in oocyte fertilization and embryo growth. This represents the first step in the identification of new possible diagnostic and, possibly, therapeutic markers in the field of apparently idiopathic male infertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (L.M.M.); (A.E.C.)
| | | | | | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (L.M.M.); (A.E.C.)
| | | |
Collapse
|
49
|
Crespi BJ. Why and How Imprinted Genes Drive Fetal Programming. Front Endocrinol (Lausanne) 2020; 10:940. [PMID: 32117048 PMCID: PMC7025584 DOI: 10.3389/fendo.2019.00940] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023] Open
Abstract
Imprinted genes mediate fetal and childhood growth and development, and early growth patterns drive fetal programming effects. However, predictions and evidence from the kinship theory of imprinting have yet to be directly integrated with data on fetal programming and risks of metabolic disease. I first define paternal-gene and maternal-gene optima with regard to early human growth and development. Next, I review salient evidence with regard to imprinted gene effects on birth weight, body composition, trajectories of feeding and growth, and timing of developmental stages, to evaluate why and how imprinted gene expression influences risks of metabolic disease in later life. I find that metabolic disease risks derive primarily from maternal gene biases that lead to reduced placental efficacy, low birth weight, low relative muscle mass, high relative white fat, increased abdominal adiposity, reduced pancreatic β-cell mass that promotes insulin resistance, reduced appetite and infant sucking efficacy, catch-up fat deposition from family foods after weaning, and early puberty. Paternal gene biases, by contrast, may contribute to metabolic disease via lower rates of brown fat thermiogenesis, and through favoring more rapid postnatal catch-up growth after intrauterine growth restriction from environmental causes. These disease risks can be alleviated through dietary and pharmacological alterations that selectively target imprinted gene expression and relevant metabolic pathways. The kinship theory of imprinting, and mother-offspring conflict more generally, provide a clear predictive framework for guiding future research on fetal programming and metabolic disease.
Collapse
Affiliation(s)
- Bernard J. Crespi
- Department of Biological Sciences and Human Evolutionary Studies Program, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
50
|
|