1
|
Franzolin AML, Fioretto MN, Ribeiro IT, Maciel FA, Barata LA, Vitali PM, Magosso N, Fagundes FL, Emílio-Silva MT, Hiruma Lima CA, Scarano WR, Justulin LA. Maternal protein restriction compromises hepatic phenotype and antioxidant defense in postweaning male rats, while females exhibit resilience. Biochem Biophys Res Commun 2025; 766:151873. [PMID: 40300334 DOI: 10.1016/j.bbrc.2025.151873] [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: 01/29/2025] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
The Developmental Origins of Health and Disease (DOHaD) concept postulates that maternal malnutrition can program offspring for dysfunction of multiple systems, including the liver. Maternal Protein Restriction (MPR) is a maternal malnutrition model that dysregulates catabolic hormones early in life, with long-term consequences on offspring such as hypertension and reproductive system cancers. Furthermore, studies evaluating sex-specific differences are scarce, especially considering the consequences of MPR on early life. Here, we investigated the impacts of MPR on hepatic phenotypic and molecular aspects of male and female rats at postnatal day (PND)21. The rats were divided into two groups: CTR, from dams that consumed a normal-protein diet (17 % protein), or GLLP, from dams that consumed a low-protein diet (6 % protein) throughout gestation and lactation. Our results demonstrated that MPR leads to an increase in collagen fibers, glycogen, and peroxiredoxin 1, in addition to a decrease in reticular fibers, mast cells, GSH, and MDA in the liver of male rats. In females, a reduction of reticular fibers and protein expression of hepatic peroxiredoxin 4 was observed. By contrasting these results with in silico analyses, we suggest that the main altered mechanisms in males are associated with oxidative stress, glycogen metabolism, and inflammatory responses. In females, a subtle dysregulation of antioxidant activity within the extracellular matrix was noted. Therefore, this work demonstrates sex-specific hepatic differences in post-weaning rats exposed to MPR, highlighting possible maternal modulations that lead males to be more affected, which may generate long-term effects on hepatic and systemic health.
Collapse
Affiliation(s)
| | - Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Isabelle Tenori Ribeiro
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Flávia Alessandra Maciel
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Luisa Annibal Barata
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Pedro Menchini Vitali
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Natália Magosso
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Felipe Leonardo Fagundes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Maycon Tavares Emílio-Silva
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Clélia Akiko Hiruma Lima
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil.
| |
Collapse
|
2
|
Ponsuksili S, Li S, Siengdee P, Hadlich F, Trakooljul N, Oster M, Reyer H, Wimmers K. DNA methylation in adipocyte differentiation of porcine mesenchymal stem cells and the impact of the donor metabolic type. Genomics 2025; 117:111050. [PMID: 40306557 DOI: 10.1016/j.ygeno.2025.111050] [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: 02/12/2024] [Revised: 07/26/2024] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
The impact of metabolic donor mesenchymal stem cells (MSCs) on DNA methylation, a critical epigenetic mechanism, significantly regulates adipogenesis. In this study, we investigated epigenetic changes during differentiation of synovial MSCs (SMSCs) from two pig breeds differing in metabolic performance (German Landrace (DL) and Angeln Saddleback (AS)). Stimulation of SMSCs to differentiate into adipocytes in vitro revealed several differentially methylated loci and regions, particularly on gene promoter regions, at day 7 and 14. AS breeds, known for higher fat deposition, exhibited more hypermethylation compared to DL. Furthermore, we utilized differentially methylated regions associated with the adipogenic process and breed, especially those in promoter regions, for predicting transcription factor motifs. This study provides insights into the DNA methylation landscape during adipogenesis in pigs of different metabolic types, revealing its role in regulating cell fate and donor memory retention in culture.
Collapse
Affiliation(s)
- Siriluck Ponsuksili
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Shuaichen Li
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Puntita Siengdee
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany; Chulabhorn Graduate Institute, Program in Applied Biological Sciences, Chulabhorn Royal Academy, Kamphaeng Phet 6 Road, Laksi, Bangkok 10210, Thailand
| | - Frieder Hadlich
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Nares Trakooljul
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Michael Oster
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany; Faculty of Agricultural and Environmental Sciences, University of Rostock, Justus-von-Liebig-Weg 6b, 18059 Rostock, Germany
| |
Collapse
|
3
|
Jiao P, Lu H, Hao L, Degen AA, Cheng J, Yin Z, Mao S, Xue Y. Nutrigenetic and Epigenetic Mechanisms of Maternal Nutrition-Induced Glucolipid Metabolism Changes in the Offspring. Nutr Rev 2025; 83:728-748. [PMID: 38781288 DOI: 10.1093/nutrit/nuae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Maternal nutrition during pregnancy regulates the offspring's metabolic homeostasis, including insulin sensitivity and the metabolism of glucose and lipids. The fetus undergoes a crucial period of plasticity in the uterus; metabolic changes in the fetus during pregnancy caused by maternal nutrition not only influence fetal growth and development but also have a long-term or even life-long impact for the offspring. Epigenetic modifications, such as DNA methylation, histone modification, and non-coding RNAs, play important roles in intergenerational and transgenerational effects. In this context, this narrative review comprehensively summarizes and analyzes the molecular mechanisms underlying how maternal nutrition, including a high-fat diet, polyunsaturated fatty acid diet, methyl donor nutrient supplementation, feed restriction, and protein restriction during pregnancy, impacts the genes involved in glucolipid metabolism in the liver, adipose tissue, hypothalamus, muscle, and oocytes of the offspring in terms of the epigenetic modifications. This will provide a foundation for the further exploration of nutrigenetic and epigenetic mechanisms for integrative mother-child nutrition and promotion of the offspring's health through the regulation of maternal nutrition during pregnancy. Note: This paper is part of the Nutrition Reviews Special Collection on Precision Nutrition.
Collapse
Affiliation(s)
- Peng Jiao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Huizhen Lu
- Biotechnology Center, Anhui Agricultural University, Hefei, China
| | - Lizhuang Hao
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Plateau Yak Research Center, Qinghai Academy of Science and Veterinary Medicine of Qinghai University, Xining, China
| | - A Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jianbo Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Shengyong Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yanfeng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
4
|
Davenport BN, Williams A, Regnault TRH, Jones HN, Wilson RL. Placenta hIGF1 nanoparticle treatment in guinea pigs mitigates FGR-associated fetal sex-dependent effects on liver metabolism-related signaling pathways. Am J Physiol Endocrinol Metab 2025; 328:E395-E409. [PMID: 39907801 DOI: 10.1152/ajpendo.00440.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/03/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
Fetal development in an adverse in utero environment significantly increases the risk of developing metabolic diseases in later life, including dyslipidemia, nonalcoholic fatty liver diseases, and diabetes. The aim of this study was to determine whether improving the in utero fetal growth environment with a placental nanoparticle gene therapy would ameliorate fetal growth restriction (FGR)-associated dysregulation of fetal hepatic lipid and glucose metabolism-related signaling pathways. Using the guinea pig maternal nutrient restriction (MNR) model of placental insufficiency and FGR, placenta efficiency and fetal weight were significantly improved following three administrations of a nonviral polymer-based nanoparticle gene therapy to the placenta from mid-pregnancy (gestational day 35) until gestational day 52. The nanoparticle gene therapy transiently increased expression of human insulin-like growth factor 1 (hIGF1) in placenta trophoblast. Fetal liver tissue was collected near-term at gestational day 60. Fetal sex-specific differences in liver gene and protein expression of profibrosis and glucose metabolism-related factors were demonstrated in sham-treated FGR fetuses but not observed in FGR fetuses who received placental hIGF1 nanoparticle treatment. Increased plasma bilirubin, an indirect measure of hepatic activity, was also demonstrated with placental hIGF1 nanoparticle treatment. We speculate that the changes in liver gene and protein expression and increased liver activity that result in similar expression profiles to appropriately growing control fetuses might confer protection against increased susceptibility to aberrant liver physiology in later life. Overall, this work opens avenues for future research assessing the translational prospect of mitigating FGR-induced metabolic derangements.NEW & NOTEWORTHY A placenta-specific nonviral polymer-based nanoparticle gene therapy that improves placenta nutrient transport and near-term fetal weight ameliorates growth restriction-associated changes to fetal liver activity, and cholesterol and glucose/nutrient homeostasis genes/proteins that might confer protection against increased susceptibility to aberrant liver physiology in later life. This knowledge may have implications toward removing predispositions that increase the risk of metabolic diseases, including diabetes, dyslipidemia, and nonalcoholic fatty liver disease in later life.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Alyssa Williams
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Timothy R H Regnault
- Departments of Obstetrics and Gynaecology, Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
5
|
Gyllenhammer LE, Boyle KE. New Frontiers: Umbilical Cord Mesenchymal Stem Cells Uncover Developmental Roots and Biological Underpinnings of Obesity Susceptibility. Curr Obes Rep 2025; 14:10. [PMID: 39814984 PMCID: PMC11735562 DOI: 10.1007/s13679-024-00599-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE OF REVIEW To review evidence supporting human umbilical cord mesenchymal stem cells (UC-MSC) as an innovative model system advancing obesity precision medicine. RECENT FINDINGS Obesity prevalence is increasing rapidly and exposures during fetal development can impact individual susceptibility to obesity. UC-MSCs exhibit heterogeneous phenotypes associated with maternal exposures and predictive of child cardiometabolic outcomes. This recent evidence supports UC-MSCs as a precision model serving three purposes: (1) as a mechanistic tool to interrogate biological underpinnings of obesity in human studies, (2) as a sensitive index of early life causes and determinants of obesity, and (3) as a marker and transducer of susceptibility, highlighting populations most at risk for future obesity. Data from UC-MSCs emphasize nutrient sensing and lipid partitioning as phenotypes most relevant to neonatal and early childhood adiposity and implicate a role for these cell-autonomous features of mesodermal tissues in the biological underpinnings of obesity.
Collapse
Affiliation(s)
- Lauren E Gyllenhammer
- Department of Pediatrics, School of Medicine, University of California, Irvine, CA, USA.
| | - Kristen E Boyle
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA.
| |
Collapse
|
6
|
Davies LR, Kristensen TN, Sørensen JG, Loeschcke V, Schou MF. Nutritional stress in larvae induces adaptive responses that transcend generations in males of a model insect. J Exp Biol 2025; 228:jeb247972. [PMID: 39820330 PMCID: PMC11832117 DOI: 10.1242/jeb.247972] [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: 05/03/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025]
Abstract
The ability of organisms to cope with poor quality nutrition is essential for their persistence. For species with a short generation time, the nutritional environments can transcend generations, making it beneficial for adults to prime their offspring to particular diets. However, our understanding of adaptive generational responses, including those to diet quality, are still very limited. Here, we used the vinegar fly, Drosophila melanogaster, to investigate whether females developing as larvae on a nutritionally poor diet produce offspring that are primed for nutrient deficiencies in the following generations. We found that females developed on low-quality diets produced offspring that, on similarly low-quality diets, had both increased egg-to-adult viability and starvation tolerance compared with offspring of females experiencing a nutrient-rich diet. When testing the persistence of such generational priming, we found that just one generation of high-quality diet is sufficient to erase the signal of priming. A global transcriptomic profile analysis on male offspring suggests that the observed phenotypic priming is not a constitutive transcriptomic adjustment of adults; instead, offspring are probably primed as larvae, enabling them to initiate an adaptive response as adults when exposed to low-quality diets. Our results support that generational priming is an important adaptive mechanism that enables organisms to cope with transient nutritional fluctuations.
Collapse
Affiliation(s)
- Lucy Rebecca Davies
- Department of Biology, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Biological and Environmental Science, University of Jyväskylä, 40500 Jyväskylä, Finland
| | - Torsten N. Kristensen
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg East, Denmark
| | | | - Volker Loeschcke
- Department of Biology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Mads F. Schou
- Department of Biology, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Biology, Lund University, 223 62 Lund, Sweden
| |
Collapse
|
7
|
Sinclair KD. Developmental epigenetics: Understanding genetic and sexually dimorphic responses to parental diet and outcomes following assisted reproduction. J Dairy Sci 2024:S0022-0302(24)01392-4. [PMID: 39701526 DOI: 10.3168/jds.2024-25811] [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: 10/02/2024] [Accepted: 11/17/2024] [Indexed: 12/21/2024]
Abstract
The developmental integrity and wellbeing of offspring are influenced by events that occur in utero, particularly around the time of conception. While extraneous factors such as environmental temperature and exposure to environmental chemicals can each have a bearing on these events, the epigenetic mechanisms that direct cellular differentiation during early development in ruminants are best described for studies which have investigated the effects of parental nutrition or pregnancy outcomes following assisted reproduction. In this article the case is made that the genetic constitution of an individual directs epigenetic responses to environmental stimuli, and consideration in this regard is also given to the origins of sexual dimorphism and mechanisms of germline intergenerational inheritance. These aspects are considered in the context of epigenetic modifications that take place during the normal course of gametogenesis and embryogenesis, and again following either dietary or procedural interventions such as embryo culture. A recurring feature of such interventions, irrespective of species, is that one carbon metabolic pathways are invariably disrupted, and this affects the provision of methyl groups for chromatin and RNA methylation. Inter-specific variation in how these pathways operate, both within the liver and in germ cells, indicates that ruminants may be particularly sensitive in this regard. Recent advances in genomic technologies should enable rapid progress in these areas. Knowledge gained can be integrated into breed improvement programs and used to tailor management practices to specific breeds and strains (including sexes) within breeds. Ultimately, consideration should be given to integrating metagenomics into analyses of genetic-directed epigenetic programming of animal development.
Collapse
Affiliation(s)
- Kevin D Sinclair
- School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, UK, LE12 5RD.
| |
Collapse
|
8
|
Diniz F, Edgington-Giordano F, Ngo NYN, Caspi G, El-Dahr SS, Tortelote GG. Morphometric analysis of the intergenerational effects of protein restriction on nephron endowment in mice. Heliyon 2024; 10:e39552. [PMID: 39498088 PMCID: PMC11533620 DOI: 10.1016/j.heliyon.2024.e39552] [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: 08/09/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Background Parental nutritional status is crucial in shaping offspring's kidney development. However, the association between a protein-restrictive diet and its intergenerational impact on kidney development remains unclear. Methods We conducted multigenerational morphometric measurements to investigate the effects of parental protein deprivation on offspring kidney development across four generations. F0 mice were divided into two groups and fed a normal protein diet (NPD) or a low-protein diet (LPD) for three weeks before mating and continued these diets throughout gestation and lactation. Body weight (BW), kidney weight (KW), KW/BW ratio, nephron counts, and blood pressure were assessed in F1 pups. To examine paternal effects, we bred CD1 females on an NPD with males on an LPD. BW, KW, KW/BW, and nephron counts were measured at P20. To measure the transgenerational effect of parental LPD on kidney development, F1 offspring (from parents on LPD) were fed NPD upon weaning. These F1 offspring were bred at 6 weeks of age to produce F2, F3 and F4 generations. Kidney metrics were evaluated across generations. Results The average body weight of P0 pups from parents on NPD was 1.61g, while pups from parental LPD weighed an average of 0.869g, a decrease of 54 % (p = 6.9e-11, Wilcoxon test). F1 from parental LPD have significantly smaller kidneys than the control, with an average combined kidney weight of 0.0082g versus 0.0129g, a 37 % decrease (p = 3.2e-02, Wilcoxon test). P20 BW and KW remained low in LPD offspring. These effects persisted for 4 generations (F1 to F4) with an average glomerular count reduction of roughly 20 %. F3 and F4 showed wider variability in glomerular counts but were not statistically significant compared to controls. Conclusions Both maternal and paternal LPD significantly affected offspring nephron endowment. Our study underscores the complex nature of nutritional transgenerational effects on kidney development, emphasizing the importance of both maternal and paternal dietary impacts on kidney development and the developmental origin of adult disease.
Collapse
Affiliation(s)
- Fabiola Diniz
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Francesca Edgington-Giordano
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nguyen Yen Nhi Ngo
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Gal Caspi
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Samir S. El-Dahr
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Giovane G. Tortelote
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| |
Collapse
|
9
|
Ribeiro IT, Fioretto MN, Dos Santos SAA, Colombelli KT, Portela LMF, Niz Alvarez MV, de Magalhães Padilha P, Delgado AQ, Marques MVLSG, Bosqueiro JR, Seiva FRF, Barbisan LF, de Andrade Paes AM, Zambrano E, Justulin LA. Maternal protein restriction combined with postnatal sugar consumption alters liver proteomic profile and metabolic pathways in adult male offspring rats. Mol Cell Endocrinol 2024; 592:112316. [PMID: 38880278 DOI: 10.1016/j.mce.2024.112316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/18/2024]
Abstract
This study investigated the impact of maternal protein restriction (MPR) and early postnatal sugar consumption (SUG) on the liver health of adult male descendant rats. Male offspring of mothers fed a normal protein diet (NPD) or a low protein diet (LPD) were divided into four groups: Control (CTR), Sugar Control (CTR + SUG), LPD during gestation and lactation (GLLP), and LPD with sugar (GLLP + SUG). Sugar consumption (10% glucose diluted in water) began after weaning on day 21 (PND 21), and at 90 days (PND 90), rats were sacrificed for analysis. Sugar intake reduced food intake and increased water consumption in CTR + SUG and GLLP + SUG compared to CTR and GLLP. GLLP and GLLP + SUG groups showed lower body weight and total and retroperitoneal fat compared to CTR and CTR + SUG. CTR + SUG and GLLP + SUG groups exhibited hepatocyte vacuolization associated with increased hepatic glycogen content compared to CTR and GLLP. Hepatic catalase activity increased in GLLP compared to CTR. Proteomic analysis identified 223 differentially expressed proteins (DEPs) among experimental groups. While in the GLLP group, the DEPs enriched molecular pathways related to cellular stress, glycogen metabolic pathways were enriched in the GLLP + SUG and CTR + SUG groups. The association of sugar consumption amplifies the effects of MPR, deregulating molecular mechanisms related to metabolism and the antioxidant system.
Collapse
Affiliation(s)
- Isabelle Tenori Ribeiro
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Matheus Naia Fioretto
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcantara Dos Santos
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Ketlin Thassiani Colombelli
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Luiz Marcos Frediani Portela
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | | | - Pedro de Magalhães Padilha
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Aislan Quintiliano Delgado
- Department of Physical Education, Institute of Biosciences, Sao Paulo State University, Bauru, SP, Brazil
| | | | - José Roberto Bosqueiro
- Department of Physical Education, Institute of Biosciences, Sao Paulo State University, Bauru, SP, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luís Fernando Barbisan
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | | | - Elena Zambrano
- Department Reproductive Biology, Salvador Zubirán National Institute of Medical Sciences and Nutrition, Mexico City, Mexico; Facultad de Química, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Luis Antonio Justulin
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil.
| |
Collapse
|
10
|
Huang W, Hu W, Fang M, Zhang Q, Zhang Y, Wang H. Impacts of prenatal environmental exposures on fetal-placental-maternal bile acid homeostasis and long-term health in offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116929. [PMID: 39213751 DOI: 10.1016/j.ecoenv.2024.116929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
During pregnancy, the maternal body undergoes a series of adaptative physiological changes, leading to a slight increase in serum bile acid (BA) levels. Although the fetal liver can synthesize BAs since the first trimester through the alternative pathway, the BA metabolic system is immature in the fetus. Compared to adults, the fetus has a distinct composition of BA pool and limited expression of BA synthesis enzymes and transporters. Besides, the "enterohepatic circulation" of BAs is absent in fetus. Thus, fetal BAs need to be transported to the mother through the placenta for further metabolism and excretion, and maternal BAs can also be transported to the fetus. That is what we call the "fetal-placental-maternal BA circulation". Various BA transporters and nuclear receptors are essential for maintaining the balance of this BA circulation to ensure normal fetal development. However, prenatal adverse environments can alter fetal BA metabolism, resulting in intrauterine developmental abnormalities and susceptibility to a variety of adult chronic diseases. This review summarizes the current understanding of the fetal-placental-maternal BA circulation and discusses the effects of prenatal adverse environments on this particular BA circulation, aiming to provide a theoretical basis for exploring early prevention and treatment strategies for BA metabolism-associated adverse pregnancy outcomes and long-term impairments.
Collapse
Affiliation(s)
- Wen Huang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wen Hu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Man Fang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qi Zhang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| |
Collapse
|
11
|
Lin X, Han H, Wang N, Wang C, Qi M, Wang J, Liu G. The Gut Microbial Regulation of Epigenetic Modification from a Metabolic Perspective. Int J Mol Sci 2024; 25:7175. [PMID: 39000282 PMCID: PMC11241073 DOI: 10.3390/ijms25137175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Obesity is a global health challenge that has received increasing attention in contemporary research. The gut microbiota has been implicated in the development of obesity, primarily through its involvement in regulating various host metabolic processes. Recent research suggests that epigenetic modifications may serve as crucial pathways through which the gut microbiota and its metabolites contribute to the pathogenesis of obesity and other metabolic disorders. Hence, understanding the interplay between gut microbiota and epigenetic mechanisms is crucial for elucidating the impact of obesity on the host. This review primarily focuses on the understanding of the relationship between the gut microbiota and its metabolites with epigenetic mechanisms in several obesity-related pathogenic mechanisms, including energy dysregulation, metabolic inflammation, and maternal inheritance. These findings could serve as novel therapeutic targets for probiotics, prebiotics, and fecal microbiota transplantation tools in treating metabolic disruptions. It may also aid in developing therapeutic strategies that modulate the gut microbiota, thereby regulating the metabolic characteristics of obesity.
Collapse
Affiliation(s)
- Xingtong Lin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Hui Han
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Nan Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Chengming Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Ming Qi
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Gang Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
12
|
Manav N, Jit BP, Kataria B, Sharma A. Cellular and epigenetic perspective of protein stability and its implications in the biological system. Epigenomics 2024; 16:879-900. [PMID: 38884355 PMCID: PMC11370918 DOI: 10.1080/17501911.2024.2351788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/30/2024] [Indexed: 06/18/2024] Open
Abstract
Protein stability is a fundamental prerequisite in both experimental and therapeutic applications. Current advancements in high throughput experimental techniques and functional ontology approaches have elucidated that impairment in the structure and stability of proteins is intricately associated with the cause and cure of several diseases. Therefore, it is paramount to deeply understand the physical and molecular confounding factors governing the stability of proteins. In this review article, we comprehensively investigated the evolution of protein stability, examining its emergence over time, its relationship with organizational aspects and the experimental methods used to understand it. Furthermore, we have also emphasized the role of Epigenetics and its interplay with post-translational modifications (PTMs) in regulating the stability of proteins.
Collapse
Affiliation(s)
- Nisha Manav
- Department of Biochemistry, All India Institute of Medical Sciences New Delhi, Ansari Nagar, 110029, India
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences New Delhi, Ansari Nagar, 110029, India
| | - Babita Kataria
- Department of Medical Oncology, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, 124105, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences New Delhi, Ansari Nagar, 110029, India
- Department of Biochemistry, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, 124105, India
| |
Collapse
|
13
|
Faienza MF, Urbano F, Anaclerio F, Moscogiuri LA, Konstantinidou F, Stuppia L, Gatta V. Exploring Maternal Diet-Epigenetic-Gut Microbiome Crosstalk as an Intervention Strategy to Counter Early Obesity Programming. Curr Issues Mol Biol 2024; 46:4358-4378. [PMID: 38785533 PMCID: PMC11119222 DOI: 10.3390/cimb46050265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/21/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Alterations in a mother's metabolism and endocrine system, due to unbalanced nutrition, may increase the risk of both metabolic and non-metabolic disorders in the offspring's childhood and adulthood. The risk of obesity in the offspring can be determined by the interplay between maternal nutrition and lifestyle, intrauterine environment, epigenetic modifications, and early postnatal factors. Several studies have indicated that the fetal bowel begins to colonize before birth and that, during birth and nursing, the gut microbiota continues to change. The mother's gut microbiota is primarily transferred to the fetus through maternal nutrition and the environment. In this way, it is able to impact the establishment of the early fetal and neonatal microbiome, resulting in epigenetic signatures that can possibly predispose the offspring to the development of obesity in later life. However, antioxidants and exercise in the mother have been shown to improve the offspring's metabolism, with improvements in leptin, triglycerides, adiponectin, and insulin resistance, as well as in the fetal birth weight through epigenetic mechanisms. Therefore, in this extensive literature review, we aimed to investigate the relationship between maternal diet, epigenetics, and gut microbiota in order to expand on current knowledge and identify novel potential preventative strategies for lowering the risk of obesity in children and adults.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, 70124 Bari, Italy
| | - Flavia Urbano
- Giovanni XXIII Pediatric Hospital, 70126 Bari, Italy; (F.U.); (L.A.M.)
| | - Federico Anaclerio
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.A.); (F.K.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | | | - Fani Konstantinidou
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.A.); (F.K.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.A.); (F.K.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.A.); (F.K.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
14
|
Saavedra LPJ, Piovan S, Moreira VM, Gonçalves GD, Ferreira ARO, Ribeiro MVG, Peres MNC, Almeida DL, Raposo SR, da Silva MC, Barbosa LF, de Freitas Mathias PC. Epigenetic programming for obesity and noncommunicable disease: From womb to tomb. Rev Endocr Metab Disord 2024; 25:309-324. [PMID: 38040983 DOI: 10.1007/s11154-023-09854-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Several epidemiological, clinical and experimental studies in recent decades have shown the relationship between exposure to stressors during development and health outcomes later in life. The characterization of these susceptible phases, such as preconception, gestation, lactation and adolescence, and the understanding of factors that influence the risk of an adult individual for developing obesity, metabolic and cardiovascular diseases, is the focus of the DOHaD (Developmental Origins of Health and Disease) research line. In this sense, advancements in molecular biology techniques have contributed significantly to the understanding of the mechanisms underlying the observed phenotypes, their morphological and physiological alterations, having as a main driving factor the epigenetic modifications and their consequent modulation of gene expression. The present narrative review aimed to characterize the different susceptible phases of development and associated epigenetic modifications, and their implication in the development of non-communicable diseases. Additionally, we provide useful insights into interventions during development to counteract or prevent long-term programming for disease susceptibility.
Collapse
Affiliation(s)
- Lucas Paulo Jacinto Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Silvano Piovan
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Gessica Dutra Gonçalves
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maria Natália Chimirri Peres
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Scarlett Rodrigues Raposo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Mariane Carneiro da Silva
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Letícia Ferreira Barbosa
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil.
| |
Collapse
|
15
|
Awata K, Shoji H, Arai Y, Santosa I, Tokita K, Murano Y, Shimizu T. Maternal Protein Restriction Inhibits Insulin Signaling and Insulin Resistance in the Skeletal Muscle of Young Adult Rats. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2024; 70:142-151. [PMID: 39430205 PMCID: PMC11487360 DOI: 10.14789/jmj.jmj23-0029-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/12/2024] [Indexed: 10/22/2024]
Abstract
Objectives Infants with fetal growth restriction (FGR) are at a risk of developing metabolic syndromes in adulthood. We hypothesized that skeletal muscle degeneration by nutrition-restricted FGR results in abnormal insulin signaling and epigenetic changes. Material and Methods To develop a protein-restricted FGR model, rats were fed a low-protein diet (7% protein) during the gestational period; rats fed a normal diet (20% protein) were used as controls. At 8 and 12 weeks of age, the pups were subjected to oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) to evaluate insulin resistance. At 12 weeks, the mRNA and protein levels of insulin signaling pathway molecules in the skeletal muscles were examined. DNA methylation of promoters was detected. DNA extracted from skeletal muscles was used as a template for methylation-specific PCR analysis of GLUT4. Results The body weight of FGR rats from birth to 8 weeks was significantly lower than that of the controls; no significant difference was observed between the groups at 12 weeks. In the OGTT and ITT, the incremental area under the curve (iAUC) was significantly higher in FGR rats than in the controls at 12 weeks. The mRNA and protein levels of Akt2 and GLUT4 in the plantar muscles were significantly lower in FGR rats than in the controls. GLUT4 methylation was comparable between the groups. Conclusions Protein-restricted FGR rats showed insulin resistance and altered insulin signaling in skeletal muscles after 12 weeks. However, we could not demonstrate the involvement of DNA methylation in this model.
Collapse
|
16
|
Bokor S, Csölle I, Felső R, Vass RA, Funke S, Ertl T, Molnár D. Dietary nutrients during gestation cause obesity and related metabolic changes by altering DNA methylation in the offspring. Front Endocrinol (Lausanne) 2024; 15:1287255. [PMID: 38449848 PMCID: PMC10916691 DOI: 10.3389/fendo.2024.1287255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024] Open
Abstract
Growing evidence shows that maternal nutrition from preconception until lactation has an important effect on the development of non-communicable diseases in the offspring. Biological responses to environmental stress during pregnancy, including undernutrition or overnutrition of various nutrients, are transmitted in part by DNA methylation. The aim of the present narrative review is to summarize literature data on altered DNA methylation patterns caused by maternal macronutrient or vitamin intake and its association with offspring's phenotype (obesity and related metabolic changes). With our literature search, we found evidence for the association between alterations in DNA methylation pattern of different genes caused by maternal under- or overnutrition of several nutrients (protein, fructose, fat, vitamin D, methyl-group donor nutrients) during 3 critical periods of programming (preconception, pregnancy, lactation) and the development of obesity or related metabolic changes (glucose, insulin, lipid, leptin, adiponectin levels, blood pressure, non-alcoholic fatty liver disease) in offspring. The review highlights that maternal consumption of several nutrients could individually affect the development of offspring's obesity and related metabolic changes via alterations in DNA methylation.
Collapse
Affiliation(s)
- Szilvia Bokor
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| | - Ildikó Csölle
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| | - Regina Felső
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| | - Réka A. Vass
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, Pécs, Hungary
- Obstetrics and Gynecology, Magyar Imre Hospital Ajka, Ajka, Hungary
| | - Simone Funke
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, Pécs, Hungary
| | - Tibor Ertl
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, Pécs, Hungary
| | - Dénes Molnár
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| |
Collapse
|
17
|
Raghubeer S. The influence of epigenetics and inflammation on cardiometabolic risks. Semin Cell Dev Biol 2024; 154:175-184. [PMID: 36804178 DOI: 10.1016/j.semcdb.2023.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Cardiometabolic diseases include metabolic syndrome, obesity, type 2 diabetes mellitus, and hypertension. Epigenetic modifications participate in cardiometabolic diseases through several pathways, including inflammation, vascular dysfunction, and insulin resistance. Epigenetic modifications, which encompass alterations to gene expression without mutating the DNA sequence, have gained much attention in recent years, since they have been correlated with cardiometabolic diseases and may be targeted for therapeutic interventions. Epigenetic modifications are greatly influenced by environmental factors, such as diet, physical activity, cigarette smoking, and pollution. Some modifications are heritable, indicating that the biological expression of epigenetic alterations may be observed across generations. Moreover, many patients with cardiometabolic diseases present with chronic inflammation, which can be influenced by environmental and genetic factors. The inflammatory environment worsens the prognosis of cardiometabolic diseases and further induces epigenetic modifications, predisposing patients to the development of other metabolism-associated diseases and complications. A deeper understanding of inflammatory processes and epigenetic modifications in cardiometabolic diseases is necessary to improve our diagnostic capabilities, personalized medicine approaches, and the development of targeted therapeutic interventions. Further understanding may also assist in predicting disease outcomes, especially in children and young adults. This review describes epigenetic modifications and inflammatory processes underlying cardiometabolic diseases, and further discusses advances in the research field with a focus on specific points for interventional therapy.
Collapse
Affiliation(s)
- Shanel Raghubeer
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health & Wellness Sciences, Cape Peninsula University of Technology, South Africa.
| |
Collapse
|
18
|
Chen Y, Wang H. The changes in adrenal developmental programming and homeostasis in offspring induced by glucocorticoids exposure during pregnancy. VITAMINS AND HORMONES 2024; 124:463-490. [PMID: 38408809 DOI: 10.1016/bs.vh.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Clinically, synthetic glucocorticoids are often used to treat maternal and fetal related diseases, such as preterm birth and autoimmune diseases. Although its clinical efficacy is positive, it will expose the fetus to exogenous glucocorticoids. Adverse environments during pregnancy (e.g., exogenous glucocorticoids exposure, malnutrition, infection, hypoxia, and stress) can lead to fetal overexposure to endogenous maternal glucocorticoids. Basal glucocorticoids levels in utero are crucial in determining fetal tissue maturation and its postnatal fate. As the synthesis and secretion organ of glucocorticoids, the adrenal development is crucial for the growth and development of the body. Studies have found that glucocorticoids exposure during pregnancy could cause abnormal fetal adrenal development, which could last after birth or even adulthood. As the key organ of fetal-originated adult disease, the adrenal developmental programming has a profound impact on the health of offspring, which can lead to many chronic diseases in adulthood. However, the aberrant adrenal development in offspring caused by glucocorticoids exposure during pregnancy and its intrauterine programming mechanism have not been systematically clarified. Therefore, this review summarizes recent research progress on the short and long-term hazards of aberrant adrenal development induced by glucocorticoids exposure during pregnancy, which is of great significance for the analysis of aberrant adrenal development and clarify the intrauterine origin mechanism of fetal-originated adult disease.
Collapse
Affiliation(s)
- Yawen Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, P.R. China.
| |
Collapse
|
19
|
Ersoy U, Kanakis I, Alameddine M, Pedraza-Vazquez G, Ozanne SE, Peffers MJ, Jackson MJ, Goljanek-Whysall K, Vasilaki A. Lifelong dietary protein restriction accelerates skeletal muscle loss and reduces muscle fibre size by impairing proteostasis and mitochondrial homeostasis. Redox Biol 2024; 69:102980. [PMID: 38064763 PMCID: PMC10755587 DOI: 10.1016/j.redox.2023.102980] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 01/01/2024] Open
Abstract
The early life environment significantly affects the development of age-related skeletal muscle disorders. However, the long-term effects of lactational protein restriction on skeletal muscle are still poorly defined. Our study revealed that male mice nursed by dams fed a low-protein diet during lactation exhibited skeletal muscle growth restriction. This was associated with a dysregulation in the expression levels of genes related to the ribosome, mitochondria and skeletal muscle development. We reported that lifelong protein restriction accelerated loss of type-IIa muscle fibres and reduced muscle fibre size by impairing mitochondrial homeostasis and proteostasis at 18 months of age. However, feeding a normal-protein diet following lactational protein restriction prevented accelerated fibre loss and fibre size reduction in later life. These findings provide novel insight into the mechanisms by which lactational protein restriction hinders skeletal muscle growth and includes evidence that lifelong dietary protein restriction accelerated skeletal muscle loss in later life.
Collapse
Affiliation(s)
- Ufuk Ersoy
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK
| | - Ioannis Kanakis
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK; Chester Medical School, Faculty of Medicine and Life Sciences, University of Chester, Chester, UK
| | - Moussira Alameddine
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK
| | - Gibran Pedraza-Vazquez
- Department of Physiology, School of Medicine and REMEDI, CMNHS, University of Galway, Galway, Ireland
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge, UK
| | - Mandy Jayne Peffers
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK
| | - Malcolm J Jackson
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK
| | - Katarzyna Goljanek-Whysall
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK; Department of Physiology, School of Medicine and REMEDI, CMNHS, University of Galway, Galway, Ireland
| | - Aphrodite Vasilaki
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), The MRC - Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool, UK.
| |
Collapse
|
20
|
Tando Y, Matsui Y. Inheritance of environment-induced phenotypic changes through epigenetic mechanisms. ENVIRONMENTAL EPIGENETICS 2023; 9:dvad008. [PMID: 38094661 PMCID: PMC10719065 DOI: 10.1093/eep/dvad008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 03/08/2024]
Abstract
Growing evidence suggests that epigenetic changes through various parental environmental factors alter the phenotypes of descendants in various organisms. Environmental factors, including exposure to chemicals, stress and abnormal nutrition, affect the epigenome in parental germ cells by different epigenetic mechanisms, such as DNA methylation, histone modification as well as small RNAs via metabolites. Some current remaining questions are the causal relationship between environment-induced epigenetic changes in germ cells and altered phenotypes of descendants, and the molecular basis of how the abnormal epigenetic changes escape reprogramming in germ cells. In this review, we introduce representative examples of intergenerational and transgenerational inheritance of phenotypic changes through parental environmental factors and the accompanied epigenetic and metabolic changes, with a focus on animal species. We also discuss the molecular mechanisms of epigenomic inheritance and their possible biological significance.
Collapse
Affiliation(s)
- Yukiko Tando
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Yasuhisa Matsui
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
21
|
Zhang H, Wang Y, Zhang X, Zhang L, Zhao X, Xu Y, Wang P, Liang X, Xue M, Liang H. Maternal Folic Acid Supplementation during Pregnancy Prevents Hepatic Steatosis in Male Offspring of Rat Dams Fed High-Fat Diet, Which Is Associated with the Regulation of Gut Microbiota. Nutrients 2023; 15:4726. [PMID: 38004120 PMCID: PMC10675082 DOI: 10.3390/nu15224726] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Maternal dietary patterns during pregnancy have been demonstrated to impact the structure of the gut microbiota in offspring, altering their susceptibility to diseases. This study is designed to elucidate whether the impact of folic acid supplementation during pregnancy on hepatic steatosis in male offspring of rat dams exposed to a high-fat diet (HFD) is related to gut-liver axis homeostasis. In this study, female rats were administered a HFD and simultaneously supplemented with 5 mg/kg folic acid throughout their pregnancy. Histopathological examination showed that folic acid supplementation effectively ameliorated hepatic lipid accumulation and inflammatory infiltrate in male offspring subjected to a maternal HFD. Maternal folic acid supplementation reduced the abundance of Desulfobacterota and the Firmicutes/Bacteroidota (F/B) ratio in male offspring. The expression of tight junction proteins in the colon was significantly upregulated, and the serum LPS level was significantly reduced. Furthermore, there was a notable reduction in the hepatic expression of the TLR4/NF-κB signaling pathway and subsequent inflammatory mediators. Spearman's correlation analysis revealed significant associations between hepatic inflammation-related indices and several gut microbiota, particularly Desulfobacterota and Lactobacillus. With a reduction in hepatic inflammation, the expression of PPAR-α was upregulated, and the expression of SREBP-1c and its downstream lipid metabolism-related genes was downregulated. In summary, folic acid supplementation during pregnancy modulates gut microbiota and enhances intestinal barrier integrity in male offspring of HFD dams. This helps reduce the LPS leakage and suppress the expression of TLR4/NF-κB pathway in the liver, thereby improving lipid metabolism disorders, and alleviating hepatic steatosis.
Collapse
Affiliation(s)
- Huaqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Yutong Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Xinyu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Li Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Xuenuo Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Yan Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Peng Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Xi Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University, Qingdao 266071, China;
| | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China; (H.Z.); (Y.W.); (X.Z.); (L.Z.); (X.Z.); (Y.X.); (P.W.); (X.L.)
| |
Collapse
|
22
|
Mussai EX, Lofft ZA, Vanderkruk B, Boonpattrawong N, Miller JW, Smith A, Bottiglieri T, Devlin AM. Folic acid supplementation in a mouse model of diabetes in pregnancy alters insulin sensitivity in female mice and beta cell mass in offspring. FASEB J 2023; 37:e23200. [PMID: 37773756 DOI: 10.1096/fj.202301491r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023]
Abstract
Epidemiological studies have reported discrepant findings on the relationship between folic acid intake during pregnancy and risk for gestational diabetes mellitus (GDM). To begin to understand how folic acid impacts metabolic health during pregnancy, we determined the effects of excess folic acid supplementation (5× recommendation) on maternal and fetal offspring metabolic health. Using a mouse (female C57BL/6J) model of diet-induced diabetes in pregnancy (western diet) and control mice, we show that folic acid supplementation improved insulin sensitivity in the female mice fed the western diet and worsened insulin sensitivity in control mice. We found no unmetabolized folic acid in liver from supplemented mice suggesting the metabolic effects of folic acid supplementation are not due to unmetabolized folic acid. Male fetal (gestational day 18.5) offspring from folic acid supplemented dams (western and control) had greater beta cell mass and density than those from unsupplemented dams; this was not observed in female offspring. Differential sex-specific hepatic gene expression profiles were observed in the fetal offspring from supplemented dams but this differed between western and controls. Our findings suggest that folic acid supplementation affects insulin sensitivity in female mice, but is dependent on their metabolic phenotype and has sex-specific effects on offspring pancreas and liver.
Collapse
Affiliation(s)
- Ei-Xia Mussai
- Department of Obstetrics and Gynecology, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Zoe A Lofft
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ben Vanderkruk
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicha Boonpattrawong
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua W Miller
- Department of Nutritional Sciences, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Andre Smith
- Department of Nutritional Sciences, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | | | - Angela M Devlin
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
23
|
Abstract
Unlike genetic changes, epigenetics modulates gene expression without stable modification of the genome. Even though all cells, including sperm and egg, have an epigenome pattern, most of these modifications occur during lifetime and interestingly, some of them, are reversible. Lifestyle and especially nutrients as well as diet regimens are presently gaining importance due to their ability to affect the epigenome. On the other hand, since the epigenome profoundly affects gene expression profile it can be speculated that the epigenome could modulate individual response to nutrients. Recent years have thus seen growing interest on nutrients, macronutrients ratio and diet regimens capable to affect the epigenetic pattern. In fact, while genetic alterations are mostly detrimental at the individual level, reshaping the epigenome may be a feasible strategy to positively counteract the detrimental effect of aging. Here, I review nutrient consumption and diet regimens as a possible strategy to counteract aging-driven epigenome derangement.
Collapse
Affiliation(s)
- Mario G Mirisola
- STeBiCeF Department, Università di Palermo, Building 16, Viale delle Scienze, 90128 Palermo, Italy
| |
Collapse
|
24
|
Li N, Li J, Wang H, Qiao Y, Li W, Gao M, Liu E, Yu Z, Hu G, Fang Z, Leng J, Yang X. Serum Sulfur-Containing Amino Acids and Risk of Maternal Gestational Diabetes and Adverse Growth Patterns in Offspring. Nutrients 2023; 15:4089. [PMID: 37764871 PMCID: PMC10537007 DOI: 10.3390/nu15184089] [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: 08/17/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND To estimate associations of sulfur-containing amino acids (SAAs) in the early trimester of pregnancy and gestational diabetes mellitus (GDM) and estimate associations of maternal SAAs with adverse growth patterns in offspring. METHODS We established a 1:1 matched case-control study (n = 486) from our cohort of pregnant women, and 401 children were followed up at ages 1 to 8 years. We conducted binary conditional logistic regression to estimate the risk associations of serum SAAs with GDM. Multinomial logistic regression was implemented to explore associations of maternal SAAs with adverse growth patterns in the offspring. RESULTS High serum methionine and cystine were independently associated with increased GDM risk (OR: 1.92, 95%CI: 1.18-3.13 and 2.69, 1.59-4.53). Conversely, a low level of serum taurine was independently associated with increased GDM risk (2.61, 1.64-4.16). Maternal high cystine and low taurine were also associated with an increased risk of persistent obesity growth pattern (POGP) in offspring (OR: 2.79, 95%CI: 1.09-7.17 and 3.92, 1.11-13.89) and the effect was largely independent of GDM. CONCLUSIONS High serum methionine, cystine and low serum taurine in the early trimester of pregnancy were associated with a greatly increased risk of GDM. Maternal high cystine and low taurine were associated with elevated risk of offspring POGP, largely independent of GDM.
Collapse
Affiliation(s)
- Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (N.L.); (J.L.); (H.W.); (M.G.)
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (N.L.); (J.L.); (H.W.); (M.G.)
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (N.L.); (J.L.); (H.W.); (M.G.)
| | - Yijuan Qiao
- Project Office, Tianjin Women and Children’s Health Center, Tianjin 300070, China; (Y.Q.); (W.L.); (E.L.)
| | - Weiqin Li
- Project Office, Tianjin Women and Children’s Health Center, Tianjin 300070, China; (Y.Q.); (W.L.); (E.L.)
| | - Ming Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (N.L.); (J.L.); (H.W.); (M.G.)
| | - Enqing Liu
- Project Office, Tianjin Women and Children’s Health Center, Tianjin 300070, China; (Y.Q.); (W.L.); (E.L.)
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, NS 15000, Canada;
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA;
| | - Zhongze Fang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Junhong Leng
- Project Office, Tianjin Women and Children’s Health Center, Tianjin 300070, China; (Y.Q.); (W.L.); (E.L.)
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (N.L.); (J.L.); (H.W.); (M.G.)
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Increasing bone mineral accrual during childhood might delay the onset of osteoporosis. We discuss the scientific evidence for early life approaches to optimising skeletal health. RECENT FINDINGS There is an ever-growing body of evidence from observational studies suggesting associations between early life exposures, particularly during foetal development, and bone mineral density (BMD). The findings of such studies are often heterogeneous, and for some exposures, for example, maternal smoking and alcohol intake in pregnancy or age at conception, intervention studies are not feasible. The most frequently studied exposures in intervention studies are calcium or vitamin D supplementation in pregnancy, which overall suggest positive effects on offspring childhood BMD. Maternal calcium and/or vitamin D supplementation during pregnancy appear to have positive effects on offspring BMD during early childhood, but further long-term follow-up is required to demonstrate persistence of the effect into later life.
Collapse
Affiliation(s)
- Rebecca J. Moon
- MRC Lifecourse Epidemiology Centre, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
- Paediatric Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Natasha L. Citeroni
- MRC Lifecourse Epidemiology Centre, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| | | | - Nicholas C. Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
26
|
Fernando KK, Craig JM, Dawson SL. Relationships between the maternal prenatal diet and epigenetic state in infants: a systematic review of human studies. J Dev Orig Health Dis 2023; 14:540-555. [PMID: 37496159 DOI: 10.1017/s2040174423000211] [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] [Indexed: 07/28/2023]
Abstract
Most human studies investigating the relationship between maternal diet in pregnancy and infant epigenetic state have focused on macro- and micro-nutrient intake, rather than the whole diet. This makes it difficult to translate the evidence into practical prenatal dietary recommendations.To review the evidence on how the prenatal diet relates to the epigenetic state of infants measured in the first year of life via candidate gene or genome-wide approaches.Following the PRISMA guidelines, this systematic literature search was completed in August 2020, and updated in August 2021 and April 2022. Studies investigating dietary supplementation were excluded. Risk of bias was assessed, and the certainty of results was analysed with consideration of study quality and validity.Seven studies were included, encompassing 6852 mother-infant dyads. One study was a randomised controlled trial and the remaining six were observational studies. There was heterogeneity in dietary exposure measures. Three studies used an epigenome-wide association study (EWAS) design and four focused on candidate genes from cord blood samples. All studies showed inconsistent associations between maternal dietary measures and DNA methylation in infants. Effect sizes of maternal diet on DNA methylation ranged from very low (< 1%) to high (> 10%). All studies had limitations and were assessed as having moderate to high risk of bias.The evidence presented here provides very low certainty that dietary patterns in pregnancy relate to epigenetic state in infants. We recommend that future studies maximise sample sizes and optimise and harmonise methods of dietary measurement and pipelines of epigenetic analysis.
Collapse
Affiliation(s)
- Kathya K Fernando
- Department of Immunology & Pathology, Alfred Health and Monash University, Melbourne, Australia
| | - Jeffrey M Craig
- Epigenetics, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Australia
| | - Samantha L Dawson
- Epigenetics, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Australia
| |
Collapse
|
27
|
Muroya S, Otomaru K, Oshima K, Oshima I, Ojima K, Gotoh T. DNA Methylation of Genes Participating in Hepatic Metabolisms and Function in Fetal Calf Liver Is Altered by Maternal Undernutrition during Gestation. Int J Mol Sci 2023; 24:10682. [PMID: 37445858 DOI: 10.3390/ijms241310682] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
This study aimed to elucidate the effects of maternal undernutrition (MUN) on epigenetic modification of hepatic genes in Japanese Black fetal calves during gestation. Using a previously established experimental design feeding the dams with 60% (LN) or 120% (HN) of their global nutritional requirements during the 8.5-month gestational period, DNA methylation in the fetal liver was analyzed with reduced representation bisulfite sequencing (RRBS). The promoters and gene bodies in the LN fetuses were hypomethylated compared to HN fetuses. Pathway analysis showed that the genes with DMR in the exon/intron in the LN group were associated with pathways involved in Cushing syndrome, gastric acid secretion, and aldosterone synthesis and secretion. Promoter hypomethylation in the LN group was frequently observed in genes participating in various signaling pathways (thyroid hormone, Ras/Rap1, PIK3-Akt, cAMP), fatty acid metabolism, and cholesterol metabolism. The promoter hypomethylated genes ALPL and GNAS were upregulated in the LN group, whereas the promoter hypermethylated genes GRB10 and POR were downregulated. The intron/exon hypomethylated genes IGF2, IGF2R, ACAD8, TAT, RARB, PINK1, and SOAT2 were downregulated, whereas the hypermethylated genes IGF2BP2, NOS3, and NR2F1 were upregulated. Collectively, MUN alters the promoter and gene body methylation of genes associated with hepatic metabolisms (energy, cholesterol, mitochondria) and function, suggesting an impact of altered gene methylation on the dysregulation of gene expression in the fetal liver.
Collapse
Affiliation(s)
- Susumu Muroya
- Division of Animal Products Research, NARO Institute of Livestock and Grassland Science (NILGS), Tsukuba 305-0901, Ibaraki, Japan
| | - Konosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Kazunaga Oshima
- Division of Year-Round Grazing Research, NARO Western Region Agricultural Research Center, 60 Yoshinaga, Ohda 694-0013, Shimane, Japan
| | - Ichiro Oshima
- Department of Agricultural Sciences and Natural Resources, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Koichi Ojima
- Division of Animal Products Research, NARO Institute of Livestock and Grassland Science (NILGS), Tsukuba 305-0901, Ibaraki, Japan
| | - Takafumi Gotoh
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| |
Collapse
|
28
|
Shafique S, Wolpert SH, Philbrook NA, Winn LM. Gestational exposure to triphenyl phosphate induces epigenetic modifications in C57Bl/6 fetal liver. Birth Defects Res 2023; 115:338-347. [PMID: 36369707 DOI: 10.1002/bdr2.2121] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/26/2022] [Accepted: 10/18/2022] [Indexed: 11/14/2022]
Abstract
INTRODUCTION Triphenyl phosphate (TPHP) is a chemical flame retardant and plasticizer which is added to consumer and industrial products. The developmental origins of health and disease hypothesis postulate that in utero exposures can have later-in-life effects on the developing fetus and can alter fetal gene expression. This study aimed to determine whether epigenetic modifications occurred following in utero TPHP exposure in mice and whether these changes were dose and/or sex-dependent. METHODS Pregnant C57Bl/6 mice were treated with 0, 5, 25, or 50 mg/kg of TPHP on gestational days (GD) 8, 10, 12, and 14 via intraperitoneal injection and fetal livers were collected on GD 19. Changes in the levels of acetylation of H3 and H4, as well as methylation of H3K9 and global DNA methylation were assessed in the fetal livers by western blot. RESULTS Results showed that there was a significant decrease in fetal DNA methylation following in utero exposure to 50 mg/kg TPHP compared to the control (0 mg/kg) independent of the sex of the fetus. While there were no significant alterations compared to controls in any histone modifications at any dose or sex following in utero TPHP exposure, we did note a decrease (t test, p = .025) in the levels of acetylated H3 in males versus females following a maternal dose of 25 mg/kg. The monomethylated H3K9 levels were also increased in females versus males following exposure to TPHP at 5 mg/kg (p = .018) and 25 mg/kg (p = .027) when analyzed via unpaired t tests, although not significantly different from controls. DISCUSSION The results suggest that gestational TPHP exposure can induce epigenetic modifications in murine fetal tissue. Specifically, global DNA methylation levels were downregulated in response to TPHP. Additionally, males appear to be more sensitive to TPHP-induced histone modifications than females. These data support the need for further studies investigating the impacts of gestational TPHP exposure on the developing fetus.
Collapse
Affiliation(s)
- Sidra Shafique
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sydney H Wolpert
- School of Environmental Studies, Queen's University, Kingston, Ontario, Canada
| | - Nicola A Philbrook
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Louise M Winn
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,School of Environmental Studies, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
29
|
Švorcová J. Transgenerational Epigenetic Inheritance of Traumatic Experience in Mammals. Genes (Basel) 2023; 14:120. [PMID: 36672861 PMCID: PMC9859285 DOI: 10.3390/genes14010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, we have seen an increasing amount of evidence pointing to the existence of a non-genetic heredity of the effects of events such as separation from parents, threat to life, or other traumatising experiences such as famine. This heredity is often mediated by epigenetic regulations of gene expression and may be transferred even across several generations. In this review, we focus on studies which involve transgenerational epigenetic inheritance (TEI), with a short detour to intergenerational studies focused on the inheritance of trauma or stressful experiences. The reviewed studies show a plethora of universal changes which stress exposure initiates on multiple levels of organisation ranging from hormonal production and the hypothalamic-pituitary-adrenal (HPA) axis modulation all the way to cognition, behaviour, or propensity to certain psychiatric or metabolic disorders. This review will also provide an overview of relevant methodology and difficulties linked to implementation of epigenetic studies. A better understanding of these processes may help us elucidate the evolutionary pathways which are at work in the course of emergence of the diseases and disorders associated with exposure to trauma, either direct or in a previous generation.
Collapse
Affiliation(s)
- Jana Švorcová
- Department of Philosophy and History of Science, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| |
Collapse
|
30
|
Morgan BL, Donohue K. Parental methylation mediates how progeny respond to environments of parents and of progeny themselves. ANNALS OF BOTANY 2022; 130:883-899. [PMID: 36201313 PMCID: PMC9758305 DOI: 10.1093/aob/mcac125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND AIMS Environments experienced by both parents and offspring influence progeny traits, but the epigenetic mechanisms that regulate the balance of parental vs. progeny control of progeny phenotypes are not known. We tested whether DNA methylation in parents and/or progeny mediates responses to environmental cues experienced in both generations. METHODS Using Arabidopsis thaliana, we manipulated parental and progeny DNA methylation both chemically, via 5-azacytidine, and genetically, via mutants of methyltransferase genes, then measured progeny germination responses to simulated canopy shade in parental and progeny generations. KEY RESULTS We first found that germination of offspring responded to parental but not seed demethylation. We further found that parental demethylation reversed the parental effect of canopy in seeds with low (Cvi-1) to intermediate (Col) dormancy, but it obliterated the parental effect in seeds with high dormancy (Cvi-0). Demethylation did so by either suppressing germination of seeds matured under white-light (Cvi-1) or under canopy (Cvi-0), or by increasing the germination of seeds matured under canopy (Col). Disruption of parental methylation also prevented seeds from responding to their own light environment in one genotype (Cvi-0, most dormant), but it enabled seeds to respond to their own environment in another genotype (Cvi-1, least dormant). Using mutant genotypes, we found that both CG and non-CG DNA methylation were involved in parental effects on seed germination. CONCLUSIONS Parental methylation state influences seed germination more strongly than does the progeny's own methylation state, and it influences how seeds respond to environments of parents and progeny in a genotype-specific manner.
Collapse
Affiliation(s)
- Britany L Morgan
- University Program in Ecology Duke University, Durham, NC 27705, USA
- Center for Agricultural Synthetic Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Kathleen Donohue
- University Program in Ecology Duke University, Durham, NC 27705, USA
- Biology Department, Duke University, Durham, NC 27705, USA
| |
Collapse
|
31
|
Rajamoorthi A, LeDuc CA, Thaker VV. The metabolic conditioning of obesity: A review of the pathogenesis of obesity and the epigenetic pathways that "program" obesity from conception. Front Endocrinol (Lausanne) 2022; 13:1032491. [PMID: 36329895 PMCID: PMC9622759 DOI: 10.3389/fendo.2022.1032491] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Understanding the developmental origins of health and disease is integral to overcome the global tide of obesity and its metabolic consequences, including atherosclerotic cardiovascular disease, type 2 diabetes, hyperlipidemia, and nonalcoholic fatty liver disease. The rising prevalence of obesity has been attributed, in part, to environmental factors including the globalization of the western diet and unhealthy lifestyle choices. In this review we argue that how and when such exposures come into play from conception significantly impact overall risk of obesity and later health outcomes. While the laws of thermodynamics dictate that obesity is caused by an imbalance between caloric intake and energy expenditure, the drivers of each of these may be laid down before the manifestation of the phenotype. We present evidence over the last half-century that suggests that the temporospatial evolution of obesity from intrauterine life and beyond is, in part, due to the conditioning of physiological processes at critical developmental periods that results in maladaptive responses to obesogenic exposures later in life. We begin the review by introducing studies that describe an association between perinatal factors and later risk of obesity. After a brief discussion of the pathogenesis of obesity, including the systemic regulation of appetite, adiposity, and basal metabolic rate, we delve into the mechanics of how intrauterine, postnatal and early childhood metabolic environments may contribute to adult obesity risk through the process of metabolic conditioning. Finally, we detail the specific epigenetic pathways identified both in preclinical and clinical studies that synergistically "program" obesity.
Collapse
Affiliation(s)
- Ananthi Rajamoorthi
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - Charles A. LeDuc
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
- The Naomi Berrie Diabetes Center, Columbia University IRVING Medical Center, New York, NY, United States
| | - Vidhu V. Thaker
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
- The Naomi Berrie Diabetes Center, Columbia University IRVING Medical Center, New York, NY, United States
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
32
|
Ormazabal V, Nair S, Carrión F, Mcintyre HD, Salomon C. The link between gestational diabetes and cardiovascular diseases: potential role of extracellular vesicles. Cardiovasc Diabetol 2022; 21:174. [PMID: 36057662 PMCID: PMC9441052 DOI: 10.1186/s12933-022-01597-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Extracellular vesicles are critical mediators of cell communication. They encapsulate a variety of molecular cargo such as proteins, lipids, and nucleic acids including miRNAs, lncRNAs, circular RNAs, and mRNAs, and through transfer of these molecular signals can alter the metabolic phenotype in recipient cells. Emerging studies show the important role of extracellular vesicle signaling in the development and progression of cardiovascular diseases and associated risk factors such as type 2 diabetes and obesity. Gestational diabetes mellitus (GDM) is hyperglycemia that develops during pregnancy and increases the future risk of developing obesity, impaired glucose metabolism, and cardiovascular disease in both the mother and infant. Available evidence shows that changes in maternal metabolism and exposure to the hyperglycemic intrauterine environment can reprogram the fetal genome, leaving metabolic imprints that define life-long health and disease susceptibility. Understanding the factors that contribute to the increased susceptibility to metabolic disorders of children born to GDM mothers is critical for implementation of preventive strategies in GDM. In this review, we discuss the current literature on the fetal programming of cardiovascular diseases in GDM and the impact of extracellular vesicle (EV) signaling in epigenetic programming in cardiovascular disease, to determine the potential link between EV signaling in GDM and the development of cardiovascular disease in infants.
Collapse
Affiliation(s)
- Valeska Ormazabal
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia.,Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile
| | - Soumyalekshmi Nair
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - H David Mcintyre
- Mater Research, Faculty of Medicine, University of Queensland, Mater Health, South Brisbane, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia. .,Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
33
|
A maternal low-protein diet during gestation induces hepatic autophagy-related gene expression in a sex-specific manner in Sprague-Dawley rats. Br J Nutr 2022; 128:592-603. [PMID: 34511147 PMCID: PMC9346618 DOI: 10.1017/s0007114521003639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study investigates the mechanism by which maternal protein restriction induces hepatic autophagy-related gene expression in the offspring of rats. Pregnant Sprague-Dawley rats were fed either a control diet (C, 18 % energy from protein) or a low-protein diet (LP, 8·5 % energy from protein) during gestation, followed by the control diet during lactation and post-weaning. Liver tissue was collected from the offspring at postnatal day 38 and divided into four groups according to sex and maternal diet (F-C, F-LP, M-C and M-LP) for further analysis. Autophagy-related mRNA and protein levels were determined by real-time PCR and Western blotting, respectively. In addition, chromatin immunoprecipitation (ChIP) was performed to investigate the interactions between transcription factors and autophagy-related genes. Protein levels of p- eukaryotic translation initiation factor 2a and activating transcription factor 4 (ATF4) were increased only in the female offspring born to dams fed the LP diet. Correlatively, the mRNA expression of hepatic autophagy-related genes including Map1lc3b, P62/Sqstm1, Becn1, Atg3, Atg7 and Atg10 was significantly greater in the F-LP group than in the F-C group. Furthermore, ChIP results showed greater ATF4 and C/EBP homology protein (CHOP) binding at the regions of a set of autophagy-related genes in the F-LP group than in the F-C group. Our data demonstrated that a maternal LP diet transcriptionally programmed hepatic autophagy-related gene expression only in female rat offspring. This transcriptional programme involved the activation of the eIF2α/ATF4 pathway and intricate regulation by transcription factors ATF4 and CHOP.
Collapse
|
34
|
Chavira-Suárez E, Reyes-Castro LA, López-Tenorio II, Vargas-Hernández L, Rodríguez-González GL, Chavira R, Zárate-Segura P, Domínguez-López A, Vadillo-Ortega F, Zambrano E. Sex-differential RXRα gene methylation effects on mRNA and protein expression in umbilical cord of the offspring rat exposed to maternal obesity. Front Cell Dev Biol 2022; 10:892315. [PMID: 36072345 PMCID: PMC9442673 DOI: 10.3389/fcell.2022.892315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Maternal obesity (MO) induces negative consequences in the offspring development. Adiposity phenotype is associated with maternal diet at early pregnancy and DNA methylation marks in the RXRα promotor at birth. Glucocorticoids play an important role in the regulation of metabolism through the activation of nuclear hormone receptors such as the RXRα protein. The aim of the study was to analyze steroid hormone changes at the end of pregnancy in the obese mother and RXRα gene methylation in the umbilical cord. For this purpose, in a well-established MO model, female Wistar rats were fed either standard chow (controls: C) or high-fat obesogenic diet (MO) before and during pregnancy to evaluate at 19 days of gestation (19 dG): 1) maternal concentration of circulating steroid hormones in MO and C groups, 2) maternal and fetal weights, 3) analysis of correlation between hormones concentration and maternal and fetal weights, 4) DNA methylation status of a single locus of RXRα gene near the early growth response (EGR-1) protein DNA binding site, and 5) RXRα mRNA and protein expressions in umbilical cords. Our results demonstrate that at 19 dG, MO body weight before and during pregnancy was higher than C; MO progesterone and corticosterone serum concentrations were higher and estradiol lower than C. There were not differences in fetal weight between male and female per group, therefore averaged data was used; MO fetal weight was lower than C. Positive correlations were found between progesterone and corticosterone with maternal weight, and estradiol with fetal weight, while negative correlation was observed between corticosterone and fetal weight. Additionally, male umbilical cords from MO were hypermethylated in RXRα gene compared to male C group, without differences in the female groups; mRNA and protein expression of RXRα were decreased in F1 male but not in female MO compared to C. In conclusion, MO results in dysregulation of circulating steroid hormones of the obese mothers and low fetal weight in the F1, modifying DNA methylation of RXRα gene as well as RXRα mRNA and protein expression in the umbilical cord in a sex-dependent manner.
Collapse
Affiliation(s)
- Erika Chavira-Suárez
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, México
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, México
| | - Luis Antonio Reyes-Castro
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Itzel Ivonn López-Tenorio
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, México
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, México
| | - Lilia Vargas-Hernández
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, México
- Instituto Mexicano del Seguro Social, Hospital de Ginecología y Obstetricia No. 4 Luis Castelazo Ayala, Mexico City, México
| | - Guadalupe L. Rodríguez-González
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Roberto Chavira
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Paola Zárate-Segura
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, México
| | | | - Felipe Vadillo-Ortega
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, México
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, México
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
- *Correspondence: Elena Zambrano,
| |
Collapse
|
35
|
Li P, He L, Lan Y, Fang J, Fan Z, Li Y. Intrauterine Growth Restriction Induces Adulthood Chronic Metabolic Disorder in Cardiac and Skeletal Muscles. Front Nutr 2022; 9:929943. [PMID: 35938117 PMCID: PMC9354130 DOI: 10.3389/fnut.2022.929943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Although population-based studies of intrauterine growth restriction (IUGR) demonstrated a series of postnatal complications, several studies identified that IUGR could definitely cause dysfunction of metabolism of cardiac and skeletal muscles in the perinatal period and early life. However, it is still unknown if such metabolic alternation would remain for long term or not, and whether normal protein diet administration postnatally would protect the IUGR offsprings from a “catch-up growth” and be able to reverse the premature metabolic remodeling. Materials and Methods We established an IUGR rat model with pregnant rats and a low-protein diet, and the developmental phenotypes had been carefully recorded. The cardiac and skeletal muscles had been collected to undergo RNA-seq. Results According to a series of comparisons of transcriptomes among various developmental processes, programmed metabolic dysfunction and chronic inflammation activity had been identified by transcriptome sequencing in IUGR offsprings, even such rats presented a normal developmental curve or body weight after normal postnatal diet feeding. Conclusion The data revealed that IUGR had a significant adverse impact on long-term cardiovascular function in rats, even they exhibit good nutritional status. So that, the fetal stage adverse events would encode the lifelong disease risk, which could hide in young age. This study remaindered that the research on long-term molecular changes is important, and only nutrition improvement would not totally reverse the damage of IUGR.
Collapse
Affiliation(s)
- Ping Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatrics, West China Second University Hospital, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lewei He
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yue Lan
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jie Fang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatrics, West China Second University Hospital, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Jie Fang,
| | - Zhenxin Fan
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- *Correspondence: Zhenxin Fan,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatrics, West China Second University Hospital, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Titcombe P, Murray R, Hewitt M, Antoun E, Cooper C, Inskip HM, Holbrook JD, Godfrey KM, Lillycrop K, Hanson M, Barton SJ. Human non-CpG methylation patterns display both tissue-specific and inter-individual differences suggestive of underlying function. Epigenetics 2022; 17:653-664. [PMID: 34461806 PMCID: PMC9235887 DOI: 10.1080/15592294.2021.1950990] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/25/2021] [Accepted: 06/29/2021] [Indexed: 12/29/2022] Open
Abstract
DNA methylation (DNAm) in mammals is mostly examined within the context of CpG dinucleotides. Non-CpG DNAm is also widespread across the human genome, but the functional relevance, tissue-specific disposition, and inter-individual variability has not been widely studied. Our aim was to examine non-CpG DNAm in the wider methylome across multiple tissues from the same individuals to better understand non-CpG DNAm distribution within different tissues and individuals and in relation to known genomic regulatory features.DNA methylation in umbilical cord and cord blood at birth, and peripheral venous blood at age 12-13 y from 20 individuals from the Southampton Women's Survey cohort was assessed by Agilent SureSelect methyl-seq. Hierarchical cluster analysis (HCA) was performed on CpG and non-CpG sites and stratified by specific cytosine environment. Analysis of tissue and inter-individual variation was then conducted in a second dataset of 12 samples: eight muscle tissues, and four aliquots of cord blood pooled from two individuals.HCA using methylated non-CpG sites showed different clustering patterns specific to the three base-pair triplicate (CNN) sequence. Analysis of CAC sites with non-zero methylation showed that samples clustered first by tissue type, then by individual (as observed for CpG methylation), while analysis using non-zero methylation at CAT sites showed samples grouped predominantly by individual. These clustering patterns were validated in an independent dataset using cord blood and muscle tissue.This research suggests that CAC methylation can have tissue-specific patterns, and that individual effects, either genetic or unmeasured environmental factors, can influence CAT methylation.
Collapse
Affiliation(s)
- Philip Titcombe
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Robert Murray
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Matthew Hewitt
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Elie Antoun
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Hazel M Inskip
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Joanna D Holbrook
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Karen Lillycrop
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Mark Hanson
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sheila J Barton
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| |
Collapse
|
37
|
Krstic N, Bishop N, Curtis B, Cooper C, Harvey N, Lilycrop K, Murray R, Owen R, Reilly G, Skerry T, Borg S. Early life vitamin D depletion and mechanical loading determine methylation changes in the RUNX2, RXRA, and osterix promoters in mice. GENES & NUTRITION 2022; 17:7. [PMID: 35619053 PMCID: PMC9137183 DOI: 10.1186/s12263-022-00711-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/11/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Early life vitamin D exposure is linked to later skeletal health with maternal vitamin D status in pregnancy associated with neonatal bone mass. The MAVIDOS study has demonstrated that vitamin D supplementation leads to reduced RXRA DNA methylation. Mice exposed to early life vitamin D deficiency have reduced bone mass and bone accrual in response to mechanical loading. Using the tibiae of these mice, we have examined the effect of diet and mechanical loading on the DNA methylation of promoters of genetic loci important for bone growth and development and their association with bone strength. RESULTS Mechanical loading of mouse tibiae leads to a reduction of RXRA DNA methylation. Early life vitamin D deficiency is associated with altered methylation of osterix and Runx2 in these bones. Tibia strength was also demonstrated to be associated with a change in DNA methylation status in CpGs of the vitamin D receptor (VDR), ostrix, and RXRA genes. CONCLUSIONS We have shown for the first time that mechanical loading of bone and early life vitamin D deficiency leads to changes in the epigenome of this tissue in key genes in the vitamin D and osteoblast differentiation pathway.
Collapse
Affiliation(s)
- Nevena Krstic
- Biological Sciences and NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| | - Nick Bishop
- Department of Oncology & Metabolism, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Beth Curtis
- MRC Lifecourse Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, UK
- Rheumatology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- NIHR Oxford Biomedical Research Unit, University of Oxford, Oxford, UK
| | - Nick Harvey
- MRC Lifecourse Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, UK
- Rheumatology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Karen Lilycrop
- Biological Sciences and NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| | - Robert Murray
- Biological Sciences and NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
| | - Robert Owen
- Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield, UK
- INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, UK
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Gwen Reilly
- Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield, UK
- INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, UK
| | - Tim Skerry
- Department of Oncology & Metabolism, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Steph Borg
- Department of Oncology & Metabolism, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
38
|
Xie L, Jiao Z, Zhang H, Wang T, Qin J, Zhang S, Luo M, Lu M, Yao B, Wang H, Xu D. Altered hippocampal GR/KCC2 signaling mediates susceptibility to convulsion in male offspring following dexamethasone exposure during pregnancy in rats. Toxicol Lett 2022; 364:12-23. [PMID: 35595036 DOI: 10.1016/j.toxlet.2022.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/02/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Epidemiological research suggests that convulsions may have an intrauterine developmental origin related to the application of dexamethasone, an artificially synthesized glucocorticoid. Here, using a rat animal model of prenatal dexamethasone exposure (PDE) we confirm that PDE can cause susceptibility to convulsions in male offspring and explore the epigenetic programming mechanism underlying this effect related to intrauterine type 2K+-Cl- cotransporter (KCC2). Wistar rats were injected with dexamethasone (0.2mg/kg/d) subcutaneously during the gestational days (GD) 9-20 and part of the offspring was given lithium pilocarpine (LiPC) at postnatal week 10. Our results showed that male offspring of the PDE+LiPC group exhibited convulsions susceptibility, as well as increased hippocampal gamma-aminobutyric acid (GABA) and intracellular chloride ions level and decreased GABA receptor expression. The offspring also showed a decrease of hippocampal KCC2 H3K14ac levels and KCC2 expression. PDE male fetal rats (GD20) showed similar changes to male offspring after birth and exhibited an increased expression of glucocorticoid receptor (GR) and histone deacetylase type 2 (HDAC2). We observed effects consistent with those observed in PDE fetal rats following in vitro dexamethasone treatment of the fetal rat hippocampal neuron H19-7 cell line, and the effects could be reversed by treatment with a GR inhibitor (RU486) or HDAC2 inhibitor (romidepsin). Taken together, this study confirmed that PDE causes a reduction of H3K14ac levels in the KCC2 promoter region caused by activation of fetal hippocampal GR-HDAC2-KCC2 signaling. We proposed that this abnormal epigenetic modification is the mechanism underlying offspring convulsions susceptibility. CATEGORIES: mechanism of toxicity.
Collapse
Affiliation(s)
- Lulu Xie
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhexiao Jiao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Haiju Zhang
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tingting Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jiaxin Qin
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Mingcui Luo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Mengxi Lu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| | - Dan Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
39
|
Vipin VA, Blesson CS, Yallampalli C. Maternal low protein diet and fetal programming of lean type 2 diabetes. World J Diabetes 2022; 13:185-202. [PMID: 35432755 PMCID: PMC8984567 DOI: 10.4239/wjd.v13.i3.185] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/30/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Maternal nutrition is found to be the key factor that determines fetal health in utero and metabolic health during adulthood. Metabolic diseases have been primarily attributed to impaired maternal nutrition during pregnancy, and impaired nutrition has been an immense issue across the globe. In recent years, type 2 diabetes (T2D) has reached epidemic proportion and is a severe public health problem in many countries. Although plenty of research has already been conducted to tackle T2D which is associated with obesity, little is known regarding the etiology and pathophysiology of lean T2D, a variant of T2D. Recent studies have focused on the effects of epigenetic variation on the contribution of in utero origins of lean T2D, although other mechanisms might also contribute to the pathology. Observational studies in humans and experiments in animals strongly suggest an association between maternal low protein diet and lean T2D phenotype. In addition, clear sex-specific disease prevalence was observed in different studies. Consequently, more research is essential for the understanding of the etiology and pathophysiology of lean T2D, which might help to develop better disease prevention and treatment strategies. This review examines the role of protein insufficiency in the maternal diet as the central driver of the developmental programming of lean T2D.
Collapse
Affiliation(s)
- Vidyadharan Alukkal Vipin
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Chellakkan Selvanesan Blesson
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
- Family Fertility Center, Texas Children's Hospital, Houston, TX 77030, United States
| | - Chandra Yallampalli
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
40
|
Maternal Protein Restriction and Post-Weaning High-Fat Feeding Alter Plasma Amino Acid Profiles and Hepatic Gene Expression in Mice Offspring. Foods 2022; 11:foods11050753. [PMID: 35267386 PMCID: PMC8909731 DOI: 10.3390/foods11050753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/25/2022] [Accepted: 03/03/2022] [Indexed: 12/04/2022] Open
Abstract
Maternal undernutrition during pregnancy is closely associated with epigenetic changes in the child, and it affects the development of obesity throughout the child’s life. Here, we investigate the effect of fetal low protein exposure and post-weaning high-fat consumption on plasma amino acid profiles and hepatic gene expression. Mother C57BL/6J mice were fed a 20% (CN) or 9% (LP) casein diet during pregnancy. After birth, the male offspring of both these groups were fed a high-fat diet (HF) from 6 to 32 weeks. At 32 weeks, the final body weight between the two groups remained unchanged, but the LP-HF group showed markedly higher white fat weight and plasma leptin levels. The LP-HF group exhibited a significant increase in the concentrations of isoleucine, leucine, histidine, phenylalanine, serine, and tyrosine. However, no differences were observed in the lipid content in the liver. According to the hepatic gene expression analysis, the LP-HF group significantly upregulated genes involved in the chromatin modification/organization pathways. Thus, maternal low protein and a post-weaning high-fat environment contributed to severe obesity states and changes in gene expression related to hepatic chromatin modification in offspring. These findings provide novel insights for the prevention of lifestyle-related diseases at the early life stage.
Collapse
|
41
|
Koemel NA, Senior AM, Dissanayake HU, Ross J, McMullan RL, Kong Y, Phang M, Hyett J, Raubenheimer D, Gordon A, Simpson SJ, Skilton MR. Maternal dietary fatty acid composition and newborn epigenetic aging-a geometric framework approach. Am J Clin Nutr 2022; 115:118-127. [PMID: 34591100 DOI: 10.1093/ajcn/nqab318] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Maternal nutrition is associated with epigenetic and cardiometabolic risk factors in offspring. Research in humans has primarily focused on assessing the impact of individual nutrients. OBJECTIVES We sought to assess the collective impact of maternal dietary MUFAs, PUFAs, and SFAs on epigenetic aging and cardiometabolic risk markers in healthy newborn infants using a geometric framework approach. METHODS Body fatness (n = 162), aortic intima-media thickness (aIMT; n = 131), heart rate variability (n = 118), and epigenetic age acceleration (n = 124) were assessed in newborn infants. Maternal dietary intake was cross-sectionally assessed in the immediate postpartum period via a validated 80-item self-administered FFQ. Generalized additive models were used to explore interactive associations of nutrient intake, with results visualized as response surfaces. RESULTS After adjustment for total energy intake, maternal age, gestational age, and sex there was a 3-way interactive association of MUFAs, PUFAs, and SFAs (P = 0.001) with newborn epigenetic aging. This suggests that the nature of each fat class association depends upon one another. Response surfaces revealed MUFAs were positively associated with newborn epigenetic age acceleration only at proportionately lower intakes of SFAs or PUFAs. We also demonstrate a potential beneficial association of omega-3 (n-3) PUFAs with newborn epigenetic age acceleration (P = 0.008). There was no significant association of fat class with newborn aIMT, heart rate variability, or body fatness. CONCLUSIONS In this study, we demonstrated an association between maternal dietary fat class composition and epigenetic aging in newborns. Future research should consider other characteristics such as the source of maternal dietary fatty acids.
Collapse
Affiliation(s)
- Nicholas A Koemel
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Hasthi U Dissanayake
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Sleep Research Group, The University of Sydney, Sydney, Australia
| | - Jason Ross
- CSIRO Health and Biosecurity, Sydney, Australia
| | - Rowena L McMullan
- Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Yang Kong
- Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Melinda Phang
- Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Jon Hyett
- Sydney Institute for Women, Children and their Families, Sydney Local Health District, Sydney, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Adrienne Gordon
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Institute for Women, Children and their Families, Sydney Local Health District, Sydney, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Michael R Skilton
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, The University of Sydney, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Sydney Institute for Women, Children and their Families, Sydney Local Health District, Sydney, Australia
| |
Collapse
|
42
|
Antoun E, Issarapu P, di Gravio C, Shrestha S, Betts M, Saffari A, Sahariah SA, Sankareswaran A, Arumalla M, Prentice AM, Fall CHD, Silver MJ, Chandak GR, Lillycrop KA. DNA methylation signatures associated with cardiometabolic risk factors in children from India and The Gambia: results from the EMPHASIS study. Clin Epigenetics 2022; 14:6. [PMID: 35000590 PMCID: PMC8744249 DOI: 10.1186/s13148-021-01213-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/08/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The prevalence of cardiometabolic disease (CMD) is rising globally, with environmentally induced epigenetic changes suggested to play a role. Few studies have investigated epigenetic associations with CMD risk factors in children from low- and middle-income countries. We sought to identify associations between DNA methylation (DNAm) and CMD risk factors in children from India and The Gambia. RESULTS Using the Illumina Infinium HumanMethylation 850 K Beadchip array, we interrogated DNAm in 293 Gambian (7-9 years) and 698 Indian (5-7 years) children. We identified differentially methylated CpGs (dmCpGs) associated with systolic blood pressure, fasting insulin, triglycerides and LDL-Cholesterol in the Gambian children; and with insulin sensitivity, insulinogenic index and HDL-Cholesterol in the Indian children. There was no overlap of the dmCpGs between the cohorts. Meta-analysis identified dmCpGs associated with insulin secretion and pulse pressure that were different from cohort-specific dmCpGs. Several differentially methylated regions were associated with diastolic blood pressure, insulin sensitivity and fasting glucose, but these did not overlap with the dmCpGs. We identified significant cis-methQTLs at three LDL-Cholesterol-associated dmCpGs in Gambians; however, methylation did not mediate genotype effects on the CMD outcomes. CONCLUSION This study identified cardiometabolic biomarkers associated with differential DNAm in Indian and Gambian children. Most associations were cohort specific, potentially reflecting environmental and ethnic differences.
Collapse
Affiliation(s)
- Elie Antoun
- School of Medicine, University of Southampton, Southampton, UK
| | - Prachand Issarapu
- Genomic Research On Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Chiara di Gravio
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Smeeta Shrestha
- Genomic Research On Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore, India
| | - Modupeh Betts
- MRC Unit The Gambia at the London, School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Ayden Saffari
- MRC Unit The Gambia at the London, School of Hygiene and Tropical Medicine, London, UK
| | | | - Alagu Sankareswaran
- Genomic Research On Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Manisha Arumalla
- Genomic Research On Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Andrew M Prentice
- MRC Unit The Gambia at the London, School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Caroline H D Fall
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Matt J Silver
- MRC Unit The Gambia at the London, School of Hygiene and Tropical Medicine, London, UK
| | - Giriraj R Chandak
- Genomic Research On Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Karen A Lillycrop
- School of Medicine, University of Southampton, Southampton, UK.
- Biological Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
43
|
Lite C, Raja GL, Juliet M, Sridhar VV, Subhashree KD, Kumar P, Chakraborty P, Arockiaraj J. In utero exposure to endocrine-disrupting chemicals, maternal factors and alterations in the epigenetic landscape underlying later-life health effects. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 89:103779. [PMID: 34843942 DOI: 10.1016/j.etap.2021.103779] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
Widespread persistence of endocrine-disrupting chemicals (EDCs) in the environment has mandated the need to study their potential effects on an individual's long-term health after both acute and chronic exposure periods. In this review article a particular focus is given on in utero exposure to EDCs in rodent models which resulted in altered epigenetic programming and transgenerational effects in the offspring causing disrupted reproductive and metabolic phenotypes. The literature to date establishes the impact of transgenerational effects of EDCs potentially associated with epigenetic mediated mechanisms. Therefore, this review aims to provide a comprehensive overview of epigenetic programming and it's regulation in mammals, primarily focusing on the epigenetic plasticity and susceptibility to exogenous hormone active chemicals during the early developmental period. Further, we have also in depth discussed the epigenetic alterations associated with the exposure to selected EDCs such as Bisphenol A (BPA), di-2-ethylhexyl phthalate (DEHP) and vinclozlin upon in utero exposure especially in rodent models.
Collapse
Affiliation(s)
- Christy Lite
- Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India.
| | - Glancis Luzeena Raja
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - Melita Juliet
- Department of Oral and Maxillofacial Surgery, SRM Kattankulathur Dental College and Hospital, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - Vasisht Varsh Sridhar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - K Divya Subhashree
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - Praveen Kumar
- Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Paromita Chakraborty
- Environmental Science and Technology Laboratory, Department of Chemical Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India.
| |
Collapse
|
44
|
Abstract
The intestinal tract is the entry gate for nutrients and symbiotic organisms, being in constant contact with external environment. DNA methylation is one of the keys to how environmental conditions, diet and nutritional status included, shape functionality in the gut and systemically. This review aims to summarise findings on the importance of methylation to gut development, differentiation and function. Evidence to date on how external factors such as diet, dietary supplements, nutritional status and microbiota modifications modulate intestinal function through DNA methylation is also presented.
Collapse
|
45
|
Vickers MH. Early life nutrition and neuroendocrine programming. Neuropharmacology 2021; 205:108921. [PMID: 34902348 DOI: 10.1016/j.neuropharm.2021.108921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022]
Abstract
Alterations in the nutritional environment in early life can significantly increase the risk for obesity and a range of development of metabolic disorders in offspring in later life, effects that can be passed onto future generations. This process, termed development programming, provides the framework of the developmental origins of health and disease (DOHaD) paradigm. Early life nutritional compromise including undernutrition, overnutrition or specific macro/micronutrient deficiencies, results in a range of adverse health outcomes in offspring that can be further exacerbated by a poor postnatal nutritional environment. Although the mechanisms underlying programming remain poorly defined, a common feature across the phenotypes displayed in preclinical models is that of altered wiring of neuroendocrine circuits that regulate satiety and energy balance. As such, altered maternal nutritional exposures during critical early periods of developmental plasticity can result in aberrant hardwiring of these circuits with lasting adverse consequences for the offspring. There is also increasing evidence around the role of an altered epigenome and the gut-brain axis in mediating some of the central programming effects observed. Further, although such programming was once considered to result in a permanent change in developmental trajectory, there is evidence, at least from preclinical models, that programming can be reversed via targeted nutritional manipulations during early development. Further work is required at a mechanistic level to allow for identification for early markers of later disease risk, delineation of sex-specific effects and pathways to implementation of strategies aimed at breaking the transgenerational transmission of disease.
Collapse
Affiliation(s)
- M H Vickers
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand.
| |
Collapse
|
46
|
Epigenetic Modifications at the Center of the Barker Hypothesis and Their Transgenerational Implications. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182312728. [PMID: 34886453 PMCID: PMC8656758 DOI: 10.3390/ijerph182312728] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/15/2021] [Accepted: 11/29/2021] [Indexed: 02/08/2023]
Abstract
Embryo/fetal nutrition and the environment in the reproductive tract influence the subsequent risk of developing adult diseases and disorders, as formulated in the Barker hypothesis. Metabolic syndrome, obesity, heart disease, and hypertension in adulthood have all been linked to unwanted epigenetic programing in embryos and fetuses. Multiple studies support the conclusion that environmental challenges, such as a maternal low-protein diet, can change one-carbon amino acid metabolism and, thus, alter histone and DNA epigenetic modifications. Since histones influence gene expression and the program of embryo development, these epigenetic changes likely contribute to the risk of adult disease onset not just in the directly affected offspring, but for multiple generations to come. In this paper, we hypothesize that the effects of parental nutritional status on fetal epigenetic programming are transgenerational and warrant further investigation. Numerous studies supporting this hypothesis are reviewed, and potential research techniques to study these transgenerational epigenetic effects are offered.
Collapse
|
47
|
Wang Y, Wang X, Long Q, Liu Y, Yin T, Sirota I, Ren F, Gu Z, Luo J. Reducing embryonic mtDNA copy number alters epigenetic profile of key hepatic lipolytic genes and causes abnormal lipid accumulation in adult mice. FEBS J 2021; 288:6828-6843. [PMID: 34258867 DOI: 10.1111/febs.16121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022]
Abstract
Adverse fetal environment, in particular a shortage or excess of nutrients, is associated with increased risks of metabolic diseases later in life. However, the molecular mechanisms underlying this developmental origin of adult diseases remain unclear. Here, we directly tested the role of mitochondrial stress in mediating fetal programming in mice by enzymatically depleting mtDNA in zygotes. mtDNA-targeted plasmid microinjection is used to reduce embryonic mtDNA copy number directly, followed by embryo transfer. Mice with reduced zygote mtDNA copy number were born morphologically normal and showed no accelerated body weight gain. However, at 5 months of age these mice showed markedly increased hepatic lipidosis and became glucose-intolerant. Hepatic mRNA and protein expressions of peroxisome proliferator-activated receptor α (Pparα), a key transcriptional regulator of lipid metabolism, were significantly decreased as a result of increased DNA methylation in its proximal regulatory region. These results indicate that perturbation of mitochondrial function around the periconceptional period causes hypermethylation and thus suppressed expression of PPARα in fetal liver, leading to impaired hepatic lipid metabolism. Our findings provide the first direct evidence that mitochondrial stress mediates epigenetic changes associated with fetal programming of adult diseases in a mammalian system.
Collapse
Affiliation(s)
- Yakun Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Qiaoming Long
- Cam-Su Mouse Genomic Resource Center, Soochow University, China
| | - Yuanwu Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Tao Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Inna Sirota
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Junjie Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
48
|
Higa R, Leonardi ML, Jawerbaum A. Intrauterine Programming of Cardiovascular Diseases in Maternal Diabetes. Front Physiol 2021; 12:760251. [PMID: 34803741 PMCID: PMC8595320 DOI: 10.3389/fphys.2021.760251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Maternal diabetes is a prevalent pathology that increases the risk of cardiovascular diseases in the offspring, the heart being one of the main target organs affected from the fetal stage until the adult life. Metabolic, pro-oxidant, and proinflammatory alterations in the fetal heart constitute the first steps in the adverse fetal programming of cardiovascular disease in the context of maternal diabetes. This review discusses both human and experimental studies addressing putative mechanisms involved in this fetal programming of heart damage in maternal diabetes. These include cardiac epigenetic changes, alterations in cardiac carbohydrate and lipid metabolism, damaging effects caused by a pro-oxidant and proinflammatory environment, alterations in the cardiac extracellular matrix remodeling, and specific signaling pathways. Putative actions to prevent cardiovascular impairments in the offspring of mothers with diabetes are also discussed.
Collapse
Affiliation(s)
- Romina Higa
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Laboratory of Reproduction and Metabolism, CONICET-Universidad de Buenos Aires, CEFYBO, Buenos Aires, Argentina
| | - María Laura Leonardi
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Laboratory of Reproduction and Metabolism, CONICET-Universidad de Buenos Aires, CEFYBO, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Laboratory of Reproduction and Metabolism, CONICET-Universidad de Buenos Aires, CEFYBO, Buenos Aires, Argentina
| |
Collapse
|
49
|
Cai S, Quan S, Yang G, Chen M, Ye Q, Wang G, Yu H, Wang Y, Qiao S, Zeng X. Nutritional Status Impacts Epigenetic Regulation in Early Embryo Development: A Scoping Review. Adv Nutr 2021; 12:1877-1892. [PMID: 33873200 PMCID: PMC8483970 DOI: 10.1093/advances/nmab038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/03/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
With the increasing maternal age and the use of assisted reproductive technology in various countries worldwide, the influence of epigenetic modification on embryonic development is increasingly notable and prominent. Epigenetic modification disorders caused by various nutritional imbalance would cause embryonic development abnormalities and even have an indelible impact on health in adulthood. In this scoping review, we summarize the main epigenetic modifications in mammals and the synergies among different epigenetic modifications, especially DNA methylation, histone acetylation, and histone methylation. We performed an in-depth analysis of the regulation of various epigenetic modifications on mammals from zygote formation to cleavage stage and blastocyst stage, and reviewed the modifications of key sites and their potential molecular mechanisms. In addition, we discuss the effects of nutrition (protein, lipids, and one-carbon metabolism) on epigenetic modification in embryos and emphasize the importance of various nutrients in embryonic development and epigenetics during pregnancy. Failures in epigenetic regulation have been implicated in mammalian and human early embryo loss and disease. With the use of reproductive technologies, it is becoming even more important to establish developmentally competent embryos. Therefore, it is essential to evaluate the extent to which embryos are sensitive to these epigenetic modifications and nutrition status. Understanding the epigenetic regulation of early embryo development will help us make better use of reproductive technologies and nutrition regulation to improve reproductive health in mammals.
Collapse
Affiliation(s)
- Shuang Cai
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Shuang Quan
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Guangxin Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Meixia Chen
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Qianhong Ye
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Gang Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Yuming Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Bio-feed Additives, China Agricultural University, Beijing, China
| |
Collapse
|
50
|
Leese HJ, McKeegan PJ, Sturmey RG. Amino Acids and the Early Mammalian Embryo: Origin, Fate, Function and Life-Long Legacy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:9874. [PMID: 34574797 PMCID: PMC8467587 DOI: 10.3390/ijerph18189874] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022]
Abstract
Amino acids are now recognised as having multiple cellular functions in addition to their traditional role as constituents of proteins. This is well-illustrated in the early mammalian embryo where amino acids are now known to be involved in intermediary metabolism, as energy substrates, in signal transduction, osmoregulation and as intermediaries in numerous pathways which involve nitrogen metabolism, e.g., the biosynthesis of purines, pyrimidines, creatine and glutathione. The amino acid derivative S-adenosylmethionine has emerged as a universal methylating agent with a fundamental role in epigenetic regulation. Amino acids are now added routinely to preimplantation embryo culture media. This review examines the routes by which amino acids are supplied to the early embryo, focusing on the role of the oviduct epithelium, followed by an outline of their general fate and function within the embryo. Functions specific to individual amino acids are then considered. The importance of amino acids during the preimplantation period for maternal health and that of the conceptus long term, which has come from the developmental origins of health and disease concept of David Barker, is discussed and the review concludes by considering the potential utility of amino acid profiles as diagnostic of embryo health.
Collapse
Affiliation(s)
- Henry J. Leese
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| | - Paul J. McKeegan
- Centre for Anatomical and Human Sciences, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| | - Roger G. Sturmey
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
- Division of Developmental Biology and Medicine, The University of Manchester, St Mary’s Hospital, Manchester M13 9WL, UK
| |
Collapse
|