1
|
Na ES. Epigenetic Mechanisms of Obesity: Insights from Transgenic Animal Models. Life (Basel) 2025; 15:653. [PMID: 40283207 PMCID: PMC12028693 DOI: 10.3390/life15040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Obesity is a chronic disease with prevalence rates that have risen dramatically over the past four decades. This increase is not due to changes in the human genome but rather to environmental factors that promote maladaptive physiological responses. Emerging evidence suggests that external influences, such as high-fat diets, modify the epigenome-the interface between genes and the environment-leading to persistent alterations in energy homeostasis. This review explores the role of epigenetic mechanisms in obesity, emphasizing insights from transgenic animal models and clinical studies. Additionally, we discuss the evolution of obesity research from homeostatic to allostatic frameworks, highlighting key neuroendocrine regulators of energy balance.
Collapse
Affiliation(s)
- Elisa S Na
- School of Social Work, Psychology, & Philosophy, Texas Woman's University, Denton, TX 76209, USA
| |
Collapse
|
2
|
Jiao P, Lu H, Hao L, Degen AA, Cheng J, Yin Z, Mao S, Xue Y. Nutrigenetic and Epigenetic Mechanisms of Maternal Nutrition-Induced Glucolipid Metabolism Changes in the Offspring. Nutr Rev 2025; 83:728-748. [PMID: 38781288 DOI: 10.1093/nutrit/nuae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Maternal nutrition during pregnancy regulates the offspring's metabolic homeostasis, including insulin sensitivity and the metabolism of glucose and lipids. The fetus undergoes a crucial period of plasticity in the uterus; metabolic changes in the fetus during pregnancy caused by maternal nutrition not only influence fetal growth and development but also have a long-term or even life-long impact for the offspring. Epigenetic modifications, such as DNA methylation, histone modification, and non-coding RNAs, play important roles in intergenerational and transgenerational effects. In this context, this narrative review comprehensively summarizes and analyzes the molecular mechanisms underlying how maternal nutrition, including a high-fat diet, polyunsaturated fatty acid diet, methyl donor nutrient supplementation, feed restriction, and protein restriction during pregnancy, impacts the genes involved in glucolipid metabolism in the liver, adipose tissue, hypothalamus, muscle, and oocytes of the offspring in terms of the epigenetic modifications. This will provide a foundation for the further exploration of nutrigenetic and epigenetic mechanisms for integrative mother-child nutrition and promotion of the offspring's health through the regulation of maternal nutrition during pregnancy. Note: This paper is part of the Nutrition Reviews Special Collection on Precision Nutrition.
Collapse
Affiliation(s)
- Peng Jiao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Huizhen Lu
- Biotechnology Center, Anhui Agricultural University, Hefei, China
| | - Lizhuang Hao
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Plateau Yak Research Center, Qinghai Academy of Science and Veterinary Medicine of Qinghai University, Xining, China
| | - A Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jianbo Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Shengyong Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yanfeng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
3
|
Rodrigues FDS, Jantsch J, de Farias Fraga G, Dias VS, Pereira Medeiros C, Wickert F, Schroder N, Giovernardi M, Guedes RP. Cannabidiol partially rescues behavioral, neuroinflammatory and endocannabinoid dysfunctions stemming from maternal obesity in the adult offspring. Neuropharmacology 2025; 262:110196. [PMID: 39447736 DOI: 10.1016/j.neuropharm.2024.110196] [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: 05/21/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024]
Abstract
Maternal obesity is known to increase the risk of psychiatric disorders, such as anxiety, depression, schizophrenia and autism spectrum disorder in the offspring. While preventive measures are well-documented, practical approaches for addressing the damages once they are already established are limited. We have recently demonstrated the interplay between maternal obesity and treatment with cannabidiol (CBD) on neuroinflammation and peripheral metabolic disturbances during adolescence, however, it is known that both factors tend to vary throughout life. Therefore, here we investigated the potential of CBD to mitigate these alterations in the adult offspring of obese dams. Female Wistar rats were fed a cafeteria diet for 12 weeks prior to mating, and during gestation and lactation. Offspring received CBD (50 mg/kg) for 3 weeks from the 70th day of life. Behavioral tests assessed anxiety-like manifestations and social behavior, while neuroinflammatory and endocannabinoid markers were evaluated in the hypothalamus, prefrontal cortex (PFC) and hippocampus, as well as the biochemical profile in the plasma. CBD treatment attenuated maternal obesity-induced anxiety-like and social behavioral alterations, restoring exacerbated astrocytic and microglial markers in the hypothalamus, PFC and hippocampus of the offspring, as well as endocannabinoid levels in the PFC, with notable sex differences. Additionally, CBD attenuated plasma glucose and lipopolysaccharides (LPS) concentrations in females. These findings underscore the persistent influence of maternal obesity on the offspring's health, encompassing metabolic irregularities and behavioral impairments, as well as the role of the endocannabinoid system in mediating these outcomes across the lifespan.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Pereira Medeiros
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Wickert
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Nadja Schroder
- Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcia Giovernardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Rio Grande do Sul, Brazil.
| |
Collapse
|
4
|
El Sharkawy M, Felix JF, Grote V, Voortman T, Jaddoe VWV, Koletzko B, Küpers LK. Animal and plant protein intake during infancy and childhood DNA methylation: a meta-analysis in the NutriPROGRAM consortium. Epigenetics 2024; 19:2299045. [PMID: 38198623 PMCID: PMC10793674 DOI: 10.1080/15592294.2023.2299045] [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: 03/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Higher early-life animal protein intake is associated with a higher childhood obesity risk compared to plant protein intake. Differential DNA methylation may represent an underlying mechanism. METHODS We analysed associations of infant animal and plant protein intakes with DNA methylation in early (2-6 years, N = 579) and late (7̄-12 years, N = 604) childhood in two studies. Study-specific robust linear regression models adjusted for relevant confounders were run, and then meta-analysed using a fixed-effects model. We also performed sex-stratified meta-analyses. Follow-up analyses included pathway analysis and eQTM look-up. RESULTS Infant animal protein intake was not associated with DNA methylation in early childhood, but was associated with late-childhood DNA methylation at cg21300373 (P = 4.27 × 10¯8, MARCHF1) and cg10633363 (P = 1.09 × 10¯7, HOXB9) after FDR correction. Infant plant protein intake was associated with early-childhood DNA methylation at cg25973293 (P = 2.26 × 10-7, C1orf159) and cg15407373 (P = 2.13 × 10-7, MBP) after FDR correction. There was no overlap between the findings from the animal and plant protein analyses. We did not find enriched functional pathways at either time point using CpGs associated with animal and plant protein. These CpGs were not previously associated with childhood gene expression. Sex-stratified meta-analyses showed sex-specific DNA methylation associations for both animal and plant protein intake. CONCLUSION Infant animal protein intake was associated with DNA methylation at two CpGs in late childhood. Infant plant protein intake was associated with DNA methylation in early childhood at two CpGs. A potential mediating role of DNA methylation at these CpGs between infant protein intake and health outcomes requires further investigation.
Collapse
Affiliation(s)
- Mohammed El Sharkawy
- Division of Metabolic and Nutritional Medicine, Department of Pediatrics, Dr. Von Hauner Children’s Hospital, LMU University Hospital Munich, Munich, Germany
- Munich Medical Research School, Faculty of Medicine, LMU - Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Janine F. Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Veit Grote
- Division of Metabolic and Nutritional Medicine, Department of Pediatrics, Dr. Von Hauner Children’s Hospital, LMU University Hospital Munich, Munich, Germany
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W. V. Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Department of Pediatrics, Dr. Von Hauner Children’s Hospital, LMU University Hospital Munich, Munich, Germany
| | - Leanne K. Küpers
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
5
|
Zhang L, Zhang S, Zou W, Hu Y, Gao Y, Zhang J, Zheng J. Maternal high-fat diet regulates offspring hepatic ABCG5 expression and cholesterol metabolism via the gut microbiota and its derived butyrate. Clin Sci (Lond) 2024; 138:1039-1054. [PMID: 39136693 DOI: 10.1042/cs20240997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
Maternal high-fat diet intake has profound effects on the long-term health of offspring, predisposing them to a higher susceptibility to obesity and metabolic dysfunction-associated steatotic liver disease. However, the detailed mechanisms underlying the role of a maternal high-fat diet in hepatic lipid accumulation in offspring, especially at the weaning age, remain largely unclear. In this study, female C57BL/6J mice were randomly assigned to either a high-fat diet or a control diet, and lipid metabolism parameters were assessed in male offspring at weaning. Gut microbiota analysis and targeted metabolomics of short-chain fatty acids (SCFAs) in these offspring were further performed. Both in vivo and in vitro studies were conducted to explore the role of butyrate in hepatic cholesterol excretion in the liver and HepG2 cells. Our results showed that maternal high-fat feeding led to obesity and dyslipidemia, and exacerbated hepatic lipid accumulation in the livers of offspring at weaning. We observed significant decreases in the abundance of the Firmicutes phylum and the Allobaculum genus, known as producers of SCFAs, particularly butyrate, in the offspring of dams fed a high-fat diet. Additionally, maternal high-fat diet feeding markedly decreased serum butyrate levels and down-regulated ATP-binding cassette transporters G5 (ABCG5) in the liver, accompanied by decreased phosphorylated AMP-activated protein kinase (AMPK) and histone deacetylase 5 (HADC5) expressions. Subsequent in vitro studies revealed that butyrate could induce ABCG5 activation and alleviate lipid accumulation via the AMPK-pHDAC5 pathway in HepG2 cells. Moreover, knockdown of HDAC5 up-regulated ABCG5 expression and promoted cholesterol excretion in HepG2 cells. In conclusion, our study provides novel insights into how maternal high-fat diet feeding inhibits hepatic cholesterol excretion and down-regulates ABCG5 through the butyrate-AMPK-pHDAC5 pathway in offspring at weaning.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Shixuan Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Wenyu Zou
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing 100034, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
6
|
Alameddine M, Altinpinar AE, Ersoy U, Kanakis I, Myrtziou I, Ozanne SE, Goljanek-Whysall K, Vasilaki A. Effect of Lactational Low-Protein Diet on Skeletal Muscle during Adulthood and Ageing in Male and Female Mouse Offspring. Nutrients 2024; 16:2926. [PMID: 39275242 PMCID: PMC11397042 DOI: 10.3390/nu16172926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Sarcopenia is characterised by the loss of skeletal muscle mass and function, which leads to a high risk of increased morbidity and mortality. Maternal malnutrition has been linked to impaired development of skeletal muscle of the offspring; however, there are limited studies that report the long-term effect of a maternal low-protein diet during lactation on the ageing of skeletal muscles. This study aimed to examine how a maternal low-protein diet (LPD) during lactation affects skeletal muscle ageing in the offspring. Pups born from control mothers were lactated by mothers fed with an LPD. Post-weaning, mice were either maintained on an LPD or switched to a control, normal-protein diet (NPD). In males, an LPD mainly affected the size of the myofibres without a major effect on fibre number and led to reduced grip strength in ageing mice (24 months). Female mice from mothers on an LPD had a lower body and muscle weight at weaning but caught up with control mice at 3 months. During ageing, the muscle weight, myofibre number and survival rate of female pups were significantly affected. These findings highlight the effect of an LPD during lactation on skeletal muscle ageing, the lifespan of offspring and the importance of sexual dimorphism in response to dietary challenges.
Collapse
Affiliation(s)
- Moussira Alameddine
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Atilla Emre Altinpinar
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK
| | - Ufuk Ersoy
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK
| | - Ioannis Kanakis
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK
- Chester Medical School, Faculty of Health, Medicine and Society, University of Chester, Chester CH1 4BJ, UK
| | - Ioanna Myrtziou
- Chester Medical School, Faculty of Health, Medicine and Society, University of Chester, Chester CH1 4BJ, UK
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Welcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge CB2 0QQ, UK
| | - Katarzyna Goljanek-Whysall
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK
- Department of Physiology, School of Medicine and REMEDI, CMNHS, University of Galway, H91 TK33 Galway, Ireland
| | - Aphrodite Vasilaki
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
7
|
Pang W, Zhang B, Zhang J, Chen T, Han Q, Yang Z. Effects of maternal advanced lipoxidation end products diet on the glycolipid metabolism and gut microbiota in offspring mice. Front Nutr 2024; 11:1421848. [PMID: 38962449 PMCID: PMC11220281 DOI: 10.3389/fnut.2024.1421848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/30/2024] [Indexed: 07/05/2024] Open
Abstract
Introduction Dietary advanced lipoxidation end products (ALEs), which are abundant in heat-processed foods, could induce lipid metabolism disorders. However, limited studies have examined the relationship between maternal ALEs diet and offspring health. Methods To investigate the transgenerational effects of ALEs, a cross-generation mouse model was developed. The C57BL/6J mice were fed with dietary ALEs during preconception, pregnancy and lactation. Then, the changes of glycolipid metabolism and gut microbiota of the offspring mice were analyzed. Results Maternal ALEs diet not only affected the metabolic homeostasis of dams, but also induced hepatic glycolipid accumulation, abnormal liver function, and disturbance of metabolism parameters in offspring. Furthermore, maternal ALEs diet significantly upregulated the expression of TLR4, TRIF and TNF-α proteins through the AMPK/mTOR/PPARα signaling pathway, leading to dysfunctional glycolipid metabolism in offspring. In addition, 16S rRNA analysis showed that maternal ALEs diet was capable of altered microbiota composition of offspring, and increased the Firmicutes/Bacteroidetes ratio. Discussion This study has for the first time demonstrated the transgenerational effects of maternal ALEs diet on the glycolipid metabolism and gut microbiota in offspring mice, and may help to better understand the adverse effects of dietary ALEs.
Collapse
Affiliation(s)
- Wenwen Pang
- School of Medicine, Nankai University, Tianjin, China
| | - Bowei Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Junshi Zhang
- Department of Hematology, Oncology Center, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Tianyi Chen
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiurong Han
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Yang
- Department of Clinical Laboratory, Tianjin Union Medical Center, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Yin J, Zhang N, Feng Y, Meng Q, Zhang T, Hong R, Zhang X. Exposure to famine during early life and the risk of MAFLD during adulthood: evidence from the China Multi-Ethnic Cohort (CMEC) study. BMJ PUBLIC HEALTH 2024; 2:e000114. [PMID: 40018122 PMCID: PMC11812743 DOI: 10.1136/bmjph-2023-000114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 03/04/2024] [Indexed: 03/01/2025]
Abstract
Background Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common chronic liver disease, affecting nearly one-third of the global population. The relationship between early-life famine exposure and MAFLD remains unclear in the multiethnic region of less-developed southwest China. Methods A total of 18 558 participants who came from the baseline survey of the China Multi-Ethnic Cohort Study in Yunnan were included. Participants were divided into four groups according to birth year, including non-exposed (1962-1978 and 1939-1943), fetal exposed (1959-1961), childhood exposed (1949-1958) and adolescence exposed (1943-1949). Logistic regression analysis was used to explore the relationship between famine exposure in early life and the risk of MAFLD in adulthood. Results Experiencing the shock of early-life exposure to famine would affect adulthood MAFLD. Exposure to famine during fetal life and childhood increased the risk of MAFLD in adulthood, with this association being particularly pronounced in Bai populations. Moreover, famine exposure in males during fetal life raised the risk of MAFLD in adulthood. Conclusion We suggest that adequate nutrition in early life may be beneficial in preventing MAFLD in adulthood. The prevention of chronic liver disease should adopt a whole-life strategy by extending the prevention window beginning from fetal life.
Collapse
Affiliation(s)
- Jianzhong Yin
- Baoshan College of Traditional Chinese Medicine, Baoshan, Yunnan, China
- Kunming Medical University, Kunming, Yunnan, China
| | - Nichang Zhang
- Baoshan College of Traditional Chinese Medicine, Baoshan, Yunnan, China
| | - Yuemei Feng
- Kunming Medical University, Kunming, Yunnan, China
| | - Qiong Meng
- Kunming Medical University, Kunming, Yunnan, China
| | - Teng Zhang
- Baoshan College of Traditional Chinese Medicine, Baoshan, Yunnan, China
| | - Rudan Hong
- Kunming Medical University, Kunming, Yunnan, China
| | - Xuehui Zhang
- Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
9
|
Li Y, Chen Y, Lin B, Liu Z, Xu F, Song D, Che K, Chen D, Su Y, Li W, Xu W. A maternal low-protein diet impaired glucose metabolism and altered the lncRNA profiles of islets in adult offspring. J Nutr Biochem 2024; 128:109618. [PMID: 38462210 DOI: 10.1016/j.jnutbio.2024.109618] [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: 11/21/2023] [Revised: 02/10/2024] [Accepted: 03/05/2024] [Indexed: 03/12/2024]
Abstract
A maternal low-protein diet during pregnancy can increase children's susceptibility to diabetes mellitus in adulthood. However, whether long noncoding RNAs (lncRNAs) in islets participate in the development of diabetes in adult offspring following maternal protein restriction is not fully understood. Female mice were fed a low-protein (LP) diet or control diet throughout gestation and lactation. The male offspring were then randomly divided into two groups according to maternal diet: offspring from control diet group dams (Ctrl group) and offspring from LP group dams (LP group). We observed the glucose metabolism of adult offspring. A lncRNA microarray was constructed for the islets from the LP group and Ctrl group to explore the differently expressed lncRNAs. Gene ontology enrichment and Kyoto Encyclopedia of Genes and Genomes analyses were subsequently used to predict the functions of the differently expressed lncRNAs. The body weight from birth to 12 weeks of age was significantly lower in the LP offspring. Adult LP offspring exhibited impaired glucose tolerance and decreased insulin secretion, consistent with the reduction in β-cell proliferation. According to the lncRNA microarray, four lncRNAs, three upregulated lncRNAs, and one downregulated lncRNA were differently expressed in LP offspring islets compared with Ctrl offspring. Gene ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that these differentially expressed lncRNAs were mostly associated with the hypoxia-inducible factor-1α signaling pathway. Additionally, we validated the expression of these four differentially expressed lncRNAs via quantitative real-time polymerase chain reaction. Our findings demonstrated the expression patterns of lncRNAs in islets from adult offspring of mothers who consumed a maternal low-protein diet.
Collapse
Affiliation(s)
- Yanli Li
- Department of Endocrinology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yalan Chen
- Key Laboratory of Diabetology of Guangdong Province, Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Beisi Lin
- Key Laboratory of Diabetology of Guangdong Province, Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ziyu Liu
- Department of Rheumatology, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fen Xu
- Key Laboratory of Diabetology of Guangdong Province, Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dalong Song
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kanshou Che
- Guangzhou Medical University, Guangzhou, China
| | - Danrui Chen
- Key Laboratory of Diabetology of Guangdong Province, Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanna Su
- Key Laboratory of Diabetology of Guangdong Province, Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wangen Li
- Department of Endocrinology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Wen Xu
- Key Laboratory of Diabetology of Guangdong Province, Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
10
|
Pérez-Coria M, Vázquez-Rivera GE, Gómez-García EF, Mendoza-Carrera F. Sex differences in fetal kidney reprogramming: the case in the renin-angiotensin system. Pediatr Nephrol 2024; 39:645-653. [PMID: 37572115 DOI: 10.1007/s00467-023-06112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/14/2023]
Abstract
During the early stages of the development of the living multiorgan systems, genome modifications other than sequence variation occur that guide cell differentiation and organogenesis. These modifications are known to operate as a fetal programming code during this period, and recent research indicates that there are some tissue-specific codes in organogenesis whose effects may persist after birth until adulthood. Consequently, the events that disrupt the pre-established epigenetic pattern could induce shifts in organ physiology, with implications on health from birth or later in adult life. Chronic kidney disease (CKD) is one of the main causes of mortality worldwide; its etiology is multifactorial, but diabetes, obesity, and hypertension are the main causes of CKD in adults, although there are other risk factors that are mainly associated with an individual's lifestyle. Recent studies suggest that fetal reprogramming in the developing kidney could be implicated in the susceptibility to kidney disease in both childhood and adulthood. Some epigenetic modifications, such as genome methylation status, dysregulation of miRNA, and histone coding alterations in genes related to the regulation of the renin-angiotensin axis, a common denominator in CKD, may have originated during fetal development. This review focuses on epigenetic changes during nephrogenesis and their repercussions on kidney health and disease. In addition, the focus is on the influence of environmental factors during pregnancy, such as maternal metabolic diseases and dietary and metabolic conditions, as well as some sex differences in fetal kidney reprogramming during which dysregulation of the renin-angiotensin system is involved.
Collapse
Affiliation(s)
- Mariana Pérez-Coria
- Molecular Medicine Division, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada # 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Gloria Elizabeth Vázquez-Rivera
- Molecular Medicine Division, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada # 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Erika Fabiola Gómez-García
- Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Tijuana, Baja California, Mexico
| | - Francisco Mendoza-Carrera
- Molecular Medicine Division, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada # 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
11
|
Elgazzaz M, Berdasco C, Garai J, Baddoo M, Lu S, Daoud H, Zabaleta J, Mauvais-Jarvis F, Lazartigues E. Maternal Western diet programs cardiometabolic dysfunction and hypothalamic inflammation via epigenetic mechanisms predominantly in the male offspring. Mol Metab 2024; 80:101864. [PMID: 38159883 PMCID: PMC10806294 DOI: 10.1016/j.molmet.2023.101864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE Maternal exposure during pregnancy is a strong determinant of offspring health outcomes. Such exposure induces changes in the offspring epigenome resulting in gene expression and functional changes. In this study, we investigated the effect of maternal Western hypercaloric diet (HCD) programming during the perinatal period on neuronal plasticity and cardiometabolic health in adult offspring. METHODS C57BL/6J dams were fed HCD for 1 month prior to mating with regular diet (RD) sires and kept on the same diet throughout pregnancy and lactation. At weaning, offspring were maintained on either HCD or RD for 3 months resulting in 4 treatment groups that underwent cardiometabolic assessments. DNA and RNA were extracted from the hypothalamus to perform whole genome methylation, mRNA, and miRNA sequencing followed by bioinformatic analyses. RESULTS Maternal programming resulted in male-specific hypertension and hyperglycemia, with both males and females showing increased sympathetic tone to the vasculature. Surprisingly, programmed male offspring fed HCD in adulthood exhibited lower glucose levels, less insulin resistance, and leptin levels compared to non-programmed HCD-fed male mice. Hypothalamic genes involved in inflammation and type 2 diabetes were targeted by differentially expressed miRNA, while genes involved in glial and astrocytic differentiation were differentially methylated in programmed male offspring. These data were supported by our findings of astrogliosis, microgliosis and increased microglial activation in programmed males in the paraventricular nucleus (PVN). Programming induced a protective effect in male mice fed HCD in adulthood, resulting in lower protein levels of hypothalamic TGFβ2, NF-κB2, NF-κBp65, Ser-pIRS1, and GLP1R compared to non-programmed HCD-fed males. Although TGFβ2 was upregulated in male mice exposed to HCD pre- or post-natally, only blockade of the brain TGFβ receptor in RD-HCD mice improved glucose tolerance and a trend to weight loss. CONCLUSIONS Our study shows that maternal HCD programs neuronal plasticity in the offspring and results in male-specific hypertension and hyperglycemia associated with hypothalamic inflammation in mechanisms and pathways distinct from post-natal HCD exposure. Together, our data unmask a compensatory role of HCD programming, likely via priming of metabolic pathways to handle excess nutrients in a more efficient way.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA; Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Clara Berdasco
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Jone Garai
- Department of Interdisciplinary Oncology and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine/Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shiping Lu
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hisham Daoud
- School of Computer and Cyber Sciences, Augusta University, Augusta, GA 30901, USA
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Franck Mauvais-Jarvis
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA; Department of Medicine, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Eric Lazartigues
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA; Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
12
|
Zhang L, Zou W, Hu Y, Wu H, Gao Y, Zhang J, Zheng J. Maternal high-calorie diet feeding programs hepatic cholesterol metabolism and Abca1 promoter methylation in the early life of offspring. J Nutr Biochem 2023; 122:109449. [PMID: 37748622 DOI: 10.1016/j.jnutbio.2023.109449] [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: 01/30/2023] [Revised: 07/19/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023]
Abstract
Maternal high-calorie diet feeding can dramatically increase the susceptibility of metabolic diseases in offspring. However, whether maternal high-calorie diet feeding can program hepatic cholesterol metabolism in the early life of offspring is less understood, and the epigenetic mechanisms underlying this intergenerational effect, especially during the early life of offspring, are unknown. Female C57BL/6J mice were randomly assigned to a high-calorie diet or control diet before and during gestation, and lactation. Lipid metabolism was evaluated in male offspring at weaning. Gene expressions and quantitative methylation levels of key genes associated with hepatic cholesterol metabolism were further evaluated in offspring at weaning age. We found that maternal high-calorie diet feeding resulted in higher body weight, hypercholesterolemia, elevated total cholesterol in liver homogenates, and fat deposits in the liver in offspring at weaning. For key genes that regulate cholesterol metabolism in liver, we showed lower Hmgcr and Ldlr, and higher Abca1 mRNA and protein expressions in offspring from dams fed with high-calorie diet at weaning age. We further found that maternal high-calorie diet feeding significantly decreased Abca1 methylation level in offspring, with lower methylation levels of both CpG 11 and CpG 22 sites. Interestingly, we found that Abca1 methylation level was negatively associated with hepatic Abca1 mRNA expression in offspring from dams fed with high-calorie diet and controls. However, the expressions of key genes associated with hepatic cholesterol metabolism were not significant between fetuses of dams fed with high-calorie diet and control diet. In conclusion, our results indicate that maternal high-calorie diet feeding results in aberrant lipid metabolism, including hypercholesterolemia and fat deposits in the liver of offspring as early as weaning age. Furthermore, maternal high-calorie feeding can program hepatic cholesterol metabolism and Abca1 methylation in the early life of offspring.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Wenyu Zou
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing, China
| | - Honghua Wu
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China.
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
13
|
Kim J, Choi A, Kwon YH. Maternal low-protein diet alters hepatic lipid accumulation and gene expression related to glucose metabolism in young adult mouse offspring fed a postweaning high-fat diet. Biochem Biophys Res Commun 2023; 682:193-198. [PMID: 37820455 DOI: 10.1016/j.bbrc.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/26/2023] [Accepted: 10/01/2023] [Indexed: 10/13/2023]
Abstract
Maternal consumption of low-protein (LP) diet during pregnancy has been demonstrated to increase the chances of adult offspring developing metabolic syndrome, and this risk can be exacerbated when the postnatal diets do not align with the prenatal conditions. However, in our previous study, focusing on serum parameters and gene expression patterns within adipose tissue, we discovered the presence of "healthy obesity" in young adult offspring from dams that were fed an LP, as a response to a postweaning high-fat (HF) diet. Here, we subsequently investigated the role played by the liver and skeletal muscle in alleviation of insulin resistance in male offspring that were fed either control (C/C group) or HF diet (C/HF and LP/HF groups) for 22 weeks. While a postweaning HF diet increased liver weight and hepatic triglyceride (TG) and cholesterol levels in offspring of control dams, these levels were lower in the LP/HF group compared to the C/HF group. Analysis of the liver transcriptome identified 430 differentially expressed genes (DEGs) in the LP/HF and C/HF comparison. Especially, downregulated DEGs were enriched in carbohydrate metabolism and the levels of DEGs were significantly correlated with the levels of markers for serum glucose homeostasis and hepatic lipid accumulation. In the LP/HF group compared to the C/HF group, there was a decrease in the gastrocnemius muscle weight, while no differences were observed in gene expression levels associated with muscle fiber phenotype, mitochondrial function, and inflammation. In conclusion, maternal LP diet induced changes in lipid and glucose metabolism within the liver, similar to what was observed in adipose tissue, while there were no alterations in metabolic functions in the skeletal muscle in young offspring mice fed an HF diet. Further research that investigating the enduring impact of nutritional transition on offspring is essential to gain a comprehensive grasp of developmental programming throughout their entire lifespan.
Collapse
Affiliation(s)
- Juhae Kim
- Department of Food and Nutrition, Seoul National University, Seoul, South Korea
| | - Alee Choi
- Department of Food and Nutrition, Seoul National University, Seoul, South Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, Seoul, South Korea; Research Institute of Human Ecology, Seoul National University, Seoul, South Korea.
| |
Collapse
|
14
|
do Carmo JM, Dai X, Aitken N, Larson KM, Omoto ACM, Gulke RR, Wang Z, Li X, Mouton AJ, Hall JE, da Silva AA. Sex differences in weight gain, blood pressure control, and responses to melanocortin-4 receptor antagonism in offspring from lean and obese parents. Am J Physiol Regul Integr Comp Physiol 2023; 325:R401-R410. [PMID: 37519251 PMCID: PMC10639017 DOI: 10.1152/ajpregu.00106.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/07/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
We examined potential sex differences in appetite and blood pressure (BP) responses to melanocortin-4 receptor (MC4R) blockade in offspring from lean and obese parents. Offspring from normal (N) diet-fed parents were fed N (NN) or high-fat (H) diets (NH) from weaning until adulthood. Offspring from obese H diet-fed parents were also fed N (HN) or H diets (HH). Adult male and female offspring were implanted with BP telemetry probes and intracerebroventricular cannulas to infuse MC4R antagonist or vehicle. Infusion of the MC4R antagonist SHU-9119 (1 nmol/h) for 7 days caused larger increases in calorie intake and body weight in obese compared with lean offspring. In male offspring, HH and HN groups exhibited higher baseline BP compared with NN and NH, and HH showed a greater reduction in BP during SHU-9119 infusion. In female offspring, HH also showed higher baseline BP and greater reduction in BP during MC4R blockade. SHU-9119 reduced heart rate in all groups, but reductions were more pronounced in offspring from lean parents. Combined α and β-adrenergic blockade reduced BP more in male HH offspring compared with NN controls. Losartan reduced BP more in male NH, HN, and HH offspring compared with NN controls. Losartan and α- and β-adrenergic blockade reduced BP similarly in all female groups. These results suggest that endogenous MC4R activity contributes to elevated BP in obese offspring from obese parents. Our findings also indicate important sex differences in the mechanisms of BP control in male and female offspring of obese parents.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Xuemei Dai
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Nikaela Aitken
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Kylie M Larson
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Ana C M Omoto
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Rodrigo R Gulke
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Centro Universitário Barão de Mauá, Ribeirao Preto, Brazil
| | - Zhen Wang
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Xuan Li
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alan J Mouton
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - John E Hall
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
15
|
Zhang L, Zou W, Hu Y, Wu H, Gao Y, Zhang J, Zheng J. Maternal voluntary wheel running modulates glucose homeostasis, the gut microbiota and its derived fecal metabolites in offspring. Clin Sci (Lond) 2023; 137:1151-1166. [PMID: 37505199 PMCID: PMC10412464 DOI: 10.1042/cs20230372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/14/2023] [Accepted: 07/28/2023] [Indexed: 07/29/2023]
Abstract
Maternal overnutrition can dramatically increase the susceptibility of offspring to metabolic diseases, whereas maternal exercise may improve glucose metabolism in offspring. However, the underlying mechanism programming the intergenerational effects of maternal exercise on the benefits of glucose metabolism has not been fully elaborated. C57BL/6 female mice were randomly assigned to four subgroups according to a diet and exercise paradigm before and during pregnancy as follows: NC (fed with normal chow diet and sedentary), NCEx (fed with normal chow diet and running), HF (fed with high-fat diet and sedentary), and HFEx (fed with high-fat diet and running). Integrative 16S rDNA sequencing and mass spectrometry-based metabolite profiling were synchronously performed to characterize the effects of maternal exercise on the gut microbiota composition and metabolite alterations in offspring. Maternal exercise, acting as a natural pharmaceutical intervention, prevented deleterious effects on glucose metabolism in offspring. 16S rDNA sequencing revealed remarkable changes in the gut microbiota composition in offspring. Metabolic profiling indicated multiple altered metabolites, which were enriched in butanoate metabolism signaling in offspring. We further found that maternal exercise could mediate gene expression related to intestinal gluconeogenesis in offspring. In conclusion, our study indicated that maternal running significantly improved glucose metabolism in offspring and counteracted the detrimental effects of maternal high-fat feeding before and during pregnancy. We further demonstrated that maternal voluntary wheel running could integratively program the gut microbiota composition and fecal metabolite changes and then regulate butanoate metabolism and mediate intestinal gluconeogenesis in offspring.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Wenyu Zou
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing 100034, China
| | - Honghua Wu
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
16
|
Rodrigues FDS, Jantsch J, Fraga GDF, Dias VS, Eller S, De Oliveira TF, Giovenardi M, Guedes RP. Cannabidiol treatment improves metabolic profile and decreases hypothalamic inflammation caused by maternal obesity. Front Nutr 2023; 10:1150189. [PMID: 36969815 PMCID: PMC10033544 DOI: 10.3389/fnut.2023.1150189] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionThe implications of maternal overnutrition on offspring metabolic and neuroimmune development are well-known. Increasing evidence now suggests that maternal obesity and poor dietary habits during pregnancy and lactation can increase the risk of central and peripheral metabolic dysregulation in the offspring, but the mechanisms are not sufficiently established. Furthermore, despite many studies addressing preventive measures targeted at the mother, very few propose practical approaches to treat the damages when they are already installed.MethodsHere we investigated the potential of cannabidiol (CBD) treatment to attenuate the effects of maternal obesity induced by a cafeteria diet on hypothalamic inflammation and the peripheral metabolic profile of the offspring in Wistar rats.ResultsWe have observed that maternal obesity induced a range of metabolic imbalances in the offspring in a sex-dependant manner, with higher deposition of visceral white adipose tissue, increased plasma fasting glucose and lipopolysaccharides (LPS) levels in both sexes, but the increase in serum cholesterol and triglycerides only occurred in females, while the increase in plasma insulin and the homeostatic model assessment index (HOMA-IR) was only observed in male offspring. We also found an overexpression of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNFα), interleukin (IL) 6, and interleukin (IL) 1β in the hypothalamus, a trademark of neuroinflammation. Interestingly, the expression of GFAP, a marker for astrogliosis, was reduced in the offspring of obese mothers, indicating an adaptive mechanism to in utero neuroinflammation. Treatment with 50 mg/kg CBD oil by oral gavage was able to reduce white adipose tissue and revert insulin resistance in males, reduce plasma triglycerides in females, and attenuate plasma LPS levels and overexpression of TNFα and IL6 in the hypothalamus of both sexes.DiscussionTogether, these results indicate an intricate interplay between peripheral and central counterparts in both the pathogenicity of maternal obesity and the therapeutic effects of CBD. In this context, the impairment of internal hypothalamic circuitry caused by neuroinflammation runs in tandem with the disruptions of important metabolic processes, which can be attenuated by CBD treatment in both ends.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiago Franco De Oliveira
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- *Correspondence: Renata Padilha Guedes,
| |
Collapse
|
17
|
Loss of hypothalamic Furin affects POMC to proACTH cleavage and feeding behavior in high-fat diet-fed mice. Mol Metab 2022; 66:101627. [PMID: 36374777 PMCID: PMC9664468 DOI: 10.1016/j.molmet.2022.101627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE The hypothalamus regulates feeding and glucose homeostasis through the balanced action of different neuropeptides, which are cleaved and activated by the proprotein convertases PC1/3 and PC2. However, the recent association of polymorphisms in the proprotein convertase FURIN with type 2 diabetes, metabolic syndrome, and obesity, prompted us to investigate the role of FURIN in hypothalamic neurons controlling glucose and feeding. METHODS POMC-Cre+/- mice were bred with Furinfl/fl mice to generate conditional knockout mice with Furin-deletion in neurons expressing proopiomelanocortin (POMCFurKO), and Furinfl/fl mice were used as controls. POMCFurKO and controls were periodically monitored on both normal chow diet and high fat diet (HFD) for body weight and glucose tolerance by established in-vivo procedures. Food intake was measured in HFD-fed FurKO and controls. Hypothalamic Pomc mRNA was measured by RT-qPCR. ELISAs quantified POMC protein and resulting peptides in the hypothalamic extracts of POMCFurKO mice and controls. The in-vitro processing of POMC was studied by biochemical techniques in HEK293T and CHO cell lines lacking FURIN. RESULTS In control mice, Furin mRNA levels were significantly upregulated on HFD feeding, suggesting an increased demand for FURIN activity in obesogenic conditions. Under these conditions, the POMCFurKO mice were hyperphagic and had increased body weight compared to Furinfl/fl mice. Moreover, protein levels of POMC were elevated and ACTH concentrations markedly reduced. Also, the ratio of α-MSH/POMC was decreased in POMCFurKO mice compared to controls. This indicates that POMC processing was significantly reduced in the hypothalami of POMCFurKO mice, highlighting for the first time the involvement of FURIN in the cleavage of POMC. Importantly, we found that in vitro, the first stage in processing where POMC is cleaved into proACTH was achieved by FURIN but not by PC1/3 or the other proprotein convertases in cell lines lacking a regulated secretory pathway. CONCLUSIONS These results suggest that FURIN processes POMC into proACTH before sorting into the regulated secretory pathway, challenging the dogma that PC1/3 and PC2 are the only convertases responsible for POMC cleavage. Furthermore, its deletion affects feeding behaviors under obesogenic conditions.
Collapse
|
18
|
Huang X, Zhuo Y, Jiang D, Zhu Y, Fang Z, Che L, Lin Y, Xu S, Hua L, Zou Y, Huang C, Li L, Wu D, Feng B. Maternal Low-Protein Diet during Puberty and Adulthood Aggravates Lipid Metabolism of Their Offspring Fed a High-Fat Diet in Mice. Nutrients 2022; 14:nu14194057. [PMID: 36235710 PMCID: PMC9570549 DOI: 10.3390/nu14194057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
A maternal low-protein (LP) diet during gestation and/or lactation results in metabolic syndrome in their offspring. Here, we investigated the effect of maternal LP diet during puberty and adulthood on the metabolic homeostasis of glucose and lipids in offspring. Female mice were fed with normal-protein (NP) diet or a LP diet for 11 weeks. Male offspring were then fed with a high-fat diet (NP-HFD and LP-HFD groups) or standard chow diet (NP-Chow and LP-Chow groups) for 4 months. Results showed that maternal LP diet during puberty and adulthood did not alter the insulin sensitivity and hepatic lipid homeostasis of their offspring under chow diet, but aggravated insulin resistance, hepatic steatosis, and hypercholesterolemia of offspring in response to a post-weaning HFD. Accordingly, transcriptomics study with offspring’s liver indicated that several genes related to glucose and lipid metabolism, including lipoprotein lipase (Lpl), long-chain acyl-CoA synthetase 1 (Acsl1), Apoprotein A1 (Apoa1), major urinary protein 19 (Mup19), cholesterol 7α hydroxylase (Cyp7a1) and fibroblast growth factor 1 (Fgf1), were changed by maternal LP diet. Taken together, maternal LP diet during puberty and adulthood could disarrange the expression of metabolic genes in the liver of offspring and aggravate insulin resistance and hepatic steatosis in offspring fed a HFD.
Collapse
Affiliation(s)
- Xiaohua Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Dandan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yingguo Zhu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Lixia Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence:
| |
Collapse
|
19
|
Zheng J, Zhang L, Gao Y, Wu H, Zhang J. The dynamic effects of maternal high-calorie diet on glycolipid metabolism and gut microbiota from weaning to adulthood in offspring mice. Front Nutr 2022; 9:941969. [PMID: 35928844 PMCID: PMC9343994 DOI: 10.3389/fnut.2022.941969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Dysbiosis of gut microbiota can contribute to the progression of diabetes and obesity. Previous studies have shown that maternal high-fat (HF) diet during the perinatal period can alter the microbiota and induce metabolic disorders at weaning. However, whether dysbiosis of gut microbiota and metabolism could be recovered by a normal diet after weaning and the dynamic changes of gut microbiota have not been fully studied. In this study, C57BL/6J female mice were fed with a normal chow (NC) or HF diet for 4 weeks preconception, during gestation, and until pup weaning. After weaning, male offspring were fed with an NC diet until 9 weeks of age. The microbiota of offspring at weaning and 9 weeks of age was collected for 16S rRNA gene amplicon sequencing. We found that dams fed with an HF diet showed glucose intolerance after lactation. Compared with the offspring from NC dams, the offspring from HF dams exhibited a higher body weight, hyperglycemia, glucose intolerance, hyperinsulinemia, hypercholesterolemia, and leptin resistance and lower adiponectin at weaning. Fecal analysis indicated altered microbiota composition between the offspring of the two groups. The decrease in favorable bacteria (such as norank f Bacteroidales S24-7 group) and increase in unfavorable bacteria (such as Lachnoclostridium and Desulfovibrio) were strongly associated with a disturbance of glucose and lipid metabolism. After 6 weeks of normal diet, no difference in body weight, glucose, and lipid profiles was observed between the offspring of the two groups. However, the microbiota composition of offspring in the HF group was still different from that in the NC group, and microbiota diversity was lower in offspring of the HF group. The abundance of Lactobacillus was lower in the offspring of the HF group. In conclusion, a maternal HF diet can induce metabolic homeostasis and gut microbiota disturbance in offspring at weaning. Gut microbiota dysbiosis can persist into adulthood in the offspring, which might have a role in the promotion of susceptibility to obesity and diabetes in the later life of the offspring.
Collapse
Affiliation(s)
- Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Honghua Wu
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
20
|
Zhang Y, Pu J, Ding Y, Wu L, Yin Y, Sun M, Gu Y, Zhang D, Zhang Z, Zheng Q, He Q, Xu T, He Y, Su H, Zhou X, Li L, Ye Y, Li J, Xu Z. Sex Differences at Early Old Stage in Glycolipid Metabolism and Fatty Liver in Offspring Prenatally Exposed to Chinese Great Famine. Front Nutr 2022; 9:913966. [PMID: 35811949 PMCID: PMC9257182 DOI: 10.3389/fnut.2022.913966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAbout 50 years ago, Chinese Great Famine (CGF) affected the entire population in China, and its long-term influence on the offspring has attracted significant attention for research. However, information on possible metabolic differences between sexes is limited. This study explored whether there might be sex differences in the risks of development of glucolipid metabolic dysfunction and fatty liver following prenatal exposure to CGF.Materials and MethodsThere were 11,417 subjects around 55 years of age (6,661 women and 4,756 men). They were divided as the exposed group in which the fetal stage was in CGF, and the unexposed group included those born after CGF. Analysis focused on comparisons between sexes.ResultsCompared to the unexposed group, the BMI and triglyceride (P < 0.05) in men were higher in exposed group, while waist circumference and blood sugar (P < 0.05) in the exposed women were significantly higher. With the ages being properly balanced, the risks of glycolipid metabolic dysfunction were significantly higher in both men and women in the exposed than in the unexposed group (P < 0.001). Prenatal exposure to CGF significantly increased risks of abnormal BMI (P < 0.001, 95% CI: 2.305–2.93), blood sugar (P < 0.05, 95% CI: 1.050–1.401), triglycerides (P < 0.05, 95% CI: 1.006–1.245), and fatty liver (P < 0.001, 95% CI: 1.121–1.390) in men, and increased risks of abnormal blood sugar (P < 0.05, 95% CI: 1.024–1.689) and positive urine sugar (P < 0.05, 95% CI: 1.062–6.211) in women. Height and body weight were either the same or higher in the exposed subjects compared with the unexposed ones, regardless of sexes.ConclusionThis study is the first to identify sex differences in the long-term effects of CGF on metabolism and fatty liver. Importance of the findings include the benefits of prescribing medicine for the early prevention of certain diseases for each sex before aging based on the differences revealed. This study also shows “catch-up growth” in the offspring prenatally exposed to CGF as possible mechanisms underlying the long-term effects.
Collapse
Affiliation(s)
- Yumeng Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianhong Pu
- The Center of Management, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Ding
- Department of Preventive Medicine, College of Clinical Medicine, Suzhou Vocational Health College, Suzhou, China
| | - Lei Wu
- Suzhou Industrial Park Centers for Disease Control and Prevention, Suzhou, China
| | - Yongxiang Yin
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Mingya Sun
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ying Gu
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Daiyi Zhang
- The Center of Management, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ze Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiutong Zheng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qinyuan He
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yun He
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongyu Su
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiuwen Zhou
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Ye
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Jingyang Li
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- *Correspondence: Zhice Xu,
| |
Collapse
|
21
|
Simões-Alves AC, Arcoverde-Mello APFC, Campos JDO, Wanderley AG, Leandro CVG, da Costa-Silva JH, de Oliveira Nogueira Souza V. Cardiometabolic Effects of Postnatal High-Fat Diet Consumption in Offspring Exposed to Maternal Protein Restriction In Utero. Front Physiol 2022; 13:829920. [PMID: 35620602 PMCID: PMC9127546 DOI: 10.3389/fphys.2022.829920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/29/2022] [Indexed: 01/01/2023] Open
Abstract
In recent decades, the high incidence of infectious and parasitic diseases has been replaced by a high prevalence of chronic and degenerative diseases. Concomitantly, there have been profound changes in the behavior and eating habits of families around the world, characterizing a "nutritional transition" phenomenon, which refers to a shift in diet in response to modernization, urbanization, or economic development from undernutrition to the excessive consumption of hypercaloric and ultra-processed foods. Protein malnutrition that was a health problem in the first half of the 20th century has now been replaced by high-fat diets, especially diets high in saturated fat, predisposing consumers to overweight and obesity. This panorama points us to the alarming coexistence of both malnutrition and obesity in the same population. In this way, individuals whose mothers were undernourished early in pregnancy and then exposed to postnatal hyperlipidic nutrition have increased risk factors for developing metabolic dysfunction and cardiovascular diseases in adulthood. Thus, our major aim was to review the cardiometabolic effects resulting from postnatal hyperlipidic diets in protein-restricted subjects, as well as to examine the epigenetic repercussions occasioned by the nutritional transition.
Collapse
Affiliation(s)
- Aiany Cibelle Simões-Alves
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - Ana Paula Fonseca Cabral Arcoverde-Mello
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - Jéssica de Oliveira Campos
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | | | - Carol Virginia Gois Leandro
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - João Henrique da Costa-Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - Viviane de Oliveira Nogueira Souza
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| |
Collapse
|
22
|
Franzago M, Porreca A, D’Ardes M, Di Nicola M, Di Tizio L, Liberati M, Stuppia L, Vitacolonna E. The Obesogenic Environment: Epigenetic Modifications in Placental Melanocortin 4 Receptor Gene Connected to Gestational Diabetes and Smoking. Front Nutr 2022; 9:879526. [PMID: 35571924 PMCID: PMC9100829 DOI: 10.3389/fnut.2022.879526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Background Maternal metabolic insults as well as Gestational Diabetes Mellitus (GDM) influence the fetal health and may affect ‘offspring’s susceptibility to chronic diseases via epigenetic modifications. GDM, the most common metabolic disorder in pregnancy, can be considered the result of complex interactions between genetic and environmental factors. A critical point in this view is the identification of genes which are epigenetically modified under the influence of GDM. The melanocortin 4 receptor (MC4R) gene plays a crucial role in nutritional health by suppressing appetite and participating in energy control regulation. The correlations between pregnant ‘women’s metabolic profiles and placental epigenetic modifications of this gene have been poorly investigated. Objective The aim of this study was to evaluate the effect of GDM and maternal clinical parameters at the third trimester of pregnancy to DNA methylation levels in the placenta at CpG sites of MC4R gene. Design and Methods Socio-demographic and clinical characteristics, Mediterranean diet adherence, smoking habits, and physical activity were assessed at the third trimester of pregnancy of 60 Caucasian pregnant women, of which 33 with GDM. Clinical parameters of the newborns were recorded at birth. MC4R DNA methylation on maternal and fetal sides of the placenta was analyzed using bisulfite pyrosequencing. Results MC4R DNA methylation levels at CpG1 and CpG2 were lower on the fetal side of the placenta in GDM-affected women than in non-GDM-affected recruits (p = 0.033). Moreover, DNA methylation levels on the maternal side at CpG1 were positively related to glucose concentration at 2-h oral glucose tolerance test (OGTT). On the other hand, CpG2 DNA methylation was positively related to both 1-h and 2-h during OGTT. Maternal DNA methylation level at CpG2 was also associated with low density lipoprotein cholesterol (LDL-C) at the third trimester of pregnancy (rho = 0.340, p < 0.05), while CpG1 methylation was negatively related to maternal weight variations at delivery (rho = −0.316, p < 0.05). Significant associations between MC4R DNA methylation on the maternal side and lipid profile at third trimester of pregnancy in women smokers were found. Conclusion Our results suggest that MC4R methylation profile in the placenta is related to maternal metabolic and nutritional conditions, potentially affecting fetal programming and the future metabolic health of the newborn.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
- Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti, Italy
| | - Annamaria Porreca
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Mario D’Ardes
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Luciano Di Tizio
- Department of Obstetrics and Gynaecology, SS. Annunziata Hospital, “G. d’Annunzio” University, Chieti, Italy
| | - Marco Liberati
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti, Italy
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, “G. d’Annunzio” University, Chieti, Italy
- Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti, Italy
- *Correspondence: Ester Vitacolonna,
| |
Collapse
|