1
|
Noriega L, Yang CY, Wang CH. Brown Fat and Nutrition: Implications for Nutritional Interventions. Nutrients 2023; 15:4072. [PMID: 37764855 PMCID: PMC10536824 DOI: 10.3390/nu15184072] [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: 08/02/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Brown and beige adipocytes are renowned for their unique ability to generate heat through a mechanism known as thermogenesis. This process can be induced by exposure to cold, hormonal signals, drugs, and dietary factors. The activation of these thermogenic adipocytes holds promise for improving glucose metabolism, reducing fat accumulation, and enhancing insulin sensitivity. However, the translation of preclinical findings into effective clinical therapies poses challenges, warranting further research to identify the molecular mechanisms underlying the differentiation and function of brown and beige adipocytes. Consequently, research has focused on the development of drugs, such as mirabegron, ephedrine, and thyroid hormone, that mimic the effects of cold exposure to activate brown fat activity. Additionally, nutritional interventions have been explored as an alternative approach to minimize potential side effects. Brown fat and beige fat have emerged as promising targets for addressing nutritional imbalances, with the potential to develop strategies for mitigating the impact of metabolic diseases. Understanding the influence of nutritional factors on brown fat activity can facilitate the development of strategies to promote its activation and mitigate metabolic disorders.
Collapse
Affiliation(s)
- Lloyd Noriega
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chih-Hao Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
2
|
Molière S, Jaulin A, Tomasetto CL, Dali-Youcef N. Roles of Matrix Metalloproteinases and Their Natural Inhibitors in Metabolism: Insights into Health and Disease. Int J Mol Sci 2023; 24:10649. [PMID: 37445827 DOI: 10.3390/ijms241310649] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-activated peptidases that can be classified into six major classes, including gelatinases, collagenases, stromelysins, matrilysins, membrane type metalloproteinases, and other unclassified MMPs. The activity of MMPs is regulated by natural inhibitors called tissue inhibitors of metalloproteinases (TIMPs). MMPs are involved in a wide range of biological processes, both in normal physiological conditions and pathological states. While some of these functions occur during development, others occur in postnatal life. Although the roles of several MMPs have been extensively studied in cancer and inflammation, their function in metabolism and metabolic diseases have only recently begun to be uncovered, particularly over the last two decades. This review aims to summarize the current knowledge regarding the metabolic roles of metalloproteinases in physiology, with a strong emphasis on adipose tissue homeostasis, and to highlight the consequences of impaired or exacerbated MMP actions in the development of metabolic disorders such as obesity, fatty liver disease, and type 2 diabetes.
Collapse
Affiliation(s)
- Sébastien Molière
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- Department of Radiology, Strasbourg University Hospital, Hôpital de Hautepierre, Avenue Molière, 67200 Strasbourg, France
- Breast and Thyroid Imaging Unit, ICANS-Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Amélie Jaulin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Catherine-Laure Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
| | - Nassim Dali-Youcef
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Illkirch, 67400 Illkirch-Graffenstaden, France
- Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale, U1258, 67400 Illkirch-Graffenstaden, France
- Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
- Laboratoire de Biochimie et Biologie Moléculaire, Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, 67000 Strasbourg, France
| |
Collapse
|
3
|
Attaallah A, Elmrazeky AR, El-Beltagy AEFBM, Abdelaziz KK, Soliman MF. Modulatory role of Coriandrum sativum (coriander) extract against diabetic complications on the gonads of female rats and their offspring. Tissue Cell 2023; 83:102127. [PMID: 37331322 DOI: 10.1016/j.tice.2023.102127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/10/2023] [Accepted: 06/06/2023] [Indexed: 06/20/2023]
Abstract
It is well known that diabetes is associated with impairment of ovarian and testicular structure and function. Coriander (Coriandrum sativum L.) is identified as one of the oldest herbal plants valued for its nutritional and medicinal properties. This work is mainly designed to evaluate the possible modulatory role of dry coriander fruit extract against gonadal impairments associated with diabetes in female rats and their pups. Twenty-four pregnant rats were divided into four groups (n = 6): group I served as control, group II was administered daily with coriander fruit extract (250 mg/kg b.wt), group III was injected interaperitoneally with a single dose of streptozotocin (STZ) (80 mg/kg b.wt), and group IV was injected with single dose of STZ and post administered coriander extract. The experiment was conducted from the 4th day of gestation till the end of weaning. At the end of the experiment, the mothers' rats and their offspring were weighed, sacrificed, the ovaries from mothers as well as ovaries and testes from offspring were immediately excised, and processed for histological, immunohistochemical and evaluation of apoptosis and transforming growth factorβ (TGF-β). Also, blood samples were collected and analyzed to estimate the levels of sex hormones as well as antioxidants.In STZ induced diabetes in mother's rats and their offspring, the ovarian sections revealed severe histopathological signs included several atretic follicles, dilated and congested blood capillaries. Additionally, the testicular sections of offspring appeared with destructive seminiferous tubules. Immunohistochemically, the ovarian sections displayed weak to negative expression for calretinin marker however the testicular sections showed strong expression for Bax protein (apoptotic marker) and weak to negative expression for Ki67 protein (proliferative marker). Also, the mean % values of positively expressed cells for TGF-β and annexin-v markers (late and early apoptosis indicator) were significantly elevated in the ovarian and testicular tissues of STZ-induced group of mother's rats and their pups if compared with control. Further results revealed that the levels of insulin, FSH, LH, estrogen, SOD and CAT were significantly decreased if compared with control however the levels of MDA and NO were significantly elevated. Administration of coriander fruit extract to diabetic rats successfully alleviated most of the altered histological, immunohistochemical, biochemical, and apoptotic changes induced by diabetes. Coriandrum sativum fruit extract has a powerful ameliorative role against STZ-induced diabetic gonadal dysfunctions in female rats and their offspring.
Collapse
Affiliation(s)
- Amany Attaallah
- Zoology Department, Faculty of Science, Damanhur University, Egypt.
| | | | | | | | - Mona Fm Soliman
- Histology Department, Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
4
|
Jeje SO, Adenawoola M, Abosede C. Gestational Nutrition as a Predisposing Factor to Obesity Onset in Offspring: Role for Involvement of Epigenetic Mechanism. Niger J Physiol Sci 2022; 37:1-7. [PMID: 35947841 DOI: 10.54548/njps.v37i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
Maternal lifestyle has been implicated as a predisposing factor in the development of metabolic disorders in adulthood. This lifestyle includes the immediate environment, physical activity and nutrition. Maternal nutrition has direct influence on the developmental programming through biochemical alterations and can lead to modifications in the fetal genome through epigenetic mechanisms. Imbalance in basic micro or macro nutrients due to famine or food deficiency during delicate gestational periods can lead to onset of metabolic syndrome including obesity. A major example is the Dutch famine which led to a serious metabolic disorder in adulthood of affected infants. Notably due to gene variants, individualized responses to nutritional deficiencies are unconventional, therefore intensifying the need to study nutritional genomics during fetal programming. Epigenetic mechanisms can cause hereditary changes without changing the DNA sequence; the major mechanisms include small non-coding RNAs, histone modifications and most stable of all is DNA methylation. The significance association between obesity and DNA methylation is through regulation of genes implicated in lipid and glucose metabolism either directly or indirectly by hypomethylation or hypermethylation. Examples include CPT1A, APOA2, ADRB3 and POMC. Any maternal exposure to malnutrition or overnutrition that can affect genes regulating major metabolic pathways in the fetus, will eventually cause underlying changes that can predispose or cause the onset of metabolic disorder in adulthood. In this review, we examined the interaction between nutrition during gestation and epigenetic programming of metabolic syndrome.
Collapse
|
5
|
Shokri M, Rizebandi M, Karimi P, Badfar G, Nasirkandy MP, Kalvandi G, Rahmati S. Association between Maternal Birth Weight and Gestational Diabetes Mellitus: A Systematic Review and Meta-Analysis. J Obstet Gynaecol India 2022; 72:125-133. [PMID: 35492858 PMCID: PMC9008117 DOI: 10.1007/s13224-022-01645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022] Open
Abstract
Background and Aims Gestational diabetes mellitus is one of the most important issue related to health status of mothers and their children throughout life. This meta-analysis has been conducted to assess relationship between maternal birth weight and gestational diabetes. Methods and Results This article is written using PRISMA guideline for systematic review and meta-analysis. We searched epidemiological studies without a time limit from following databases-Scopus, PubMed, Science Direct, Embase, Web of Science, CINAHL, Cochrane, EBSCO, and Google Scholar search engine using MESH keywords. Heterogeneity was determined using the Cochran Q test and I 2 index. Data were analyzed using comprehensive meta-analysis, version 2. The significance level of the tests was considered as P < 0.05. Results The result of combining ten studies with a sample size of 228,409 cases using a fixed-effect model showed that low maternal birth weight increased the risk of gestational diabetes mellitus (1.71 [95% CI 1.43-2.06, P < 0.001]). In addition, the result of combining nine studies with a sample size of 227,805 cases using a random-effects model showed that macrosomia did not increase the risk of gestational diabetes mellitus, and there was no significant relationship between them (1.04 [95% CI 0.79-1.38; p value: 0.730]). Conclusion The results of this systematic review and meta-analysis showed that low maternal birth weight could be a risk factor for gestational diabetes in adulthood.
Collapse
Affiliation(s)
- Mehdi Shokri
- Department of Pediatrics, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Mahsa Rizebandi
- Department of Internal Medicine, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Parviz Karimi
- Department of Pediatrics, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Gholamreza Badfar
- Department of Pediatric, Faculty of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Marzieh Parizad Nasirkandy
- Department of Obstetrics and Gynecology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Kalvandi
- Associate Professor of Pediatrics Gastroenterology, Department of Pediatrics, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shoboo Rahmati
- Student Research Committee, Department of Epidemiology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Li Y, Pollock CA, Saad S. Aberrant DNA Methylation Mediates the Transgenerational Risk of Metabolic and Chronic Disease Due to Maternal Obesity and Overnutrition. Genes (Basel) 2021; 12:genes12111653. [PMID: 34828259 PMCID: PMC8624316 DOI: 10.3390/genes12111653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/02/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is a rapidly evolving universal epidemic leading to acute and long-term medical and obstetric health issues, including increased maternal risks of gestational diabetes, hypertension and pre-eclampsia, and the future risks for offspring's predisposition to metabolic diseases. Epigenetic modification, in particular DNA methylation, represents a mechanism whereby environmental effects impact on the phenotypic expression of human disease. Maternal obesity or overnutrition contributes to the alterations in DNA methylation during early life which, through fetal programming, can predispose the offspring to many metabolic and chronic diseases, such as non-alcoholic fatty liver disease, obesity, diabetes, and chronic kidney disease. This review aims to summarize findings from human and animal studies, which support the role of maternal obesity in fetal programing and the potential benefit of altering DNA methylation to limit maternal obesity related disease in the offspring.
Collapse
Affiliation(s)
- Yan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China;
| | - Carol A. Pollock
- Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2065, Australia;
| | - Sonia Saad
- Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2065, Australia;
- Correspondence:
| |
Collapse
|
7
|
Goodrich JM, Hector EC, Tang L, LaBarre JL, Dolinoy DC, Mercado-Garcia A, Cantoral A, Song PX, Téllez-Rojo MM, Peterson KE. Integrative Analysis of Gene-Specific DNA Methylation and Untargeted Metabolomics Data from the ELEMENT Cohort. Epigenet Insights 2020; 13:2516865720977888. [PMID: 33354655 PMCID: PMC7734565 DOI: 10.1177/2516865720977888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022] Open
Abstract
Epigenetic modifications, such as DNA methylation, influence gene expression and cardiometabolic phenotypes that are manifest in developmental periods in later life, including adolescence. Untargeted metabolomics analysis provide a comprehensive snapshot of physiological processes and metabolism and have been related to DNA methylation in adults, offering insights into the regulatory networks that influence cellular processes. We analyzed the cross-sectional correlation of blood leukocyte DNA methylation with 3758 serum metabolite features (574 of which are identifiable) in 238 children (ages 8-14 years) from the Early Life Exposures in Mexico to Environmental Toxicants (ELEMENT) study. Associations between these features and percent DNA methylation in adolescent blood leukocytes at LINE-1 repetitive elements and genes that regulate early life growth (IGF2, H19, HSD11B2) were assessed by mixed effects models, adjusting for sex, age, and puberty status. After false discovery rate correction (FDR q < 0.05), 76 metabolites were significantly associated with LINE-1 DNA methylation, 27 with HSD11B2, 103 with H19, and 4 with IGF2. The ten identifiable metabolites included dicarboxylic fatty acids (five associated with LINE-1 or H19 methylation at q < 0.05) and 1-octadecanoyl-rac-glycerol (q < 0.0001 for association with H19 and q = 0.04 for association with LINE-1). We then assessed the association between these ten known metabolites and adiposity 3 years later. Two metabolites, dicarboxylic fatty acid 17:3 and 5-oxo-7-octenoic acid, were inversely associated with measures of adiposity (P < .05) assessed approximately 3 years later in adolescence. In stratified analyses, sex-specific and puberty-stage specific (Tanner stage = 2 to 5 vs Tanner stage = 1) associations were observed. Most notably, hundreds of statistically significant associations were observed between H19 and LINE-1 DNA methylation and metabolites among children who had initiated puberty. Understanding relationships between subclinical molecular biomarkers (DNA methylation and metabolites) may increase our understanding of genes and biological pathways contributing to metabolic changes that underlie the development of adiposity during adolescence.
Collapse
Affiliation(s)
- Jaclyn M Goodrich
- Deptartment of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Emily C Hector
- Deptartment of Biostatistics, University of Michigan, Ann Arbor, MI, USA.,Deptartment of Statistics, North Carolina State University, USA
| | - Lu Tang
- Deptartment of Biostatistics, University of Pittsburgh, USA
| | - Jennifer L LaBarre
- Deptartment of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Deptartment of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA.,Deptartment of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Adriana Mercado-Garcia
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Alejandra Cantoral
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Peter Xk Song
- Deptartment of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Martha Maria Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Karen E Peterson
- Deptartment of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA.,Deptartment of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Abstract
In order to better understand the events that precede and precipitate the onset of type 2 diabetes (T2DM), several nutritional animal models have been developed. These models are generated by manipulating the diet of either the animal itself, or its mother during her pregnancy, and in comparison to traditional genetic and knock out models, have the advantage that they more accurately reflect the etiology of human T2DM. This chapter will discuss some of the most widely used nutritional models of T2DM: Diet-induced obesity (DIO) in adult rodents, and studies of offspring of mothers fed a low-protein, high-fat and/or high-sugar diet during pregnancy and/or lactation. Several common mechanisms have been identified through which these nutritional manipulations can lead to metabolic disease, including pancreatic beta-cell dysfunction, impaired insulin signaling in skeletal muscle, and the excess accumulation of visceral adipose tissue and consequent deposition of nonesterified fatty acids in peripheral tissues. In addition, there is an emerging concept that obesity/poor quality diets result in increased production and release of pro-inflammatory cytokines from adipose tissue leading to a state of chronic low-grade inflammation, and that this is likely to represent an important link between obesity/diet and metabolic dysfunction. The following chapter will discuss the most common nutritional models of T2DM in experimental animals, their application, and relationship to human etiology, and will highlight the important insights these models have provided into the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Beverly Sara Mühlhäusler
- Food and Nutrition Research Group, Department of Food and Wine Sciences, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia.
- FOODplus Research Centre, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia.
- CSIRO, Health and Biosecurity, Adelaide, SA, Australia.
| | - Carla Toop
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sheridan Gentili
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
9
|
Kim M. DNA methylation: a cause and consequence of type 2 diabetes. Genomics Inform 2019; 17:e38. [PMID: 31896238 PMCID: PMC6944052 DOI: 10.5808/gi.2019.17.4.e38] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022] Open
Abstract
DNA methylation is a relatively stable epigenetic modification that can regulate and stabilize gene expression patterns and hence establish cell identity. Because metabolic intermediates are key factors of DNA methylation and demethylation, perturbations in metabolic homeostasis can trigger alterations in cell-specific patterns of DNA methylation and contribute to disease development, including type 2 diabetes (T2D). During the past decade, genome-wide DNA methylation studies of T2D have expanded our knowledge of the molecular mechanisms underlying T2D. This review summarizes case-control studies of the DNA methylome of T2D and discusses DNA methylation as both a cause and consequence of T2D. Therefore, DNA methylation has potential as a promising T2D biomarker that can be applied to the development of therapeutic strategies for T2D.
Collapse
Affiliation(s)
- Mirang Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| |
Collapse
|
10
|
|
11
|
Plasticity of histone modifications around Cidea and Cidec genes with secondary bile in the amelioration of developmentally-programmed hepatic steatosis. Sci Rep 2019; 9:17100. [PMID: 31745102 PMCID: PMC6863835 DOI: 10.1038/s41598-019-52943-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
We recently reported that a treatment with tauroursodeoxycholic acid (TUDCA), a secondary bile acid, improved developmentally-deteriorated hepatic steatosis in an undernourishment (UN, 40% caloric restriction) in utero mouse model after a postnatal high-fat diet (HFD). We performed a microarray analysis and focused on two genes (Cidea and Cidec) because they are enhancers of lipid droplet (LD) sizes in hepatocytes and showed the greatest up-regulation in expression by UN that were completely recovered by TUDCA, concomitant with parallel changes in LD sizes. TUDCA remodeled developmentally-induced histone modifications (dimethylation of H3K4, H3K27, or H3K36), but not DNA methylation, around the Cidea and Cidec genes in UN pups only. Changes in these histone modifications may contribute to the markedly down-regulated expression of Cidea and Cidec genes in UN pups, which was observed in the alleviation of hepatic fat deposition, even under HFD. These results provide an insight into the future of precision medicine for developmentally-programmed hepatic steatosis by targeting histone modifications.
Collapse
|
12
|
Barrón-Cabrera E, Ramos-Lopez O, González-Becerra K, Riezu-Boj JI, Milagro FI, Martínez-López E, Martínez JA. Epigenetic Modifications as Outcomes of Exercise Interventions Related to Specific Metabolic Alterations: A Systematic Review. Lifestyle Genom 2019; 12:25-44. [PMID: 31546245 DOI: 10.1159/000503289] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chronic diseases arise as a consequence of an unhealthy lifestyle primarily characterized by physical inactivity and unbalanced diets. Regular physical activity can improve health, and there is consistent evidence that these improvements may be the result of epigenetic modifications. OBJECTIVE To identify epigenetic modificationsas outcomes of exercise interventions related to specific metabolic alterations. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols (PRISMA-P) methodology for manuscript research and preparation was followed using PubMed and EBSCO databases for literature review. Out of 2,638 articles identified, only 34 articles met the inclusion criteria. RESULTS The sections of the review were organized by metabolic alterations in which studies were grouped according to healthy, diseased, and trained individuals. Resistance exercise in humans induced epigenetic changes in pathways associated with energy metabolism and insulin sensitivity, contributing to healthy skeletal muscle. Endurance exercise also caused modifications in biomarkers associated to metabolic alterations through changes in DNA methylation and the expression of specific miRNAs. However, both resistance and endurance exercise are necessary to obtain a better physiological adaptation and a combination of both seems to be needed to properly tackle the increasing prevalence of non-communicable pathologies. CONCLUSION Given the heterogeneity and complexity of the existing literature, it is currently not possible to propose a specific recommendation about the type, intensity, or duration of exercise that could be beneficial for different subsets of the population (healthy, diseased, and/or trained). Nevertheless, this review highlights the importance of exercise for health and shows the need to perform more research in this emerging area to identify epigenetic biomarkers that could serve as indicators of exercise adaptations.
Collapse
Affiliation(s)
- Elisa Barrón-Cabrera
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara, Mexico
| | - Omar Ramos-Lopez
- Centre for Nutrition Research, Department of Nutrition, Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Spain.,Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana, Mexico
| | - Karina González-Becerra
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara, Mexico
| | - Jose Ignacio Riezu-Boj
- Centre for Nutrition Research, Department of Nutrition, Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Fermin I Milagro
- Centre for Nutrition Research, Department of Nutrition, Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Carlos III Health Institute, Madrid, Spain
| | - Erika Martínez-López
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara, Mexico
| | - Jose Alfredo Martínez
- Centre for Nutrition Research, Department of Nutrition, Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Spain, .,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain, .,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Carlos III Health Institute, Madrid, Spain, .,Madrid Institute of Advanced Studies (IMDEA Food), Madrid, Spain,
| |
Collapse
|
13
|
Guo J, Li B, Zuo Z, Chen M, Wang C. Maternal Supplementation with β‐Carotene During Pregnancy Disturbs Lipid Metabolism and Glucose Homoeostasis in F1 Female Mice. Mol Nutr Food Res 2019; 63:e1900072. [DOI: 10.1002/mnfr.201900072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Jiaojiao Guo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Bingshui Li
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Meng Chen
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem ResearchXiamen University Xiamen 36110 P. R. China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem ResearchXiamen University Xiamen 36110 P. R. China
| |
Collapse
|
14
|
Ozkan H, Topsakal S, Ozmen O. Investigation of the diabetic effects of maternal high-glucose diet on rats. Biomed Pharmacother 2019; 110:609-617. [DOI: 10.1016/j.biopha.2018.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 11/30/2018] [Accepted: 12/02/2018] [Indexed: 12/18/2022] Open
|
15
|
Azizi S, Shamshirian A, Alizadeh-Navaei R, Jafarpour H, Asemi Z, Tamtaji OR, Vaziri MS, Homayounfar R, Rezaei Shahmirzadi A, Alipoor R. A Genetic Association Study of MTHFR C677T Polymorphism with Risk of Metabolic Syndrome: A Systematic Review and Meta-Analysis. Galen Med J 2019; 8:e1472. [PMID: 34466514 PMCID: PMC8343661 DOI: 10.31661/gmj.v8i0.1472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/23/2019] [Accepted: 02/08/2019] [Indexed: 02/05/2023] Open
Abstract
Methylenetetrahydrofolate reductase (MTHFR) is an enzyme that plays a crucial role as a methyl-group donor in demethylation of homocysteine. The aim of this systematic review and meta-analysis was to study the relationship between MTHFR gene polymorphism and metabolic syndrome (MS). We used search engines and databases such as Science Direct, Google Scholar, Embase, Cochrane Library, and PubMed to identify eligible studies up to 2018. The articles were studied based on keywords including MTHFR, mutation, variant, and polymorphism in combination with MS. Data was analyzed using Comprehensive Meta-Analysis version 2.2.064 software. After extracting the data from seven articles, the total number of subjects was 1280 in the patient group and 1374 in the control group. The odds ratio was estimated to be 1.078 for the allele model of T vs. C (95% confidence interval [CI]: 1.626-0.715), 1.157 for the allele model of CC vs. CT (95% CI: 0.829-1.615), 1.020 for the allele model of CT + TT vs. CC (95% CI: 1.611-0.646) and 0.799 for the allele model of TT vs. CC + CT (95% CI: 1.185- 0.539). As well, the results showed no statistically significant correlation between polymorphism genotypes of the MTHFR gene and MS (P<0.05). In general, this study showed that the presence of C677T polymorphism in the MTHFR gene has no effect on the incidence of MS.
Collapse
Affiliation(s)
- Soheil Azizi
- Department of Laboratory Sciences, Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amir Shamshirian
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamed Jafarpour
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Omid Reza Tamtaji
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sadegh Vaziri
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Reza Homayounfar
- Non-Communicable Disease Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Reza Alipoor
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Non-Communicable Disease Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Correspondence to: Reza Alipoor, Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran Telephone Number: +989174462124 Email Address:
| |
Collapse
|
16
|
Sharma H, Kumar P, Deshmukh RR, Bishayee A, Kumar S. Pentacyclic triterpenes: New tools to fight metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 50:166-177. [PMID: 30466975 DOI: 10.1016/j.phymed.2018.09.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/25/2018] [Accepted: 09/03/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Metabolic syndrome is a combination of dysregulated cardiometabolic risk factors characterized by dyslipidemia, impaired glucose tolerance, insulin resistance, inflammation, obesity as well as hypertension. These factors are tied to the increased risk for type-II diabetes and cardiovascular diseases including myocardial infarction in patients with metabolic syndrome. PURPOSE To review the proposed molecular mechanisms of pentacyclic triterpenes for their potential use in the metabolic syndrome. METHODS PubMed, Science Direct, and Google Scholar database were searched from commencement to April 2018. Following keywords were searched in the databases with varying combinations: "metabolic syndrome", "pentacyclic triterpenes", "transcription factors", "protein kinase", "lipogenesis", "adipogenesis", "lipolysis", "fatty acids", "gluconeogenesis", "cardiovascular", "mitochondria", "oxidative stress", "pancreas", "hepatic cells", "skeletal muscle", "3T3-L1", "C2C12", "obesity", "inflammation", "insulin resistance", "glucose uptake", "clinical studies" and "bioavailability". RESULTS Pentacyclic triterpenes, such as asiatic acid, ursolic acid, oleanolic acid, 18β-glycyrrhetinic acid, α,β-amyrin, celastrol, carbenoxolone, corosolic acid, maslinic acid, bardoxolone methyl and lupeol downregulate several metabolic syndrome components by regulating transcription factors, protein kinases and enzyme involved in the adipogenesis, lipolysis, fatty acid oxidation, insulin resistance, mitochondria biogenesis, gluconeogenesis, oxidative stress and inflammation. CONCLUSION In vitro and in vivo studies suggests that pentacyclic triterpenes effectively downregulate various factors related to metabolic syndrome. These phytochemicals may serve as promising candidates for clinical trials for the management of metabolic syndrome.
Collapse
Affiliation(s)
- Hitender Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136 119 Haryana, India
| | - Pushpander Kumar
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136 119 Haryana, India
| | - Rahul R Deshmukh
- School of Pharmacy, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Sunil Kumar
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136 119 Haryana, India.
| |
Collapse
|
17
|
Devaux CA, Raoult D. The Microbiological Memory, an Epigenetic Regulator Governing the Balance Between Good Health and Metabolic Disorders. Front Microbiol 2018; 9:1379. [PMID: 29997595 PMCID: PMC6028609 DOI: 10.3389/fmicb.2018.01379] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022] Open
Abstract
If the transmission of biological information from one generation to the next is based on DNA, most heritable phenotypic traits such as chronic metabolic diseases, are not linked to genetic variation in DNA sequences. Non-genetic heritability might have several causes including epigenetic, parental effect, adaptive social learning, and influence of the ecological environment. Distinguishing among these causes is crucial to resolve major phenotypic enigmas. Strong evidence indicates that changes in DNA expression through various epigenetic mechanisms can be linked to parent-offspring resemblance in terms of sensitivity to metabolic diseases. Among non-genetic heritable traits, early nutrition could account for a long term deviant programming of genes expression responsible for metabolic diseases in adulthood. Nutrition could shape an inadequate gut microbiota (dysbiosis), triggering epigenetic deregulation of transcription which can be observed in chronic metabolic diseases. We review herein the evidence that dysbiosis might be a major cause of heritable epigenetic patterns found to be associated with metabolic diseases. By taking into account the recent advances on the gut microbiome, we have aggregated together different observations supporting the hypothesis that the gut microbiota could promote the molecular crosstalk between bacteria and surrounding host cells which controls the pathological epigenetic signature. We introduce for the first time the concept of "microbiological memory" as the main regulator of the epigenetic signatures, thereby indicating that different causes of non-genetic heritability can interact in complex pathways to produce inheritance.
Collapse
Affiliation(s)
- Christian A. Devaux
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix-Marseille University, Marseille, France
- Centre National de la Recherche Scientifique, Marseille, France
| | - Didier Raoult
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix-Marseille University, Marseille, France
| |
Collapse
|
18
|
Kermani ES, Nazari Z, Mehdizadeh M, Shahbazi M, Golalipour MJ. Gestational diabetes influences the expression of hypertrophic genes in left ventricle of rat's offspring. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:525-528. [PMID: 29922434 PMCID: PMC6000218 DOI: 10.22038/ijbms.2018.25116.6233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Objective(s): Gestational diabetes increases the risk of congenital heart disease in the offspring, but the molecular mechanism underlying this process remains unclear. Therefore, the current study was conducted to assess the effects of induced gestational diabetes on expression of some involved genes in cardiac hypertrophy in the offspring of diabetic rats. Materials and Methods: Diabetes was induced in 40 adult Wistar rats by intraperitoneal injection of 45 mg/kg of streptozotocin. The day of appearance of the vaginal plug was assumed as day zero of gestation for inducing diabetes. After pregnancy, the offspring was maintained until they reach the age of 12 weeks. Then, their hearts were excised and were sectioned for molecular study. We analyzed the expression pattern of some hypertrophic genes by the quantitative real-time RT-PCR. Results: The mRNA expression levels of all studied genes including c-jun, c-fos, c-myc, alpha-myosin heavy chain (α-MHC), atrial natriuretic factor (ANF) and β-MHC, which are important in cardiomyocyte hypertrophy, were higher in the offspring of the diabetic group compared to controls. Significant differences were found for β-MHC and c-myc with P<0.01 and for α-MHC and c-fos with P<0.05. Conclusion: Gestational diabetes upregulates expression of c-jun, c-fos c-myc, α-MHC, ANF and β-MHC genes that are involved in cardiac hypertrophy in the offspring of diabetic rats.
Collapse
Affiliation(s)
- Elia Saragard Kermani
- Department of Anatomical sciences, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Nazari
- Gorgan Congenital Malformations Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Mehdizadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Shahbazi
- Molecular Genetic Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Jafar Golalipour
- Gorgan Congenital Malformations Research Center, Department of Anatomical Sciences, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
19
|
Donat-Vargas C, Åkesson A, Tornevi A, Wennberg M, Sommar J, Kiviranta H, Rantakokko P, Bergdahl IA. Persistent Organochlorine Pollutants in Plasma, Blood Pressure, and Hypertension in a Longitudinal Study. Hypertension 2018; 71:1258-1268. [PMID: 29712742 PMCID: PMC5959216 DOI: 10.1161/hypertensionaha.117.10691] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/22/2017] [Accepted: 03/07/2018] [Indexed: 11/16/2022]
Abstract
Persistent organochlorine pollutants (POPs) have shown to be involved in the atherosclerotic process and to cause endothelial cell dysfunction. To assess longitudinally whether plasma concentrations of different POPs were associated with blood pressure and risk of hypertension in middle-aged women and men. Study subjects were 850 participants in the VIP (Västerbotten Intervention Programme) with 2 blood samples and blood pressure measurements, 10 years apart, during 1990 to 2003 (baseline) and during 2000 to 2013 (follow-up). Dioxin-like and nondioxin-like polychlorinated biphenyls (DL-PCBs, NDL-PCBs) and p,p'-dichlorodiphenyldichloroethylene (DDE) were measured. Associations were assessed using generalized estimating equations. At baseline sampling 49% and at follow-up 64% had hypertension. DL-PCBs and DDE, but not NDL-PCBs or hexachlorobenzene, were associated with hypertension. Only the association for DL-PCBs remained statistically significant after lipid-standardization and adjustment for body mass index and total serum lipids. The multivariable-adjusted odds ratio of hypertension based on repeated measurements were 1.52 (95% confidence interval, 1.08-2.13) for DL-PCBs (third versus first tertile of lipid-standardized POPs). In stratified adjusted analyses, odds ratio for those born after 1950 increased to 3.99 (95% confidence interval, 2.15-7.43), whereas no association was observed among those born earlier. Based on repeated measurements, the accumulated exposure to DL-PCBs and DDE, although less clear for the latter, may disrupt the normal blood pressure levels and increase the odds of hypertension. Moreover, individuals experiencing early-life POP exposure may be at elevated risk of vascular POP effects.
Collapse
Affiliation(s)
- Carolina Donat-Vargas
- From the Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (C.D.-V., A.A.)
| | - Agneta Åkesson
- From the Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (C.D.-V., A.A.)
| | - Andreas Tornevi
- Department of Public Health and Clinical Medicine, Occupational and Environmental Medicine (A.T., J.S., I.A.B.)
| | | | - Johan Sommar
- Department of Public Health and Clinical Medicine, Occupational and Environmental Medicine (A.T., J.S., I.A.B.)
| | - Hannu Kiviranta
- Umeå University, Sweden; and Department for Health Security, Environmental Health Unit, National Institute for Health and Welfare, Kuopio, Finland (H.K., P.R.)
| | - Panu Rantakokko
- Umeå University, Sweden; and Department for Health Security, Environmental Health Unit, National Institute for Health and Welfare, Kuopio, Finland (H.K., P.R.)
| | - Ingvar A Bergdahl
- Department of Public Health and Clinical Medicine, Occupational and Environmental Medicine (A.T., J.S., I.A.B.)
| |
Collapse
|
20
|
Mendelson MM, Johannes R, Liu C, Huan T, Yao C, Miao X, Murabito JM, Dupuis J, Levy D, Benjamin EJ, Lin H. Epigenome-Wide Association Study of Soluble Tumor Necrosis Factor Receptor 2 Levels in the Framingham Heart Study. Front Pharmacol 2018; 9:207. [PMID: 29740313 PMCID: PMC5928448 DOI: 10.3389/fphar.2018.00207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/23/2018] [Indexed: 12/24/2022] Open
Abstract
Background: Transmembrane tumor necrosis factor (TNF) receptors are involved in inflammatory, apoptotic, and proliferative processes. In the bloodstream, soluble TNF receptor II (sTNFR2) can modify the inflammatory response of immune cells and is predictive of cardiovascular disease risk. We hypothesize that sTNFR2 is associated with epigenetic modifications of circulating leukocytes, which may relate to the pathophysiology underlying atherogenic risk. Methods: We conducted an epigenome-wide association study of sTNFR2 levels in the Framingham Heart Study Offspring cohort (examination 8; 2005-2008). sTNFR2 was quantitated by enzyme immunoassay and DNA methylation by microarray. The concentration of sTNFR2 was loge-transformed and outliers were excluded. We conducted linear mixed effects models to test the association between sTNFR2 level and methylation at over 400,000 CpGs, adjusting for age, sex, BMI, smoking, imputed cell count, technical covariates, and accounting for familial relatedness. Results: The study sample included 2468 participants (mean age: 67 ± 9 years, 52% women, mean sTNFR2 level 2661 ± 1078 pg/ml). After accounting for multiple testing, we identified 168 CpGs (P < 1.2 × 10-7) that were differentially methylated in relation to sTNFR2. A substantial proportion (27 CpGs; 16%) are in the major histocompatibility complex region and in loci overrepresented for antigen binding molecular functions (P = 1.7 × 10-4) and antigen processing and presentation biological processes (P = 1.3 × 10-8). Identified CpGs are enriched in active regulatory regions and associated with expression of 48 cis-genes (±500 kb) in whole blood (P < 1.1 × 10-5) that coincide with genes identified in GWAS of diseases of immune dysregulation (inflammatory bowel disease, type 1 diabetes, IgA nephropathy). Conclusion: Differentially methylated loci in leukocytes associated with sTNF2 levels reside in active regulatory regions, are overrepresented in antigen processes, and are linked to inflammatory diseases.
Collapse
Affiliation(s)
- Michael M Mendelson
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Department of Cardiology, Boston Children's Hospital, Boston, MA, United States.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Roby Johannes
- Hebrew SeniorLife, Harvard Medical School, Boston, MA, United States
| | - Chunyu Liu
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Tianxiao Huan
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Chen Yao
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Xiao Miao
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Joanne M Murabito
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Josée Dupuis
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Daniel Levy
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Emelia J Benjamin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Section of Cardiovascular Medicine and Preventive Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Honghuang Lin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States.,Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
21
|
Liver metabolism in adult male mice offspring: consequences of a maternal, paternal or both maternal and paternal high-fructose diet. J Dev Orig Health Dis 2018; 9:450-459. [DOI: 10.1017/s2040174418000235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractThe study aimed to evaluate the consequences of the consumption of a high-fructose diet (HFR; fructose was responsible for 45% of the energy from carbohydrates) by the mother, the father, or both on C57BL/6 adult male offspring. Non-consanguineous parents received the diet (HFR or control, C) from 8 weeks before mating until weaning (n=10 fathers and n=10 mothers on each diet). After weaning, only the C diet was offered to offspring. The groups were formed by one male randomly taken from each litter. The offspring groups were identified according to the mother’s diet (the first letter), then the father’s diet (the second letter), that is, C/C, C/HFR, HFR/C, HFR/HFR (n=10 per group). The parents exhibited the following characteristics: compared with those of the C group, the HFR parents had higher blood pressure (BP), enlarged liver, increased hepatic triacylglycerol content, hypercholesterolemia, hypertriglyceridemia, high plasma leptin and low adiponectin. The offspring exhibited the following characteristics: compared with the C/C group, the HFR/HFR group had high BP. The C/HFR, HFR/C and HFR/HFR showed elevated uric acid and leptin levels and diminished adiponectin. The HFR/HFR group showed liver inflammation (increased NFκB, SOCS3, JNK, TNF-α, IL1-β and IL6 levels). Likewise, SREBP-1c and FAS were upregulated. In conclusion, the consumption of a HFR by the mother and/or father is associated with adverse effects on liver metabolism in adult male offspring. When both mother and father are fed a HFR, the adverse effects on the offspring are more severe.
Collapse
|
22
|
Singh G, Singh V, Sobolewski M, Cory-Slechta DA, Schneider JS. Sex-Dependent Effects of Developmental Lead Exposure on the Brain. Front Genet 2018; 9:89. [PMID: 29662502 PMCID: PMC5890196 DOI: 10.3389/fgene.2018.00089] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 11/23/2022] Open
Abstract
The role of sex as an effect modifier of developmental lead (Pb) exposure has until recently received little attention. Lead exposure in early life can affect brain development with persisting influences on cognitive and behavioral functioning, as well as, elevated risks for developing a variety of diseases and disorders in later life. Although both sexes are affected by Pb exposure, the incidence, manifestation, and severity of outcomes appears to differ in males and females. Results from epidemiologic and animal studies indicate significant effect modification by sex, however, the results are not consistent across studies. Unfortunately, only a limited number of human epidemiological studies have included both sexes in independent outcome analyses limiting our ability to draw definitive conclusions regarding sex-differentiated outcomes. Additionally, due to various methodological differences across studies, there is still not a good mechanistic understanding of the molecular effects of lead on the brain and the factors that influence differential responses to Pb based on sex. In this review, focused on prenatal and postnatal Pb exposures in humans and animal models, we discuss current literature supporting sex differences in outcomes in response to Pb exposure and explore some of the ideas regarding potential molecular mechanisms that may contribute to sex-related differences in outcomes from developmental Pb exposure. The sex-dependent variability in outcomes from developmental Pb exposure may arise from a combination of complex factors, including, but not limited to, intrinsic sex-specific molecular/genetic mechanisms and external risk factors including sex-specific responses to environmental stressors which may act through shared epigenetic pathways to influence the genome and behavioral output.
Collapse
Affiliation(s)
- Garima Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Vikrant Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Jay S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
23
|
Ornellas F, Carapeto PV, Mandarim‐de‐Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
24
|
Ornellas F, Carapeto PV, Mandarim-de-Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J Pediatr (Rio J) 2017; 93:551-559. [PMID: 28822233 DOI: 10.1016/j.jped.2017.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. SOURCES A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. SUMMARY OF FINDINGS Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. CONCLUSIONS Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Priscila V Carapeto
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil.
| | - Marcia B Aguila
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
25
|
Bajpeyi S, Covington JD, Taylor EM, Stewart LK, Galgani JE, Henagan TM. Skeletal Muscle PGC1α -1 Nucleosome Position and -260 nt DNA Methylation Determine Exercise Response and Prevent Ectopic Lipid Accumulation in Men. Endocrinology 2017; 158:2190-2199. [PMID: 28398573 PMCID: PMC5505213 DOI: 10.1210/en.2017-00051] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/04/2017] [Indexed: 01/13/2023]
Abstract
Endurance exercise has been shown to improve lipid oxidation and increase mitochondrial content in skeletal muscle, two features that have shown dependence on increased expression of the peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α). It is also hypothesized that exercise-related alterations in PGC1α expression occur through epigenetic regulation of nucleosome positioning in association with differential DNA methylation status within the PGC1α promoter. In this study, we show that when primary human myotubes from obese patients with type 2 diabetes are exposed to lipolytic stimulus (palmitate, forskolin, inomycin) in vitro, nucleosome occupancy surrounding the -260 nucleotide (nt) region, a known regulatory DNA methylation site, is reduced. This finding is reproduced in vivo in the vastus lateralis from 11 healthy males after a single, long endurance exercise bout in which participants expended 650 kcal. Additionally, we show a significant positive correlation between fold change of PGC1α messenger RNA expression and -1 nucleosome repositioning away from the -260 nt methylation site in skeletal muscle tissue following exercise. Finally, we found that when exercise participants are divided into high and low responders based on the -260 nt methylation status, the -1 nucleosome is repositioned away from the regulatory -260 nt methylation site in high responders, those exhibiting a significant decrease in -260 nt methylation, but not in low responders. Additionally, high but not low responders showed a significant decrease in intramyocellular lipid content after exercise. These findings suggest a potential target for epigenetic modification of the PGC1α promoter to stimulate the therapeutic effects of endurance exercise in skeletal muscle.
Collapse
Affiliation(s)
- Sudip Bajpeyi
- Department of Kinesiology, University of Texas at El Paso, El Paso, Texas 79968
| | - Jeffrey D. Covington
- Laboratory of Skeletal Muscle Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Erin M. Taylor
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907
| | - Laura K. Stewart
- Rocky Mountain Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, Colorado 80639
| | - Jose E. Galgani
- Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Tara M. Henagan
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
26
|
Ibrahim KG, Lembede BW, Chivandi E, Erlwanger K. Erythrocyte osmotic fragility and general health status of adolescent Sprague Dawley rats supplemented with Hibiscus sabdariffa aqueous calyx extracts as neonates followed by a high-fructose diet post-weaning. J Anim Physiol Anim Nutr (Berl) 2017; 102:114-121. [PMID: 28299849 DOI: 10.1111/jpn.12665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/28/2016] [Indexed: 11/29/2022]
Abstract
High-fructose diets (HFD) can cause oxidative damage to tissues including erythrocyte cell membranes. Hibiscus sabdariffa (HS) has protective antioxidant properties. Rats were used to investigate whether the consumption of HS by neonates would result in long-term effects on their erythrocyte osmotic fragility (EOF) and general health when later fed a high-fructose diet post-weaning through adolescence. Eighty of four-day-old Sprague Dawley rat pups were divided randomly into three treatment groups. The controls (n = 27) received distilled water at 10 ml/kg b. w, while the other groups received either 50 mg/kg (n = 28) or 500 mg/kg (n = 25) of an HS aqueous calyx extract orally till post-natal day 14. The rats in each group were weaned and divided into two subgroups; one continued on normal rat chow, and the other received fructose (20% w/v) in their drinking water for 30 days. Blood was collected in heparinised tubes and added to serially diluted (0.0-0.85%) phosphate-buffered saline to determine the EOF. Clinical markers of health status were determined with an automated chemical analyser. HS extracts did not programme metabolism in the growing rats to alter their general health and EOF in response to the HFD.
Collapse
Affiliation(s)
- K G Ibrahim
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg, South Africa.,Department of Physiology, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - B W Lembede
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg, South Africa
| | - E Chivandi
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg, South Africa
| | - K Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg, South Africa
| |
Collapse
|
27
|
Sales VM, Ferguson-Smith AC, Patti ME. Epigenetic Mechanisms of Transmission of Metabolic Disease across Generations. Cell Metab 2017; 25:559-571. [PMID: 28273478 PMCID: PMC5404272 DOI: 10.1016/j.cmet.2017.02.016] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Both human and animal studies indicate that environmental exposures experienced during early life can robustly influence risk for adult disease. Moreover, environmental exposures experienced by parents during either intrauterine or postnatal life can also influence the health of their offspring, thus initiating a cycle of disease risk across generations. In this Perspective, we focus on epigenetic mechanisms in germ cells, including DNA methylation, histone modification, and non-coding RNAs, which collectively may provide a non-genetic molecular legacy of prior environmental exposures and influence transcriptional regulation, developmental trajectories, and adult disease risk in offspring.
Collapse
Affiliation(s)
- Vicencia Micheline Sales
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center and Harvard Medical School, One Joslin Place, Sixth Floor, Boston, MA 02215, USA
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center and Harvard Medical School, One Joslin Place, Sixth Floor, Boston, MA 02215, USA.
| |
Collapse
|
28
|
Schenkel LC, Rodenhiser D, Siu V, McCready E, Ainsworth P, Sadikovic B. Constitutional Epi/Genetic Conditions: Genetic, Epigenetic, and Environmental Factors. J Pediatr Genet 2017; 6:30-41. [PMID: 28180025 PMCID: PMC5288004 DOI: 10.1055/s-0036-1593849] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 04/14/2016] [Indexed: 12/12/2022]
Abstract
There are more than 4,000 phenotypes for which the molecular basis is at least partly known. Though defects in primary DNA structure constitute a major cause of these disorders, epigenetic disruption is emerging as an important alternative mechanism in the etiology of a broad range of congenital and developmental conditions. These include epigenetic defects caused by either localized (in cis) genetic alterations or more distant (in trans) genetic events but can also include environmental effects. Emerging evidence suggests interplay between genetic and environmental factors in the epigenetic etiology of several constitutional "epi/genetic" conditions. This review summarizes our broadening understanding of how epigenetics contributes to pediatric disease by exploring different classes of epigenomic disorders. It further challenges the simplistic dogma of "DNA encodes RNA encodes protein" to best understand the spectrum of factors that can influence genetic traits in a pediatric population.
Collapse
Affiliation(s)
- Laila C. Schenkel
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
| | - David Rodenhiser
- Children's Health Research Institute, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
- Department of Pediatrics, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
| | - Victoria Siu
- Children's Health Research Institute, London, Ontario, Canada
- Department of Pediatrics, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peter Ainsworth
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
- Department of Pediatrics, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| |
Collapse
|
29
|
Methylome-wide Association Study of Atrial Fibrillation in Framingham Heart Study. Sci Rep 2017; 7:40377. [PMID: 28067321 PMCID: PMC5220313 DOI: 10.1038/srep40377] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 12/05/2016] [Indexed: 12/18/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, but little is known about the molecular mechanisms associated with AF arrhythmogenesis. DNA methylation is an important epigenetic mechanism that regulates gene expression and downstream biological processes. We hypothesize that DNA methylation might play an important role in the susceptibility to develop AF. A total of 2,639 participants from the Offspring Cohort of Framingham Heart Study were enrolled in the current study. These participants included 183 participants with prevalent AF and 220 with incident AF during up to 9 years follow up. Genome-wide methylation was profiled using the Illumina Infinium HumanMethylation450 BeadChip on blood-derived DNA collected during the eighth examination cycle (2005-2008). Two CpG sites were significantly associated with prevalent AF, and five CpGs were associated with incident AF after correction for multiple testing (FDR < 0.05). Fourteen previously reported genome-wide significant AF-related SNP were each associated with at least one CpG site; the most significant association was rs6490029 at the CUX2 locus and cg10833066 (P = 9.5 × 10-279). In summary, we performed genome-wide methylation profiling in a community-based cohort and identified seven methylation signatures associated with AF. Our study suggests that DNA methylation might play an important role in AF arrhythmogenesis.
Collapse
|
30
|
Lucas ES, Watkins AJ. The Long-Term Effects of the Periconceptional Period on Embryo Epigenetic Profile and Phenotype; The Paternal Role and His Contribution, and How Males Can Affect Offspring's Phenotype/Epigenetic Profile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1014:137-154. [PMID: 28864989 DOI: 10.1007/978-3-319-62414-3_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The number of adults afflicted with heart disease, obesity and diabetes, central components of metabolic disorder, has grown rapidly in recent decades, affecting up to one quarter of the world's population. Typically, these diseases are attributed to lifestyle factors such as poor diet, lack of exercise and smoking. However, studies have now identified strong associations between patterns of growth during foetal and neonatal life and an increase predisposition towards developing heart disease, obesity and diabetes in adult life. While the connection between a mother's diet and the long-term health of her offspring has been studied in great detail, our understanding of whether offspring health might be affected by a father's diet remains limited. Greater insight into the impact that paternal nutrition has on sperm quality, epigenetic status and potential offspring programming mechanisms is needed to redress this parental-programming knowledge imbalance. Disturbances in paternal reproductive epigenetic status represents one key mechanism linking paternal diet with the programing of offspring development and adult health, as many enzymatic processes involved in epigenetic regulation use metabolic intermediates to modify DNA and histones. Here, poor paternal nutrition could result in perturbed sperm and testicular epigenetic status, impacting on post-fertilisation gene transcriptional regulation and protein expression in offspring tissues, resulting in increased incidences of metabolic disorder in adult life.
Collapse
Affiliation(s)
- Emma S Lucas
- Division of Reproductive Health, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| | - Adam J Watkins
- Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
31
|
Xu W, Wang F, Yu Z, Xin F. Epigenetics and Cellular Metabolism. GENETICS & EPIGENETICS 2016; 8:43-51. [PMID: 27695375 PMCID: PMC5038610 DOI: 10.4137/geg.s32160] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 01/03/2023]
Abstract
Living eukaryotic systems evolve delicate cellular mechanisms for responding to various environmental signals. Among them, epigenetic machinery (DNA methylation, histone modifications, microRNAs, etc.) is the hub in transducing external stimuli into transcriptional response. Emerging evidence reveals the concept that epigenetic signatures are essential for the proper maintenance of cellular metabolism. On the other hand, the metabolite, a main environmental input, can also influence the processing of epigenetic memory. Here, we summarize the recent research progress in the epigenetic regulation of cellular metabolism and discuss how the dysfunction of epigenetic machineries influences the development of metabolic disorders such as diabetes and obesity; then, we focus on discussing the notion that manipulating metabolites, the fuel of cell metabolism, can function as a strategy for interfering epigenetic machinery and its related disease progression as well.
Collapse
Affiliation(s)
- Wenyi Xu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Zhongsheng Yu
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Fengjiao Xin
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| |
Collapse
|
32
|
Measurement of Histone Methylation Dynamics by One-Carbon Metabolic Isotope Labeling and High-energy Collisional Dissociation Methylation Signature Ion Detection. Sci Rep 2016; 6:31537. [PMID: 27530234 PMCID: PMC4987619 DOI: 10.1038/srep31537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that cellular metabolites and nutrition levels control epigenetic modifications, including histone methylation. However, it is not currently possible to measure the metabolic control of histone methylation. Here we report a novel detection method to monitor methyl transfer from serine to histones through the one-carbon metabolic pathway, using stable-isotope labeling and detection of lysine methylation signature ions generated in high-energy-dissociation (HCD) tandem mass spectrometry. This method is a long-needed tool to study the metabolic control of histone methylation.
Collapse
|
33
|
Gaetani S, Cassano T. From Obesity Resistance to Obesity Prediction and Prevention? Front Neurosci 2016; 10:369. [PMID: 27554849 PMCID: PMC4977311 DOI: 10.3389/fnins.2016.00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/27/2016] [Indexed: 11/17/2022] Open
Affiliation(s)
- Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of RomeRome, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
- *Correspondence: Tommaso Cassano
| |
Collapse
|
34
|
Carraro JCC, Mansego ML, Milagro FI, Chaves LO, Vidigal FC, Bressan J, Martínez JA. LINE-1 and inflammatory gene methylation levels are early biomarkers of metabolic changes: association with adiposity. Biomarkers 2016; 21:625-32. [DOI: 10.3109/1354750x.2016.1171904] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Maria Luisa Mansego
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, CIBERobn, Fisiopatología De La Obesidad Y La Nutrición, Institute of Health Carlos III, Pamplona, Madrid, Spain
| | - Fermin Ignacio Milagro
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, CIBERobn, Fisiopatología De La Obesidad Y La Nutrición, Institute of Health Carlos III, Pamplona, Madrid, Spain
| | - Larissa Oliveira Chaves
- Department of Nutrition and Health, Universidade Federal De Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal De Viçosa, Viçosa, Minas Gerais, Brazil
| | - J. Alfredo Martínez
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, CIBERobn, Fisiopatología De La Obesidad Y La Nutrición, Institute of Health Carlos III, Pamplona, Madrid, Spain
| |
Collapse
|
35
|
Bala C, Craciun AE, Hancu N. UPDATING THE CONCEPT OF METABOLICALLY HEALTHY OBESITY. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2016; 12:197-205. [PMID: 31149087 PMCID: PMC6535297 DOI: 10.4183/aeb.2016.197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity is a well-recognized risk factor for type 2 diabetes, cardiovascular disease, and several types of cancer. However, a proportion of the obese individuals display a significantly lower risk for metabolic complications than expected for their degree of body mass index, and this subtype of obesity was described as "metabolically healthy obesity" (MHO). No universally accepted criteria for the diagnosis of MHO exists and the prevalence of this subtype of obesity varies largely according to criteria used. Broadly, MHO is characterized by a lower amount of visceral fat, a more favorable inflammatory profile, and less insulin resistance as compared to the metabolically unhealthy obesity. Currently, controversies exist regarding the risk of cardiovascular events and all-cause mortality associated with MHO as compared to metabolically-healthy non-obese individuals. Further research is needed in order to identify the MHO phenotype and if MHO is truly healthy for a long period of time or if it is a transient state from normal metabolic/normal weight to abnormal metabolic/obese state. This review will discuss the MHO definition criteria; the differences between MHO and metabolically unhealthy obesity; the possible underlying mechanisms and clinical implications of MHO.
Collapse
Affiliation(s)
| | - A-E. Craciun
- “Iuliu Haţieganu” University of Medicine and Pharmacy, Dept. of Diabetes, Nutrition and Metabolic Diseases, Cluj-Napoca, Romania
| | | |
Collapse
|
36
|
Costa TBD, Morais NGD, Pedrosa ALF, De Albuquerque SDCG, De Castro MCAB, Pereira VRA, Cavalcanti MDP, De Castro CMMB. Neonatal malnutrition programs the oxidant function of macrophages in response to Candida albicans. Microb Pathog 2016; 95:68-76. [PMID: 27001703 DOI: 10.1016/j.micpath.2016.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 02/17/2016] [Accepted: 02/21/2016] [Indexed: 10/22/2022]
Abstract
Experimental maternal nutrition restriction models are used to investigate short or long-term consequences of nutritional deficiency on puppies' growth. By assuming that the immune function is directly related to host's nutritional status, the current study aims to investigate the effects of neonatal malnutrition on oxidative stress and on the cell death of the alveolar macrophage after in vitro infection by Candida albicans. Wistar rats were suckled by mothers fed on diets containing 17% protein (Nourished group) or 8% protein (Malnourished group) in the current assay. Both groups received the standard diet used in the vivarium until adulthood, after weaning. The results showed that the offspring from mothers fed on low-protein diet presented lower body weight from 5 days of life on. Their low weight remained until adulthood when it was compared to that of rats in the nourished group. Superoxide and nitric oxide production was lower in malnourished animals and it was accompanied by low inducible nitric oxide synthase gene expression levels in systems in which the alveolar macrophages were challenged by immunogenic stimulus. No significant differences were observed in comparisons performed between the nourished and malnourished groups in any of the analyzed cell viability (apoptosis/necrosis) parameters. The fungal inoculum-stimulated system induced higher oxidative stress and cell death by necrosis. The current study demonstrated that dietary restriction during lactation alters the oxidant function of alveolar macrophages in puppies; It happens from the gene transcription step to the release of mediators, thus compromising the host's defenses against Candida albicans. It raises the possibility that Candida albicans may cease to be a commensal fungus to become a pathogen in offspring that have suffered nutritional deficiency during critical developmental periods, due to impaired immune responses.
Collapse
Affiliation(s)
- Thacianna Barreto Da Costa
- Department of Tropical Medicine, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil; Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil.
| | - Natália Gomes De Morais
- College of Medicine, Federal University of São Francisco Valley, Avenida José de Sá Maniçoba - Centro, Petrolina, Pernambuco, 56304-917, Brazil
| | - Amanda Lúcia F Pedrosa
- Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Suênia Da Cunha G De Albuquerque
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Maria Carolina A B De Castro
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Valéria Rêgo A Pereira
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Milena De Paiva Cavalcanti
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Célia Maria M B De Castro
- Department of Tropical Medicine, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil; Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| |
Collapse
|
37
|
Lopez-Jaramillo P. The Role of Adiponectin in Cardiometabolic Diseases: Effects of Nutritional Interventions. J Nutr 2016; 146:422S-426S. [PMID: 26764331 DOI: 10.3945/jn.114.202432] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 06/01/2015] [Indexed: 12/15/2022] Open
Abstract
Adiponectin is an adipocyte-derived hormone abundantly present in plasma that exerts its effects through the activation of 3 receptors. Its concentrations are negatively regulated by the accumulation of visceral fat, and clinical studies implicate hypoadiponectinemia in the pathogenesis of diabetes mellitus type 2, coronary artery disease, hypertension, and left ventricular hypertrophy. In contrast, high concentrations of adiponectin are associated with a decreased risk of coronary artery disease, with an improvement in the differentiation of preadipocytes into adipocytes, and with increased endothelial nitric oxide production. Therefore, adiponectin appears to be an important molecule involved in limiting the pathogenesis of obesity-linked disorders, and it may have potential benefits in the treatment and prevention of cardiovascular disease. Caloric restriction, moderate alcohol consumption, and consuming a Mediterranean diet increase adiponectin concentrations, and current evidence suggests a positive, dose-dependent relation between ω-3 (n-3) fatty acid intake and circulating concentrations of adiponectin. Recently, it was reported that the administration of aged garlic extract and a single food intervention with pistachios can increase adiponectin concentrations in individuals with metabolic syndrome. Moreover, the Mediterranean diet is associated with higher adiponectin concentrations. Additional studies are needed to evaluate the potential benefits of increasing adiponectin by nutritional interventions in the treatment and prevention of cardiometabolic diseases.
Collapse
Affiliation(s)
- Patricio Lopez-Jaramillo
- Metabolic Syndrome, Prediabetes, and Diabetes Clinic, Research Institute, Santander Ophthalmological Foundation Carlos Ardila Lulle Clinic (FOSCAL), and Masira Institute, Medical School, University of Santander, Bucaramanga, Colombia
| |
Collapse
|
38
|
Da Costa TB, De Morais NG, De Lira JMB, De Almeida TM, Gonçalves-De-Albuquerque SDC, Pereira VRA, De Paiva Cavalcanti M, De Castro CMMB. Relation between neonatal malnutrition and gene expression: inflammasome function in infections caused by Candida Albicans. Eur J Nutr 2015; 56:693-704. [PMID: 26658898 DOI: 10.1007/s00394-015-1113-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 11/22/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE To investigate the effects of neonatal malnutrition followed by nutritional replacement on the signaling mechanisms developed by the inflammasome complex by analyzing the expression of the targeted TLR2, TLR4, NLRP3, caspase-1 and release of IL-1β and IL-18 by alveolar macrophages infected in vitro with Candida albicans. METHODS Male Wistar rats (n = 24), 90-120 days, were suckled by mothers whose diet during lactation contained 17 % protein in the nourish group and 8 % protein in the malnourished group. After weaning, both groups were fed a normal protein diet. Macrophages were obtained after tracheostomy, through the collection of bronchoalveolar lavage fluid. The quantification of the expression levels of targets (TLR2, TLR4, NLRP3 and caspase-1) was performed by real-time RT-PCR. Production of cytokines was performed by ELISA. RESULTS The malnourished animals during lactation showed reduced body weight from the fifth day of life, remaining until adulthood. Further, the model applied malnutrition induced a lower expression of TLR4 and caspase-1. The quantification of the TLR2 and NLRP3, as well as the release of IL-1β and IL-18, was not different between groups of animals nourished and malnourished. The system challenged with Candida albicans showed high expression levels of all targets in the study. CONCLUSIONS The tests demonstrate nutritional restriction during critical periods of development, although nutritional supplementation may compromise defense patterns in adulthood in a timely manner, preserving distinct signaling mechanism, so that the individual does not become widely vulnerable to infections by opportunistic pathogens.
Collapse
Affiliation(s)
- Thacianna Barreto Da Costa
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil. .,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil.
| | - Natália Gomes De Morais
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil.,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Joana Maria Bezerra De Lira
- Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| | - Thays Miranda De Almeida
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Campus Federal University of Pernambuco, Recife, Brazil
| | | | - Valéria Rêgo Alves Pereira
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Campus Federal University of Pernambuco, Recife, Brazil
| | - Milena De Paiva Cavalcanti
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Campus Federal University of Pernambuco, Recife, Brazil
| | - Célia Maria Machado Barbosa De Castro
- Department of Tropical Medicine, Federal University of Pernambuco, Recife, Brazil.,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco, 50670-901, Brazil
| |
Collapse
|
39
|
Milagro FI, Riezu-Boj JI, Martinez JA. Epigenetic Determinants of Weight Management: Methylation Signatures. Curr Nutr Rep 2015. [DOI: 10.1007/s13668-015-0140-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Padzys GS, Ondo JP, Omouenze LP, Zongo S. Diabetes in Sub-Saharan Africa: Distribution Based on Social Status in Libreville (Gabon). Ethn Dis 2015; 25:459-62. [PMID: 26676185 PMCID: PMC4671434 DOI: 10.18865/ed.25.4.459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Many researchers continue to believe that urbanization is a major contributor to diabetes. We seek to demonstrate that the social status associated with urbanization has an impact on the prevalence of diabetes in Libreville, Gabon in sub-Saharan Africa. METHODS Our study was conducted in Libreville, the capital of Gabon; the city has a population of 397,000. Our study analyzed data from the registries of patients hospitalized in 2013 in the main diabetes center in Libreville. RESULTS The results revealed that, for 2013, 798 patients were hospitalized with diabetes at a prevalence of .2%. We found differences (P<.05) between women (423) and men (375). Mean age for women was 52.02 years and 48.88 years for men. The number of existing cases hospitalized was significantly more than new cases. All levels of society were represented in our study: students (42); military (36); administratives (99); technicians (180); unemployed (295); and retired (146). The results showed that the unemployed (36%), particularly women (29.40%) are most affected by diabetes. CONCLUSIONS Our results show the impact of social status on the increase of diabetes in Libreville. We found that urbanization, associated with insecurity especially in women, had an effect on the prevalence of diabetes in Libreville. These results indicate that, apart from the non-modifiable factors (age, race, ethnicity), insecurity is a modifiable factor that should be taken into account.
Collapse
Affiliation(s)
- Guy S. Padzys
- 1. Université des Sciences et Technique de Masuku BP; 943 Franceville, Gabon
| | - Joseph P. Ondo
- 1. Université des Sciences et Technique de Masuku BP; 943 Franceville, Gabon
| | - Linda P. Omouenze
- 1. Université des Sciences et Technique de Masuku BP; 943 Franceville, Gabon
| | - Sylvie Zongo
- 1. Université des Sciences et Technique de Masuku BP; 943 Franceville, Gabon
| |
Collapse
|
41
|
Mabasa L, Cho K, Choi WS, Crane CL, Cho K, Singh RK, Park CS. Suppression of Mammary Carcinogenesis Through Early Exposure to Dietary Lipotropes Occurs Primarily In Utero. Nutr Cancer 2015; 67:1276-82. [DOI: 10.1080/01635581.2015.1087039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
42
|
Zhang N. Epigenetic modulation of DNA methylation by nutrition and its mechanisms in animals. ACTA ACUST UNITED AC 2015; 1:144-151. [PMID: 29767106 PMCID: PMC5945948 DOI: 10.1016/j.aninu.2015.09.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/01/2015] [Indexed: 12/21/2022]
Abstract
It is well known that phenotype of animals may be modified by the nutritional modulations through epigenetic mechanisms. As a key and central component of epigenetic network, DNA methylation is labile in response to nutritional influences. Alterations in DNA methylation profiles can lead to changes in gene expression, resulting in diverse phenotypes with the potential for decreased growth and health. Here, I reviewed the biological process of DNA methylation that results in the addition of methyl groups to DNA; the possible ways including methyl donors, DNA methyltransferase (DNMT) activity and other cofactors, the critical periods including prenatal, postnatal and dietary transition periods, and tissue specific of epigenetic modulation of DNA methylation by nutrition and its mechanisms in animals.
Collapse
Affiliation(s)
- Naifeng Zhang
- Feed Research Institute of Chinese Academy of Agricultural Sciences, Key Laboratory of Feed Biotechnology of the Ministry of Agriculture, Beijing 100081, China
| |
Collapse
|
43
|
VanderJagt TA, Neugebauer MH, Morgan M, Bowden DW, Shah VO. Epigenetic profiles of pre-diabetes transitioning to type 2 diabetes and nephropathy. World J Diabetes 2015; 6:1113-1121. [PMID: 26265998 PMCID: PMC4530325 DOI: 10.4239/wjd.v6.i9.1113] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/04/2015] [Accepted: 07/14/2015] [Indexed: 02/05/2023] Open
Abstract
AIM: To examine DNA methylation profiles in a longitudinal comparison of pre-diabetes mellitus (Pre-DM) subjects who transitioned to type 2 diabetes mellitus (T2DM).
METHODS: We performed DNA methylation study in bisulphite converted DNA from Pre-DM (n = 11) at baseline and at their transition to T2DM using Illumina Infinium HumanMethylation27 BeadChip, that enables the query of 27578 individual cytosines at CpG loci throughout the genome, which are focused on the promoter regions of 14495 genes.
RESULTS: There were 694 CpG sites hypomethylated and 174 CpG sites hypermethylated in progression from Pre-DM to T2DM, representing putative genes involved in glucose and fructose metabolism, inflammation, oxidative and mitochondrial stress, and fatty acid metabolism. These results suggest that this high throughput platform is able to identify hundreds of prospective CpG sites associated with diverse genes that may reflect differences in Pre-DM compared with T2DM. In addition, there were CpG hypomethylation changes associated with a number of genes that may be associated with development of complications of diabetes, such as nephropathy. These hypomethylation changes were observed in all of the subjects.
CONCLUSION: These data suggest that some epigenomic changes that may be involved in the progression of diabetes and/or the development of complications may be apparent at the Pre-DM state or during the transition to diabetes. Hypomethylation of a number of genes related to kidney function may be an early marker for developing diabetic nephropathy.
Collapse
|
44
|
Szarc vel Szic K, Declerck K, Vidaković M, Vanden Berghe W. From inflammaging to healthy aging by dietary lifestyle choices: is epigenetics the key to personalized nutrition? Clin Epigenetics 2015; 7:33. [PMID: 25861393 PMCID: PMC4389409 DOI: 10.1186/s13148-015-0068-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/09/2015] [Indexed: 01/12/2023] Open
Abstract
The progressively older population in developed countries is reflected in an increase in the number of people suffering from age-related chronic inflammatory diseases such as metabolic syndrome, diabetes, heart and lung diseases, cancer, osteoporosis, arthritis, and dementia. The heterogeneity in biological aging, chronological age, and aging-associated disorders in humans have been ascribed to different genetic and environmental factors (i.e., diet, pollution, stress) that are closely linked to socioeconomic factors. The common denominator of these factors is the inflammatory response. Chronic low-grade systemic inflammation during physiological aging and immunosenescence are intertwined in the pathogenesis of premature aging also defined as ‘inflammaging.’ The latter has been associated with frailty, morbidity, and mortality in elderly subjects. However, it is unknown to what extent inflammaging or longevity is controlled by epigenetic events in early life. Today, human diet is believed to have a major influence on both the development and prevention of age-related diseases. Most plant-derived dietary phytochemicals and macro- and micronutrients modulate oxidative stress and inflammatory signaling and regulate metabolic pathways and bioenergetics that can be translated into stable epigenetic patterns of gene expression. Therefore, diet interventions designed for healthy aging have become a hot topic in nutritional epigenomic research. Increasing evidence has revealed that complex interactions between food components and histone modifications, DNA methylation, non-coding RNA expression, and chromatin remodeling factors influence the inflammaging phenotype and as such may protect or predispose an individual to many age-related diseases. Remarkably, humans present a broad range of responses to similar dietary challenges due to both genetic and epigenetic modulations of the expression of target proteins and key genes involved in the metabolism and distribution of the dietary constituents. Here, we will summarize the epigenetic actions of dietary components, including phytochemicals, and macro- and micronutrients as well as metabolites, that can attenuate inflammaging. We will discuss the challenges facing personalized nutrition to translate highly variable interindividual epigenetic diet responses to potential individual health benefits/risks related to aging disease.
Collapse
Affiliation(s)
- Katarzyna Szarc vel Szic
- Lab Protein Science, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Ken Declerck
- Lab Protein Science, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Melita Vidaković
- Department of Molecular Biology, Institute for Biological Research, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Wim Vanden Berghe
- Lab Protein Science, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
45
|
Lopez-Jaramillo P, Gomez-Arbelaez D, Sotomayor-Rubio A, Mantilla-Garcia D, Lopez-Lopez J. Maternal undernutrition and cardiometabolic disease: a Latin American perspective. BMC Med 2015; 13:41. [PMID: 25858591 PMCID: PMC4346113 DOI: 10.1186/s12916-015-0293-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 02/12/2015] [Indexed: 02/07/2023] Open
Abstract
The current epidemic of obesity and cardiometabolic diseases in developing countries is described as being driven by socioeconomic inequalities. These populations have a greater vulnerability to cardiometabolic diseases due to the discrepancy between the maternal undernutrition and its consequence, low-birth weight progeny, and the subsequent modern lifestyles which are associated with socioeconomic and environmental changes that modify dietary habits, discourage physical activity and encourage sedentary behaviors. Maternal undernutrition can generate epigenetic modifications, with potential long-term consequences. Throughout life, people are faced with the challenge of adapting to changes in their environment, such as excessive intake of high energy density foods and sedentary behavior. However, a mismatch between conditions experienced during fetal programming and current environmental conditions will make adaptation difficult for them, and will increase their susceptibility to obesity and cardiovascular diseases. It is important to conduct research in the Latin American context, in order to define the best strategies to prevent the epidemic of cardiometabolic diseases in the region.
Collapse
|
46
|
Henagan TM, Stefanska B, Fang Z, Navard AM, Ye J, Lenard NR, Devarshi PP. Sodium butyrate epigenetically modulates high-fat diet-induced skeletal muscle mitochondrial adaptation, obesity and insulin resistance through nucleosome positioning. Br J Pharmacol 2015; 172:2782-98. [PMID: 25559882 DOI: 10.1111/bph.13058] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 11/24/2014] [Accepted: 12/15/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Sodium butyrate (NaB), an epigenetic modifier, is effective in promoting insulin sensitivity. The specific genomic loci and mechanisms underlying epigenetically induced obesity and insulin resistance and the targets of NaB are not fully understood. EXPERIMENTAL APPROACH The anti-diabetic and anti-obesity effects of NaB treatment were measured by comparing phenotypes and physiologies of C57BL/6J mice fed a low-fat diet (LF), high-fat diet (HF) or high-fat diet plus NaB (HF + NaB) for 10 weeks. We determined a possible mechanism of NaB action through induction of beneficial skeletal muscle mitochondrial adaptations and applied microccocal nuclease digestion with sequencing (MNase-seq) to assess whole genome differences in nucleosome occupancy or positioning and to identify associated epigenetic targets of NaB. KEY RESULTS NaB prevented HF diet-induced increases in body weight and adiposity without altering food intake or energy expenditure, improved insulin sensitivity as measured by glucose and insulin tolerance tests, and decreased respiratory exchange ratio. In skeletal muscle, NaB increased the percentage of type 1 fibres, improved acylcarnitine profiles as measured by metabolomics and produced a chromatin structure, determined by MNase-seq, similar to that seen in LF. Targeted analysis of representative nuclear-encoded mitochondrial genes showed specific repositioning of the -1 nucleosome in association with altered gene expression. CONCLUSIONS AND IMPLICATIONS NaB treatment may be an effective pharmacological approach for type 2 diabetes and obesity by inducing -1 nucleosome repositioning within nuclear-encoded mitochondrial genes, causing skeletal muscle mitochondrial adaptations that result in more complete β-oxidation and a lean, insulin sensitive phenotype.
Collapse
Affiliation(s)
- Tara M Henagan
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Barbara Stefanska
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Zhide Fang
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Alexandra M Navard
- Neurosignaling Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Natalie R Lenard
- Department of Sciences, Our Lady of the Lake College, Baton Rouge, LA, USA
| | - Prasad P Devarshi
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
47
|
Slyvka Y, Zhang Y, Nowak FV. Epigenetic effects of paternal diet on offspring: emphasis on obesity. Endocrine 2015; 48:36-46. [PMID: 24997644 DOI: 10.1007/s12020-014-0328-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/05/2014] [Indexed: 12/21/2022]
Abstract
Overnutrition, obesity, and the rise in associated comorbidities are widely recognized as preventable challenges to global health. Behavioral, metabolic, and epigenetic influences that alter the epigenome, when passed on to offspring, can increase their risk of developing an altered metabolic profile. This review is focused on the role of paternal inheritance as demonstrated by clinical, epidemiological, and experimental models. Development of additional experimental models that resemble the specific epigenetic sensitive situations in human studies will be essential to explore paternally induced trans-generational effects that are mediated, primarily, by epigenetic effects. Further elucidation of epigenetic marks will help identify preventive and therapeutic targets, which in combination with healthy lifestyle choices, can diminish the growing tide of obesity, type 2 diabetes, and other related disorders.
Collapse
Affiliation(s)
- Yuriy Slyvka
- Department of Biomedical Sciences, HCOM, Ohio University, Athens, OH, 45701, USA
| | | | | |
Collapse
|
48
|
Khaire A, Rathod R, Kemse N, Kale A, Joshi S. Supplementation with omega-3 fatty acids during gestation and lactation to a vitamin B12-deficient or -supplemented diet improves pregnancy outcome and metabolic variables in Wistar rats. Reprod Fertil Dev 2015; 27:341-50. [DOI: 10.1071/rd13306] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/17/2013] [Indexed: 11/23/2022] Open
Abstract
Maternal vitamin B12 deficiency leads to an adverse pregnancy outcome and increases the risk for developing diabetes and metabolic syndrome in mothers in later life. Our earlier studies have demonstrated that vitamin B12 and n-3 polyunsaturated fatty acids (PUFA) are interlinked in the one carbon cycle. The present study for the first time examines the effect of maternal n-3 PUFA supplementation to vitamin B12 deficient or supplemented diets on pregnancy outcome, fatty-acid status and metabolic variables in Wistar rats. Pregnant dams were assigned to one of the following groups: control, vitamin B12 deficient, vitamin B12 supplemented, vitamin B12 deficient + n-3 PUFA or vitamin B12 supplemented + n-3 PUFA. The amount of vitamin B12 in the supplemented group was 0.50 μg kg–1 diet and n-3 PUFA was alpha linolenic acid (ALA) 1.68, eicosapentaenoic acid 5.64, docosahexaenoic acid (DHA) 3.15 (g per 100 g fatty acids per kg diet). Our findings indicate that maternal vitamin B12 supplementation did not affect the weight gain of dams during pregnancy but reduced litter size and weight and was ameliorated by n-3 PUFA supplementation. Vitamin B12 deficiency or supplementation resulted in a low percentage distribution of plasma arachidonic acid and DHA. n-3 PUFA supplementation to these diets improved the fatty-acid status. Vitamin B12 deficiency resulted in higher homocysteine and insulin levels, which were normalised by supplementation with either vitamin B12 or n-3 PUFA. Our study suggests that maternal vitamin B12 status is critical in determining pregnancy outcome and metabolic variables in dams and that supplementation with n-3 PUFA is beneficial.
Collapse
|
49
|
PGC1α -1 Nucleosome Position and Splice Variant Expression and Cardiovascular Disease Risk in Overweight and Obese Individuals. PPAR Res 2014; 2014:895734. [PMID: 25614734 PMCID: PMC4295622 DOI: 10.1155/2014/895734] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/26/2014] [Indexed: 11/17/2022] Open
Abstract
PGC1α, a transcriptional coactivator, interacts with PPARs and others to regulate skeletal muscle metabolism. PGC1α undergoes splicing to produce several mRNA variants, with the NTPGC1α variant having a similar biological function to the full length PGC1α (FLPGC1α). CVD is associated with obesity and T2D and a lower percentage of type 1 oxidative fibers and impaired mitochondrial function in skeletal muscle, characteristics determined by PGC1α expression. PGC1α expression is epigenetically regulated in skeletal muscle to determine mitochondrial adaptations, and epigenetic modifications may regulate mRNA splicing. We report in this paper that skeletal muscle PGC1α -1 nucleosome (-1N) position is associated with splice variant NTPGC1α but not FLPGC1α expression. Division of participants based on the -1N position revealed that those individuals with a -1N phased further upstream from the transcriptional start site (UP) expressed lower levels of NTPGC1α than those with the -1N more proximal to TSS (DN). UP showed an increase in body fat percentage and serum total and LDL cholesterol. These findings suggest that the -1N may be a potential epigenetic regulator of NTPGC1α splice variant expression, and -1N position and NTPGC1α variant expression in skeletal muscle are linked to CVD risk. This trial is registered with clinicaltrials.gov, identifier NCT00458133.
Collapse
|
50
|
Vaněčková I, Maletínská L, Behuliak M, Nagelová V, Zicha J, Kuneš J. Obesity-related hypertension: possible pathophysiological mechanisms. J Endocrinol 2014; 223:R63-78. [PMID: 25385879 DOI: 10.1530/joe-14-0368] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hypertension is one of the major risk factors of cardiovascular diseases, but despite a century of clinical and basic research, the discrete etiology of this disease is still not fully understood. The same is true for obesity, which is recognized as a major global epidemic health problem nowadays. Obesity is associated with an increasing prevalence of the metabolic syndrome, a cluster of risk factors including hypertension, abdominal obesity, dyslipidemia, and hyperglycemia. Epidemiological studies have shown that excess weight gain predicts future development of hypertension, and the relationship between BMI and blood pressure (BP) appears to be almost linear in different populations. There is no doubt that obesity-related hypertension is a multifactorial and polygenic trait, and multiple potential pathogenetic mechanisms probably contribute to the development of higher BP in obese humans. These include hyperinsulinemia, activation of the renin-angiotensin-aldosterone system, sympathetic nervous system stimulation, abnormal levels of certain adipokines such as leptin, or cytokines acting at the vascular endothelial level. Moreover, some genetic and epigenetic mechanisms are also in play. Although the full manifestation of both hypertension and obesity occurs predominantly in adulthood, their roots can be traced back to early ontogeny. The detailed knowledge of alterations occurring in the organism of experimental animals during particular critical periods (developmental windows) could help to solve this phenomenon in humans and might facilitate the age-specific prevention of human obesity-related hypertension. In addition, better understanding of particular pathophysiological mechanisms might be useful in so-called personalized medicine.
Collapse
Affiliation(s)
- Ivana Vaněčková
- Institute of PhysiologyAcademy of Sciences of the Czech Republic v.v.i., Videnska 1083, 14220 Prague 4, Czech RepublicInstitute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic
| | - Lenka Maletínská
- Institute of PhysiologyAcademy of Sciences of the Czech Republic v.v.i., Videnska 1083, 14220 Prague 4, Czech RepublicInstitute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic
| | - Michal Behuliak
- Institute of PhysiologyAcademy of Sciences of the Czech Republic v.v.i., Videnska 1083, 14220 Prague 4, Czech RepublicInstitute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic
| | - Veronika Nagelová
- Institute of PhysiologyAcademy of Sciences of the Czech Republic v.v.i., Videnska 1083, 14220 Prague 4, Czech RepublicInstitute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic
| | - Josef Zicha
- Institute of PhysiologyAcademy of Sciences of the Czech Republic v.v.i., Videnska 1083, 14220 Prague 4, Czech RepublicInstitute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of PhysiologyAcademy of Sciences of the Czech Republic v.v.i., Videnska 1083, 14220 Prague 4, Czech RepublicInstitute of Organic Chemistry and BiochemistryAcademy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic
| |
Collapse
|