1
|
Han W, Song Z, Shan D, Shi Q. Fetal origins of obesity: a novel pathway of regulating appetite neurons in the hypothalamus of growth-restricted rat offspring. Arch Gynecol Obstet 2024; 309:2411-2419. [PMID: 37378669 PMCID: PMC11147910 DOI: 10.1007/s00404-023-07108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE Fetal growth restriction causes a series of sequelae, some of which, such as hyperphagia, reduced satiety and postnatal obesity, are believed to be associated with embryonic hypothalamic neurons impairment. The mechanisms underlying the linkage of fetal brain injuries to break the energy homeostasis have not been elucidated completely. Here, we aim to investigate the effect of intrauterine energy restriction on remodeling appetite neurons in the hypothalamus of fetal and postnatal infant rats. METHODS Low-protein (8%) diet combined with 75% energy restriction was used to establish an animal model. Rats offspring brain tissues, harvested from embryo day 18 and postnatal infant day 1, were sampled for dependent regulator analyses and master neuron assessment. RESULTS Growth-restricted rats showed the increased expression of Bsx and NPY in the hypothalamus as well as remodeling hypothalamic neurons differentiation compared to controls. Intriguingly, in cells cultured in vitro test, we found that activated effects of Bsx and NPY could be exacerbated by DNMT1 inhibitor. CONCLUSIONS In embryonic and early postnatal stage of FGR rats, we detected high concentrations of orexigenic neurons in the hypothalamus. DNMT1 activity is correlated with early embryonic neurogenesis by mediating the expression of Bsx and NPY. It may be one of the reasons for the abnormal development of the appetite regulation pathway and higher susceptibility to obesity in FGR offspring.
Collapse
Affiliation(s)
- Weiling Han
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yao Jia Yuan Road, Chao Yang District, Beijing, 100026, China
| | - Zhaoyi Song
- STI-Zhilian Research Institute for Innovation and Digital Health, Beijing, China
| | - Dan Shan
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yao Jia Yuan Road, Chao Yang District, Beijing, 100026, China
- Department of Obstetrics and Gynecology, The People's Hospital of Yongcheng, Dongcheng District, Yongcheng City, Henan Province, China
| | - Qingyun Shi
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yao Jia Yuan Road, Chao Yang District, Beijing, 100026, China.
| |
Collapse
|
2
|
Arabshomali A, Bazzazzadehgan S, Mahdi F, Shariat-Madar Z. Potential Benefits of Antioxidant Phytochemicals in Type 2 Diabetes. Molecules 2023; 28:7209. [PMID: 37894687 PMCID: PMC10609456 DOI: 10.3390/molecules28207209] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The clinical relationship between diabetes and inflammation is well established. Evidence clearly indicates that disrupting oxidant-antioxidant equilibrium and elevated lipid peroxidation could be a potential mechanism for chronic kidney disease associated with type 2 diabetes mellitus (T2DM). Under diabetic conditions, hyperglycemia, especially inflammation, and increased reactive oxygen species generation are bidirectionally associated. Inflammation, oxidative stress, and tissue damage are believed to play a role in the development of diabetes. Although the exact mechanism underlying oxidative stress and its impact on diabetes progression remains uncertain, the hyperglycemia-inflammation-oxidative stress interaction clearly plays a significant role in the onset and progression of vascular disease, kidney disease, hepatic injury, and pancreas damage and, therefore, holds promise as a therapeutic target. Evidence strongly indicates that the use of multiple antidiabetic medications fails to achieve the normal range for glycated hemoglobin targets, signifying treatment-resistant diabetes. Antioxidants with polyphenols are considered useful as adjuvant therapy for their potential anti-inflammatory effect and antioxidant activity. We aimed to analyze the current major points reported in preclinical, in vivo, and clinical studies of antioxidants in the prevention or treatment of inflammation in T2DM. Then, we will share our speculative vision for future diabetes clinical trials.
Collapse
Affiliation(s)
- Arman Arabshomali
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA; (A.A.); (S.B.)
| | - Shadi Bazzazzadehgan
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA; (A.A.); (S.B.)
| | - Fakhri Mahdi
- Department of BioMolecular Sciences, Division of Pharmacology, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Zia Shariat-Madar
- Department of BioMolecular Sciences, Division of Pharmacology, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| |
Collapse
|
3
|
Wang H, Akbari-Alavijeh S, Parhar RS, Gaugler R, Hashmi S. Partners in diabetes epidemic: A global perspective. World J Diabetes 2023; 14:1463-1477. [PMID: 37970124 PMCID: PMC10642420 DOI: 10.4239/wjd.v14.i10.1463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 09/01/2023] [Indexed: 10/09/2023] Open
Abstract
There is a recent increase in the worldwide prevalence of both obesity and diabetes. In this review we assessed insulin signaling, genetics, environment, lipid metabolism dysfunction and mitochondria as the major determinants in diabetes and to identify the potential mechanism of gut microbiota in diabetes diseases. We searched relevant articles, which have key information from laboratory experiments, epidemiological evidence, clinical trials, experimental models, meta-analysis and review articles, in PubMed, MEDLINE, EMBASE, Google scholars and Cochrane Controlled Trial Database. We selected 144 full-length articles that met our inclusion and exclusion criteria for complete assessment. We have briefly discussed these associations, challenges, and the need for further research to manage and treat diabetes more efficiently. Diabetes involves the complex network of physiological dysfunction that can be attributed to insulin signaling, genetics, environment, obesity, mitochondria and stress. In recent years, there are intriguing findings regarding gut microbiome as the important regulator of diabetes. Valid approaches are necessary for speeding medical advances but we should find a solution sooner given the burden of the metabolic disorder - What we need is a collaborative venture that may involve laboratories both in academia and industries for the scientific progress and its application for the diabetes control.
Collapse
Affiliation(s)
- Huan Wang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, Liaoning Province, China
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Safoura Akbari-Alavijeh
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Ranjit S Parhar
- Department of Biological and Medical Research, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Randy Gaugler
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Sarwar Hashmi
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
- Research and Diagnostics, Ghazala and Sanya Hashmi Foundation, Holmdel, NJ 07733, United States
| |
Collapse
|
4
|
Ntambi JM, Liu X, Burhans MS, ALjohani A, Selen ES, Kalyesubula M, Assadi-Porter F. Hepatic oleate regulates one-carbon metabolism during high carbohydrate feeding. Biochem Biophys Res Commun 2023; 651:62-69. [PMID: 36791500 PMCID: PMC9992055 DOI: 10.1016/j.bbrc.2023.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Obesity is a major risk factor for type 2 diabetes, coronary heart disease, and strok. These diseases are associated with profound alterations in gene expression in metabolic tissues. Epigenetic-mediated regulation of gene expression is one mechanism through which environmental factors, such as diet, modify gene expression and disease predisposition. However, epigenetic control of gene expression in obesity and insulin resistance is not fully characterized. We discovered that liver-specific stearoyl-CoA desaturase-1 (Scd1) knockout mice (LKO) fed a high-carbohydrate low-fat diet exhibit dramatic changes in hepatic gene expression and metabolites of the folate cycle and one-carbon metabolism respectively for the synthesis of S-adenosylmethionine (SAM). LKO mice show an increased ratio of S-adenosylmethionine to S-adenosylhomocysteine, a marker for increased cellular methylation capacity. Furthermore, expression of DNA and histone methyltransferase genes is up-regulated while the mRNA and protein levels of the non-DNA methyltransferases including phosphatidylethanolamine methyltransferase (PEMT), Betaine homocysteine methyltransferase (Bhmt), and the SAM-utilizing enzymes such as glycine-N-methyltransferase (Gnmt) and guanidinoacetate methyltransferase (Gamt) are generally down-regulated. Feeding LKO mice a high carbohydrate diet supplemented with triolein, but not tristearin, and increased endogenous hepatic synthesis of oleate but not palmitoleate in Scd1 global knockout mice normalized one carbon gene expression and metabolite levels. Additionally, changes in one carbon gene expression are independent of the PGC-1α-mediated ER stress response previously reported in the LKO mice. Together, these results highlight the important role of oleate in maintaining one-carbon cycle homeostasis and point to observed changes in one-carbon metabolism as a novel mediator of the Scd1 deficiency-induced liver phenotype.
Collapse
Affiliation(s)
- James M Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Xueqing Liu
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Maggie S Burhans
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Ahmed ALjohani
- College of Science and Health Professions, King Saudi Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Ebru Selin Selen
- Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Mugagga Kalyesubula
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Fariba Assadi-Porter
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
5
|
Ribas-Aulinas F, Ribo S, Casas E, Mourin-Fernandez M, Ramon-Krauel M, Diaz R, Lerin C, Kalko SG, Vavouri T, Jimenez-Chillaron JC. Intergenerational Inheritance of Hepatic Steatosis in a Mouse Model of Childhood Obesity: Potential Involvement of Germ-Line microRNAs. Nutrients 2023; 15:nu15051241. [PMID: 36904241 PMCID: PMC10005268 DOI: 10.3390/nu15051241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Childhood obesity increases the risk of developing metabolic syndrome later in life. Moreover, metabolic dysfunction may be inherited into the following generation through non-genomic mechanisms, with epigenetics as a plausible candidate. The pathways involved in the development of metabolic dysfunction across generations in the context of childhood obesity remain largely unexplored. We have developed a mouse model of early adiposity by reducing litter size at birth (small litter group, SL: 4 pups/dam; control group, C: 8 pups/dam). Mice raised in small litters (SL) developed obesity, insulin resistance and hepatic steatosis with aging. Strikingly, the offspring of SL males (SL-F1) also developed hepatic steatosis. Paternal transmission of an environmentally induced phenotype strongly suggests epigenetic inheritance. We analyzed the hepatic transcriptome in C-F1 and SL-F1 mice to identify pathways involved in the development of hepatic steatosis. We found that the circadian rhythm and lipid metabolic process were the ontologies with highest significance in the liver of SL-F1 mice. We explored whether DNA methylation and small non-coding RNAs might be involved in mediating intergenerational effects. Sperm DNA methylation was largely altered in SL mice. However, these changes did not correlate with the hepatic transcriptome. Next, we analyzed small non-coding RNA content in the testes of mice from the parental generation. Two miRNAs (miR-457 and miR-201) appeared differentially expressed in the testes of SL-F0 mice. They are known to be expressed in mature spermatozoa, but not in oocytes nor early embryos, and they may regulate the transcription of lipogenic genes, but not clock genes, in hepatocytes. Hence, they are strong candidates to mediate the inheritance of adult hepatic steatosis in our murine model. In conclusion, litter size reduction leads to intergenerational effects through non-genomic mechanisms. In our model, DNA methylation does not seem to play a role on the circadian rhythm nor lipid genes. However, at least two paternal miRNAs might influence the expression of a few lipid-related genes in the first-generation offspring, F1.
Collapse
Affiliation(s)
| | - Sílvia Ribo
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Eduard Casas
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | | | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana G. Kalko
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Tanya Vavouri
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- School of Medicine, University of Barcelona, L’Hospitalet, 08907 Barcelona, Spain
- Correspondence: or ; Tel.: +34-934024267
| |
Collapse
|
6
|
Chu DT, Bui NL, Vu Thi H, Nguyen Thi YV. Role of DNA methylation in diabetes and obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:153-170. [PMID: 37019591 DOI: 10.1016/bs.pmbts.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Due to the fact that the upward trend of several metabolic disorders such as diabetes and obesity, in individuals especially monozygotic twins, who are under the same effects from the environment, are not similar, the role of epigenetic elements like DNA methylation needs taking into account. In this chapter, emerging scientific evidence supporting the strong relationship between changes in DNA methylation and those diseases' development was summarized. Changing in the expression level of diabetes/obesity-related genes through being silenced by methylation can be the underlying mechanism of this phenomenon. Genes with abnormal methylation status are potential biomarkers for early prediction and diagnosis. Moreover, methylation-based molecular targets should be investigated as a new treatment for both T2D and obesity.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam.
| | - Nhat-Le Bui
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Hue Vu Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Yen-Vy Nguyen Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| |
Collapse
|
7
|
Harary D, Akinyemi A, Charron MJ, Fuloria M. Fetal Growth and Intrauterine Epigenetic Programming of Obesity and Cardiometabolic Disease. Neoreviews 2022; 23:e363-e372. [PMID: 35641462 PMCID: PMC10100845 DOI: 10.1542/neo.23-6-e363] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Epidemiologic studies have shown an association between an adverse intrauterine environment (eg, exposure to malnutrition) and an increased risk of developing cardiometabolic disease in adulthood. These studies laid the foundation for the developmental origins of health and disease hypothesis, which states that limited nutrient supply to the fetus results in physiologic and metabolic adaptations that favor survival but result in unfavorable consequences in the offspring if there is excess nutrition after birth. This discrepancy in the pre- and postnatal milieus, perceived as stress by the offspring, may confer an increased risk of developing cardiometabolic disease later in life. Thus, early life exposures result in programming or changes in cellular memory that have effects on health throughout the life course. One of the mechanisms by which programming occurs is via epigenetic modifications of genes, processes that result in functionally relevant changes in genes (ie, gene expression) without an alteration in the genotype. In this review, we will describe how fetal exposures, including under- and overnutrition, affect neonatal and childhood growth and the future risk for cardiometabolic disease.
Collapse
Affiliation(s)
- David Harary
- Department of Pediatrics, Division of Neonatology, Children's Hospital at Montefiore, Bronx, NY
| | | | - Maureen J Charron
- Departments of †Biochemistry
- Obstetrics & Gynecology and Women's Health, and
- Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY
| | - Mamta Fuloria
- Department of Pediatrics, Division of Neonatology, Children's Hospital at Montefiore, Bronx, NY
| |
Collapse
|
8
|
Abstract
Introduction: Fatty liver disease, defined by the presence of liver fat infiltration, is part of a cluster of disorders that occur in the context of metabolic syndrome. Epigenetic factors - defined as stable and heritable changes in gene expression without changes in the DNA sequence - may not only play an important role in the disease development in adulthood, but they may start exerting their influence in the prenatal stage.Areas covered: By using systems biology approaches, we review the main epigenetic modifications and highlight their likely roles in the pathogenesis of nonalcoholic fatty liver disease.Expert opinion: Knowledge of the mechanisms by which epigenetic modifications participate in complex disorders would not only help scientists find novel therapeutic strategies but could also aid in implementing preventive care measures at gestation.
Collapse
Affiliation(s)
- Carlos Jose Pirola
- School of Medicine, Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina.,Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council (Conicet)-university of Buenos Aires. Institute of Medical Research (IDIM)
| | - Silvia Sookoian
- School of Medicine, Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina.,Department of Clinical and Molecular Hepatology, National Scientific and Technical Research Council (CONICET)-University of Buenos Aires. Institute of Medical Research (IDIM), Buenos Aires, Argentina
| |
Collapse
|
9
|
Genetically modified mice for research on human diseases: A triumph for Biotechnology or a work in progress? THE EUROBIOTECH JOURNAL 2022. [DOI: 10.2478/ebtj-2022-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
Abstract
Genetically modified mice are engineered as models for human diseases. These mouse models include inbred strains, mutants, gene knockouts, gene knockins, and ‘humanized’ mice. Each mouse model is engineered to mimic a specific disease based on a theory of the genetic basis of that disease. For example, to test the amyloid theory of Alzheimer’s disease, mice with amyloid precursor protein genes are engineered, and to test the tau theory, mice with tau genes are engineered. This paper discusses the importance of mouse models in basic research, drug discovery, and translational research, and examines the question of how to define the “best” mouse model of a disease. The critiques of animal models and the caveats in translating the results from animal models to the treatment of human disease are discussed. Since many diseases are heritable, multigenic, age-related and experience-dependent, resulting from multiple gene-gene and gene-environment interactions, it will be essential to develop mouse models that reflect these genetic, epigenetic and environmental factors from a developmental perspective. Such models would provide further insight into disease emergence, progression and the ability to model two-hit and multi-hit theories of disease. The summary examines the biotechnology for creating genetically modified mice which reflect these factors and how they might be used to discover new treatments for complex human diseases such as cancers, neurodevelopmental and neurodegenerative diseases.
Collapse
|
10
|
Tasin FR, Ahmed A, Halder D, Mandal C. On-going consequences of in utero exposure of Pb: An epigenetic perspective. J Appl Toxicol 2022; 42:1553-1569. [PMID: 35023172 DOI: 10.1002/jat.4287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 11/08/2022]
Abstract
Epigenetic modifications by toxic heavy metals are one of the intensively investigated fields of modern genomic research. Among a diverse group of heavy metals, lead (Pb) is an extensively distributed toxicant causing an immense number of abnormalities in the developing fetus via a wide variety of epigenetic changes. As a divalent cation, Pb can readily cross the placental membrane and the fetal blood brain barrier leading to far-reaching alterations in DNA methylation patterns, histone protein modifications and micro-RNA expression. Over recent years, several human cohorts and animal model studies have documented hyper- and hypo-methylation of developmental genes along with altered DNA methyl-transferase expression by in utero Pb exposure in a dose-, duration- and sex-dependent manner. Modifications in the expression of specific histone acetyltransferase enzymes along with histone acetylation and methylation levels have been reported in rodent and murine models. Apart from these, down-regulation and up-regulation of certain microRNAs crucial for fetal development have been shown to be associated with in utero Pb exposure in human placenta samples. All these modifications in the developing fetus during the prenatal and perinatal stages reportedly caused severe abnormalities in early or adult age, such as - impaired growth, obesity, autism, diabetes, cardiovascular diseases, risks of cancer development and Alzheimer's disease. In this review, currently available information on Pb-mediated alterations in the fetal epigenome is summarized. Further research on Pb-induced epigenome modification will help to understand the mechanisms in detail and will enable us to formulate safety guidelines for pregnant women and developing children.
Collapse
Affiliation(s)
- Fahim Rejanur Tasin
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Asif Ahmed
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Debasish Halder
- Rare Disease research center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
11
|
Burchat N, Sharma P, Ye H, Komakula SSB, Dobrzyn A, Vartanian V, Lloyd RS, Sampath H. Maternal Transmission of Human OGG1 Protects Mice Against Genetically- and Diet-Induced Obesity Through Increased Tissue Mitochondrial Content. Front Cell Dev Biol 2021; 9:718962. [PMID: 34604220 PMCID: PMC8480284 DOI: 10.3389/fcell.2021.718962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Obesity and related metabolic disorders are pressing public health concerns, raising the risk for a multitude of chronic diseases. Obesity is multi-factorial disease, with both diet and lifestyle, as well as genetic and developmental factors leading to alterations in energy balance. In this regard, a novel role for DNA repair glycosylases in modulating risk for obesity has been previously established. Global deletion of either of two different glycosylases with varying substrate specificities, Nei-like endonuclease 1 (NEIL1) or 8-oxoguanine DNA glycosylase-1 (OGG1), both predispose mice to diet-induced obesity (DIO). Conversely, enhanced expression of the human OGG1 gene renders mice resistant to obesity and adiposity. This resistance to DIO is mediated through increases in whole body energy expenditure and increased respiration in adipose tissue. Here, we report that hOGG1 expression also confers resistance to genetically-induced obesity. While Agouti obese (Ay/a) mice are hyperphagic and consequently develop obesity on a chow diet, hOGG1 expression in Ay/a mice (Ay/aTg) prevents increased body weight, without reducing food intake. Instead, obesity resistance in Ay/aTg mice is accompanied by increased whole body energy expenditure and tissue mitochondrial content. We also report for the first time that OGG1-mediated obesity resistance in both the Ay/a model and DIO model requires maternal transmission of the hOGG1 transgene. Maternal, but not paternal, transmission of the hOGG1 transgene is associated with obesity resistance and increased mitochondrial content in adipose tissue. These data demonstrate a critical role for OGG1 in modulating energy balance through changes in adipose tissue function. They also demonstrate the importance of OGG1 in modulating developmental programming of mitochondrial content and quality, thereby determining metabolic outcomes in offspring.
Collapse
Affiliation(s)
- Natalie Burchat
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Priyanka Sharma
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Hong Ye
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Sai Santosh Babu Komakula
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States.,Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Agnieszka Dobrzyn
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States.,Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Vladimir Vartanian
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| | - R Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Harini Sampath
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States.,Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States.,Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| |
Collapse
|
12
|
Capparelli R, Iannelli D. Role of Epigenetics in Type 2 Diabetes and Obesity. Biomedicines 2021; 9:977. [PMID: 34440181 PMCID: PMC8393970 DOI: 10.3390/biomedicines9080977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022] Open
Abstract
Epigenetic marks the genome by DNA methylation, histone modification or non-coding RNAs. Epigenetic marks instruct cells to respond reversibly to environmental cues and keep the specific gene expression stable throughout life. In this review, we concentrate on DNA methylation, the mechanism often associated with transgenerational persistence and for this reason frequently used in the clinic. A large study that included data from 10,000 blood samples detected 187 methylated sites associated with body mass index (BMI). The same study demonstrates that altered methylation results from obesity (OB). In another study the combined genetic and epigenetic analysis allowed us to understand the mechanism associating hepatic insulin resistance and non-alcoholic disease in Type 2 Diabetes (T2D) patients. The study underlines the therapeutic potential of epigenetic studies. We also account for seemingly contradictory results associated with epigenetics.
Collapse
Affiliation(s)
- Rosanna Capparelli
- Department of Agriculture Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy
| | - Domenico Iannelli
- Department of Agriculture Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy
| |
Collapse
|
13
|
Vannuchi N, Jamar G, Pisani L, Braga ARC, de Rosso VV. Chemical composition, bioactive compounds extraction, and observed biological activities from jussara (Euterpe edulis): The exotic and endangered Brazilian superfruit. Compr Rev Food Sci Food Saf 2021; 20:3192-3224. [PMID: 34125477 DOI: 10.1111/1541-4337.12775] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 03/11/2021] [Accepted: 05/03/2021] [Indexed: 12/24/2022]
Abstract
In this article, we reviewed studies on the fruits of the jussara palm (Euterpe edulis Martius), an endangered Brazilian Atlantic Forest palm tree, also coined as "Superfruit." We summarized the chemical components of the pulp and observed biological activities in murine and humans, as well as the best practices involving the extraction of its target compounds, bioavailability, and stability of extracts. Jussara has shown a rich phenolic profile that justifies its antioxidant properties, in addition to a considerable lipidic and energetic value. As the main feature, the fruit possesses large amounts of anthocyanins that can be commercially explored as a food additive or cosmetic colorants. Recent studies emphasized jussara's antioxidant, anti-inflammatory, and cardioprotective capabilities via reshaping of the gut microbiota. Further knowledge is needed to establish bioavailability and optimal serving size, as many of its antioxidant compounds go under chemical bioconversion in the intestinal tract. While extraction of phenolic compounds, anthocyanins, and oils have interesting results, more studies are required in order to reduce the use of conventional organic solvents and improve their stability and shelf life when added to food products, an area in which nanotechnology seems promising.
Collapse
Affiliation(s)
- Nicholas Vannuchi
- Department of Biosciences, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim 136, Santos, Brazil
| | - Giovana Jamar
- Department of Biosciences, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim 136, Santos, Brazil
| | - Luciana Pisani
- Department of Biosciences, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim 136, Santos, Brazil
| | - Anna Rafaela Cavalcante Braga
- Department of Biosciences, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim 136, Santos, Brazil.,Department of Chemical Engineering, Universidade Federal de São Paulo (UNIFESP), Rua São Nicolau 210, Diadema, Sao Paulo, Brazil
| | - Veridiana Vera de Rosso
- Department of Biosciences, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim 136, Santos, Brazil.,Nutrition and Food Service Research Center, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim 136, Santos, Brazil
| |
Collapse
|
14
|
Ghaneifar Z, Yousefi Z, Tajik F, Nikfar B, Ghalibafan F, Abdollahi E, Momtazi-Borojeni AA. The potential therapeutic effects of curcumin on pregnancy complications: Novel insights into reproductive medicine. IUBMB Life 2020; 72:2572-2583. [PMID: 33107698 DOI: 10.1002/iub.2399] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 10/11/2020] [Indexed: 01/13/2023]
Abstract
Pregnancy complications including preeclampsia, preterm birth, intrauterine growth restriction, and gestational diabetes are the main adverse reproductive outcomes. Excessive inflammation and oxidative stress play crucial roles in the pathogenesis of pregnancy disorders. Curcumin, the main polyphenolic compound derived from Curcuma longa, is mainly known by its anti-inflammatory and antioxidant properties. There are in vitro and in vivo reports revealing the preventive and ameliorating effects of curcumin against pregnancy complications. Here, we aimed to seek mechanisms underlying the modulatory effects of curcumin on dysregulated inflammatory and oxidative responses in various pregnancy complications.
Collapse
Affiliation(s)
- Zahra Ghaneifar
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fatemeh Tajik
- Faculty of medicine, Azad University of Tehran, Tehran, Iran
| | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghalibafan
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Abdollahi
- Department of Medical Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Kaji N, Takagi Y, Matsuda S, Takahashi A, Fujio S, Asai F. Effects of liraglutide on metabolic syndrome in WBN/Kob diabetic fatty rats supplemented with a high-fat diet. Animal Model Exp Med 2020; 3:62-68. [PMID: 32318661 PMCID: PMC7167233 DOI: 10.1002/ame2.12106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Liraglutide, a GLP-1 receptor agonist, has recently been used to treat metabolic syndrome (MS) because of its anti-diabetic and anti-obesity effects. We have previously shown that Wistar Bonn Kobori diabetic and fatty (WBN/Kob-Lepr fa , WBKDF) rats fed a high-fat diet (HFD) developed MS including marked obesity, hyperglycemia, and dyslipidemia. To obtain further information on WBKDF-HFD rats as a severe MS model, we performed a pharmacological investigation into the anti-MS effects of liraglutide in this model. METHODS Seven-week-old male WBKDF-HFD rats were allocated to three groups (N = 8 in each group): a vehicle group, a low-dose liraglutide group, and a high-dose liraglutide group. They received subcutaneous injections of either saline or liraglutide at doses of 75 or 300 μg/kg body weight once daily for 4 weeks. RESULTS Results showed that liraglutide treatment reduced body weight gain and food intake in a dose-dependent manner. The marked hyperglycemia and the glucose tolerance were also significantly ameliorated in the liraglutide-treated groups. Moreover, liraglutide also reduced the plasma triglyceride concentration and liver fat accumulation. CONCLUSIONS The present study demonstrated that liraglutide could significantly alleviate MS in WBKDF-HFD rats, and the reaction to liraglutide is similar to human patients with MS. WBKDF-HFD rats are therefore considered to be a useful model for research on severe human MS.
Collapse
Affiliation(s)
- Noriyuki Kaji
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineAzabu UniversityKanagawaJapan
| | - Yoshiichi Takagi
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineAzabu UniversityKanagawaJapan
| | - Satomi Matsuda
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineAzabu UniversityKanagawaJapan
| | - Anna Takahashi
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineAzabu UniversityKanagawaJapan
| | - Sakurako Fujio
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineAzabu UniversityKanagawaJapan
| | - Fumitoshi Asai
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineAzabu UniversityKanagawaJapan
| |
Collapse
|
16
|
Reeves J, Smith C, Dierenfeld ES, Whitehouse-Tedd K. Captivity-induced metabolic programming in an endangered felid: implications for species conservation. Sci Rep 2020; 10:3630. [PMID: 32107441 PMCID: PMC7046719 DOI: 10.1038/s41598-020-60577-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/12/2020] [Indexed: 11/08/2022] Open
Abstract
Reintroduction of captive-bred individuals into the wild is an important conservation activity. However, environmental conditions can influence developmental programming, potentially causing metabolic disorders in adults. These effects are investigated here for the first time in an endangered species. Using body weight and feed intake data for Iberian lynx (Lynx pardinus) (n = 22), we compared the growth of captive versus wild born and/or reared individuals. Captive-born individuals gained weight as a function of calorie intake, unlike wild-born individuals. When compared with females reared in the wild, captive-reared females achieved a larger body size, without evidence of obesity. Captivity-associated changes to metabolic programming may compromise survival in the wild if an increased body size incurs a greater energy requirement. Large body size may also confer a competitive advantage over smaller, wild-born individuals, disrupting the social organisation of existing wild populations, and inferring long-term implications for the phenotypic composition of wild populations.
Collapse
Affiliation(s)
- Jessica Reeves
- Iberian Lynx Captive Breeding Centre "El Acebuche", Parque Nacional de Doñana, Matalascañas, 21760, Huelva, Spain
| | - Carl Smith
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, NG25 0QF, Nottinghamshire, United Kingdom
- Department of Ecology & Vertebrate Zoology, University of Łódź, 12/16 Banacha Street, 90-237, Łódź, Poland
- Institute of Vertebrate Biology, Academy of Sciences of the Czech Republic, Květná 8, 603 65, Brno, Czech Republic
| | - Ellen S Dierenfeld
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, NG25 0QF, Nottinghamshire, United Kingdom
- Ellen S. Dierenfeld LLC, St. Louis, MO, United States of America
| | - Katherine Whitehouse-Tedd
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, NG25 0QF, Nottinghamshire, United Kingdom.
| |
Collapse
|
17
|
Huerta-Cervantes M, Peña-Montes DJ, Montoya-Pérez R, Trujillo X, Huerta M, López-Vázquez MÁ, Olvera-Cortés ME, Saavedra-Molina A. Gestational Diabetes Triggers Oxidative Stress in Hippocampus and Cerebral Cortex and Cognitive Behavior Modifications in Rat Offspring: Age- and Sex-Dependent Effects. Nutrients 2020; 12:nu12020376. [PMID: 32023917 PMCID: PMC7071266 DOI: 10.3390/nu12020376] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/04/2020] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Gestational diabetes (GD) has been linked with an increased risk of developing metabolic disorders and behavioral abnormalities in the offspring. Oxidative stress is strongly associated with neurodegeneration and cognitive disruption. In the offspring brains in a GD experimental rat model, increased oxidative stress in the prenatal and postnatal stages was reported. However, long-term alterations to offspring behavior and oxidative stress, caused by changes in the cerebral cortex and hippocampus, remain unclear. In this study, we evaluated the effect of GD on young and adult male and female rat offspring in metabolic parameters, cognitive behavior, and oxidative stress. GD was induced using streptozotocin in dams. Next, the offspring were evaluated at two and six months of age. Anxiety-like behavior was evaluated using the elevated plus maze and open field maze; spatial learning and short-term memory were evaluated using the Morris water maze and radial maze, respectively. We determined oxidative stress biomarkers (reactive oxygen species (ROS), lipid peroxidation and glutathione status) and antioxidant enzymes (superoxide dismutase and catalase) in the brain of offspring. We observed that male GD offspring showed a reduced level of anxiety at both ages as they spent less time in the closed arms of the elevated plus maze at adult age ((P = 0.019, d = 1.083 ( size effect)) and spent more time in the open area of an open field (P = 0.0412, d = 0.743) when young and adult age (P = 0.018, d = 0.65). Adult female GD offspring showed a reduced level of anxiety (P = 0.036; d = 0.966), and young female GD offspring showed a deficiency in spatial learning (P = 0.0291 vs. control, d = 3.207). Adult male GD offspring showed a deficiency in short-term memory (P = 0.017, d = 1.795). We found an increase in ROS and lipid peroxidation, a disruption in the glutathione status, and decreased activity of catalase and superoxide dismutase (P < 0.05 vs. control, d > 1.0), in the cerebral cortex and hippocampus of male and female GD offspring. GD altered metabolism; male offspring of both ages and adult females showed a high level of triglycerides and a lower level of high-density lipoprotein-cholesterol (P < 0.05 vs. control, d > 1.0). Young and adult female offspring displayed higher insulin levels (P < 0.05, d > 1.0). These results suggest that gestational diabetes modifies oxidative stress and cognitive behavior in an age- and sex-dependent manner.
Collapse
Affiliation(s)
- Maribel Huerta-Cervantes
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, 58030 Morelia, Mich., Mexico; (M.H.-C.); (D.J.P.-M.); (R.M.-P.)
| | - Donovan J. Peña-Montes
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, 58030 Morelia, Mich., Mexico; (M.H.-C.); (D.J.P.-M.); (R.M.-P.)
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045 Colima, Colima, Mexico; (X.T.); (M.H.)
| | - Rocío Montoya-Pérez
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, 58030 Morelia, Mich., Mexico; (M.H.-C.); (D.J.P.-M.); (R.M.-P.)
| | - Xóchitl Trujillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045 Colima, Colima, Mexico; (X.T.); (M.H.)
| | - Miguel Huerta
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045 Colima, Colima, Mexico; (X.T.); (M.H.)
| | - Miguel Ángel López-Vázquez
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, 58341 Morelia, Mich., Mexico;
| | - María Esther Olvera-Cortés
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, 58341 Morelia, Mich., Mexico;
- Correspondence: (A.S.-M.); (M.E.O-C.); Tel.: +52-443-326-5790 (A.S.-M.); + 52-443-322-2600 (M.E.O-C.)
| | - Alfredo Saavedra-Molina
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, 58030 Morelia, Mich., Mexico; (M.H.-C.); (D.J.P.-M.); (R.M.-P.)
- Correspondence: (A.S.-M.); (M.E.O-C.); Tel.: +52-443-326-5790 (A.S.-M.); + 52-443-322-2600 (M.E.O-C.)
| |
Collapse
|
18
|
Contreras RE, Schriever SC, Pfluger PT. Physiological and Epigenetic Features of Yoyo Dieting and Weight Control. Front Genet 2019; 10:1015. [PMID: 31921275 PMCID: PMC6917653 DOI: 10.3389/fgene.2019.01015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022] Open
Abstract
Obesity and being overweight have become a worldwide epidemic affecting more than 1.9 billion adults and 340 million children. Efforts to curb this global health burden by developing effective long-term non-surgical weight loss interventions continue to fail due to weight regain after weight loss. Weight cycling, often referred to as Yoyo dieting, is driven by physiological counter-regulatory mechanisms that aim at preserving energy, i.e. decreased energy expenditure, increased energy intake, and impaired brain-periphery communication. Models based on genetically determined set points explained some of the weight control mechanisms, but exact molecular underpinnings remained elusive. Today, gene–environment interactions begin to emerge as likely drivers for the obesogenic memory effect associated with weight cycling. Here, epigenetic mechanisms, including histone modifications and DNA methylation, appear as likely factors that underpin long-lasting deleterious adaptations or an imprinted obesogenic memory to prevent weight loss maintenance. The first part summarizes our current knowledge on the physiology of weight cycling by discussing human and murine studies on the Yoyo-dieting phenomenon and physiological adaptations associated with weight loss and weight re-gain. The second part provides an overview on known associations between obesity and epigenetic modifications. We further interrogate the roles of epigenetic mechanisms in the CNS control of cognitive functions as well as reward and addictive behaviors, and subsequently discuss whether such mechanisms play a role in weight control. The final two parts describe major opportunities and challenges associated with studying epigenetic mechanisms in the CNS with its highly heterogenous cell populations, and provide a summary of recent technological advances that will help to delineate whether an obese memory is based upon epigenetic mechanisms.
Collapse
Affiliation(s)
- Raian E Contreras
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Centre for Diabetes Research (DZD), Neuherberg, Germany.,Neurobiology of Diabetes, TUM School of Medicine, Technische Universität München, Munich, Germany
| | - Sonja C Schriever
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Centre for Diabetes Research (DZD), Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.,German Centre for Diabetes Research (DZD), Neuherberg, Germany.,Neurobiology of Diabetes, TUM School of Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
19
|
Lopez-Tello J, Arias-Alvarez M, Gonzalez-Bulnes A, Sferuzzi-Perri AN. Models of Intrauterine growth restriction and fetal programming in rabbits. Mol Reprod Dev 2019; 86:1781-1809. [PMID: 31538701 DOI: 10.1002/mrd.23271] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/02/2019] [Indexed: 12/23/2022]
Abstract
Intrauterine growth restriction (IUGR) affects approximately 10% of human pregnancies globally and has immediate and life-long consequences for offspring health. However, the mechanisms underlying the pathogenesis of IUGR and its association with later health and disease outcomes are poorly understood. To address these knowledge gaps, the use of experimental animals is critically important. Since the 50's different environmental, pharmacological, and surgical manipulations have been performed in the rabbit to improve our knowledge of the control of fetal growth, fetal responses to IUGR, and mechanisms by which offspring may be programmed by an adverse gestational environment. The purpose of this review is therefore to summarize the utility of the rabbit as a model for IUGR research. It first summarizes the knowledge of prenatal and postnatal development in the rabbit and how these events relate to developmental milestones in humans. It then describes the methods used to induce IUGR in rabbits and the knowledge gained about the mechanisms determining prenatal and postnatal outcomes of the offspring. Finally, it discusses the application of state of the art approaches in the rabbit, including high-resolution ultrasound, magnetic resonance imaging, and gene targeting, to gain a deeper integrative understanding of the physiological and molecular events governing the development of IUGR. Overall, we hope to engage and inspire investigators to employ the rabbit as a model organism when studying pregnancy physiology so that we may advance our understanding of mechanisms underlying IUGR and its consequences in humans and other mammalian species.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Maria Arias-Alvarez
- Department of Animal Production. Veterinary Faculty, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | | | - Amanda N Sferuzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Programming mediated by fatty acids affects uncoupling protein 1 (UCP-1) in brown adipose tissue. Br J Nutr 2019; 120:619-627. [PMID: 30176958 DOI: 10.1017/s0007114518001629] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Brown adipose tissue (BAT) has recently been given more attention for the part it plays in obesity. BAT can generate great amounts of heat through thermogenesis by the activation of uncoupling protein 1 (UCP-1), which can be regulated by many environmental factors such as diet. Moreover, the build-up of BAT relates to maternal nutritional changes during pregnancy and lactation. However, at present, there is a limited number of studies looking at maternal nutrition and BAT development, and it seems that the research trend in this field has been considerably declining since the 1980s. There is much to discover yet about the role of different fatty acids on the development of BAT and the activation of UCP-1 during the fetal and the postnatal periods of life. A better understanding of the impact of nutritional intervention on the epigenetic regulation of BAT could lead to new preventive care for metabolic diseases such as obesity. It is important to know in which circumstances lipids could programme BAT during pregnancy and lactation. The modification of maternal dietary fatty acids, amount and composition, during pregnancy and lactation might be a promising strategy for the prevention of obesity in the offspring and future generations.
Collapse
|
21
|
Abstract
AIM Type 2 diabetes (T2DM) is a complex disease. Interactions between genetic susceptible variants and environmental cues results in the development of this heterogenous disease. Having an understanding of the genetics of T2DM may lead to a better perspective and management of the pathogenesis contributing to T2DM. MATERIALS AND METHODS Published primary and secondary sources were reviewed covering the keywords "genetics + type 2 diabetes" using PubMed and Google Scholar as the main databases. Full articles were considered when the title and the abstract was found to be sufficiently related to the review's aim. RESULTS Various genetic aspects of T2DM were summarised including a general understanding of the heritability and heterogeneity of T2DM. Furthermore, an explanation of the different genetic modalities that can be used to identify T2DM susceptible genes was provided. CONCLUSION In this day and era, researchers and healthcare professionals working in the field of metabolic disorders should have an understanding of T2DM genetics. The future lies in preventive and management action plans targeting the combination of genetics and environmental risk factors for a better health outcome.
Collapse
Affiliation(s)
- Sarah Cuschieri
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| |
Collapse
|
22
|
Alam I, Ali F, Zeb F, Almajwal A, Fatima S, Wu X. Relationship of nutrigenomics and aging: Involvement of DNA methylation. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2019. [DOI: 10.1016/j.jnim.2019.100098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
23
|
Menting MD, Mintjens S, van de Beek C, Frick CJ, Ozanne SE, Limpens J, Roseboom TJ, Hooijmans CR, van Deutekom AW, Painter RC. Maternal obesity in pregnancy impacts offspring cardiometabolic health: Systematic review and meta-analysis of animal studies. Obes Rev 2019; 20:675-685. [PMID: 30633422 PMCID: PMC6849816 DOI: 10.1111/obr.12817] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/19/2018] [Accepted: 11/06/2018] [Indexed: 12/24/2022]
Abstract
Obesity before and during pregnancy leads to reduced offspring cardiometabolic health. Here, we systematically reviewed animal experimental evidence of maternal obesity before and during pregnancy and offspring anthropometry and cardiometabolic health. We systematically searched Embase and Medline from inception until January 2018. Eligible publications compared offspring of mothers with obesity to mothers with a normal weight. We performed meta-analyses and subgroup analyses. We also examined methodological quality and publication bias. We screened 2543 publications and included 145 publications (N = 21 048 animals, five species). Essential methodological details were not reported in the majority of studies. We found evidence of publication bias for birth weight. Offspring of mothers with obesity had higher body weight (standardized mean difference (SMD) 0.76 [95% CI 0.60;0.93]), fat percentage (0.99 [0.64;1.35]), systolic blood pressure (1.33 [0.75;1.91]), triglycerides (0.64 [0.42;0.86], total cholesterol (0.46 [0.18;0.73]), glucose level (0.43 [0.24;0.63]), and insulin level (0.81 [0.61;1.02]) than offspring of control mothers, but similar birth weight. Sex, age, or species did not influence the effect of maternal obesity on offspring's cardiometabolic health. Obesity before and during pregnancy reduces offspring cardiometabolic health in animals. Future intervention studies should investigate whether reducing obesity prior to conception could prevent these detrimental programming effects and improve cardiometabolic health of future generations.
Collapse
Affiliation(s)
- M D Menting
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - S Mintjens
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pediatrics, Department of Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - C van de Beek
- Department of Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - C J Frick
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - S E Ozanne
- MRC Metabolic Diseases Unit and Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - J Limpens
- Department of Research Support-Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - T J Roseboom
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - C R Hooijmans
- Department for Health Evidence Unit SYRCLE, Department of Anesthesiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A W van Deutekom
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Cardiology, Amsterdam, The Netherlands
| | - R C Painter
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Ding N, Maiuri AR, O'Hagan HM. The emerging role of epigenetic modifiers in repair of DNA damage associated with chronic inflammatory diseases. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2019; 780:69-81. [PMID: 31395351 PMCID: PMC6690501 DOI: 10.1016/j.mrrev.2017.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 09/25/2017] [Accepted: 09/27/2017] [Indexed: 12/15/2022]
Abstract
At sites of chronic inflammation epithelial cells are exposed to high levels of reactive oxygen species (ROS), which can contribute to the initiation and development of many different human cancers. Aberrant epigenetic alterations that cause transcriptional silencing of tumor suppressor genes are also implicated in many diseases associated with inflammation, including cancer. However, it is not clear how altered epigenetic gene silencing is initiated during chronic inflammation. The high level of ROS at sites of inflammation is known to induce oxidative DNA damage in surrounding epithelial cells. Furthermore, DNA damage is known to trigger several responses, including recruitment of DNA repair proteins, transcriptional repression, chromatin modifications and other cell signaling events. Recruitment of epigenetic modifiers to chromatin in response to DNA damage results in transient covalent modifications to chromatin such as histone ubiquitination, acetylation and methylation and DNA methylation. DNA damage also alters non-coding RNA expression. All of these alterations have the potential to alter gene expression at sites of damage. Typically, these modifications and gene transcription are restored back to normal once the repair of the DNA damage is completed. However, chronic inflammation may induce sustained DNA damage and DNA damage responses that result in these transient covalent chromatin modifications becoming mitotically stable epigenetic alterations. Understanding how epigenetic alterations are initiated during chronic inflammation will allow us to develop pharmaceutical strategies to prevent or treat chronic inflammation-induced cancer. This review will focus on types of DNA damage and epigenetic alterations associated with chronic inflammatory diseases, the types of DNA damage and transient covalent chromatin modifications induced by inflammation and oxidative DNA damage and how these modifications may result in epigenetic alterations.
Collapse
Affiliation(s)
- Ning Ding
- Medical Sciences Program, School of Medicine, Indiana University, Bloomington, IN 47405, USA
| | - Ashley R Maiuri
- Medical Sciences Program, School of Medicine, Indiana University, Bloomington, IN 47405, USA
| | - Heather M O'Hagan
- Medical Sciences Program, School of Medicine, Indiana University, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
25
|
Franzago M, Fraticelli F, Stuppia L, Vitacolonna E. Nutrigenetics, epigenetics and gestational diabetes: consequences in mother and child. Epigenetics 2019; 14:215-235. [PMID: 30865571 PMCID: PMC6557546 DOI: 10.1080/15592294.2019.1582277] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gestational Diabetes Mellitus (GDM) is the most common metabolic condition during pregnancy and may result in short- and long-term complications for both mother and offspring. The complexity of phenotypic outcomes seems influenced by genetic susceptibility, nutrient-gene interactions and lifestyle interacting with clinical factors. There is strong evidence that not only the adverse genetic background but also the epigenetic modifications in response to nutritional and environmental factors could influence the maternal hyperglycemia in pregnancy and the foetal metabolic programming. In this view, the correlation between epigenetic modifications and their transgenerational effects represents a very interesting field of study. The present review gives insight into the role of gene variants and their interactions with nutrients in GDM. In addition, we provide an overview of the epigenetic changes and their role in the maternal-foetal transmission of chronic diseases. Overall, the knowledge of epigenetic modifications induced by an adverse intrauterine and perinatal environment could shed light on the potential pathophysiological mechanisms of long-term disease development in the offspring and provide useful tools for their prevention.
Collapse
Affiliation(s)
- Marica Franzago
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy.,b Molecular Genetics, Unit , CeSI-Met , Chieti , Italy
| | - Federica Fraticelli
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| | - Liborio Stuppia
- b Molecular Genetics, Unit , CeSI-Met , Chieti , Italy.,c Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| | - Ester Vitacolonna
- a Department of Medicine and Aging, School of Medicine and Health Sciences , "G. d'Annunzio" University, Chieti-Pescara , Chieti , Italy
| |
Collapse
|
26
|
Li X, Tu P, Umar M, Liu Q, Luo W, Yang X, Zhu J, Kong D, Li M. A study on molecular mechanisms of adiposis induced by long-term treatment of high-fat and high-sucrose in C57BL/6J mice. Physiol Res 2019; 68:75-87. [PMID: 30433796 DOI: 10.33549/physiolres.933830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Adiposis is reputed as a twin disease of type 2 diabetes and greatly harmful to human health. In order to understand the molecular mechanisms of adiposis, the changes of physiological, pathological, epigenetic and correlative gene expression were investigated during the adiposis development of C57BL/6J mice induced by long time (9 months) high-fat and high-sucrose diet (HFSD) sustainably. The results showed that mRNA transcription level of the Leptin, Glut4 and Glut2 genes have been obviously changed, which exhibit a negative correlation with methylation on their promoter DNA. The results also revealed that HFSD induced higher level of DNA methyltransferase 1 (DNMT1) in fat tissue might play important role in regulating the changes of methylation pattern on Glut4 and Leptin genes, and which might be one of the molecular mechanisms for the adiposis development.
Collapse
Affiliation(s)
- X Li
- Life Science College, Nankai University, Nankai District, Tianjin, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Longo M, Alrais M, Tamayo EH, Ferrari F, Facchinetti F, Refuerzo JS, Blackwell SC, Sibai BM. Vascular and metabolic profiles in offspring born to pregnant mice with metabolic syndrome treated with inositols. Am J Obstet Gynecol 2019; 220:279.e1-279.e9. [PMID: 30521799 DOI: 10.1016/j.ajog.2018.11.1101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 11/11/2018] [Accepted: 11/24/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Inositols (INOs) supplementation during pregnancy, specifically the combination of myo-inositol (MI) and D-chiro-inositol (DCI), has been reported to improve vascular parameters in women with gestational diabetes mellitus. We demonstrated previously that offspring born to pregnant mice lacking the endothelial nitric oxide synthase (eNOS+/-) gene have hypertension (HTN) as adults and, when fed a high-fat diet (HFD), develop a metabolic syndrome (MS) phenotype. OBJECTIVE Our aim was to evaluate whether INOs treatment in pregnancy complicated by MS improves the vascular and metabolic profile in mice offspring programmed in utero to develop HTN and MS. MATERIALS AND METHODS Heterozygous eNOS+/- mice fed an HFD manifest a MS phenotype. Female eNOS+/- mice with MS were bred with a wild-type (WT) male. On gestational day 1, pregnant females were randomly allocated to receive either a mixture of INOs (MI/DCI: 7.2/0.18 mg/mL) or water as placebo until delivery. The female offspring obtained were genotyped and categorized as: WT (genetically normal, with eNOS gene) and eNOS+/- offspring (genetically modified, heterozygous for eNOS gene). Both offspring developed in an abnormal uterine environment due to maternal MS. At 9-10 weeks of age, the offspring underwent a glucose tolerance test (GTT) and systolic blood pressure (SBP) measurement. The mice were then sacrificed, and the carotid arteries were isolated for evaluation of vascular responses. Responses to phenylephrine (PE), in the presence and absence of a nonspecific nitric oxide inhibitor (N-nitro-L-arginine methyl ester [L-NAME]), the vasodilator acetylcholine (ACh), and sodium nitroprusside (SNP) were assessed. RESULTS The GTT showed lower glucose levels in both eNOS+/-INOs (P = .03) and WT-INOs (P = .05) offspring born to MS dams on INOs supplementation compared to offspring born to untreated dams. SBP was higher in eNOS+/- offspring compared to WT (169 ± 7 vs 142 ± 9 mm Hg, respectively, P = .04) and INOs treatment decreased SBP in WT-INOs (110 ± 10 mm Hg, P = .01) but not in eNOS+/-INOs offspring. Maximal (%Max) contractile response to PE was higher in eNOS+/- offspring born to MS dams and was decreased in those born to MS dams treated with INOs (%Max, eNOS+/-, 123 ± 7 vs eNOS+/-INOs, 82 ± 11 mm Hg, P = .007). No differences were seen in PE contractile responses in WT offspring born to MS dams treated or not treated with INOs (WT, 92 ± 4 vs WT-INOs, 75 ± 7). The L-NAME response was decreased in eNOS+/-INOs and WT-INOs offspring compared to untreated ones. The ACh vasorelaxation was impaired in eNOS+/- and WT offspring born to MS dams, and maternal INOs treatment improved offspring vascular relaxation in both offspring (P = .01 and P = .03, respectively). No differences were seen in response to SNP. CONCLUSION Inositols supplementation improved glucose tolerance, SBP, and vascular responses in adult eNOS+/- and WT offspring born to dams with MS. Interestingly, WT born to MS dams show an altered vascular profile similar to eNOS+/- offspring and exhibit an improved response to INOs treatment. Our findings suggest that the benefits of INOs treatment are more pronounced in offspring exposed to environmental factors in utero, and less likely in those due to genetic factors.
Collapse
|
28
|
Jacquet A, Barbeau D, Arnaud J, Hijazi S, Hazane-Puch F, Lamarche F, Quiclet C, Couturier K, Fontaine E, Moulis JM, Demeilliers C. Impact of maternal low-level cadmium exposure on glucose and lipid metabolism of the litter at different ages after weaning. CHEMOSPHERE 2019; 219:109-121. [PMID: 30537584 DOI: 10.1016/j.chemosphere.2018.11.137] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 06/09/2023]
Abstract
Cadmium (Cd) is a metal which may participate in the development of type II diabetes even if Cd exposure levels are mild. However, experimental studies focusing on daily environmentally relevant doses are scarce, particularly for glucose metabolism of the offspring of chronically exposed mothers. The aim is to measure the impact of maternal low level Cd exposure on glucose and lipid metabolism of offspring. Female rats were exposed to 0, 50 or 500 μg.kg-1.d-1 of CdCl2, 21 days before mating and during 21 days of gestation and 21 days of lactation. Pups exposure was organized in 3 groups (control, Cd1, Cd2) according to renal dams' Cd burden. Parameters of glucose and lipid metabolisms were measured for the pups on post-natal day 21, 26 and 60. Maternal Cd exposure led to significant amounts of Cd in the liver and kidney of pups. At weaning, insulin secretion upon glucose stimulation was unchanged, but the removal of circulating glucose was slower for pups born from the lowest impregnated dams (Cd1). Five days after, glucose tolerance of all groups was identical. Thus, this loss of insulin sensitivity was reversed, in part by increased adiponectin secretion for the Cd1 group. Furthermore, pups from dams accumulating the highest levels of Cd (Cd2) exhibited a compensatory increased insulin pancreatic secretion, together with increased circulating non-esterified fatty acids, indicating the establishment of insulin resistance, 2 months after birth. This study has demonstrated the influence of maternal exposure to low levels of Cd on glucose homeostasis in the offspring that might increase the risk of developing type II diabetes later in life.
Collapse
Affiliation(s)
- Adeline Jacquet
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France
| | - Damien Barbeau
- Grenoble University Hospital, Institute of Biology and Pathology, Grenoble, France; EPSP-TIMC UMR CNRS 5525, Grenoble, France
| | - Josiane Arnaud
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France; Grenoble University Hospital, Institute of Biology and Pathology, Grenoble, France
| | - Samer Hijazi
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France
| | - Florence Hazane-Puch
- Grenoble University Hospital, Institute of Biology and Pathology, Grenoble, France
| | | | - Charline Quiclet
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France
| | - Karine Couturier
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France
| | - Eric Fontaine
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France; Grenoble University Hospital, Institute of Biology and Pathology, Grenoble, France
| | - Jean-Marc Moulis
- Univ. Grenoble Alpes, Inserm U1055, LBFA, 38000 Grenoble, France; CEA-DRF-BIG, Grenoble, France
| | | |
Collapse
|
29
|
Unveiling the Role of DNA Methylation in Kidney Transplantation: Novel Perspectives toward Biomarker Identification. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1602539. [PMID: 30766879 PMCID: PMC6350635 DOI: 10.1155/2019/1602539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/30/2018] [Indexed: 12/13/2022]
Abstract
The burden of chronic kidney disease is dramatically rising, making it a major public health concern worldwide. Kidney transplantation is now the best treatment for patients with end-stage renal disease. Although kidney transplantation may improve survival and quality of life, its long-term results are hampered by immune- and/or non-immune-mediated complications. Thus, the identification of transplanted patients with a higher risk of posttransplant complications has become a big challenge for public health. However, current biomarkers of posttransplant complications have a poor predictive value, rising the need to explore novel approaches for the management of transplant patient. In this review we summarize the emerging literature about DNA methylation in kidney transplant complications, in order to highlight its perspectives toward biomarker identification. In the forthcoming future the monitoring of DNA methylation in kidney transplant patients could become a plausible strategy toward the prevention and/or treatment of kidney transplant complications.
Collapse
|
30
|
Supplementation of Juçara Berry (Euterpe edulis Mart.) Modulates Epigenetic Markers in Monocytes from Obese Adults: A Double-Blind Randomized Trial. Nutrients 2018; 10:nu10121899. [PMID: 30513988 PMCID: PMC6315800 DOI: 10.3390/nu10121899] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022] Open
Abstract
Nutrigenomics is an emerging field in obesity since epigenetic markers can be modified by environmental factors including diet. Considering juçara composition—rich in anthocyanins, monounsaturated fatty acids (MUFAs) and fibers—it has the potential for epigenetic modulation. We evaluated the juçara supplementation modulating the serum fatty acids profile and epigenetic markers in monocytes of adult obese humans. It was a randomized double-blind, controlled trial with 27 obese (Body mass index between 30.0 and 39.9 kg/m2) participants of both genders aged from 31 to 59 years, divided into juçara group (5 g juçara freeze-dried pulp) or placebo group (5 g of maltodextrin) for 6 weeks. Before and after supplementation, blood samples were collected. The serum and monocytes cells obtained were cultured and stimulated with lipopolysaccharides as proinflammatory stimulus. After 24 h of incubation, the cells and supernatants were collected and analyzed. Juçara improved the serum fatty acids profile on unsaturated fatty acids levels. The epigenetic markers evaluated were improved post-treatment. Also, the methylated DNA level was increased after treatment. We find that juçara supplementation is a predictor of methyl CpG binding proteins 2 (MeCP2) in monocytes. Concluding, juçara supplementation improved the serum fatty acids profile, modulating the epigenetic markers in monocytes from obese individuals.
Collapse
|
31
|
Martino J, Segura MT, García-Valdés L, Padilla MC, Rueda R, McArdle HJ, Budge H, Symonds ME, Campoy C. The Impact of Maternal Pre-Pregnancy Body Weight and Gestational Diabetes on Markers of Folate Metabolism in the Placenta. Nutrients 2018; 10:nu10111750. [PMID: 30428605 PMCID: PMC6266824 DOI: 10.3390/nu10111750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 01/22/2023] Open
Abstract
Dietary methyl donors, including folate, may modify the placenta and size at birth but the influence of maternal body weight has not been widely investigated. We therefore examined whether maternal or fetal folate status, together with indices of placental folate transport, were modulated by either maternal pre-pregnancy body mass index (BMI i.e., overweight: 25 ≤ BMI < 30 or obesity: BMI ≥ 30 kg/m2) and/or gestational diabetes mellitus (GD). We utilised a sub-sample of 135 pregnant women participating in the Spanish PREOBE survey for our analysis (i.e., 59 healthy normal weight, 29 overweight, 22 obese and 25 GD). They were blood sampled at 34 weeks gestation, and, at delivery, when a placental sample was taken together with maternal and cord blood. Placental gene expression of folate transporters and DNA methyltransferases (DNMT) were all measured. Folate plasma concentrations were determined with an electro-chemiluminescence immunoassay. Food diaries indicated that folate intake was unaffected by BMI or GD and, although all women maintained normal folate concentrations (i.e., 5–16 ng/mL), higher BMIs were associated with reduced maternal folate concentrations at delivery. Umbilical cord folate was not different, reflecting an increased concentration gradient between the mother and her fetus. Placental mRNA abundance for the folate receptor alpha (FOLR1) was reduced with obesity, whilst DNMT1 was increased with raised BMI, responses that were unaffected by GD. Multi-regression analysis to determine the best predictors for placental FOLR1 indicated that pre-gestational BMI had the greatest influence. In conclusion, the placenta’s capacity to maintain fetal folate supply was not compromised by either obesity or GD.
Collapse
Affiliation(s)
- Jole Martino
- Early Life Research Unit, Division of Child Health and Obstetrics and Gynaecology, Nottingham NG7 2UH, UK.
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18071 Granada, Spain.
| | - Maria Teresa Segura
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18071 Granada, Spain.
| | - Luz García-Valdés
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18071 Granada, Spain.
| | - M C Padilla
- Department of Obstetrics and Gynaecology, University of Granada, 18071 Granada, Spain.
| | | | - Harry J McArdle
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB24 3FX UK.
| | - Helen Budge
- Early Life Research Unit, Division of Child Health and Obstetrics and Gynaecology, Nottingham NG7 2UH, UK.
| | - Michael E Symonds
- Early Life Research Unit, Division of Child Health and Obstetrics and Gynaecology, Nottingham NG7 2UH, UK.
- Nottingham Digestive Disease Centre, Biomedical Research Unit, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, 18071 Granada, Spain.
| |
Collapse
|
32
|
Dechow CD, Liu WS. DNA methylation patterns in peripheral blood mononuclear cells from Holstein cattle with variable milk yield. BMC Genomics 2018; 19:744. [PMID: 30309336 PMCID: PMC6182825 DOI: 10.1186/s12864-018-5124-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/27/2018] [Indexed: 11/15/2022] Open
Abstract
Background Milk yield for Holstein cows has doubled over five decades due to genetic selection and changes to management, but the molecular mechanisms that facilitated this increase are mostly unknown. Epigenetic modifications to the cattle genome are a plausible molecular mechanism to cause variation in milk yield and our objective was to describe genome-wide DNA methylation patterns in peripheral blood mononuclear cells (PBMC) from mature Holstein dairy cows with variable milk yield. Results Whole genome MeDIP-seq was performed following DNA extraction from PBMC of 6 lactating dairy cows from 4 different herds that varied in milk yield from 13,556 kg to 23,105 kg per 305 day lactation. We describe methylation across the genome and for 13,677 protein coding genes. Repetitive element reads were primarily mapped to satellite (36.4%), SINE (29.1%), and LINE (23.7%) regions and the majority (78.4%) of CpG sites were sequenced at least once. DNA methylation was generally low upstream of genes with the nadir occurring 95 bp prior to the transcription start site (TSS). Methylation was lower in the first exon than in later exons, was highest for introns near the intron-exon junctions, and declined downstream as the distance from the gene increased. We identified 72 differentially methylated regions (DMR) between high milk yield cows and their control, and 252 DMR across herd environments. Conclusions This reference methylome for cattle with extreme variation in milk yield phenotype provides a resource to more fully evaluate relationships between DNA methylation and phenotype in populations subject to selection. The detection of DMR in cows of varying milk yield suggests potential to exploit epigenetic variation in cattle improvement programs. Electronic supplementary material The online version of this article (10.1186/s12864-018-5124-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chad D Dechow
- Department of Animal Science, College of Agricultural Sciences, The Pennsylvania State University, 324 Henning Building, University Park, State College, PA, 16802, USA.
| | - Wan-Sheng Liu
- Department of Animal Science, College of Agricultural Sciences, The Pennsylvania State University, 324 Henning Building, University Park, State College, PA, 16802, USA
| |
Collapse
|
33
|
Luo H, Chen C, Guo L, Xu Z, Peng X, Wang X, Wang J, Wang N, Li C, Luo X, Wang H, Jose PA, Fu C, Huang Y, Shi W, Zeng C. Exposure to Maternal Diabetes Mellitus Causes Renal Dopamine D 1 Receptor Dysfunction and Hypertension in Adult Rat Offspring. Hypertension 2018; 72:962-970. [PMID: 30354705 PMCID: PMC6207228 DOI: 10.1161/hypertensionaha.118.10908] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/05/2018] [Indexed: 01/01/2023]
Abstract
Epidemiological and experimental studies suggest that maternal diabetes mellitus programs hypertension that is associated with impaired sodium excretion in the adult offspring. However, the underlying mechanisms are not clear. Because dopamine receptor function is involved in the pathogenesis of hypertension, we hypothesized that impaired renal dopamine D1 receptor function is also involved in the hypertension in offspring of maternal diabetes mellitus. Maternal diabetes mellitus was induced by a single intraperitoneal injection of streptozotocin (35 mg/kg) to pregnant Sprague-Dawley rats at day 0 of gestation. Compared with the offspring of mothers injected with citrate buffer (control mother offspring), the diabetic mother offspring (DMO) had increased systolic blood pressure and impaired D1 receptor-mediated diuresis and natriuresis, accompanied by increased renal PKC (protein kinase C) expression and activity, GRK-2 (G protein-coupled receptor kinase-2) expression, D1 receptor phosphorylation, D1 receptor/Gαs uncoupling, and loss of D1 receptor-mediated inhibition of Na+-K+-ATPase activity in renal proximal tubule cells from DMO. Inhibition of PKC reduced the increased GRK-2 expression and normalized D1 receptor function in primary cultures of renal proximal tubule cells from DMO. In addition, DMO, relative to control mother offspring, in vivo, had increased oxidative stress, indicated by decreased renal glutathione and increased renal malondialdehyde and urine 8-isoprostane. Normalization of oxidative stress with tempol also normalized the renal D1 receptor phosphorylation, D1 receptor-mediated diuresis and natriuresis, and blood pressure in DMO. Our present study indicates that maternal diabetes mellitus-programed hypertension in the offspring is caused by impaired renal D1 receptor function because of oxidative stress that is mediated by increased PKC-GRK-2 activity.
Collapse
Affiliation(s)
- Hao Luo
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Caiyu Chen
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Li Guo
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Zaicheng Xu
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoyu Peng
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xinquan Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Jialiang Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Na Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chuanwei Li
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoli Luo
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Hongyong Wang
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Pedro A. Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Chunjiang Fu
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yu Huang
- Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Sha Tin, Hong Kong, China
| | - Weibin Shi
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Chongqing Key Laboratory for Hypertension, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
34
|
Tzika E, Dreker T, Imhof A. Epigenetics and Metabolism in Health and Disease. Front Genet 2018; 9:361. [PMID: 30279699 PMCID: PMC6153363 DOI: 10.3389/fgene.2018.00361] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/21/2018] [Indexed: 01/09/2023] Open
Abstract
In the next 10 years, one billion people are estimated to suffer from disabling consequences of metabolic disorders, making them the number one noncommunicable disease on a global scale by 2030. Lots of risk factors such as dietary intake, lack of exercise and other life style behaviors are considered to play a role in the development of metabolic disorders. Despite the efforts that have been undertaken to unravel their potential causes, the underlying molecular mechanisms remain elusive. Evidence suggests that the pathogenesis involves changes on chromatin and chromatin-modifying enzymes, which can contribute to a persistent dysregulated metabolic phenotype. Indeed, a rising number of studies links epigenetic alterations with the diagnosis and prognosis of metabolic disorders. A prerequisite for exploiting these findings for pharmacological intervention is a detailed understanding of how differential epigenetic modifications control cell metabolism. In this mini review, we summarize the recent advances in uncovering the interplay between epigenetics and metabolic pathways on a cellular level and highlight potential new avenues for alternative treatment strategies.
Collapse
Affiliation(s)
- Evangelia Tzika
- 4SC AG, Translational Pharmacology, Munich, Germany.,Faculty of Medicine, Ludwig Maximilians University of Munich, Munich, Germany
| | | | - Axel Imhof
- Faculty of Medicine, Ludwig Maximilians University of Munich, Munich, Germany.,Protein Analysis Unit (ZfP), Biomedical Center, Ludwig Maximilians University of Munich, Munich, Germany
| |
Collapse
|
35
|
Rodrigo S, Fauste E, de la Cuesta M, Rodríguez L, Álvarez-Millán JJ, Panadero MI, Otero P, Bocos C. Maternal fructose induces gender-dependent changes in both LXRα promoter methylation and cholesterol metabolism in progeny. J Nutr Biochem 2018; 61:163-172. [PMID: 30236873 DOI: 10.1016/j.jnutbio.2018.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/03/2018] [Accepted: 08/24/2018] [Indexed: 12/25/2022]
Abstract
Fructose consumption from added sugars correlates with the epidemic rise in obesity, metabolic syndrome and cardiovascular diseases. However, consumption of beverages containing fructose is allowed during gestation. We have investigated whether maternal fructose intake produces subsequent changes in cholesterol metabolism of progeny. Carbohydrates were supplied to pregnant rats in drinking water (10% w/v solution) throughout gestation. Adult male and female descendants from fructose-fed, control or glucose-fed mothers were studied. Male offspring from fructose-fed mothers had elevated plasma HDL-cholesterol levels, whereas female progeny from fructose-fed mothers presented lower levels of non-HDL cholesterol vs. the other two groups. Liver X-receptor (LXR), an important regulator of cholesterol metabolism, and its target genes such as scavenger receptor B1, ATP-binding cassette (ABC)G5 and cholesterol 7-alpha hydroxylase showed decreased gene expression in males from fructose-fed mothers and the opposite in the female progeny. Moreover, the expression of a number of LXRα target genes related to lipogenesis paralleled to that for LXRα expression. In accordance with this, LXRα gene promoter methylation was increased in males from fructose-fed mothers and decreased in the corresponding group of females. Surprisingly, plasma folic acid levels, an important methyl-group donor, were augmented in males from fructose-fed mothers and diminished in female offspring. Maternal fructose intake produces a fetal programming that influences, in a gender-dependent manner, the transcription factor LXRα epigenetically, and both hepatic mRNA gene expression and plasma parameters of cholesterol metabolism in adult progeny. Changes in the LXRα promoter methylation might be related to the availability of the methyl donor folate.
Collapse
Affiliation(s)
- Silvia Rodrigo
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Elena Fauste
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Maite de la Cuesta
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Lourdes Rodríguez
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | | | - María I Panadero
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Paola Otero
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Carlos Bocos
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, Madrid, Spain.
| |
Collapse
|
36
|
Villegas-Romero M, Castrejón-Téllez V, Pérez-Torres I, Rubio-Ruiz ME, Carreón-Torres E, Díaz-Díaz E, Del Valle-Mondragón L, Guarner-Lans V. Short-Term Exposure to High Sucrose Levels near Weaning Has a Similar Long-Lasting Effect on Hypertension as a Long-Term Exposure in Rats. Nutrients 2018; 10:nu10060728. [PMID: 29882756 PMCID: PMC6024587 DOI: 10.3390/nu10060728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/31/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022] Open
Abstract
Adverse conditions during early developmental stages permanently modify the metabolic function of organisms through epigenetic changes. Exposure to high sugar diets during gestation and/or lactation affects susceptibility to metabolic syndrome or hypertension in adulthood. The effect of a high sugar diet for shorter time lapses remains unclear. Here we studied the effect of short-term sucrose ingestion near weaning (postnatal days 12 and 28) (STS) and its effect after long-term ingestion, for a period of seven months (LTS) in rats. Rats receiving sucrose for seven months develop metabolic syndrome (MS). The mechanisms underlying hypertension in this model and those that underlie the effects of short-term exposure have not been studied. We explore NO and endothelin-1 concentration, endothelial nitric oxide synthase (eNOS) expression, fatty acid participation and the involvement of oxidative stress (OS) after LTS and STS. Blood pressure increased to similar levels in adult rats that received sucrose during short- and long-term glucose exposure. The endothelin-1 concentration increased only in LTS rats. eNOS and SOD2 expression determined by Western blot and total antioxidant capacity were diminished in both groups. Saturated fatty acids and arachidonic acid were only decreased in LTS rats. In conclusion, a high-sugar diet during STS increases the hypertension predisposition in adulthood to as high a level as LTS, and the mechanisms involved have similarities (participation of OS and eNOS and SOD expression) and differences (fatty acids and arachidonic acid only participate in LTS and an elevated level of endothelin-1 was only found in LTS) in both conditions. Changes in the diet during short exposure times in early developmental stages have long-lasting effects in determining hypertension susceptibility.
Collapse
Affiliation(s)
- Mariana Villegas-Romero
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| | - Vicente Castrejón-Téllez
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| | - Israel Pérez-Torres
- Department of Pathology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| | - Eulises Díaz-Díaz
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Sección XVI, Tlalpan, Mexico City 14000, Mexico.
| | - Leonardo Del Valle-Mondragón
- Department of Pharmacology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| | - Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico.
| |
Collapse
|
37
|
Epigenetic Modifications Linked to T2D, the Heritability Gap, and Potential Therapeutic Targets. Biochem Genet 2018; 56:553-574. [DOI: 10.1007/s10528-018-9863-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/16/2018] [Indexed: 12/22/2022]
|
38
|
Maternal overnutrition programs epigenetic changes in the regulatory regions of hypothalamic Pomc in the offspring of rats. Int J Obes (Lond) 2018; 42:1431-1444. [PMID: 29777232 PMCID: PMC6113193 DOI: 10.1038/s41366-018-0094-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/27/2018] [Accepted: 03/16/2018] [Indexed: 02/02/2023]
Abstract
Background and objective Maternal overnutrition has been implicated in affecting the offspring by programming metabolic disorders such as obesity and diabetes, by mechanisms that are not clearly understood. This study aimed to determine the long-term impact of maternal high-fat (HF) diet feeding on epigenetic changes in the offspring’s hypothalamic Pomc gene, coding a key factor in the control of energy balance. Further, it aimed to study the additional effects of postnatal overnutrition on epigenetic programming by maternal nutrition. Methods Eight-week-old female Sprague–Dawley rats were fed HF diet or low-fat (LF) diet for 6 weeks before mating, and throughout gestation and lactation. At postnatal day 21, samples were collected from a third offspring and the remainder were weaned onto LF diet for 5 weeks, after which they were either fed LF or HF diet for 12 weeks, resulting in four groups of offspring differing by their maternal and postweaning diet. Results With maternal HF diet, offspring at weaning had rapid early weight gain, increased adiposity, and hyperleptinemia. The programmed adult offspring, subsequently fed LF diet, retained the increased body weight. Maternal HF diet combined with offspring HF diet caused more pronounced hyperphagia, fat mass, and insulin resistance. The ARC Pomc gene from programmed offspring at weaning showed hypermethylation in the enhancer (nPE1 and nPE2) regions and in the promoter sequence mediating leptin effects. Interestingly, hypermethylation at the Pomc promoter but not at the enhancer region persisted long term into adulthood in the programmed offspring. However, there were no additive effects on methylation levels in the regulatory regions of Pomc in programmed offspring fed a HF diet. Conclusion Maternal overnutrition programs long-term epigenetic alterations in the offspring’s hypothalamic Pomc promoter. This predisposes the offspring to metabolic disorders later in life.
Collapse
|
39
|
Affiliation(s)
- Andrew P Feinberg
- From the Department of Medicine, Johns Hopkins University School of Medicine, the Department of Biomedical Engineering, Whiting School of Engineering, and the Department of Mental Health, Johns Hopkins Bloomberg School of Public Health - all in Baltimore
| |
Collapse
|
40
|
Abstract
The common relationships among a great variety of biological phenomena seem enigmatic when considered solely at the level of the phenotype. The deep connections in physiology, for example, between the effects of maternal food restriction in utero and the subsequent incidence of metabolic syndrome in offspring, the effects of microgravity on cell polarity and reproduction in yeast, stress effects on jellyfish, and their endless longevity, or the relationship between nutrient abundance and the colonial form in slime molds, are not apparent by phenotypic observation. Yet all of these phenomena are ultimately determined by the Target of Rapamycin (TOR) gene and its associated signaling complexes. In the same manner, the unfolding of evolutionary physiology can be explained by a comparable application of the common principle of cell-cell signaling extending across complex developmental and phylogenetic traits. It is asserted that a critical set of physiologic and phenotypic adaptations emanated from a few crucial, ancestral receptor gene duplications that enabled the successful terrestrial transition of vertebrates from water to land. In combination, mTor and its cognate receptors and a few crucial genetic duplications provide a mechanistic common denominator across a diverse spectrum of biological responses. The proper understanding of their purpose yields a unified concept of physiology and its evolutionary development. © 2018 American Physiological Society. Compr Physiol 8:761-771, 2018.
Collapse
Affiliation(s)
- John S Torday
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | | |
Collapse
|
41
|
Pinos H, Carrillo B, Díaz F, Chowen JA, Collado P. Differential vulnerability to adverse nutritional conditions in male and female rats: Modulatory role of estradiol during development. Front Neuroendocrinol 2018; 48:13-22. [PMID: 28754628 DOI: 10.1016/j.yfrne.2017.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/07/2017] [Accepted: 07/23/2017] [Indexed: 01/21/2023]
Abstract
Many studies have shown the importance of an adequate nutritional environment during development to optimally establish the neurohormonal circuits that regulate feeding behavior. Under- or over-nutrition during early stages of life can lead to alterations in the physiology and brain networks that control food intake, resulting in a greater vulnerability to suffer maladjustments in energy metabolism in adulthood. These alterations produced by under- or over-nourishment during development differ between males and females, as does the modulatory action that estradiol exerts on the alterations produced by malnutrition. Estradiol regulates metabolism and brain metabolic circuits through the same transcription factor pathway, STAT3, that leptin and ghrelin use to program feeding circuits. Although more research is needed to disentangle the actual role of estradiol during development on the programming of feeding circuits, a synergistic role together with leptin and/or ghrelin might be hypothesized.
Collapse
Affiliation(s)
- Helena Pinos
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain
| | - Beatriz Carrillo
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain
| | - Francisca Díaz
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda. Menéndez Pelayo, N° 65, 28009 Madrid, Spain
| | - Julie A Chowen
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda. Menéndez Pelayo, N° 65, 28009 Madrid, Spain
| | - Paloma Collado
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain.
| |
Collapse
|
42
|
Nunes LM, Otero X. Quantification of health risks in Ecuadorian population due to dietary ingestion of arsenic in rice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:27457-27468. [PMID: 28980169 DOI: 10.1007/s11356-017-0265-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/19/2017] [Indexed: 06/07/2023]
Abstract
In Ecuador alone, 500,000 people in rural areas are estimated to have been exposed to high concentrations of As from water and food, but no quantitative evaluation of health risk has yet been made. The present study quantifies exposure and health risk for the Ecuadorian population from the ingestion of arsenic in white rice. Estimated exposure is correlated with published data on tap water quality and biomarkers of exposure for the population of two towns in the metropolitan area of Quito. Estimated daily intake (EDI) of arsenic for infants living in urban areas of Ecuador is around four times that of European infants, being equal for those livings in rural areas. EDI for the population as a whole is almost twice that of Europe, but between a half and a third of that of Brazil, Bangladesh, and India. Estimated excess lifetime risk (ELTR) for adults is 3 per 10,000, while for infants varies between 10 per 10,000 in rural areas and 20 per 10,000 in urban areas. Future research on arsenic impacts on human health in Ecuador should consider in particular poor populations living in regions where environmental arsenic concentrations are highest, including cross-sectional and longitudinal epidemiologic studies.
Collapse
Affiliation(s)
- Luís Miguel Nunes
- Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, Faro, Portugal.
- Civil Engineering Research and Innovation for Sustainability, Instituto Superior Técnico, Lisbon, Portugal.
| | - Xosé Otero
- Departamento de Edafoloxía e Química Agrícola, Facultade de Bioloxía, Universidade de Santiago de Compostela, Campus Sur, 15782, Santiago de Compostela, Spain
- Departamento de Ciencia de los Alimentos y Biotecnología, Escuela Politécnica Nacional, Quito, Ecuador
| |
Collapse
|
43
|
McGovern F, Boland T, Ryan M, Sweeney T. Assessment of RNA Stability in Postmortem Tissue from New-Born Lambs. Anim Biotechnol 2017; 29:269-275. [PMID: 29172984 DOI: 10.1080/10495398.2017.1378226] [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] [Indexed: 10/18/2022]
Abstract
The recovery of high quality RNA from postmortem tissue is crucial to gene expression analyses. The acquisition of postmortem tissue has inherent time delays and, hence, understanding the temporal variation in the stability of total RNA is imperative. This experiment aimed: ( 1 ) to qualitatively and quantitatively assess the integrity of total RNA derived from a range of new-born ovine tissues (liver, spleen, thyroid, skeletal muscle, ileum, and perirenal adipose tissue) which were stored at ambient temperature until extraction at 0, 3, 6, and 9 h postmortem; and ( 2 ) to analyze the stability of the reference gene(s) and expression of specific target genes in these tissues. Postmortem sampling time resulted in variable reductions in the relative integrity number (RIN) values across the tissues, ranging from 0.9 to 1.8% in liver, spleen, skeletal muscle, and ileum to 5.7-11.1% in the thyroid and perirenal adipose tissues, respectively (P < 0.05). In conclusion, tissues with small reductions in RIN value can exhibit disproportionately large differences in the normalization factor used to calculate the target gene expression. Hence, changes in transcript abundance due to RNA degradation are not always sufficiently buffered through normalization with reference genes. The normalization factor should be presented alongside the RIN value in postmortem tissue studies.
Collapse
Affiliation(s)
- Fiona McGovern
- a UCD School of Agriculture and Food Science , University College Dublin, Lyons Research Farm , Newcastle , Dublin , Ireland.,b UCD School of Veterinary Medicine , University College Dublin , Belfield , Dublin , Ireland
| | - Tommy Boland
- a UCD School of Agriculture and Food Science , University College Dublin, Lyons Research Farm , Newcastle , Dublin , Ireland
| | - Marion Ryan
- b UCD School of Veterinary Medicine , University College Dublin , Belfield , Dublin , Ireland
| | - Torres Sweeney
- b UCD School of Veterinary Medicine , University College Dublin , Belfield , Dublin , Ireland
| |
Collapse
|
44
|
Imam MU, Ismail M. The Impact of Traditional Food and Lifestyle Behavior on Epigenetic Burden of Chronic Disease. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700043. [PMID: 31565292 PMCID: PMC6607231 DOI: 10.1002/gch2.201700043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/12/2017] [Indexed: 05/11/2023]
Abstract
Noncommunicable chronic diseases (NCCDs) are the leading causes of morbidity and mortality globally. The mismatch between present day diets and ancestral genome is suggested to contribute to the NCCDs burden, which is promoted by traditional risk factors like unhealthy diets, physical inactivity, alcohol and tobacco. However, epigenetic evidence now suggests that cumulatively inherited epigenetic modifications may have made humans more prone to the effects of present day lifestyle factors. Perinatal starvation was widespread in the 19th century. This together with more recent events like increasing consumption of western and low fiber diets, smoking, harmful use of alcohol, physical inactivity, and environmental pollutants may have programed the human epigenome for higher NCCDs risk. In this review, on the basis of available epigenetic data it is hypothesized that transgenerational effects of lifestyle factors may be contributing to the current global burden of NCCDs. Thus, there is a need to reconsider prevention strategies so that the subsequent generations will not have to pay for our sins and those of our ancestors.
Collapse
Affiliation(s)
- Mustapha U. Imam
- Precision Nutrition Innovation InstituteCollege of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Maznah Ismail
- Laboratory of Molecular BiomedicineInstitute of BioscienceUniversiti Putra MalaysiaSerdangSelangor43400Malaysia
| |
Collapse
|
45
|
A maternal high-fat, high-sucrose diet alters insulin sensitivity and expression of insulin signalling and lipid metabolism genes and proteins in male rat offspring: effect of folic acid supplementation. Br J Nutr 2017; 118:580-588. [PMID: 29056104 DOI: 10.1017/s0007114517002501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A maternal high-fat, high-sucrose (HFS) diet alters offspring glucose and lipid homoeostasis through unknown mechanisms and may be modulated by folic acid. We investigated the effect of a maternal HFS diet on glucose homoeostasis, expression of genes and proteins associated with insulin signalling and lipid metabolism and the effect of prenatal folic acid supplementation (HFS/F) in male rat offspring. Pregnant Sprague-Dawley rats were randomly fed control (CON), HFS or HFS/F diets. Offspring were weaned on CON; at postnatal day 70, fasting plasma insulin and glucose and liver and skeletal muscle gene and protein expression were measured. Treatment effects were assessed by one-way ANOVA. Maternal HFS diet induced higher fasting glucose in offspring v. HFS/F (P=0·027) and down-regulation (P<0·05) of genes coding for v-Akt murine thymoma viral oncogene homolog 2, resistin and v-Raf-1 murine leukaemia viral oncogene homolog 1 (Raf1) in offspring skeletal muscle and acetyl-CoA carboxylase (Acaca), fatty acid synthase and phosphatidylinositol-4,5-biphosphate 3-kinase, catalytic subunit β in offspring liver. Skeletal muscle neuropeptide Y and hepatic Kruppel-like factor 10 were up-regulated in HFS v. CON offspring (P<0·05). Compared with CON, Acaca and Raf1 protein expression levels were significantly lower in HFS offspring. Maternal HFS induced higher homoeostasis model of assessment index of insulin resistance v. CON (P=0·030) and HFS/F was associated with higher insulin (P=0·016) and lower glucose (P=0·025). Maternal HFS diet alters offspring insulin sensitivity and de novo hepatic lipogenesis via altered gene and protein expression, which appears to be potentiated by folate supplementation.
Collapse
|
46
|
Oelkrug R, Herrmann B, Geissler C, Harder L, Koch C, Lehnert H, Oster H, Kirchner H, Mittag J. Dwarfism and insulin resistance in male offspring caused by α1-adrenergic antagonism during pregnancy. Mol Metab 2017; 6:1126-1136. [PMID: 29031714 PMCID: PMC5641602 DOI: 10.1016/j.molmet.2017.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Maternal and environmental factors control the epigenetic fetal programming of the embryo, thereby defining the susceptibility for metabolic or endocrine disorders in the offspring. Pharmacological interventions required as a consequence of gestational problems, e.g. hypertension, can potentially interfere with correct fetal programming. As epigenetic alterations are usually only revealed later in life and not detected in studies focusing on early perinatal outcomes, little is known about the long-term epigenetic effects of gestational drug treatments. We sought to test the consequences of maternal α1-adrenergic antagonism during pregnancy, which can occur e.g. during hypertension treatment, for the endocrine and metabolic phenotype of the offspring. METHODS We treated C57BL/6NCrl female mice with the α1-adrenergic antagonist prazosin during pregnancy and analyzed the male and female offspring for endocrine and metabolic abnormalities. RESULTS Our data revealed that maternal α1-adrenergic blockade caused dwarfism, elevated body temperature, and insulin resistance in male offspring, accompanied by reduced IGF-1 serum concentrations as the result of reduced hepatic growth hormone receptor (Ghr) expression. We subsequently identified increased CpG DNA methylation at the transcriptional start site of the alternative Ghr promotor caused by the maternal treatment, which showed a strong inverse correlation to hepatic Ghr expression. CONCLUSIONS Our results demonstrate that maternal α1-adrenergic blockade can constitute an epigenetic cause for dwarfism and insulin resistance. The findings are of immediate clinical relevance as combined α/β-adrenergic blockers are first-line treatment of maternal hypertension.
Collapse
Affiliation(s)
- Rebecca Oelkrug
- Department of Molecular Endocrinology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Beate Herrmann
- Department of Molecular Endocrinology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Cathleen Geissler
- Department of Epigenetics & Metabolism/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Lisbeth Harder
- Department of Molecular Endocrinology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Christiane Koch
- Department of Chronophysiology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Hendrik Lehnert
- Department of Experimental Neuroendocrinology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Henrik Oster
- Department of Chronophysiology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Henriette Kirchner
- Department of Epigenetics & Metabolism/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Jens Mittag
- Department of Molecular Endocrinology/CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
47
|
Seki Y, Suzuki M, Guo X, Glenn AS, Vuguin PM, Fiallo A, Du Q, Ko YA, Yu Y, Susztak K, Zheng D, Greally JM, Katz EB, Charron MJ. In Utero Exposure to a High-Fat Diet Programs Hepatic Hypermethylation and Gene Dysregulation and Development of Metabolic Syndrome in Male Mice. Endocrinology 2017; 158:2860-2872. [PMID: 28911167 PMCID: PMC5659663 DOI: 10.1210/en.2017-00334] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/16/2017] [Indexed: 12/12/2022]
Abstract
Exposure to a high-fat (HF) diet in utero is associated with increased incidence of cardiovascular disease, diabetes, and metabolic syndrome later in life. However, the molecular basis of this enhanced susceptibility for metabolic disease is poorly understood. Gene expression microarray and genome-wide DNA methylation analyses of mouse liver revealed that exposure to a maternal HF milieu activated genes of immune response, inflammation, and hepatic dysfunction. DNA methylation analysis revealed 3360 differentially methylated loci, most of which (76%) were hypermethylated and distributed preferentially to hotspots on chromosomes 4 [atherosclerosis susceptibility quantitative trait loci (QTLs) 1] and 18 (insulin-dependent susceptibility QTLs 21). Interestingly, we found six differentially methylated genes within these hotspot QTLs associated with metabolic disease that maintain altered gene expression into adulthood (Arhgef19, Epha2, Zbtb17/Miz-1, Camta1 downregulated; and Ccdc11 and Txnl4a upregulated). Most of the hypermethylated genes in these hotspots are associated with cardiovascular system development and function. There were 140 differentially methylated genes that showed a 1.5-fold increase or decrease in messenger RNA levels. Many of these genes play a role in cell signaling pathways associated with metabolic disease. Of these, metalloproteinase 9, whose dysregulation plays a key role in diabetes, obesity, and cardiovascular disease, was upregulated 1.75-fold and hypermethylated in the gene body. In summary, exposure to a maternal HF diet causes DNA hypermethylation, which is associated with long-term gene expression changes in the liver of exposed offspring, potentially contributing to programmed development of metabolic disease later in life.
Collapse
Affiliation(s)
- Yoshinori Seki
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Masako Suzuki
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Xingyi Guo
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Alan Scott Glenn
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Patricia M. Vuguin
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Ariana Fiallo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Yi-An Ko
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Yiting Yu
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - John M. Greally
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Ellen B. Katz
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Maureen J. Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
- Departments of Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
48
|
Modulatory Mechanism of Polyphenols and Nrf2 Signaling Pathway in LPS Challenged Pregnancy Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8254289. [PMID: 29138679 PMCID: PMC5613688 DOI: 10.1155/2017/8254289] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/16/2017] [Indexed: 12/16/2022]
Abstract
Early embryonic loss and adverse birth outcomes are the major reproductive disorders that affect both human and animals. The LPS induces inflammation by interacting with robust cellular mechanism which was considered as a plethora of numerous reproductive disorders such as fetal resorption, preterm birth, teratogenicity, intrauterine growth restriction, abortion, neural tube defects, fetal demise, and skeletal development retardation. LPS-triggered overproduction of free radicals leads to oxidative stress which mediates inflammation via stimulation of NF-κB and PPARγ transcription factors. Flavonoids, which exist in copious amounts in nature, possess a wide array of functions; their supplementation during pregnancy activates Nrf2 signaling pathway which encounters pregnancy disorders. It was further presumed that the development of strong antioxidant uterine environment during gestation can alleviate diseases which appear at adult stages. The purpose of this review is to focus on modulatory properties of flavonoids on oxidative stress-mediated pregnancy insult and abnormal outcomes and role of Nrf2 activation in pregnancy disorders. These findings would be helpful for providing new insights in ameliorating oxidative stress-induced pregnancy disorders.
Collapse
|
49
|
Cesar HC, Pisani LP. Fatty-acid-mediated hypothalamic inflammation and epigenetic programming. J Nutr Biochem 2017; 42:1-6. [DOI: 10.1016/j.jnutbio.2016.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/09/2016] [Accepted: 08/10/2016] [Indexed: 01/21/2023]
|
50
|
Raciti GA, Spinelli R, Desiderio A, Longo M, Parrillo L, Nigro C, D'Esposito V, Mirra P, Fiory F, Pilone V, Forestieri P, Formisano P, Pastan I, Miele C, Beguinot F. Specific CpG hyper-methylation leads to Ankrd26 gene down-regulation in white adipose tissue of a mouse model of diet-induced obesity. Sci Rep 2017; 7:43526. [PMID: 28266632 PMCID: PMC5339897 DOI: 10.1038/srep43526] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/27/2017] [Indexed: 12/16/2022] Open
Abstract
Epigenetic modifications alter transcriptional activity and contribute to the effects of environment on the individual risk of obesity and Type 2 Diabetes (T2D). Here, we have estimated the in vivo effect of a fat-enriched diet (HFD) on the expression and the epigenetic regulation of the Ankyrin repeat domain 26 (Ankrd26) gene, which is associated with the onset of these disorders. In visceral adipose tissue (VAT), HFD exposure determined a specific hyper-methylation of Ankrd26 promoter at the −436 and −431 bp CpG sites (CpGs) and impaired its expression. Methylation of these 2 CpGs impaired binding of the histone acetyltransferase/transcriptional coactivator p300 to this same region, causing hypo-acetylation of histone H4 at the Ankrd26 promoter and loss of binding of RNA Pol II at the Ankrd26 Transcription Start Site (TSS). In addition, HFD increased binding of DNA methyl-transferases (DNMTs) 3a and 3b and methyl-CpG-binding domain protein 2 (MBD2) to the Ankrd26 promoter. More importantly, Ankrd26 down-regulation enhanced secretion of pro-inflammatory mediators by 3T3-L1 adipocytes as well as in human sera. Thus, in mice, the exposure to HFD induces epigenetic silencing of the Ankrd26 gene, which contributes to the adipose tissue inflammatory secretion profile induced by high-fat regimens.
Collapse
Affiliation(s)
- Gregory A Raciti
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Rosa Spinelli
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Antonella Desiderio
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Michele Longo
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Luca Parrillo
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Cecilia Nigro
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Vittoria D'Esposito
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Paola Mirra
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesca Fiory
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Vincenzo Pilone
- Bariatric and Metabolic Surgery Unit, University of Salerno, Salerno, 84084, Italy
| | - Pietro Forestieri
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Pietro Formisano
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Ira Pastan
- Laboratory of Molecular Biology (LMB), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Claudia Miele
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesco Beguinot
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|