1
|
Cai K, Chen Z, Wu J, Yu J, Chen J, Zhou X, Pan B, Xie Z, Wang Q, Li P, Liao Q. UHPLC-QTOF-MS-based metabolomics joint high-throughput RNA sequencing transcriptomics approach for the analysis of fecal and liver biological samples and application in a case study for the mechanism of Qing-Kai-Ling oral liquid in treating MASLD. Anal Bioanal Chem 2025:10.1007/s00216-025-05892-2. [PMID: 40310493 DOI: 10.1007/s00216-025-05892-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
Qing-Kai-Ling (QKL) oral liquid has been increasingly used in metabolic dysfunction-associated steatotic liver disease (MASLD). However, the specific metabolic differentials and metabolic pathway mechanisms that affect the MASLD regulated by QKL remained unclear. In this study, serum biochemical analyses and hematoxylin-eosin staining of the liver revealed QKL reduced liver injury and enhanced lipid metabolism ability, respectively. To clarify the therapeutic mechanism of the QKL in the treatment of MASLD, UHPLC-QTOF-MS non-target metabolomics and RNA-Seq high-throughput sequencing analysis were used to explore the mechanism of the QKL in the treatment of MASLD from the perspective of metabolic-gene interactions. UHPLC-QTOF-MS-based untargeted metabolomics further revealed that there were 196 common differentially expressed metabolites identified among 3 groups; QKL significantly up-regulated 44 metabolites, while 11 metabolites (including N-phenylacetylglutamic acid and glycocholic acid) were downregulated significantly. Moreover, the main metabolic pathways regulated by QKL included amino acids, peptides, bile acids, carbohydrates, linoleic acids, etc. Additionally, the result of the RNA sequencing-based transcriptomics showed that a total of 984 differential genes (DEGs) were identified and 9 important DEGs were obtained. The result of the Kyoto Encyclopedia of Genes and Genomes (KEGG) demonstrated that the 984 DEGs were linked to bile acid metabolism, glycerophospholipid metabolism, insulin resistance, AMPK signaling pathway, etc. Overall, this work was the first to show that QKL regulated metabolites and genes to alleviate MASLD by the UHPLC-QTOF-MS-based untargeted metabolomics joint high-throughput RNA sequencing-based transcriptomics analysis, providing the basis and research method for the treatment of metabolic diseases by QKL and other drugs.
Collapse
Affiliation(s)
- Kaiwei Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zihao Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jingyun Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jingtao Yu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jingheng Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaoqin Zhou
- Guangzhou Baiyunshan Mingxing Pharmaceutical Co., Ltd, Guangzhou, 510250, China
| | - Biyan Pan
- Guangzhou Baiyunshan Mingxing Pharmaceutical Co., Ltd, Guangzhou, 510250, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Shenzhen, 528406, China
| | - Qiuyun Wang
- Guangzhou Baiyunshan Mingxing Pharmaceutical Co., Ltd, Guangzhou, 510250, China
| | - Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Chevalley T, Dübi M, Fumeaux L, Merli MS, Sarre A, Schaer N, Simeoni U, Yzydorczyk C. Sexual Dimorphism in Cardiometabolic Diseases: From Development to Senescence and Therapeutic Approaches. Cells 2025; 14:467. [PMID: 40136716 PMCID: PMC11941476 DOI: 10.3390/cells14060467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
The global incidence and prevalence of cardiometabolic disorders have risen significantly in recent years. Although lifestyle choices in adulthood play a crucial role in the development of these conditions, it is well established that events occurring early in life can have an important effect. Recent research on cardiometabolic diseases has highlighted the influence of sexual dimorphism on risk factors, underlying mechanisms, and response to therapies. In this narrative review, we summarize the current understanding of sexual dimorphism in cardiovascular and metabolic diseases in the general population and within the framework of the Developmental Origins of Health and Disease (DOHaD) concept. We explore key risk factors and mechanisms, including the influence of genetic and epigenetic factors, placental and embryonic development, maternal nutrition, sex hormones, energy metabolism, microbiota, oxidative stress, cell death, inflammation, endothelial dysfunction, circadian rhythm, and lifestyle factors. Finally, we discuss some of the main therapeutic approaches, responses to which may be influenced by sexual dimorphism, such as antihypertensive and cardiovascular treatments, oxidative stress management, nutrition, cell therapies, and hormone replacement therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Catherine Yzydorczyk
- Developmental Origins of Health and Disease (DOHaD) Laboratory, Division of Pediatrics, Department Woman-Mother-Child, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (T.C.); (M.D.); (L.F.); (M.S.M.); (A.S.); (N.S.)
| |
Collapse
|
3
|
Correia Gomes D, Meza Alvarado JE, Zamora Briseño JA, Cano Sarmiento C, Camacho Morales A, Viveros Contreras R. Maternal Supplementation with Lacticaseibacillus rhamnosus GG Improves Glucose Tolerance and Modulates the Intestinal Microbiota of Offspring. Diseases 2024; 12:312. [PMID: 39727642 PMCID: PMC11726987 DOI: 10.3390/diseases12120312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024] Open
Abstract
INTRODUCTION Consuming hypercaloric diets during pregnancy induces metabolic, immune, and maternal intestinal dysbiosis disorders. These conditions are transferred to the offspring through the placenta and breastfeeding, increasing susceptibility to metabolic diseases. We investigated the effect of L. rhamnosus GG supplementation on offspring maternally programmed with a hypercaloric diet. METHODS Our study involved sixteen female Wistar rats aged ten weeks, which were divided into four groups based on their diets: control (Ctrl), cafeteria (CAF), control + probiotic (PRO), and cafeteria + probiotic (CPRO). The control + probiotic and cafeteria + probiotic groups received a daily oral administration of 250 μL of L. rhamnosus GG cell suspension (equivalent to 109 UFC) for nine weeks. The body weight of the animals was recorded weekly, and their food intake was monitored every 24 h. An oral glucose tolerance test was conducted on the offspring at seven weeks of age. At the ninth week of age, animals were euthanized, and blood, tissues, and organs were collected. RESULTS Maternal supplementation with L. rhamnosus GG decreased food intake and the average birth weight, improved glucose sensitivity, and lowered the levels of LDL, cholesterol, triglycerides, and mesenteric adipose tissue in offspring compared with the control and cafeteria groups. CONCLUSIONS Our findings indicate that supplementing with LGG during maternal programming could protect offspring from metabolic disruptions caused by a hypercaloric maternal diet.
Collapse
Affiliation(s)
- Dayane Correia Gomes
- Centro de Investigaciones Biomédicas, Doctorado en Ciencias Biomédicas, Universidad Veracruzana, Xalapa 91190, Mexico; (D.C.G.); (J.E.M.A.)
| | - José Enrique Meza Alvarado
- Centro de Investigaciones Biomédicas, Doctorado en Ciencias Biomédicas, Universidad Veracruzana, Xalapa 91190, Mexico; (D.C.G.); (J.E.M.A.)
| | | | - Cynthia Cano Sarmiento
- Food Research and Development Unit, Technological Institute of Veracruz, National Institute of Technology of Mexico, M.A. de Quevedo 2779, Veracruz 91897, Mexico;
| | - Alberto Camacho Morales
- Faculty of Medicine, Department of Biochemistry and Molecular Medicine, Autonomous University of Nuevo León, Monterrey 66455, Mexico;
| | - Rubi Viveros Contreras
- Centro de Investigaciones Biomédicas, Doctorado en Ciencias Biomédicas, Universidad Veracruzana, Xalapa 91190, Mexico; (D.C.G.); (J.E.M.A.)
| |
Collapse
|
4
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-dependent susceptibility to brain metabolic dysfunction and memory impairment in response to pre and postnatal high-fat diet. J Nutr Biochem 2024; 132:109675. [PMID: 38945454 DOI: 10.1016/j.jnutbio.2024.109675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Huang Y, Wang A, Zhou W, Li B, Zhang L, Rudolf AM, Jin Z, Hambly C, Wang G, Speakman JR. Maternal dietary fat during lactation shapes single nucleus transcriptomic profile of postnatal offspring hypothalamus in a sexually dimorphic manner in mice. Nat Commun 2024; 15:2382. [PMID: 38493217 PMCID: PMC10944494 DOI: 10.1038/s41467-024-46589-x] [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: 09/28/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
Maternal overnutrition during lactation predisposes offspring to develop metabolic diseases and exacerbates the relevant syndromes in males more than females in later life. The hypothalamus is a heterogenous brain region that regulates energy balance. Here we combined metabolic trait quantification of mother and offspring mice under low and high fat diet (HFD) feeding during lactation, with single nucleus transcriptomic profiling of their offspring hypothalamus at peak lacation to understand the cellular and molecular alterations in response to maternal dietary pertubation. We found significant expansion in neuronal subpopulations including histaminergic (Hdc), arginine vasopressin/retinoic acid receptor-related orphan receptor β (Avp/Rorb) and agouti-related peptide/neuropeptide Y (AgRP/Npy) in male offspring when their mothers were fed HFD, and increased Npy-astrocyte interactions in offspring responding to maternal overnutrition. Our study provides a comprehensive offspring hypothalamus map at the peak lactation and reveals how the cellular subpopulations respond to maternal dietary fat in a sex-specific manner during development.
Collapse
Affiliation(s)
- Yi Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Broad Institute of MIT and Harvard, Metabolism Program, Cambridge, MA, 02142, USA
| | - Anyongqi Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China
| | - Baoguo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Linshan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China
| | - Agata M Rudolf
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zengguang Jin
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China.
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK.
- China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
6
|
Inoue Y, Fukushima M, Hirasawa G, Furukawa F, Takeda H, Umatani C. Maternal High-Fat Diet Affects the Contents of Eggs and Causes Abnormal Development in the Medaka Fish. Endocrinology 2024; 165:bqae006. [PMID: 38279936 DOI: 10.1210/endocr/bqae006] [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: 10/17/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 01/29/2024]
Abstract
Maternal nutritional status can affect development and metabolic phenotypes of progeny in animals. The effects of maternal diet are thought to be mediated mainly by changes inside oocytes such as organelles, maternal RNAs, and metabolites. However, to what extent each factor contributes to offspring phenotypes remains uncertain, especially in viviparous mammalian systems, where factors other than oocytes, such as placenta and milk, need to be considered. Here, using the medaka fish as an oviparous vertebrate model, we examined whether maternal high-fat diet (mHFD) feeding affects offspring development and what kind of changes occur in the contents of mature eggs. We found that mHFD caused the high frequency of embryonic deformities of offspring, accompanied by downregulation of transcription- and translation-related genes and zygotic transcripts at the blastula stage. Transcriptomic and metabolomic analyses of mature eggs suggested decreased catabolism of amino acids and glycogen, moderate upregulation of endoplasmic reticulum stress-related genes, and elevated lipid levels in mHFD eggs. Furthermore, high-fat diet females showed a higher incidence of oocyte atresia and downregulation of egg protein genes in the liver. These data suggest that attenuated amino acid catabolism triggered by decreased yolk protein load/processing, as well as elevated lipid levels inside eggs, are the prime candidates that account for the higher incidence of embryonic deformities in mHFD offspring. Our study presents a comprehensive data on the changes inside eggs in a mHFD model of nonmammalian vertebrates and provides insights into the mechanisms of parental nutritional effects on offspring.
Collapse
Affiliation(s)
- Yusuke Inoue
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Manatsu Fukushima
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Go Hirasawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Fumiya Furukawa
- School of Marine Biosciences, Kitasato University, Kanagawa 252-0373, Japan
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Chie Umatani
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Division of Applied Biological Chemistry, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
7
|
Arslan S, Yıldıran H, Seymen CM. The Effect of Maternal High-Fat Diet on Adipose Tissue Histology and Lipid Metabolism-Related Genes Expression in Offspring Rats. Nutrients 2024; 16:150. [PMID: 38201978 PMCID: PMC10780511 DOI: 10.3390/nu16010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The developing fetus is dependent on the maternal nutritional environment. This study was conducted to determine the effects of a maternal high-fat diet (HFD) applied during pregnancy and/or lactation on the expression levels of some lipid-related genes in rat models. Half of the pregnant rats (n: 6) were fed an HFD (energy from fat: 45%), while the other half (n: 6) were fed a control diet (CD) (energy from fat, 7.7%) during the pregnancy period. During lactation, dams in both groups were divided into two subgroups, with half fed the CD and the other half fed the HFD. Thus, four groups were obtained: CD-CD, CD-HFD, HFD-CD, and HFD-HFD. At the end of lactation, all mothers and half of the offspring were sacrificed. The remaining offspring were fed a CD for five weeks. The average birth weight of the CD group offspring was found to be lower than that of the HFD group (p < 0.05). The amount of adipose tissue was highest in CD-HFD (p < 0.05), while gene expression levels were similar between groups (p > 0.05), and the most degenerative histological changes were observed in the eight-week HFD-HFD (p < 0.05). This study suggests that maternal HFD during pregnancy and lactation may increase adiposity in offspring rats, especially during the weaning period.
Collapse
Affiliation(s)
- Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Hilal Yıldıran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Cemile Merve Seymen
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey;
| |
Collapse
|
8
|
Xu Y, Yang D, Wang L, Król E, Mazidi M, Li L, Huang Y, Niu C, Liu X, Lam SM, Shui G, Douglas A, Speakman JR. Maternal High Fat Diet in Lactation Impacts Hypothalamic Neurogenesis and Neurotrophic Development, Leading to Later Life Susceptibility to Obesity in Male but Not Female Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305472. [PMID: 37867217 PMCID: PMC10724448 DOI: 10.1002/advs.202305472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Indexed: 10/24/2023]
Abstract
Early life nutrition can reprogram development and exert long-term consequences on body weight regulation. In mice, maternal high-fat diet (HFD) during lactation predisposed male but not female offspring to diet-induced obesity when adult. Molecular and cellular changes in the hypothalamus at important time points are examined in the early postnatal life in relation to maternal diet and demonstrated sex-differential hypothalamic reprogramming. Maternal HFD in lactation decreased the neurotropic development of neurons formed at the embryo stage (e12.5) and impaired early postnatal neurogenesis in the hypothalamic regions of both males and females. Males show a larger increased ratio of Neuropeptide Y (NPY) to Pro-opiomelanocortin (POMC) neurons in early postnatal neurogenesis, in response to maternal HFD, setting an obese tone for male offspring. These data provide insights into the mechanisms by which hypothalamic reprograming by early life overnutrition contributes to the sex-dependent susceptibility to obesity in adult life in mice.
Collapse
Affiliation(s)
- Yanchao Xu
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationYantai UniversityYantai264005P. R. China
| | - Elżbieta Król
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
| | - Mohsen Mazidi
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
| | - Li Li
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
| | - Yi Huang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Chaoqun Niu
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Xue Liu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Alex Douglas
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
| | - John R. Speakman
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
- China medical universityShenyang110000P. R. China
| |
Collapse
|
9
|
Karlen-Amarante M, Bassi M, Barbosa RM, Sá JM, Menani JV, Colombari E, Zoccal DB, Colombari DSA. Maternal high-fat diet changes breathing pattern and causes excessive sympathetic discharge in juvenile offspring rat. Am J Physiol Lung Cell Mol Physiol 2023; 325:L662-L674. [PMID: 37786934 DOI: 10.1152/ajplung.00013.2023] [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: 01/11/2023] [Revised: 06/28/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
Early life over-nutrition, as experienced in maternal obesity, is a risk factor for developing cardiorespiratory and metabolic diseases. Here we investigated the effects of high-fat diet (HFD) consumption on the breathing pattern and sympathetic discharge to blood vessels in juvenile offspring from dams fed with HFD (O-HFD). Adult female Holtzman rats were given a standard diet (SD) or HFD from 6 wk before gestation to weaning. At weaning (P21), the male offspring from SD dams (O-SD) and O-HFD received SD until the experimental day (P28-P45). Nerve recordings performed in decerebrated in situ preparations demonstrated that O-HFD animals presented abdominal expiratory hyperactivity under resting conditions and higher vasoconstrictor sympathetic activity levels. The latter was associated with blunted respiratory-related oscillations in sympathetic activity, especially in control animals. When exposed to elevated hypercapnia or hypoxia levels, the O-HFD animals mounted similar ventilatory and respiratory motor responses as the control animals. Hypercapnia and hypoxia exposure also increased sympathetic activity in both groups but did not reinstate the respiratory-sympathetic coupling in the O-HFD rats. In freely behaving conditions, O-HFD animals exhibited higher resting pulmonary ventilation and larger variability of arterial pressure levels than the O-SD animals due to augmented sympathetic modulation of blood vessel diameter. Maternal obesity modified the functioning of cardiorespiratory systems in offspring at a young age, inducing active expiration and sympathetic overactivity under resting conditions. These observations represent new evidence about pregnancy-related complications that lead to the development of respiratory distress and hypertension in children of obese mothers.NEW & NOTEWORTHY Maternal obesity is a risk factor for developing cardiorespiratory and metabolic diseases. This study highlights the changes on the breathing pattern and sympathetic discharge to blood vessels in juvenile offspring from dams fed with HFD. Maternal obesity modified the functioning of cardiorespiratory systems in offspring, inducing active expiration and sympathetic overactivity. These observations represent new evidence about pregnancy-related complications that lead to the development of respiratory distress and hypertension in children of obese mothers.
Collapse
Affiliation(s)
- Marlusa Karlen-Amarante
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Mirian Bassi
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Rafaela Moreira Barbosa
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Jéssica Matheus Sá
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - José Vanderlei Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | | |
Collapse
|
10
|
Inoue Y, Suzuki Y, Kunishima Y, Washio T, Morishita S, Takeda H. High-fat diet in early life triggers both reversible and persistent epigenetic changes in the medaka fish (Oryzias latipes). BMC Genomics 2023; 24:472. [PMID: 37605229 PMCID: PMC10441761 DOI: 10.1186/s12864-023-09557-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The nutritional status during early life can have enduring effects on an animal's metabolism, although the mechanisms underlying these long-term effects are still unclear. Epigenetic modifications are considered a prime candidate mechanism for encoding early-life nutritional memories during this critical developmental period. However, the extent to which these epigenetic changes occur and persist over time remains uncertain, in part due to challenges associated with directly stimulating the fetus with specific nutrients in viviparous mammalian systems. RESULTS In this study, we used medaka as an oviparous vertebrate model to establish an early-life high-fat diet (HFD) model. Larvae were fed with HFD from the hatching stages (one week after fertilization) for six weeks, followed by normal chow (NC) for eight weeks until the adult stage. We examined the changes in the transcriptomic and epigenetic state of the liver over this period. We found that HFD induces simple liver steatosis, accompanied by drastic changes in the hepatic transcriptome, chromatin accessibility, and histone modifications, especially in metabolic genes. These changes were largely reversed after the long-term NC, demonstrating the high plasticity of the epigenetic state in hepatocytes. However, we found a certain number of genomic loci showing non-reversible epigenetic changes, especially around genes related to cell signaling, liver fibrosis, and hepatocellular carcinoma, implying persistent changes in the cellular state of the liver triggered by early-life HFD feeding. CONCLUSION In summary, our data show that early-life HFD feeding triggers both reversible and persistent epigenetic changes in medaka hepatocytes. Our data provide novel insights into the epigenetic mechanism of nutritional programming and a comprehensive atlas of the long-term epigenetic state in an early-life HFD model of non-mammalian vertebrates.
Collapse
Affiliation(s)
- Yusuke Inoue
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
| | - Yuta Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan
| | - Yoshimi Kunishima
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Terumi Washio
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Shinichi Morishita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan.
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto, 603-8555, Japan.
| |
Collapse
|
11
|
Castillo P, Pomar CA, Palou A, Palou M, Picó C. Influence of Maternal Metabolic Status and Diet during the Perinatal Period on the Metabolic Programming by Leptin Ingested during the Suckling Period in Rats. Nutrients 2023; 15:nu15030570. [PMID: 36771278 PMCID: PMC9921535 DOI: 10.3390/nu15030570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
We aimed to analyze the long-term metabolic effects of leptin supplementation at physiological doses during suckling in the offspring of diet-induced obese rats, together with the potential benefits of improving maternal diet during lactation. Thus, the offspring of: dams fed standard-diet (SD) (CON-dams), dams fed western-diet (WD) before and during gestation and lactation (WD-dams), and dams fed as WD-dams but moved to SD during lactation (REV-dams) were supplemented throughout suckling with leptin or vehicle, and fed SD or WD from weaning to four months. Under SD, leptin treatment significantly improved metabolic profile and body fat accumulation, with stronger effects in the male offspring of CON-dams and REV-dams. Under WD, the offspring of WD-dams presented metabolic alterations that were not evident in the offspring of REV-dams. Moreover, leptin supplementation improved glucose homeostasis in the male offspring of REV-dams. Conversely, leptin supplementation in females born to WD-dams and fed WD from weaning resulted in impaired insulin sensitivity and increased hepatic lipid content. These results highlight the importance of a balanced maternal diet during the perinatal period, especially lactation, for the subsequent metabolic health of the offspring and for the beneficial effects of leptin supplementation during suckling, more evident in the male offspring.
Collapse
Affiliation(s)
- Pedro Castillo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-971172373
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| |
Collapse
|
12
|
Fouda S, Vennikandam MM, Pappachan JM, Fernandez CJ. Pregnancy and Metabolic-associated Fatty Liver Disease: A Clinical Update. J Clin Transl Hepatol 2022; 10:947-954. [PMID: 36304500 PMCID: PMC9547252 DOI: 10.14218/jcth.2022.00052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/15/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
The intricate relationship between metabolic-associated fatty liver disease (MAFLD) and maternal complications has rapidly become a significant health threat in pregnant women. The presence of MAFLD in pregnancy increases the maternal risk of metabolic complications and comorbidities for both mother and baby. The preexistence or development of MAFLD in pregnancy is a complex multifactorial disorder that can lead to further complications for mother and baby. Therefore, as pregnant women are severely underrepresented in clinical research, there is a great need for a fair inclusion of this group in clinical trials. This review aims to explore the effects of MAFLD during pregnancy in the context of maternal complications and outcomes and explore the effects of pregnancy on the development and progression of MAFLD within the context of maternal obesity, altered metabolic profiles, gestational diabetes and altered hormonal profiles. We also addressed potential implications for the presence of MAFLD during pregnancy and its management in the clinical setting.
Collapse
Affiliation(s)
- Sherouk Fouda
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Madhu Mathew Vennikandam
- Department of Gastroenterology and Hepatology, Sparrow Hospital, Michigan State University College of Human Medicine, Lansing, MI, USA
| | - Joseph M. Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston, UK
- Faculty of Science, Manchester Metropolitan University, Manchester, UK
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | | |
Collapse
|
13
|
Urbonaite G, Knyzeliene A, Bunn FS, Smalskys A, Neniskyte U. The impact of maternal high-fat diet on offspring neurodevelopment. Front Neurosci 2022; 16:909762. [PMID: 35937892 PMCID: PMC9354026 DOI: 10.3389/fnins.2022.909762] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
A maternal high-fat diet affects offspring neurodevelopment with long-term consequences on their brain health and behavior. During the past three decades, obesity has rapidly increased in the whole human population worldwide, including women of reproductive age. It is known that maternal obesity caused by a high-fat diet may lead to neurodevelopmental disorders in their offspring, such as autism spectrum disorder, attention deficit hyperactivity disorder, anxiety, depression, and schizophrenia. A maternal high-fat diet can affect offspring neurodevelopment due to inflammatory activation of the maternal gut, adipose tissue, and placenta, mirrored by increased levels of pro-inflammatory cytokines in both maternal and fetal circulation. Furthermore, a maternal high fat diet causes gut microbial dysbiosis further contributing to increased inflammatory milieu during pregnancy and lactation, thus disturbing both prenatal and postnatal neurodevelopment of the offspring. In addition, global molecular and cellular changes in the offspring's brain may occur due to epigenetic modifications including the downregulation of brain-derived neurotrophic factor (BDNF) expression and the activation of the endocannabinoid system. These neurodevelopmental aberrations are reflected in behavioral deficits observed in animals, corresponding to behavioral phenotypes of certain neurodevelopmental disorders in humans. Here we reviewed recent findings from rodent models and from human studies to reveal potential mechanisms by which a maternal high-fat diet interferes with the neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Gintare Urbonaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agne Knyzeliene
- Centre for Cardiovascular Science, The Queen’s Medical Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Fanny Sophia Bunn
- Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Adomas Smalskys
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
14
|
Saullo C, Cruz LLD, Damasceno DC, Volpato GT, Sinzato YK, Karki B, Gallego FQ, Vesentini G. Effects of a maternal high-fat diet on adipose tissue in murine offspring: A systematic review and meta-analysis. Biochimie 2022; 201:18-32. [PMID: 35779649 DOI: 10.1016/j.biochi.2022.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 05/10/2022] [Accepted: 06/21/2022] [Indexed: 12/09/2022]
Abstract
The aim of this systematic review and meta-analysis was to analyze the influence of a maternal and/or offspring high-fat diet (HFD) on the morphology of the offspring adipocytes and amount of food and energy consumption. The search was conducted through Pubmed, EMBASE, and Web of Science databases up to October 31st, 2021. The outcomes were extracted and pooled as a standardized mean difference with random effect models. 5,004 articles were found in the databases. Of these, only 31 were selected for this systematic review and 21 were included in the meta-analysis. A large discrepancy in the percentage of fat composing the HFD (from 14% to 62% fat content) was observed. Considering the increase of adipose tissue by hyperplasia (cell number increase) and hypertrophy (cell size increase) in HFD models, the meta-analysis showed that excessive consumption of a maternal HFD influences the development of visceral white adipose tissue in offspring, related to adipocyte hypertrophy, regardless of their HFD or control diet consumption. Upon following a long-term HFD, hyperplasia was confirmed in the offspring. When analyzing the secondary outcome in terms of the amount of food and energy consumed, there was an increase of caloric intake in the offspring fed with HFD whose mothers consumed HFD. Furthermore, the adipocyte hypertrophy in different regions of the adipose tissue is related to the sex of the pups. Thus, the adipose tissue obesity phenotypes in offspring are programmed by maternal consumption of a high-fat diet, independent of postnatal diet.
Collapse
Affiliation(s)
- Carolina Saullo
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Larissa Lopes da Cruz
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil; Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Barshana Karki
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Giovana Vesentini
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil.
| |
Collapse
|
15
|
Quek SXZ, Tan EXX, Ren YP, Muthiah M, Loo EXL, Tham EH, Siah KTH. Factors early in life associated with hepatic steatosis. World J Hepatol 2022; 14:1235-1247. [PMID: 35978672 PMCID: PMC9258263 DOI: 10.4254/wjh.v14.i6.1235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/01/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The rise in prevalence of non-alcoholic fatty liver disease (NAFLD) mirrors the obesity epidemic. NAFLD is insidious but may gradually progress from simple steatosis to steatohepatitis, fibrosis and cirrhosis and/or hepatocellular carcinoma. Intervention strategies to ameliorate developmental programming of NAFLD may be more efficacious during critical windows of developmental plasticity.
AIM To review the early developmental factors associated with NAFLD.
METHODS Databases MEDLINE via PubMed, and EMBASE and Reference Citation Analysis were searched and relevant publications up to April 30, 2021 were assessed. Original research studies that included risk factors associated with early development of NAFLD in human subjects were included. These factors include: Maternal factors, intrauterine and prenatal factors, post-natal factors, genetic and ethnic predisposition, childhood and adolescence environmental factors. Studies were excluded if they were review articles or animal studies, case reports or conference abstracts, or if NAFLD was not clearly defined and assessed radiologically.
RESULTS Of 1530 citations identified by electronic search, 420 duplicates were removed. Of the 1110 citations screened from title and abstract, 80 articles were included in the final analysis. Genetic polymorphisms such as patatin-like phospholipase domain-containing protein 3 (PNPLA3) and membrane-bound O-acyltransferase domain-containing protein 7 (MBOAT7) were associated with increased risk of NAFLD. Familial factors such as maternal obesogenic environment and parental history of hepatic steatosis was associated with offspring NAFLD. Longer duration of exclusive breastfeeding in infancy was associated with a lower risk of developing NAFLD later in life while metabolic dysfunction and/or obesity in adolescence was associated with increased risk of NAFLD. Studies relating to socioeconomic factors and its association with NAFLD reported confounding results.
CONCLUSION Maternal metabolic dysfunction during pregnancy, being exclusively breastfed for a longer time postnatally, diet and physical activity in childhood and adolescence are potential areas of intervention to decrease risk of NAFLD.
Collapse
Affiliation(s)
- Sabrina Xin Zi Quek
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore 119228, Singapore
| | - Eunice Xiang-Xuan Tan
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore 119228, Singapore
- National University Centre for Organ Transplantation, National University Health System, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yi Ping Ren
- Department of Medicine, National University Hospital, Singapore 119228, Singapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore 119228, Singapore
- National University Centre for Organ Transplantation, National University Health System, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Evelyn Xiu Ling Loo
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore 119228, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
| | - Elizabeth Huiwen Tham
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore 119228, Singapore
| | - Kewin Tien Ho Siah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
16
|
Pomar CA, Picó C, Palou A, Sánchez J. Maternal Consumption of a Cafeteria Diet during Lactation Leads to Altered Diet-Induced Thermogenesis in Descendants after Exposure to a Western Diet in Adulthood. Nutrients 2022; 14:nu14091958. [PMID: 35565926 PMCID: PMC9102879 DOI: 10.3390/nu14091958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
This study investigates the ability of a maternal cafeteria diet during lactation to program brown adipose tissue (BAT) metabolic responses to an obesogenic diet re-exposure in the adult offspring after consuming a standard diet (SD). Nursing rats were fed an SD or a cafeteria diet during lactation. Their offspring (O-C and O-CAF, respectively) were weaned onto an SD, and at 16 weeks of age they were switched to a Western diet until week 24. Gene and protein expression in BAT were measured at PN22 and at 24 weeks. At PN22, compared to controls, O-CAF rats displayed lower mRNA levels of lipogenesis-related genes (Fasn), and higher expression of genes related to lipolysis (Pnpla2), fatty acid uptake (Cd36, Lpl), and oxidation (Cpt1b). Additionally, O-CAF animals displayed increased mRNA levels of Adrb3, Ucp1, and Cidea. In adulthood, these animals maintained lower mRNA levels of lipogenesis-related genes (Pparg, Srebf1, Fasn), but displayed lower expression of genes related to fatty acid uptake (Cd36), fatty acid oxidation (Cpt1b), lipolysis (Pnpla2), Adrb3, Ucp1, and Cidea. Thus, exposure to an obesogenic diet in nursing rats can affect long-term lipid metabolism and attenuate diet-induced thermogenesis in BAT in response to a new obesogenic dietary challenge later in life.
Collapse
Affiliation(s)
- Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (A.P.); (J.S.)
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (A.P.); (J.S.)
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-971-173454
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (A.P.); (J.S.)
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, 07122 Palma, Spain; (C.A.P.); (A.P.); (J.S.)
- Health Research Institute of the Balearic Islands IdISBa, 07010 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| |
Collapse
|
17
|
Gestational Exercise Increases Male Offspring's Maximal Workload Capacity Early in Life. Int J Mol Sci 2022; 23:ijms23073916. [PMID: 35409278 PMCID: PMC8999565 DOI: 10.3390/ijms23073916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Mothers’ antenatal strategies to improve the intrauterine environment can positively decrease pregnancy-derived intercurrences. By challenging the mother–fetus unit, gestational exercise (GE) favorably modulates deleterious stimuli, such as high-fat, high-sucrose (HFHS) diet-induced adverse consequences for offspring. We aimed to analyze whether GE alters maternal HFHS-consumption effects on male offspring’s maximal workload performance (MWP) and in some skeletal muscle (the soleus—SOL and the tibialis anterior—TA) biomarkers associated with mitochondrial biogenesis and oxidative fitness. Infant male Sprague-Dawley rats were divided into experimental groups according to mothers’ dietary and/or exercise conditions: offspring of sedentary control diet-fed or HFHS-fed mothers (C–S or HFHS–S, respectively) and of exercised HFHS-fed mothers (HFHS–E). Although maternal HFHS did not significantly alter MWP, offspring from GE dams exhibited increased MWP. Lower SOL AMPk levels in HFHS–S were reverted by GE. SOL PGC-1α, OXPHOS C-I and C-IV subunits remained unaltered by maternal diet, although increased in HFHS–E offspring. Additionally, GE prevented maternal diet-related SOL miR-378a overexpression, while upregulated miR-34a expression. Decreased TA C-IV subunit expression in HFHS–S was reverted in HFHS–E, concomitantly with the downregulation of miR-338. In conclusion, GE in HFHS-fed dams increases the offspring’s MWP, which seems to be associated with the intrauterine modulation of SM mitochondrial density and functional markers.
Collapse
|
18
|
Stevanović-Silva J, Beleza J, Coxito P, Costa RC, Ascensão A, Magalhães J. Fit mothers for a healthy future: Breaking the intergenerational cycle of non-alcoholic fatty liver disease with maternal exercise. Eur J Clin Invest 2022; 52:e13596. [PMID: 34120338 DOI: 10.1111/eci.13596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022]
Abstract
UNLABELLED SPECIAL ISSUE: 'FOIEGRAS-Bioenergetic Remodelling in the Pathophysiology and Treatment of Non-Alcoholic Fatty Liver Disease'. BACKGROUND Non-alcoholic fatty liver disease (NAFLD) emerges as significant health burden worldwide. Lifestyle changes, unhealthy dietary habits and physical inactivity, can trigger NAFLD development. Persisting on these habits during pregnancy affects in utero environment and prompts a specific metabolic response in foetus resulting in offspring metabolic maladjustments potentially critical for developing NAFLD later in life. The increasing prevalence of NAFLD, particularly in children, has shifted the research focus towards preventive and therapeutic strategies. Yet, designing effective approaches that can break the NAFLD intergenerational cycle becomes even more complicated. Regular physical exercise (PE) is a powerful non-pharmacological strategy known to counteract deleterious metabolic outcomes. In this narrative review, we aimed to briefly describe NAFLD pathogenesis focusing on maternal nutritional challenge and foetal programming, and to provide potential mechanisms behind the putative intergenerational effect of PE against metabolic diseases, including liver diseases. METHODS Following detailed electronic database search, recent existing evidence about NAFLD development, intergenerational programming and gestational exercise effects was critically analysed and discussed. RESULTS PE during pregnancy could have a great potential to counteract intergenerational transmission of metabolic burden. The interplay between different PE roles-metabolic, endocrine and epigenetic-could offer a more stable in utero environment to the foetus, thus rescuing offspring vulnerability to metabolic disturbances. CONCLUSIONS The better understanding of maternal PE beneficial consequences on offspring metabolism could reinforce the importance of PE during pregnancy as an indispensable strategy in improving offspring health.
Collapse
Affiliation(s)
- Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
19
|
Thompson MD, Kang J, Faerber A, Hinrichs H, Özler O, Cowen J, Xie Y, Tarr PI, Davidson NO. Maternal obesogenic diet regulates offspring bile acid homeostasis and hepatic lipid metabolism via the gut microbiome in mice. Am J Physiol Gastrointest Liver Physiol 2022; 322:G295-G309. [PMID: 34984925 PMCID: PMC8816615 DOI: 10.1152/ajpgi.00247.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mice exposed in gestation to maternal high-fat/high-sucrose (HF/HS) diet develop altered bile acid (BA) homeostasis. We hypothesized that these reflect an altered microbiome and asked if microbiota transplanted from HF/HS offspring change hepatic BA and lipid metabolism to determine the directionality of effect. Female mice were fed HF/HS or chow (CON) for 6 wk and bred with lean males. 16S sequencing was performed to compare taxa in offspring. Cecal microbiome transplantation (CMT) was performed from HF/HS or CON offspring into antibiotic-treated mice fed chow or high fructose. BA, lipid metabolic, and gene expression analyses were performed in recipient mice. Gut microbiomes from HF/HS offspring segregated from CON offspring, with increased Firmicutes to Bacteriodetes ratios and Verrucomicrobial abundance. After CMT was performed, HF/HS-recipient mice had larger BA pools, increased intrahepatic muricholic acid, and decreased deoxycholic acid species. HF/HS-recipient mice exhibited downregulated hepatic Mrp2, increased hepatic Oatp1b2, and decreased ileal Asbt mRNA expression. HF/HS-recipient mice exhibited decreased cecal butyrate and increased hepatic expression of Il6. HF/HS-recipient mice had larger livers and increased intrahepatic triglyceride versus CON-recipient mice after fructose feeding, with increased hepatic mRNA expression of lipogenic genes including Srebf1, Fabp1, Mogat1, and Mogat2. CMT from HF/HS offspring increased BA pool and shifted the composition of the intrahepatic BA pool. CMT from HF/HS donor offspring increased fructose-induced liver triglyceride accumulation. These findings support a causal role for vertical transfer of an altered microbiome in hepatic BA and lipid metabolism in HF/HS offspring.NEW & NOTEWORTHY We utilized a mouse model of maternal obesogenic diet exposure to evaluate the effect on offspring microbiome and bile acid homeostasis. We identified shifts in the offspring microbiome associated with changes in cecal bile acid levels. Transfer of the microbiome from maternal obesogenic diet-exposed offspring to microbiome-depleted mice altered bile acid homeostasis and increased fructose-induced hepatic steatosis.
Collapse
Affiliation(s)
- Michael D. Thompson
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Jisue Kang
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Austin Faerber
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Holly Hinrichs
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Oğuz Özler
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Jamie Cowen
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Yan Xie
- 2Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Phillip I. Tarr
- 3Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Nicholas O. Davidson
- 2Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
20
|
Tajaddini A, Kendig MD, Prates KV, Westbrook RF, Morris MJ. Male Rat Offspring Are More Impacted by Maternal Obesity Induced by Cafeteria Diet than Females-Additive Effect of Postweaning Diet. Int J Mol Sci 2022; 23:ijms23031442. [PMID: 35163366 PMCID: PMC8835941 DOI: 10.3390/ijms23031442] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 01/07/2023] Open
Abstract
Maternal obesity increases the risk of health complications in offspring, but whether these effects are exacerbated by offspring exposure to unhealthy diets warrants further investigation. Female Sprague-Dawley rats were fed either standard chow (n = 15) or ‘cafeteria’ (Caf, n = 21) diets across pre-pregnancy, gestation, and lactation. Male and female offspring were weaned onto chow or Caf diet (2–3/sex/litter), forming four groups; behavioural and metabolic parameters were assessed. At weaning, offspring from Caf dams were smaller and lighter, but had more retroperitoneal (RP) fat, with a larger effect in males. Maternal Caf diet significantly increased relative expression of ACACA and Fasn in male and female weanling liver, but not CPT-1, SREBP and PGC1; PPARα was increased in males from Caf dams. Maternal obesity enhanced the impact of postweaning Caf exposure on adult body weight, RP fat, liver mass, and plasma leptin in males but not females. Offspring from Caf dams appeared to exhibit reduced anxiety-like behaviour on the elevated plus maze. Hepatic CPT-1 expression was reduced only in adult males from Caf fed dams. Post weaning Caf diet consumption did not alter liver gene expression in the adult offspring. Maternal obesity exacerbated the obesogenic phenotype produced by postweaning Caf diet in male, but not female offspring. Thus, the impact of maternal obesity on adiposity and liver gene expression appeared more marked in males. Our data underline the sex-specific detrimental effects of maternal obesity on offspring.
Collapse
Affiliation(s)
- Aynaz Tajaddini
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
| | - Michael D. Kendig
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
| | - Kelly V. Prates
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
| | | | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (A.T.); (M.D.K.); (K.V.P.)
- Correspondence:
| |
Collapse
|
21
|
Marchlewicz E, McCabe C, Djuric Z, Hoenerhoff M, Barks J, Tang L, Song PX, Peterson K, Padmanabhan V, Dolinoy DC. Gestational exposure to high fat diets and bisphenol A alters metabolic outcomes in dams and offspring, but produces hepatic steatosis only in dams. CHEMOSPHERE 2022; 286:131645. [PMID: 34426127 PMCID: PMC8595757 DOI: 10.1016/j.chemosphere.2021.131645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/29/2021] [Accepted: 07/21/2021] [Indexed: 05/07/2023]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide. Perinatal development is a critical window for altered, lifelong health trajectory, and evidence supports the role of perinatal programming in chronic metabolic diseases. To examine the impact of diet and bisphenol A (BPA) on the developmental trajectory of NAFLD in offspring, we exposed dams from pre-gestation through lactation to a human-relevant dose of oral BPA coupled with intake of high fat Western or Mediterranean-style diets. We assessed hepatic steatosis by quantifying hepatic triglycerides (TGs) and metabolic health by measuring body weight, relative organ weights, and serum hormone levels in dams and offspring at postnatal day 10 (PND10) and 10-months of age. In dams, consumption of the Western or Mediterranean diet increased hepatic TGs 1.7-2.4-fold, independent of BPA intake. Among offspring, both perinatal diet and BPA exposure had a greater impact on metabolic outcomes than on hepatic steatosis. At PND10, serum leptin levels were elevated 2.6-4.8-fold in pups exposed to the Mediterranean diet, with a trend for sex-specific effects on body and organ weights. At 10-months, sex-specific increases in organ weight and hormone levels were observed in mice perinatally exposed to Western + BPA or Mediterranean + BPA. These findings suggest lifestage-specific interaction of perinatal exposures to experimental diets and BPA on offspring metabolic health without effects on NAFLD later in life. Importantly, alterations in dam phenotype by diet and BPA exposure appear to impact offspring health trajectory, emphasizing the need to define dam diet in assessing effects of environmental exposures on offspring health.
Collapse
Affiliation(s)
- Elizabeth Marchlewicz
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Carolyn McCabe
- Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Zora Djuric
- Department of Family Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John Barks
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lu Tang
- Department of Biostatistics, University of Pittsburgh, Pittsburg, PA, USA
| | - Peter X Song
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karen Peterson
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Huang Y, Osorio Mendoza J, Li M, Jin Z, Li B, Wu Y, Togo J, Speakman JR. Impact of graded maternal dietary fat content on offspring susceptibility to high-fat diet in mice. Obesity (Silver Spring) 2021; 29:2055-2067. [PMID: 34813173 DOI: 10.1002/oby.23270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/19/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Maternal high-fat diet (HFD) increases offspring obesity, yet the impacts of different levels of maternal dietary fat have seldom been addressed. In mice, the impact of graded maternal dietary fat on offspring adiposity and offspring's later susceptibility to HFD were assessed. METHODS Lactating mice were fed diets with graded fat content from 8.3% to 66.6%. One male and one female pup from each litter were weaned onto a low-fat diet for 15 weeks. HFD (41.7%) was then introduced to half of the offspring for 12 weeks. RESULTS Offspring body weight and adiposity were positively related to maternal dietary fat content and were higher when mothers were exposed to HFD. The maternal diet effect was nonlinear and sex dependent. A maternal dietary fat of 41.7% and above exaggerated the offspring body weight gain in males but was not significant in females. Maternal 8.3% fat and 25% fat diets led to the highest daily energy expenditure and respiratory exchange ratio in offspring. Offspring fed a low-fat diet had higher daily energy expenditure and respiratory exchange ratio than those fed an HFD. CONCLUSIONS Increasing maternal dietary fat during lactation, and HFD in later life, had significant and interacting impacts on offspring obesity. Maternal diet had a bigger impact on male offspring. The effects of maternal dietary fat content were nonlinear.
Collapse
Affiliation(s)
- Yi Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | | | - Min Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zengguang Jin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- School of Basic Medical Science, Dali University, Dali, Yunnan, China
| | - Baoguo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yingga Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jacques Togo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Center for Excellence in Animal Evolution and Genetics, Kunming, Yunnan, China
| |
Collapse
|
23
|
Li Y, Pollock CA, Saad S. Aberrant DNA Methylation Mediates the Transgenerational Risk of Metabolic and Chronic Disease Due to Maternal Obesity and Overnutrition. Genes (Basel) 2021; 12:genes12111653. [PMID: 34828259 PMCID: PMC8624316 DOI: 10.3390/genes12111653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/02/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is a rapidly evolving universal epidemic leading to acute and long-term medical and obstetric health issues, including increased maternal risks of gestational diabetes, hypertension and pre-eclampsia, and the future risks for offspring's predisposition to metabolic diseases. Epigenetic modification, in particular DNA methylation, represents a mechanism whereby environmental effects impact on the phenotypic expression of human disease. Maternal obesity or overnutrition contributes to the alterations in DNA methylation during early life which, through fetal programming, can predispose the offspring to many metabolic and chronic diseases, such as non-alcoholic fatty liver disease, obesity, diabetes, and chronic kidney disease. This review aims to summarize findings from human and animal studies, which support the role of maternal obesity in fetal programing and the potential benefit of altering DNA methylation to limit maternal obesity related disease in the offspring.
Collapse
Affiliation(s)
- Yan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China;
| | - Carol A. Pollock
- Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2065, Australia;
| | - Sonia Saad
- Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2065, Australia;
- Correspondence:
| |
Collapse
|
24
|
Sekkarie A, Welsh JA, Northstone K, Stein AD, Ramakrishnan U, Vos MB. Associations of maternal diet and nutritional status with offspring hepatic steatosis in the Avon longitudinal study of parents and children. BMC Nutr 2021; 7:28. [PMID: 34233762 PMCID: PMC8265091 DOI: 10.1186/s40795-021-00433-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/22/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Priming for cardiometabolic diseases, including non-alcoholic fatty liver disease (NAFLD), is hypothesized to begin in utero. The primary objective of this study is to determine whether there is an association between maternal nutritional status and offspring NAFLD. METHODS Data come from the Avon Longitudinal Study of Parents and Children (ALSPAC) in the UK. The analytic sample included 3353 participants who had maternal information on pre-pregnancy BMI, gestational weight gain, diabetes, and free sugar intake as percent of total energy and were assessed for mild-severe hepatic steatosis at 24 years by transient elastography (controlled attenuation parameter score ≥ 248 dB/m). Multiple logistic regression was used to evaluate the association between maternal factors and offspring hepatic steatosis at 24 years. RESULTS In confounder-adjusted models the independent associations for each maternal factor with mild to severe vs low hepatic steatosis at 24 years were: pre-pregnancy overweight (OR: 1.84, 95%CL: 1.43-2.38) or obesity (OR: 2.73, 95%CL: 1.84-4.03), more than recommended gestational weight gain (OR: 1.30, 95%CL: 1.04-1.64), diabetes (OR: 1.39, 95%CI: 0.87, 2.21), and high free sugar intake during pregnancy (OR: 1.04, 95% CI: 0.82, 1.33). These associations were largely mediated by BMI at 24 years, but not by birthweight or breastfeeding. CONCLUSIONS Our results suggest that maternal nutritional status is associated with the development of NAFLD in their adult offspring, although the relationship is largely mediated by offspring BMI in adulthood.
Collapse
Affiliation(s)
- Ahlia Sekkarie
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, 1518 Clifton Rd NE, Atlanta, GA, 30322, USA.
| | - Jean A Welsh
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, 1518 Clifton Rd NE, Atlanta, GA, 30322, USA
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, 30322, USA
| | - Kate Northstone
- Population Health Science, Bristol Medical School, Bristol, BS8 2BN, UK
| | - Aryeh D Stein
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, 1518 Clifton Rd NE, Atlanta, GA, 30322, USA
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Usha Ramakrishnan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Miriam B Vos
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
25
|
Kasper P, Breuer S, Hoffmann T, Vohlen C, Janoschek R, Schmitz L, Appel S, Fink G, Hünseler C, Quaas A, Demir M, Lang S, Steffen HM, Martin A, Schramm C, Bürger M, Mahabir E, Goeser T, Dötsch J, Hucklenbruch-Rother E, Bae-Gartz I. Maternal Exercise Mediates Hepatic Metabolic Programming via Activation of AMPK-PGC1α Axis in the Offspring of Obese Mothers. Cells 2021; 10:1247. [PMID: 34069390 PMCID: PMC8158724 DOI: 10.3390/cells10051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is associated with an increased risk of hepatic metabolic dysfunction for both mother and offspring and targeted interventions to address this growing metabolic disease burden are urgently needed. This study investigates whether maternal exercise (ME) could reverse the detrimental effects of hepatic metabolic dysfunction in obese dams and their offspring while focusing on the AMP-activated protein kinase (AMPK), representing a key regulator of hepatic metabolism. In a mouse model of maternal western-style-diet (WSD)-induced obesity, we established an exercise intervention of voluntary wheel-running before and during pregnancy and analyzed its effects on hepatic energy metabolism during developmental organ programming. ME prevented WSD-induced hepatic steatosis in obese dams by alterations of key hepatic metabolic processes, including activation of hepatic ß-oxidation and inhibition of lipogenesis following increased AMPK and peroxisome-proliferator-activated-receptor-γ-coactivator-1α (PGC-1α)-signaling. Offspring of exercised dams exhibited a comparable hepatic metabolic signature to their mothers with increased AMPK-PGC1α-activity and beneficial changes in hepatic lipid metabolism and were protected from WSD-induced adipose tissue accumulation and hepatic steatosis in later life. In conclusion, this study demonstrates that ME provides a promising strategy to improve the metabolic health of both obese mothers and their offspring and highlights AMPK as a potential metabolic target for therapeutic interventions.
Collapse
Affiliation(s)
- Philipp Kasper
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Saida Breuer
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Thorben Hoffmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Lisa Schmitz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christoph Hünseler
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Alexander Quaas
- Department of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany;
| | - Münevver Demir
- Charité Campus Mitte and Campus Virchow Clinic, Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, D-13353 Berlin, Germany;
| | - Sonja Lang
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Anna Martin
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Christoph Schramm
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Martin Bürger
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, D-50937 Cologne, Germany;
| | - Tobias Goeser
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| |
Collapse
|
26
|
Verduci E, Calcaterra V, Di Profio E, Fiore G, Rey F, Magenes VC, Todisco CF, Carelli S, Zuccotti GV. Brown Adipose Tissue: New Challenges for Prevention of Childhood Obesity. A Narrative Review. Nutrients 2021; 13:1450. [PMID: 33923364 PMCID: PMC8145569 DOI: 10.3390/nu13051450] [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: 03/30/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric obesity remains a challenge in modern society. Recently, research has focused on the role of the brown adipose tissue (BAT) as a potential target of intervention. In this review, we revised preclinical and clinical works on factors that may promote BAT or browning of white adipose tissue (WAT) from fetal age to adolescence. Maternal lifestyle, type of breastfeeding and healthy microbiota can affect the thermogenic activity of BAT. Environmental factors such as exposure to cold or physical activity also play a role in promoting and activating BAT. Most of the evidence is preclinical, although in clinic there is some evidence on the role of omega-3 PUFAs (EPA and DHA) supplementation on BAT activation. Clinical studies are needed to dissect the early factors and their modulation to allow proper BAT development and functions and to prevent onset of childhood obesity.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Health Sciences, University of Milan, 20146 Milan, Italy
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Valeria Calcaterra
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Elisabetta Di Profio
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Animal Sciences for Health, Animal Production and Food Safety, University of Milan, 20133 Milan, Italy
| | - Giulia Fiore
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Vittoria Carlotta Magenes
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Carolina Federica Todisco
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, 20154 Milan, Italy; (V.C.); (E.D.P.); (G.F.); (V.C.M.); (C.F.T.); (G.V.Z.)
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, University of Milan, 20157 Milan, Italy
| |
Collapse
|
27
|
Monthé-Drèze C, Rifas-Shiman SL, Aris IM, Shivappa N, Hebert JR, Sen S, Oken E. Maternal diet in pregnancy is associated with differences in child body mass index trajectories from birth to adolescence. Am J Clin Nutr 2021; 113:895-904. [PMID: 33721014 PMCID: PMC8023853 DOI: 10.1093/ajcn/nqaa398] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/24/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Nutrition in pregnancy and accelerated childhood growth are important predictors of obesity risk. Yet, it is unknown which dietary patterns in pregnancy are associated with accelerated growth and whether there are specific periods from birth to adolescence that are most sensitive to these associations. OBJECTIVES To examine the extent to which 3 dietary indices in pregnancy [Dietary Inflammatory Index (DII), Alternate Healthy Eating Index for Pregnancy (AHEI-P), and Mediterranean Diet Score (MDS)] are associated with child BMI z-score (BMI-z) trajectories from birth to adolescence. METHODS We examined 1459 mother-child dyads from Project Viva that had FFQ data in pregnancy and ≥3 child BMI-z measurements between birth and adolescence. We used linear spline mixed-effects models to examine whether BMI-z growth rates and BMI z-scores differed by quartile of each dietary index from birth to 1 mo, 1-6 mo, 6 mo to 3 y, 3-10 y, and >10 y. RESULTS The means ± SDs for DII (range, -9 to +8 units), AHEI-P (range, 0-90 points), and MDS (range, 0-9 points) were -2.6 ± 1.4 units, 61 ± 10 points, and 4.6 ± 2.0 points, respectively. In adjusted models, children of women in the highest (vs. lowest) DII quartile had higher BMI-z growth rates between 3-10 y (β, 0.03 SD units/y; 95% CI: 0.00-0.06) and higher BMI z-scores from 7 y through 10 y. Children of women with low adherence to a Mediterranean diet had higher BMI z-scores from 3 y through 15 y. Associations of AHEI-P with growth rates and BMI z-scores from birth through adolescence were null. CONCLUSIONS A higher DII and a lower MDS in pregnancy, but not AHEI-P results, are associated with higher BMI-z trajectories during distinct growth periods from birth through adolescence. Identifying the specific dietary patterns in pregnancy associated with rapid weight gain in children could inform strategies to reduce child obesity.
Collapse
Affiliation(s)
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Izzuddin M Aris
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Nitin Shivappa
- South Carolina Statewide Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC, USA,Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC, USA
| | - James R Hebert
- South Carolina Statewide Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC, USA,Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC, USA
| | - Sarbattama Sen
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| |
Collapse
|
28
|
Saengnipanthkul S, Noh HL, Friedline RH, Suk S, Choi S, Acosta NK, Tran DA, Hu X, Inashima K, Kim AM, Lee KW, Kim JK. Maternal exposure to high-fat diet during pregnancy and lactation predisposes normal weight offspring mice to develop hepatic inflammation and insulin resistance. Physiol Rep 2021; 9:e14811. [PMID: 33769706 PMCID: PMC7995551 DOI: 10.14814/phy2.14811] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022] Open
Abstract
Increasing evidence shows a potential link between the perinatal nutrient environment and metabolic outcome in offspring. Here, we investigated the effects of maternal feeding of a high-fat diet (HFD) during the perinatal period on hepatic metabolism and inflammation in male offspring mice at weaning and in early adulthood. Female C57BL/6 J mice were fed HFD or normal chow (NC) for 4 weeks before mating and during pregnancy and lactation. The male offspring mice were weaned onto an NC diet, and metabolic and molecular experiments were performed in early adulthood. At postnatal day 21, male offspring mice from HFD-fed dams (Off-HFD) showed significant increases in whole body fat mass and fasting levels of glucose, insulin, and cholesterol compared to male offspring mice from NC-fed dams (Off-NC). The RT-qPCR analysis showed two- to fivefold increases in hepatic inflammatory markers (MCP-1, IL-1β, and F4/80) in Off-HFD mice. Hepatic expression of G6Pase and PEPCK was elevated by fivefold in the Off-HFD mice compared to the Off-NC mice. Hepatic expression of GLUT4, IRS-1, and PDK4, as well as lipid metabolic genes, CD36, SREBP1c, and SCD1 were increased in the Off-HFD mice compared to the Off-NC mice. In contrast, CPT1a mRNA levels were reduced by 60% in the Off-HFD mice. At postnatal day 70, despite comparable body weights to the Off-NC mice, Off-HFD mice developed hepatic inflammation with increased expression of MCP-1, CD68, F4/80, and CD36 compared to the Off-NC mice. Despite normal body weight, Off-HFD mice developed insulin resistance with defects in hepatic insulin action and insulin-stimulated glucose uptake in skeletal muscle and brown fat, and these metabolic effects were associated with hepatic inflammation in Off-HFD mice. Our findings indicate hidden, lasting effects of maternal exposure to HFD during pregnancy and lactation on metabolic homeostasis of normal weight offspring mice.
Collapse
Affiliation(s)
- Suchaorn Saengnipanthkul
- Division of NutritionDepartment of PediatricsFaculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Hye Lim Noh
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Randall H. Friedline
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Sujin Suk
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Stephanie Choi
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Nicholas K. Acosta
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Duy A. Tran
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Xiaodi Hu
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Kunikazu Inashima
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Allison M. Kim
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Ki Won Lee
- Department of Agricultural BiotechnologyCollege of Agricultural and Life SciencesSeoul National UniversitySeoulSouth Korea
| | - Jason K. Kim
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Division of Endocrinology, Metabolism, and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Department of Agricultural BiotechnologyCollege of Agricultural and Life SciencesSeoul National UniversitySeoulSouth Korea
| |
Collapse
|
29
|
Stevanović-Silva J, Beleza J, Coxito P, Pereira S, Rocha H, Gaspar TB, Gärtner F, Correia R, Martins MJ, Guimarães T, Martins S, Oliveira PJ, Ascensão A, Magalhães J. Maternal high-fat high-sucrose diet and gestational exercise modulate hepatic fat accumulation and liver mitochondrial respiratory capacity in mothers and male offspring. Metabolism 2021; 116:154704. [PMID: 33421507 DOI: 10.1016/j.metabol.2021.154704] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/12/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Maternal high-caloric nutrition and related gestational diabetes mellitus (GDM) are associated with a high-risk for developing metabolic complications later in life and in their offspring. In contrast, exercise is recognized as a non-pharmacological strategy against metabolic dysfunctions associated to lifestyle disorders. Therefore, we investigated whether gestational exercise delays the development of metabolic alterations in GDM mothers later in life, but also protects 6-week-old male offspring from adverse effects of maternal diet. METHODS Female Sprague-Dawley rats were fed with either control (C) or high-fat high-sucrose (HFHS) diet to induce GDM and submitted to gestational exercise during the 3 weeks of pregnancy. Male offspring were sedentary and fed with C-diet. RESULTS Sedentary HFHS-fed dams exhibited increased gestational body weight gain (p < 0.01) and glucose intolerance (p < 0.01), characteristic of GDM. Their offspring had normal glucose metabolism, but increased early-age body weight, which was reverted by gestational exercise. Gestational exercise also reduced offspring hepatic triglycerides accumulation (p < 0.05) and improved liver mitochondrial respiration capacity (p < 0.05), contributing to the recovery of liver bioenergetics compromised by maternal HFHS diet. Interestingly, liver mitochondrial respiration remained increased by gestational exercise in HFHS-fed dams despite prolonged HFHS consumption and exercise cessation. CONCLUSIONS Gestational exercise can result in liver mitochondrial adaptations in GDM animals, which can be preserved even after the exercise program cessation. Exposure to maternal GDM programs liver metabolic setting of male offspring, whereas gestational exercise appears as an important preventive tool against maternal diet-induced metabolic alterations.
Collapse
Affiliation(s)
- Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal.
| | - Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal
| | - Susana Pereira
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal; CNC - Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, 3060-197 Cantanhede, Portugal
| | - Hugo Rocha
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, 4000-053 Porto, Portugal
| | - Tiago Bordeira Gaspar
- Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal; Cancer Signalling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal; Medical Faculty of University of Porto (FMUP), 4200-139 Porto, Portugal; Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Fátima Gärtner
- Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal; Department of Molecular Pathology and Immunology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313, Porto, Portugal; Glycobiology in Cancer Group, Institute of Molecular Pathology and Immunology of University of Porto (Ipatimup), University of Porto, 4200-135 Porto, Portugal
| | - Rossana Correia
- HEMS - Histology and Electron Microscopy Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135, Porto, Portugal,; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Maria João Martins
- Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal; Department of Biomedicine, Biochemistry Unit, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Tiago Guimarães
- Department of Biomedicine, Biochemistry Unit, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; Department of Clinical Pathology, São João Hospital Centre, EPE, 4200-319 Porto, Portugal
| | - Sandra Martins
- Department of Clinical Pathology, São João Hospital Centre, EPE, 4200-319 Porto, Portugal; EPIUnit, Institute of Public Health, University of Porto, 4050-091 Porto, Portugal
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, 3060-197 Cantanhede, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal
| |
Collapse
|
30
|
Yan J, Wang D, Meng Z, Yan S, Teng M, Jia M, Li R, Tian S, Weiss C, Zhou Z, Zhu W. Effects of incremental endosulfan sulfate exposure and high fat diet on lipid metabolism, glucose homeostasis and gut microbiota in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115697. [PMID: 33070067 DOI: 10.1016/j.envpol.2020.115697] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/29/2020] [Accepted: 09/17/2020] [Indexed: 06/11/2023]
Abstract
The influence of pollutants on metabolic diseases such as type 2 diabetes mellitus is an emerging field in environmental medicine. Here, we explored the effects of a low-dose endosulfan sulfate (ES), a major metabolite of the pesticide endosulfan and a bio-persistent contaminant detected in environmental and human samples, on the progress of obesity and metabolic disorders. Pregnant CD-1 mice were given ES from gestational day 6 to postnatal day 21 (short-term). After weaning, male pups of exposed dams were provided with a low-fat or a high-fat diet (LFD or HFD) and assessed after an additional 12 weeks. At the same time, one group of male pups continuously received ES (long-term). Treatment with low-dose ES, short or long-term, alleviated the development of obesity and accumulation of hepatic triglycerides induced by HFD. Analysis of gene expression, metabolic profile and gut microbiome indicates that ES treatment inhibits adipogenesis induced by HFD due to enhanced lipid catabolism, fatty acid oxidation and disturbance of gut microbiota composition. However, impaired glucose and insulin homeostasis were still conserved in HFD-fed mice exposed to ES. Furthermore, ES treatment impaired glucose tolerance, affected hepatic gene expression, fatty acids composition and serum metabolic profile, as well as disturbed gut microbiota in LFD-fed mice. In conclusion, ES treatment at levels close to the accepted daily intake during fetal development directly impact glucose homeostasis, hepatic lipid metabolism, and gut microbiome dependent on the type of diet consumed. These findings provide a better understanding of the complex interactions of environmental pollutants and diet at early life stages also in the context of metabolic disease.
Collapse
Affiliation(s)
- Jin Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Dezhen Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Zhiyuan Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Sen Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Miaomiao Teng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Ming Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Ruisheng Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Sinuo Tian
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Carsten Weiss
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Campus North, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Wentao Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
31
|
Koo S, Kim M, Cho HM, Kim I. Maternal high-fructose intake during pregnancy and lactation induces metabolic syndrome in adult offspring. Nutr Res Pract 2020; 15:160-172. [PMID: 33841721 PMCID: PMC8007412 DOI: 10.4162/nrp.2021.15.2.160] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/12/2020] [Accepted: 08/30/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND/OBJECTIVES Nutritional status and food intake during pregnancy and lactation can affect fetal programming. In the current metabolic syndrome epidemic, high-fructose diets have been strongly implicated. This study investigated the effect of maternal high-fructose intake during pregnancy and lactation on the development of metabolic syndrome in adult offspring. SUBJECTS/METHODS Drinking water with or without 20% fructose was administered to female C57BL/6J mice over the course of their pregnancy and lactation periods. After weaning, pups ate regular chow. Accu-Chek Performa was used to measure glucose levels, and a tail-cuff method was used to examine systolic blood pressure. Animals were sacrificed at 7 months, their livers were excised, and sections were stained with Oil Red O and hematoxylin and eosin (H&E) staining. Kidneys were collected for gene expression analysis using quantitative real-time Polymerase chain reaction. RESULTS Adult offspring exposed to maternal high-fructose intake during pregnancy and lactation presented with heavier body weights, fattier livers, and broader areas under the curve in glucose tolerance test values than control offspring. Serum levels of alanine aminotransferase, aspartate aminotransferase, glucose, triglycerides, and total cholesterol and systolic blood pressure in the maternal high-fructose group were higher than that in controls. However, there were no significant differences in mRNA expressions of renin-angiotensin-aldosterone system genes and sodium transporter genes. CONCLUSIONS These results suggest that maternal high-fructose intake during pregnancy and lactation induces metabolic syndrome with hyperglycemia, hypertension, and dyslipidemia in adult offspring.
Collapse
Affiliation(s)
- Soohyeon Koo
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,BK21 Plus KNU Biomedical Convergence program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Mina Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hyun Min Cho
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Inkyeom Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,BK21 Plus KNU Biomedical Convergence program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
32
|
Charron MJ, Williams L, Seki Y, Du XQ, Chaurasia B, Saghatelian A, Summers SA, Katz EB, Vuguin PM, Reznik SE. Antioxidant Effects of N-Acetylcysteine Prevent Programmed Metabolic Disease in Mice. Diabetes 2020; 69:1650-1661. [PMID: 32444367 PMCID: PMC7372077 DOI: 10.2337/db19-1129] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/20/2020] [Indexed: 12/14/2022]
Abstract
An adverse maternal in utero and lactation environment can program offspring for increased risk for metabolic disease. The aim of this study was to determine whether N-acetylcysteine (NAC), an anti-inflammatory antioxidant, attenuates programmed susceptibility to obesity and insulin resistance in offspring of mothers on a high-fat diet (HFD) during pregnancy. CD1 female mice were acutely fed a standard breeding chow or HFD. NAC was added to the drinking water (1 g/kg) of the treatment cohorts from embryonic day 0.5 until the end of lactation. NAC treatment normalized HFD-induced maternal weight gain and oxidative stress, improved the maternal lipidome, and prevented maternal leptin resistance. These favorable changes in the in utero environment normalized postnatal growth, decreased white adipose tissue (WAT) and hepatic fat, improved glucose and insulin tolerance and antioxidant capacity, reduced leptin and insulin, and increased adiponectin in HFD offspring. The lifelong metabolic improvements in the offspring were accompanied by reductions in proinflammatory gene expression in liver and WAT and increased thermogenic gene expression in brown adipose tissue. These results, for the first time, provide a mechanistic rationale for how NAC can prevent the onset of metabolic disease in the offspring of mothers who consume a typical Western HFD.
Collapse
Affiliation(s)
- Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY
- Department of Medicine and Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, New York, NY
| | - Lyda Williams
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY
| | - Yoshinori Seki
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY
| | - Xiu Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY
| | - Bhagirath Chaurasia
- Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT
| | - Ellen B Katz
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY
| | - Patricia M Vuguin
- Department of Pediatrics, Columbia University Vagelos College of Physicians & Surgeons, New York, NY
| | - Sandra E Reznik
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, New York, NY
- Department of Pathology, Albert Einstein College of Medicine, New York, NY
- Department of Pharmaceutical Sciences, St. John's University, New York, NY
| |
Collapse
|
33
|
Khaire A, Wadhwani N, Madiwale S, Joshi S. Maternal fats and pregnancy complications: Implications for long-term health. Prostaglandins Leukot Essent Fatty Acids 2020; 157:102098. [PMID: 32380367 DOI: 10.1016/j.plefa.2020.102098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022]
Abstract
Pregnancy imposes increased nutritional requirements for the well being of the mother and fetus. Maternal lipid metabolism is critical for fetal development and long-term health of the offspring as it plays a key role in energy storage, tissue growth and cell signaling. Maternal fat composition is considered as a modifiable risk for abnormal lipid metabolism and glucose tolerance during pregnancy. Data derived from observational studies demonstrate that higher intake of saturated fats during pregnancy is associated with pregnancy complications (preeclampsia, gestational diabetes mellitus and preterm delivery) and poor birth outcomes (intra uterine growth retardation and large for gestational age babies). On the other hand, prenatal long chain polyunsaturated fatty acids status is shown to improve birth outome. In this article, we discuss the role of maternal lipids during pregnancy on fetal growth and development and its consequences on the health of the offspring.
Collapse
Affiliation(s)
- Amrita Khaire
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Nisha Wadhwani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Shweta Madiwale
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India.
| |
Collapse
|
34
|
Administration of Nicotinamide Mononucleotide (NMN) Reduces Metabolic Impairment in Male Mouse Offspring from Obese Mothers. Cells 2020; 9:cells9040791. [PMID: 32218167 PMCID: PMC7226525 DOI: 10.3390/cells9040791] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal obesity impacts offspring metabolism. We sought to boost mitochondrial energy metabolism using the nicotinamide adenine dinucleotide (NAD+) precursor nicotinamide mononucleotide (NMN) to treat metabolic impairment induced by maternal and long-term post weaning over-nutrition. Male offspring of lean or obese mothers, fed chow or high fat diet (HFD) for 30 weeks post-weaning, were given NMN injection, starting at 31 weeks of age, daily for 3 weeks before sacrifice. Glucose tolerance was tested at 10, 29 and 32 weeks of age to measure short and long term effects of post-weaning HFD, and NMN treatment. Plasma insulin and triglycerides, liver triglycerides and expression of mitochondrial metabolism-related genes were measured at 34 weeks. Impaired glucose tolerance due to maternal and post weaning HFD was significantly improved by only 8 days of NMN treatment. Furthermore, in offspring of obese mothers hepatic lipid accumulation was reduced due to NMN treatment by 50% and 23% in chow and HFD fed offspring respectively. Hepatic genes involved in fat synthesis, transport and uptake were reduced, while those involved in fatty acid oxidation were increased by NMN. Overall this finding suggests short term administration of NMN could be a therapeutic approach for treating metabolic disease due to maternal and post weaning over-nutrition, even in late adulthood.
Collapse
|
35
|
Kruse M, Keyhani-Nejad F, Osterhoff MA, Pfeiffer AFH. Sexually dimorphic metabolic responses to exposure of a high fat diet during pregnancy, lactation and early adulthood in Gipr -/- mice. Peptides 2020; 125:170250. [PMID: 31917165 DOI: 10.1016/j.peptides.2019.170250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/22/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022]
Abstract
Obesity has a multifactorial origin. It is known that alterations of the intra uterine milieu induce developmental programming effects leading to metabolic diseases in offspring. Obesity is diminished in mice lacking the glucose-dependent insulinotropic polypeptide receptor (Gipr-/-) when exposed to a high fat diet (HFD). We investigated whether Gipr-/- mice are still protected from obesity when additionally exposure to a HFD during pregnancy and lactation occurs. Male and female wild type (WT) and Gipr-/- offspring received either a control/ low fat diet or HFD during pregnancy and lactation and were then either left on this diet or placed on the opposite diet after weaning until 24 weeks of life. Female WT mice showed increased body weight and adiposity when exposed to a HFD during pregnancy and lactation and post-weaning compared to female WT that received the HFD after weaning only. This exacerbated effect of a HFD during pregnancy and lactation was abolished in female Gipr-/- mice. Male Gipr-/- mice were protected from obesity to a much lesser extent. Male Gipr-/- mice exposed to a HFD during pregnancy and lactation and after weaning exhibited significantly increased fed serum glucose compared to Gipr-/- mice exposed to a HFD after weaning only. In female Gipr-/- mice no differences in fed blood glucose were observed between these groups. Our data indicate that female Gipr-/- mice are more protected from obesity. This protection is preserved in female Gipr-/- mice when additional deleterious effects of a HFD occur during fetal development.
Collapse
Affiliation(s)
- Michael Kruse
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Farnaz Keyhani-Nejad
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Martin A Osterhoff
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Andreas F H Pfeiffer
- Department of Clinical Nutrition, German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Department for Endocrinology, Diabetes and Nutrition, Charité - University of Medicine, Hindenburgdamm 30, 12200 Berlin, Germany; German Center for Diabetes Research, Germany.
| |
Collapse
|
36
|
Li T, Gong H, Yuan Q, Du M, Ren F, Mao X. Supplementation of polar lipids-enriched milk fat globule membrane in high-fat diet-fed rats during pregnancy and lactation promotes brown/beige adipocyte development and prevents obesity in male offspring. FASEB J 2020; 34:4619-4634. [PMID: 32020679 DOI: 10.1096/fj.201901867rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
Promoting brown adipose tissue (BAT) function or browning of white adipose tissue (WAT) provides a defense against obesity. The aim of the study was to investigate whether maternal polar lipids-enriched milk fat globule membrane (MFGM-PL) supplementation to high-fat diet (HFD) rats during pregnancy and lactation could promote brown/beige adipogenesis and protect against HFD-induced adiposity in offspring. Female SD rats were fed a HFD for 8 weeks to induce obesity and, then, fed a HFD during pregnancy and lactation with or without MFGM-PL. Male offspring were weaned at postnatal Day 21 and then fed a HFD for 9 weeks. MFGM-PL treatment to HFD dams decreased the body weight gain and WAT mass as well as lowered the serum levels of insulin and triglycerides in male offspring at weaning. MFGM-PL+HFD offspring showed promoted thermogenic function in BAT and inguinal WAT through the upregulation of UCP1 and other thermogenic genes. In adulthood, maternal MFGM-PL supplementation reduced adiposity and increased oxygen consumption, respiratory exchange ratio, and heat production in male offspring. The enhancement of energy expenditure was correlated with elevated BAT activity and inguinal WAT thermogenic program. In conclusion, maternal MFGM-PL treatment activated thermogenesis in offspring, which exerted long-term beneficial effects against HFD-induced obesity in later life.
Collapse
Affiliation(s)
- Tiange Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Han Gong
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Qichen Yuan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Xueying Mao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| |
Collapse
|
37
|
Wang X, Yang Y, Zhu P, Wu Y, Jin Y, Yu S, Wei H, Qian M, Cao W, Xu S, Liu Y, Chen G, Zhao X. Prenatal exposure to diesel exhaust PM 2.5 programmed non-alcoholic fatty liver disease differently in adult male offspring of mice fed normal chow and a high-fat diet. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 255:113366. [PMID: 31668954 DOI: 10.1016/j.envpol.2019.113366] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/29/2019] [Accepted: 10/07/2019] [Indexed: 06/10/2023]
Abstract
Air pollution is one of the leading preventable threats to public health. Emerging evidence indicates that exposure to environmental stressors is associated with abnormal foetal development. However, how prenatal exposure to diesel exhaust PM2.5 (DEP) predisposes adult offspring to the development of non-alcoholic fatty liver disease (NAFLD) remains unclear. To examine this, C57BL/6J mice were exposed to DEP or a vehicle before conception and during pregnancy and fed normal chow or a high-fat diet. Then, the hepatic fatty accumulation in the adult male offspring and possible molecular mechanisms were assessed. Our data showed that prenatal exposure to DEP on normal chow led to hepatic steatosis in adult male offspring with normal liver function. However, prenatal DEP exposure relieved the hepatic steatosis and liver function in offspring of mice fed a high-fat diet. Furthermore, prenatal exposure to DEP on normal chow increased lipogenesis and worsened fatty acid oxidation. The counteractive effect of prenatal DEP exposure on high-fat-diet-induced hepatic steatosis was produced through upregulated adenosine 5'-monophosphate-activated protein kinase, and this improved lipogenesis and fatty acid oxidation. Collectively, prenatal exposure to DEP programmed the development of NAFLD differently in the adult male offspring of mice fed normal chow and a high-fat diet, showing the pleotrophic effects of exposure to adverse environmental factors in early life.
Collapse
Affiliation(s)
- Xiaoke Wang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Yuxue Yang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Piaoyu Zhu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Yifan Wu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Yang Jin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Shali Yu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Haiyan Wei
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Muzhou Qian
- Department of Hemodialysis, Fourth People's Hospital of Nantong City, Nantong, 226019, China
| | - Weiming Cao
- School of Humanities and Management, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shenya Xu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Yingqi Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| |
Collapse
|
38
|
Obesity during pregnancy results in maternal intestinal inflammation, placental hypoxia, and alters fetal glucose metabolism at mid-gestation. Sci Rep 2019; 9:17621. [PMID: 31772245 PMCID: PMC6879619 DOI: 10.1038/s41598-019-54098-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022] Open
Abstract
We investigated whether diet-induced changes in the maternal intestinal microbiota were associated with changes in bacterial metabolites and their receptors, intestinal inflammation, and placental inflammation at mid-gestation (E14.5) in female mice fed a control (17% kcal fat, n = 7) or a high-fat diet (HFD 60% kcal fat, n = 9; ad libitum) before and during pregnancy. Maternal diet-induced obesity (mDIO) resulted in a reduction in maternal fecal short-chain fatty acid producing Lachnospiraceae, lower cecal butyrate, intestinal antimicrobial peptide levels, and intestinal SCFA receptor Ffar3, Ffar2 and Hcar2 transcript levels. mDIO increased maternal intestinal pro-inflammatory NFκB activity, colonic CD3+ T cell number, and placental inflammation. Maternal obesity was associated with placental hypoxia, increased angiogenesis, and increased transcript levels of glucose and amino acid transporters. Maternal and fetal markers of gluconeogenic capacity were decreased in pregnancies complicated by obesity. We show that mDIO impairs bacterial metabolite signaling pathways in the mother at mid-gestation, which was associated with significant structural changes in placental blood vessels, likely as a result of placental hypoxia. It is likely that maternal intestinal changes contribute to adverse maternal and placental adaptations that, via alterations in fetal hepatic glucose handling, may impart increased risk of metabolic dysfunction in offspring.
Collapse
|
39
|
Moody L, Shao J, Chen H, Pan YX. Maternal Low-Fat Diet Programs the Hepatic Epigenome despite Exposure to an Obesogenic Postnatal Diet. Nutrients 2019; 11:nu11092075. [PMID: 31484384 PMCID: PMC6769607 DOI: 10.3390/nu11092075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity and metabolic disease present a danger to long-term health outcomes. It has been hypothesized that epigenetic marks established during early life might program individuals and have either beneficial or harmful consequences later in life. In the present study, we examined whether maternal diet alters DNA methylation and whether such modifications persist after an obesogenic postnatal dietary challenge. During gestation and lactation, male Sprague-Dawley rats were exposed to either a high-fat diet (HF; n = 10) or low-fat diet (LF; n = 10). After weaning, all animals were fed a HF diet for an additional nine weeks. There were no differences observed in food intake or body weight between groups. Hepatic DNA methylation was quantified using both methylated DNA immunoprecipitation sequencing (MeDIP-seq) and methylation-sensitive restriction enzyme sequencing (MRE-seq). Overall, 1419 differentially methylated regions (DMRs) were identified. DMRs tended to be located in CpG shores and were enriched for genes involved in metabolism and cancer. Gene expression was measured for 31 genes in these pathways. Map3k5 and Igf1r were confirmed to be differentially expressed. Finally, we attempted to quantify the functional relevance of intergenic DMRs. Using chromatin contact data, we saw that conserved DMRs were topologically associated with metabolism genes, which were associated with differential expression of Adh5, Enox1, and Pik3c3. We show that although maternal dietary fat is unable to reverse offspring weight gain in response to a postnatal obesogenic diet, early life diet does program the hepatic methylome. Epigenetic alterations occur primarily in metabolic and cancer pathways and are associated with altered gene expression, but it is unclear whether they bear consequence later in life.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Justin Shao
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Exeter High School, 1 Blue Hawk Drive, Exeter, NH 03833, USA
| | - Hong Chen
- Department of Food Science and Human Nutrition, Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yuan-Xiang Pan
- Department of Food Science and Human Nutrition, Division of Nutritional Sciences, and Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
40
|
Sadagurski M, Debarba LK, Werneck-de-Castro JP, Ali Awada A, Baker TA, Bernal-Mizrachi E. Sexual dimorphism in hypothalamic inflammation in the offspring of dams exposed to a diet rich in high fat and branched-chain amino acids. Am J Physiol Endocrinol Metab 2019; 317:E526-E534. [PMID: 31361548 PMCID: PMC6766606 DOI: 10.1152/ajpendo.00183.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Branched-chain amino acid (BCAAs: leucine, isoleucine, and valine) contribute to the development of obesity-associated insulin resistance in the context of consumption of a high-fat diet (HFD) in humans and rodents. Maternal diet is a major determinant of offspring health, and there is strong evidence that maternal HFD alters hypothalamic developmental programming and disrupts offspring energy homeostasis in rodents. In this study, we exposed pregnant and lactating C57BL/6JB female mice to either HFD, HFD with supplemented BCAA (HFD+BCAA), or standard diet (SC), and we studied offspring metabolic phenotypes. Both maternal HFD and HFD supplemented with BCAA had similar effect rendering the offspring metabolic imbalance and impairing their ability to cope with HFD when challenged during aging. The metabolic effects of HFD challenge were more profound in females, worsening female offspring ability to cope with an HFD challenge by activating hypothalamic inflammation in aging. Moreover, the sex differences in hypothalamic estrogen receptor α (ER-α) expression levels were lost in female offspring upon HFD challenge, supporting a link between ER-α levels and hypothalamic inflammation in offspring and highlighting the programming potential of hypothalamic inflammatory responses and maternal nutrition.
Collapse
Affiliation(s)
- Marianna Sadagurski
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, Michigan
| | - Lucas Kniess Debarba
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, Michigan
| | - Joao Pedro Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
| | - Abear Ali Awada
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, Michigan
| | - Tess A Baker
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
- Miami Veterans Affairs Health Care System, Miami, Florida
| |
Collapse
|
41
|
Bayandor P, Farajdokht F, Mohaddes G, Diba R, Hosseindoost M, Mehri K, Zavvari Oskuye Z, Babri S. The effect of troxerutin on anxiety- and depressive-like behaviours in the offspring of high-fat diet fed dams. Arch Physiol Biochem 2019; 125:156-162. [PMID: 29482367 DOI: 10.1080/13813455.2018.1443142] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This study aimed at investigating the metabolic and behavioural effects of troxerutin treatment in the offspring of high fat diet (HFD) fed dams. Female Wistar rats (n = 40) received normal diet (ND) or HFD for 8 weeks prior to breeding. After mating, pregnant animals were assigned to four subgroups: ND, ND + Tro (troxerutin 150 mg/kg/day), HFD, and HFD + Tro. On the 21st day, male offspring were weaned and fed ND until 12 weeks old. Behavioural tests were performed on postnatal day (PND) 80 and 90. Compared to the controls, the HFD offspring showed more anxiety- and depressive-like behaviours, higher blood glucose, cholesterol, and cortisol levels. On the other hand, chronic troxerutin administration during gestation restored metabolic and behavioural changes to normal. In summary, troxerutin improved anxiety- and depressive-like behaviours, as well as metabolic status in the offspring of the HFD fed dams. More studies are needed to determine the underlying mechanisms.
Collapse
Affiliation(s)
- Parvin Bayandor
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Fereshteh Farajdokht
- b Neurosciences Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Gisou Mohaddes
- b Neurosciences Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Roghayeh Diba
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Maryam Hosseindoost
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Keyvan Mehri
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Zohreh Zavvari Oskuye
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Shirin Babri
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
42
|
Sex-associated preventive effects of low-dose aspirin on obesity and non-alcoholic fatty liver disease in mouse offspring with over-nutrition in utero. J Transl Med 2019; 99:244-259. [PMID: 30413815 PMCID: PMC6354253 DOI: 10.1038/s41374-018-0144-2] [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: 06/04/2018] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 01/04/2023] Open
Abstract
Aspirin has been found to diminish hypertriglyceridemia and hyperglycemia in both obese rodents and patients with type 2 diabetes mellitus. We aimed to test whether low-dose aspirin can prevent obesity and the progression of non-alcoholic fatty liver disease (NAFLD) in high-risk subjects. We used offspring mice with maternal over-nutrition as a high-risk model of obesity and NAFLD. The offspring were given postnatal HF-diet and diethylnitrosamine (DEN) to induce obesity and NAFLD, and were treated with or without a low dose of aspirin for 12 weeks (ASP or CTL groups). Aspirin treatment reduced body weight gain, reversed glucose intolerance, and depressed hepatic lipid accumulation in female, but not in male mice. Female mice displayed re-sensitized insulin/Akt signaling and overactivated AMPK signaling, with enhanced level of hepatic PPAR-γ, Glut4, and Glut2, while male mice only enhanced hepatic PPAR-α and PPAR-γ levels. The female ASP mice had inhibited p44/42 MAPK activity and enhanced Pten expression, while male displayed activated p38 MAPK signaling. Furthermore, the female but not the male ASP mice reduced Wnt-signaling activity via both the epigenetic regulation of Apc expression and the post-transcriptional regulation of β-catenin degradation. In summary, our study demonstrates a sex-associated effect of low-dose aspirin on obesity and NAFLD prevention in female but not in male mice.
Collapse
|
43
|
Maternal dyslipidaemic diet induces sex-specific alterations in intestinal function and lipid metabolism in rat offspring. Br J Nutr 2019; 121:721-734. [DOI: 10.1017/s0007114519000011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractThis study investigated the effects of a maternal dyslipidaemic (DLP) diet on lipid metabolism, microbial counts in faeces and hepatic and intestinal morphology in rat offspring with respect to sex during different phases of life. Wistar rats (dams) were fed a control (CTL) or DLP during gestation and lactation. After weaning, CTL and DLP offspring were fed a standard diet. The effects of a maternal DLP on body composition, biochemical parameters, faecal microbiota and intestinal and hepatic histomorphometric characteristics in rat offspring were evaluated at 30 and 90 d of age. The DLP diet during gestation and lactation caused lower birth weight and a greater weight gain percentage at the end of the 90-d period in both male and female offspring. Female pups from DLP dams had higher liver fat levels compared with CTL (P≤0·001) at 90 d of age. Males from DLP dams had greater visceral fat weight and lower Lactobacillus spp. faecal counts at 90 d of age (P≤0·001) as well as lower faecal fat excretion (P≤0·05) and Bacteroides spp. faecal counts (P≤0·001) at 30 d of age when compared with pups from CTL dams. However, both dams and DLP pups showed damage to intestinal villi. A maternal DLP alters intestinal function and lipid metabolism in a sex-specific manner and is a potential predisposing factor for health complications in offspring from the juvenile period to the adult period.
Collapse
|
44
|
Moody L, Xu GB, Chen H, Pan YX. Epigenetic regulation of carnitine palmitoyltransferase 1 (Cpt1a) by high fat diet. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:141-152. [PMID: 30605728 DOI: 10.1016/j.bbagrm.2018.12.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022]
Abstract
Carnitine palmitoyltransferase 1 (Cpt1a) is a rate-limiting enzyme that mediates the transport of fatty acids into the mitochondria for subsequent beta-oxidation. The objective of this study was to uncover how diet mediates the transcriptional regulation of Cpt1a. Pregnant Sprague Dawley rats were exposed to either a high-fat (HF) or low-fat control diet during gestation and lactation. At weaning, male offspring received either a HF or control diet, creating 4 groups: lifelong control diet (C/C; n = 12), perinatal HF diet (HF/C; n = 9), post-weaning HF diet (C/HF; n = 10), and lifelong HF diet (HF/HF; n = 10). Only HF/HF animals had higher hepatic Cpt1a mRNA expression than C/C. Epigenetic analysis revealed reduced DNA methylation (DNAMe) and increased histone 3 lysine 4 dimethylation (H3K4Me2) upstream and within the promoter of Cpt1a in the HF/HF group. This was accompanied by increased peroxisome proliferator activated receptor alpha (PPARα) and CCAAT/enhancer binding protein beta (C/EBPβ) binding directly downstream of the Cpt1a transcription start site within the first intron. Findings were confirmed in rat hepatoma H4IIEC3 cells treated with non-esterified fatty acid (NEFA). After 12 h of NEFA treatment, there was an enrichment of SWI/SNF related matrix associated actin dependent regulator of chromatin subfamily D member 1 (BAF60a or SMARCD1) in the first intron of Cpt1a. We conclude that dietary fat elevates hepatic Cpt1a expression via a highly coordinated transcriptional mechanism involving increased H3K4Me2, reduced DNAMe, and recruitment of C/EBPβ, PPARα, PGC1α, and BAF60a to the gene.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| | - Guanying Bianca Xu
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| | - Hong Chen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America; Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.
| |
Collapse
|
45
|
Grape Juice Consumption with or without High Fat Diet during Pregnancy Reduced the Weight Gain and Improved Lipid Profile and Oxidative Stress Levels in Liver and Serum from Wistar Rats. BEVERAGES 2018. [DOI: 10.3390/beverages4040078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The aim of this study was to evaluate the effects of high fat diet with or without grape juice during the pregnancy on gestational weight gain, biochemical parameters, and oxidative stress in plasma and liver from Wistar rats. Forty-nine rats were divided into four groups: control diet group (CD), high fat diet (HFD), grape juice and control diet (PGJCD), and grape juice and high fat diet (PGJHFD). During the treatment the weight gain of the rats was tracked. They had free access to their respective diets during 42 days of treatment. After offspring weaning, the mother rats were euthanized and blood and liver were collected. The high fat diet increased the total cholesterol and triglycerides serum levels as well as carbonyl levels in the liver, however this diet reduced the high-density lipoprotein (HDL) and urea levels in serum. Grape juice consumption reduced gestational body weight gain. In liver, the juice consumption increased sulfhydryl levels and reduced the superoxide dismutase (SOD) activity and TBARS level, in serum the consumption reduced aspartate aminotransferase (AST) and TBARS. We can conclude that the consumption of a diet rich in fat can promotes harmful effects on health during pregnancy, however the consumption of grape juice seems to be an important alternative to prevent oxidative damages and to promote the improvement of health.
Collapse
|
46
|
Lomas-Soria C, Reyes-Castro LA, Rodríguez-González GL, Ibáñez CA, Bautista CJ, Cox LA, Nathanielsz PW, Zambrano E. Maternal obesity has sex-dependent effects on insulin, glucose and lipid metabolism and the liver transcriptome in young adult rat offspring. J Physiol 2018; 596:4611-4628. [PMID: 29972240 DOI: 10.1113/jp276372] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/15/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Maternal high-fat diet consumption predisposes to metabolic dysfunction in male and female offspring at young adulthood. Maternal obesity programs non-alcoholic fatty liver disease (NAFLD) in a sex-dependent manner. We demonstrate sex-dependent liver transcriptome profiles in rat offspring of obese mothers. In this study, we focused on pathways related to insulin, glucose and lipid signalling. These results improve understanding of the mechanisms by which a maternal high-fat diet affects the offspring. ABSTRACT Maternal obesity (MO) predisposes offspring (F1) to obesity, insulin resistance (IR) and non-alcoholic fatty liver disease (NAFLD). MO's effects on the F1 liver transcriptome are poorly understood. We used RNA-seq to determine the liver transcriptome of male and female F1 of MO and control-fed mothers. We hypothesized that MO-F1 are predisposed to sex-dependent adult liver dysfunction. Female Wistar rat mothers ate a control (C) or obesogenic (MO) diet from the time they were weaned through breeding at postnatal day (PND) 120, delivery and lactation. After weaning, all male and female F1 ate a control diet. At PND 110, F1 serum, liver and fat were collected to analyse metabolites, histology and liver differentially expressed genes. Male and female MO-F1 showed increased adiposity index, triglycerides, insulin and homeostatic model assessment vs. C-F1 with similar body weight and glucose serum concentrations. MO-F1 males presented greater physiological and histological NAFLD characteristics than MO-F1 females. RNA-seq revealed 1365 genes significantly changed in male MO-F1 liver and only 70 genes in female MO-F1 compared with controls. GO and KEGG analysis identified differentially expressed genes related to metabolic processes. Male MO-F1 liver showed the following altered pathways: insulin signalling (22 genes), phospholipase D signalling (14 genes), NAFLD (13 genes) and glycolysis/gluconeogenesis (7 genes). In contrast, few genes were altered in these pathways in MO-F1 females. In summary, MO programs sex-dependent F1 changes in insulin, glucose and lipid signalling pathways, leading to liver dysfunction and insulin resistance.
Collapse
Affiliation(s)
- Consuelo Lomas-Soria
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez, Tlalpan, México, D.F., 14080, México.,CONACyT, Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SZ, México
| | - Luis A Reyes-Castro
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez, Tlalpan, México, D.F., 14080, México
| | - Guadalupe L Rodríguez-González
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez, Tlalpan, México, D.F., 14080, México
| | - Carlos A Ibáñez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez, Tlalpan, México, D.F., 14080, México
| | - Claudia J Bautista
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez, Tlalpan, México, D.F., 14080, México
| | - Laura A Cox
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Peter W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA.,Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Elena Zambrano
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez, Tlalpan, México, D.F., 14080, México
| |
Collapse
|
47
|
Sen S, Rifas-Shiman SL, Shivappa N, Wirth MD, Hebert JR, Gold DR, Gillman MW, Oken E. Associations of prenatal and early life dietary inflammatory potential with childhood adiposity and cardiometabolic risk in Project Viva. Pediatr Obes 2018; 13:292-300. [PMID: 28493362 PMCID: PMC5681442 DOI: 10.1111/ijpo.12221] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 12/16/2016] [Accepted: 03/24/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Limited information exists regarding the association between early-life diet and cardiometabolic risk. OBJECTIVES Examine associations of dietary inflammatory index (DII) in pregnancy and early childhood (3-5 years) with adiposity, blood pressure and metabolic markers in mid-childhood (6-10 years). METHODS Among 992 mother-child pairs from Project Viva, a pre-birth cohort, we examined associations of DII scores with outcomes using multivariable linear regression adjusted for child age and sex and maternal age, BMI, education, parity, smoking, race and income. RESULTS Mean (SD) maternal DII in pregnancy was -2.6(1.4) units and in child DII in early childhood was 0.3(0.7). Mean mid-childhood BMI z-score was 0.40(0.98) units. In boys only, DII in early childhood was associated with higher BMIz (adjusted β = 0.16 units per unit DII, 95%CI 0.02, 0.29), waist circumference (0.93 cm; -0.07, 1.92) and skin fold thicknesses (1.12 mm; 0.01, 2.23). Dietary inflammatory index in the highest quartiles during both pregnancy and in early childhood, compared to the lowest quartiles, was associated with higher waist circumference (2.4 cm; 0.14, 4.6) in all children, and BMIz in boys (0.78 units; 0.34, 1.22). Associations with BP and metabolic markers were null. CONCLUSIONS A pro-inflammatory diet in pregnancy and early childhood may promote the development of adiposity.
Collapse
Affiliation(s)
- Sarbattama Sen
- Pediatric Newborn Medicine, Brigham and Women’s Hospital (SS)
| | - Sheryl L. Rifas-Shiman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute (SLR-S, MWG, EO)
| | - Nitin Shivappa
- South Carolina Statewide Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, University of South Carolina (NS, MDW, JRH)
- Connecting Health Innovations, LLC (NS, MDW, JRH)
| | - Michael D. Wirth
- South Carolina Statewide Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, University of South Carolina (NS, MDW, JRH)
- Connecting Health Innovations, LLC (NS, MDW, JRH)
| | - James R. Hebert
- South Carolina Statewide Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, University of South Carolina (NS, MDW, JRH)
- Connecting Health Innovations, LLC (NS, MDW, JRH)
| | - Diane R. Gold
- Channing Laboratory, Brigham and Women’s Hospital (DRG)
| | - Matthew W. Gillman
- Environmental Influences on Child Health Outcomes (ECHO) Program, Office of the Director, National Institutes of Health
| | - Emily Oken
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute (SLR-S, MWG, EO)
| |
Collapse
|
48
|
Itoh H, Kanayama N. Developmental Origins of Nonalcoholic Fatty Liver Disease (NAFLD). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1012:29-39. [PMID: 29956192 DOI: 10.1007/978-981-10-5526-3_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a hepatic manifestation of metabolic syndrome. Its prevalence is currently increasing not only in developed obese countries but also in developing countries. Recent findings from human cohorts and animal studies suggest that a nutritional imbalance in the early critical period is causatively associated with the incidence of NAFLD in later life. Based on the current theory of the developmental origins of health and disease (DOHaD), undernourishment and overnourishment in utero are both hypothesized to prime the predisposition for hepatic fat storage. Current knowledge on the developmental origins of NAFLD is introduced in this chapter.
Collapse
Affiliation(s)
- Hiroaki Itoh
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.
| | - Naohiro Kanayama
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| |
Collapse
|
49
|
Baik M, Kang HJ, Park SJ, Na SW, Piao M, Kim SY, Fassah DM, Moon YS. TRIENNIAL GROWTH AND DEVELOPMENT SYMPOSIUM: Molecular mechanisms related to bovine intramuscular fat deposition in the longissimus muscle. J Anim Sci 2017; 95:2284-2303. [PMID: 28727015 DOI: 10.2527/jas.2016.1160] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The intramuscular fat (IMF) content of the LM, also known as marbling, is particularly important in determining the price of beef in Korea, Japan, and the United States. Deposition of IMF is influenced by both genetic (e.g., breed, gender, and genotype) and nongenetic factors (e.g., castration, nutrition, stressors, animal weight, and age). Castration of bulls markedly increases deposition of IMF, resulting in improved beef quality. Here, we present a comparative gene expression approach between bulls and steers. Transcriptomic and proteomic studies have demonstrated that the combined effects of increases in lipogenesis, fatty acid uptake, and fatty acid esterification and decreased lipolysis are associated with increased IMF deposition in the LM. Several peripheral tissues (LM, adipose tissues, and the liver) are involved in lipid metabolism. Therefore, understanding the significance of the tissue network in lipid metabolism is important. Here, we demonstrate that lipid metabolism in LM tissues is crucial for IMF deposition, whereas lipid metabolism in the liver plays only a minor role. Metabolism of body fat and IMF deposition in bovine species has similarities with these processes in metabolic diseases, such as obesity in humans and rodents. Extensive studies on metabolic diseases using epigenome modification (DNA methylation, histone modification, and microRNA), microbial metagenomics, and metabolomics have been performed in humans and rodents, and new findings have been reported using these technologies. The importance of applying "omics" fields (epigenomics, metagenomics, and metabolomics) to the study of IMF deposition in cattle is described. New information on the molecular mechanisms of IMF deposition may be used to design nutritional or genetic methods to manipulate IMF deposition and to modify fatty acid composition in beef cattle. Applying nutrigenomics could maximize the expression of genetic potential of economically important traits (e.g., marbling) in animals.
Collapse
|
50
|
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: 4.1] [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
|