1
|
Zhang JK, Li XP, Tang Y, Zeng LP, Liu X, Zhang JL, Chen CY, Zheng S, Liu ZZ, Gong X, Jose PA, Guo L, Zeng CY. Semaglutide ameliorates metabolic disorders in offspring via regulation of oocyte ROS of pre-pregnancy obesity mice. Acta Pharmacol Sin 2025:10.1038/s41401-025-01501-1. [PMID: 39984623 DOI: 10.1038/s41401-025-01501-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Pre-pregnancy obesity (PPO) seriously threatens the health of both mother and offspring. Pre-pregnancy weight management is particularly important for the prevention of metabolic diseases in offspring. Semaglutide is one of the most effective glucagon-like peptide-1 agonizts for the management of obesity and metabolic diseases, but little is known about its effect on the long-term health of offspring. In this study we investigated the effects of semaglutide administered before pregnancy on the offspring health from PPO mice. PPO mice model was established by feeding with high-fat diet for 16 weeks, and then injected with semaglutide (30 nmol/kg-1·d-1, sc.) for 22 days before pregnancy. After the treatment, the mice were mated with normal males or underwent in vitro fertilization (IVF) for offspring reproduction. We showed that the semaglutide treatment not only improved the lipid and glucose metabolic disorders and fertility of PPO mice, but also significantly reversed the overweight, impaired energy balance, adipose inflammatory state, lipid and glucose metabolic disorders and insulin resistance of their IVF offspring. By conducting RNA-seq analysis, SOD activity and malondialdehyde assays in ovaries, as well as ROS staining in oocytes, we revealed that the semaglutide treatment reduced the elevated oxidative stress in ovaries and high ROS levels in oocytes from PPO mice, possibly through activating the PI3K/AKT pathway and improving the state of SOD. Interestingly, incubation of oocytes from semaglutide-treated dams with H2O2 (100 μM) in vitro during IVF blocked the protective effects of semaglultide against the metabolic disorders in the offspring. In conclusion, semaglutide treatment before pregnancy effectively alleviates obesity-related metabolic disorders in offspring. The regulation of ROS in oocytes plays a crucial role in the protective effects of semaglutide.
Collapse
Affiliation(s)
- Jun-Kai Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, Sichuan Provincial Corps Hospital of Chinese People's Armed Police Force (Sichuan Corps Hospital of PAP), Leshan, 614000, China
| | - Xiao-Ping Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410081, China
| | - Yang Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Li-Ping Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xuan Liu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Jian-Li Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Cai-Yu Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Zhi-Zhao Liu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Xue Gong
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Li Guo
- Endocrinology Department, The First Affiliated Hospital of The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China.
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
2
|
Meakin AS, Nathanielsz PW, Li C, Huber HF, Clifton VL, Wiese MD, Morrison JL. Maternal obesogenic diet during pregnancy and its impact on fetal hepatic function in baboons. Obesity (Silver Spring) 2024; 32:1910-1922. [PMID: 39210592 PMCID: PMC11421985 DOI: 10.1002/oby.24124] [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: 03/11/2024] [Revised: 06/08/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Maternal obesity (MO) increases the risk of later-life liver disease in offspring, especially in males. This may be due to impaired cytochrome P450 (CYP) enzyme activity driven by an altered maternal-fetal hormonal milieu. MO increases fetal cortisol concentrations that may increase CYP activity; however, glucocorticoid receptor (GR)-mediated signaling can be modulated by alternative GR isoform expression. We hypothesized that MO induces sex-specific changes in GR isoform expression and localization that contribute to reduced hepatic CYP activity. METHODS Nonpregnant, nulliparous female baboons were assigned to either an ad libitum control diet or a high-fat, high-energy diet (HF-HED) at 9 months pre pregnancy. At 165 days' gestation (term = 180 days), fetal liver samples were collected (n = 6/sex/group). CYP activity was quantified using functional assays, and GR was measured using quantitative RT-PCR and Western blot. RESULTS CYP3A activity was reduced in the HF-HED group, whereas CYP2B6 activity was reduced in HF-HED males only. Total GR expression was increased in the HF-HED group. Relative nuclear expression of the antagonistic GR isoform GRβ was increased in HF-HED males only. CONCLUSIONS Reduced CYP activity in HF-HED males may be driven in part by dampened hepatic-specific glucocorticoid signaling via altered GR isoform expression. These findings highlight targetable mechanisms that may reduce later-life sex-specific disease risk.
Collapse
Affiliation(s)
- Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical & Health Sciences, University of South Australia, Adelaide, SA, AUS
| | | | - Cun Li
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Hillary F. Huber
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Vicki L. Clifton
- Mater Medical Research Institute – The University of Queensland, Brisbane, QLD, AUS
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences University of South Australia, Adelaide, SA, AUS
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical & Health Sciences, University of South Australia, Adelaide, SA, AUS
| |
Collapse
|
3
|
Meakin AS, Nathanielsz PW, Li C, Clifton VL, Wiese MD, Morrison JL. Maternal obesity impacts fetal liver androgen signalling in a sex-specific manner. Life Sci 2024; 337:122344. [PMID: 38081408 DOI: 10.1016/j.lfs.2023.122344] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/22/2023]
Abstract
BACKGROUND Maternal obesity (MO) increases fetal androgen concentrations, the prevalence of macrosomia, and predisposes offspring to metabolic dysfunction in later life, especially males. These risks may be, in part, the result of increased liver-specific androgen signalling pathway activity in utero. Androgen signalling activity can be suppressed by androgen metabolism via cytochrome P450 (CYP) isoenzymes (CYP2B6, CYP3A) or through inhibition of the full-length androgen receptor (AR-FL) via the antagonistic isoform, AR-45. We hypothesised MO impairs CYP enzyme activity and AR-45 expression in male fetal livers, thereby enhancing activity of androgen signalling pathways. METHODS Nine months prior to pregnancy, nulliparous female baboons were assigned to either ad libitum control or high fat diet. At 165 day (d) gestation (term, 180 d) fetal liver was collected (n = 6/sex/group). CYP activity was quantified using functional assays; subcellular AR expression was measured using Western blot. RESULTS CYP2B6 and CYP3A activity, and nuclear expression of AR-45, was reduced in MO males only. Nuclear AR-45 expression was inversely related with fetal body weight of MO males only. CONCLUSIONS Reduced CYP2B6 and CYP3A activity in conjunction with decreased nuclear AR-45 expression may enhance liver androgen signalling in males from MO pregnancies, thereby increasing the risk of macrosomia, as well as metabolic dysfunction in later life.
Collapse
Affiliation(s)
- Ashley S Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical & Health Sciences, University of South Australia, Adelaide, SA, Australia.
| | | | - Cun Li
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Vicki L Clifton
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Michael D Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, Clinical & Health Sciences, University of South Australia, Adelaide, SA, Australia.
| |
Collapse
|
4
|
Barrera C, Castillo V, Valenzuela R, Valenzuela CA, Garcia-Diaz DF, Llanos M. Effects on Fetal Metabolic Programming and Endocannabinoid System of a Normocaloric Diet during Pregnancy and Lactation of Female Mice with Pregestational Obesity. Nutrients 2023; 15:3531. [PMID: 37630722 PMCID: PMC10458167 DOI: 10.3390/nu15163531] [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: 07/18/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Fetal programming provides explanatory mechanisms for the currently high prevalence of gestational obesity. The endocannabinoid system (ECS) participates in the regulation of energy balance, and with a high-fat diet (HFD), it is overactivated. The aim of this study was to determine the effects of a nutritional intervention during pregnancy and lactation on obese female progenitors, on metabolic alterations of the offspring and on the involvement of ECS. Female mice (C57/BL/6-F0), 45 days old, and their offspring (males) were separated according to type of diet before and during gestation and lactation: CON-F1: control diet; HFD-F1 group: HFD (fat: 60% Kcal); INT-F1 group: HFD until mating and control diet (fat: 10% Kcal) afterward. Glucose tolerance and insulin sensitivity (IS) were tested at 2 and 4 months. At 120 days, mice were sacrificed, plasma was extracted for the determination of hormones, and livers for gene expression and the protein level determination of ECS components. INT-F1 group presented a lower IS compared to CON-F1, and normal levels of adiponectin and corticosterone in relation to the HFD-F1 group. The intervention increased hepatic gene expression for fatty-acid amide hydrolase and monoacylglycerol lipase enzymes; however, these differences were not observed at the protein expression level. Our results suggest that this intervention model normalized some hormonal parameters and hepatic mRNA levels of ECS components that were altered in the offspring of progenitors with pre-pregnancy obesity.
Collapse
Affiliation(s)
- Cynthia Barrera
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (C.B.); (R.V.); (D.F.G.-D.)
| | - Valeska Castillo
- Laboratory of Nutrition and Metabolic Regulation, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 8380453, Chile;
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (C.B.); (R.V.); (D.F.G.-D.)
| | - Carina A. Valenzuela
- Eating Behavior Research Center, School of Nutrition and Dietetics, Faculty of Pharmacy, Universidad de Valparaíso Playa Ancha, Valparaíso 2360102, Chile;
| | - Diego F. Garcia-Diaz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (C.B.); (R.V.); (D.F.G.-D.)
| | - Miguel Llanos
- Laboratory of Nutrition and Metabolic Regulation, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 8380453, Chile;
| |
Collapse
|
5
|
Nagagata BA, Ajackson M, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. Obese mothers supplemented with melatonin during gestation and lactation ameliorate the male offspring's pancreatic islet cellular composition and beta-cell function. J Dev Orig Health Dis 2023; 14:490-500. [PMID: 37366144 DOI: 10.1017/s2040174423000168] [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] [Indexed: 06/28/2023]
Abstract
Melatonin supplementation to obese mothers during gestation and lactation might benefit the pancreatic islet cellular composition and beta-cell function in male offspring adulthood. C57BL/6 females (mothers) were assigned to two groups (n = 20/each) based on their consumption in control (C 17% kJ as fat) or high-fat diet (HF 49% kJ as fat). Mothers were supplemented with melatonin (Mel) (10 mg/kg daily) during gestation and lactation, or vehicle, forming the groups (n = 10/each): C, CMel, HF, and HFMel. The male offspring were studied, considering they only received the C diet after weaning until three months old. The HF mothers and their offspring showed higher body weight, glucose intolerance, insulin resistance, and low insulin sensitivity than the C ones. However, HFMel mothers and their offspring showed improved glucose metabolism and weight loss than the HF ones. Also, the offspring's higher expressions of pro-inflammatory markers and endoplasmic reticulum (ER) stress were observed in HF but reduced in HFMel. Contrarily, antioxidant enzymes were less expressed in HF but improved in HFMel. In addition, HF showed increased beta-cell mass and hyperinsulinemia but diminished in HFMel. Besides, the beta-cell maturity and identity gene expressions diminished in HF but enhanced in HFMel. In conclusion, obese mothers supplemented with melatonin benefit their offspring's islet cell remodeling and function. In addition, improving pro-inflammatory markers, oxidative stress, and ER stress resulted in better glucose and insulin levels control. Consequently, pancreatic islets and functioning beta cells were preserved in the offspring of obese mothers supplemented with melatonin.
Collapse
Affiliation(s)
- Brenda A Nagagata
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Ajackson
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Ajackson M, Nagagata BA, Marcondes-de-Castro IA, Mandarim-de-Lacerda CA, Aguila MB. Adult mice offspring of obese mothers supplemented with melatonin show lessened liver steatosis, inflammation, oxidative stress, and endoplasmic reticulum stress. Life Sci 2023; 312:121253. [PMID: 36481166 DOI: 10.1016/j.lfs.2022.121253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
AIMS To investigate, in the liver of adult offspring, the possible effects of melatonin supplementation in the obese mother during pregnancy and lactation. MAIN METHODS C57BL/6 females were fed with a control (C) or a high-fat (HF) diet and supplemented with melatonin (Mel) during the pregnancy and lactation, forming the groups: C, CMel, HF, and HFMel. After weaning until three months old, the offspring only received the C diet. KEY FINDINGS The HF mothers and their offspring showed higher body weight (BW) than the C mothers and offspring. However, at 3-mo-old, BW was reduced in HFMel vs. HF offspring. Also, plasmatic and liver lipid markers increased in HF vs. C offspring but were reduced in HFMel vs. HF offspring. Liver lipid content was lessened in HFMel vs. HF offspring by 50 %. Also, lipid metabolism, pro-inflammatory and endoplasmic reticulum (ER) stress genes were higher expressed in HF vs. C offspring but reduced in HFMel vs. HF offspring. Contrarily, beta-oxidation and antioxidant enzyme genes were less expressed in HF vs. C offspring but improved in HFMel vs. HF offspring. Finally, AMPK/mTOR pathway genes, initially dysregulated in the HF, were restored in the HFMel offspring. SIGNIFICANCE The obese mother leads to liver alterations in the offspring. Current findings demonstrated the maternal melatonin supplementation during pregnancy and lactation in adult offspring's liver. Consequently, the effects were seen in mitigating the liver's AMPK/mTOR pathway genes, lipogenesis, beta-oxidation, inflammation, oxidative stress, and ER stress, preventing liver disease progression in the offspring.
Collapse
Affiliation(s)
- Matheus Ajackson
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Brenda A Nagagata
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ilitch A Marcondes-de-Castro
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
7
|
Yang Z, Jiang J, Chen M, Huang J, Liu J, Wei X, Jia R, Song L, Sun B, Luo X, Song Q, Han Z. Sex-Specific Effects of Maternal and Post-Weaning High-Fat Diet on Adipose Tissue Remodeling and Asprosin Expression in Mice Offspring. Mol Nutr Food Res 2021; 66:e2100470. [PMID: 34933410 DOI: 10.1002/mnfr.202100470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/15/2021] [Indexed: 11/07/2022]
Abstract
SCOPE Perinatal high-fat diet (HFD) increases risk of metabolic disorders in offspring. Adipose tissue remodeling is associated with metabolic syndrome. The current study characterizes the profile of maternal HFD-induced changes in adipose tissue remodeling and adipokines expression in mice offspring. METHODS AND RESULTS Female C57BL/6 mice are fed with CHOW or HFD for 2 weeks before mating, throughout gestation and lactation. At weaning, pups are randomly fed with CHOW or HFD, resulting in eight groups according to sex and maternal diet: Male CHOW-CHOW (MCC), Male CHOW-HFD (MCH), Male HFD-CHOW (MHC), Male HFD-HFD (MHH), Female CHOW-CHOW (FCC), Female CHOW-HFD (FCH), Female HFD-CHOW (FHC), and Female HFD-HFD (FHH). Increased body weight, impaired glucose tolerance, increased adipose tissue mass and hypertrophy, and decreased circulating asprosin level are only observed in male offspring exposure to maternal HFD. Serum asprosin level negatively correlates with fasting blood glucose, serum cholesterol (CHO), and high-density lipoprotein (HDL) levels, while positively correlates with serum low-density lipoprotein (LDL) and glutamate-oxaloacetate transaminase (GOT) levels in male offspring. A combination of genetic and biochemical analyses of adipokines shows the depot- and sex-specific changes in response to maternal and/or post-weaning HFD. CONCLUSION This study's results reveal the differential metabolic changes in response to maternal and/or post-weaning HFD in male and female offspring. The effect of maternal HFD on metabolic phonotype is more obvious in male offspring, supporting the notion that males are more susceptible to HFD-induced metabolic disorders.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jianan Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Miao Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jing Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Ru Jia
- Department of Prosthodontics, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Qing Song
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhen Han
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
8
|
Oxidative Stress Profile of Mothers and Their Offspring after Maternal Consumption of High-Fat Diet in Rodents: A Systematic Review and Meta-Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9073859. [PMID: 34868458 PMCID: PMC8636978 DOI: 10.1155/2021/9073859] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/27/2021] [Accepted: 10/26/2021] [Indexed: 01/03/2023]
Abstract
Maternal exposure to the high-fat diet (HFD) during gestation or lactation can be harmful to both a mother and offspring. The aim of this systematic review was to identify and evaluate the studies with animal models (rodents) that were exposed to the high-fat diet during pregnancy and/or lactation period to investigate oxidative stress and lipid and liver enzyme profile of mothers and their offspring. The electronic search was performed in the PUBMED (Public/Publisher MEDLINE), EMBASE (Ovid), and Web of Science databases. Data from 77 studies were included for qualitative analysis, and of these, 13 studies were included for meta-analysis by using a random effects model. The pooled analysis revealed higher malondialdehyde levels in offspring of high-fat diet groups. Furthermore, the pooled analysis showed increased reactive oxygen species and lower superoxide dismutase and catalase in offspring of mothers exposed to high-fat diet during pregnancy and/or lactation. Despite significant heterogeneity, the systematic review shows oxidative stress in offspring induced by maternal HFD.
Collapse
|
9
|
Strain J, Spaans F, Serhan M, Davidge ST, Connor KL. Programming of weight and obesity across the lifecourse by the maternal metabolic exposome: A systematic review. Mol Aspects Med 2021; 87:100986. [PMID: 34167845 DOI: 10.1016/j.mam.2021.100986] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022]
Abstract
Exposome research aims to comprehensively understand the multiple environmental exposures that influence human health. To date, much of exposome science has focused on environmental chemical exposures and does not take a lifecourse approach. The rising prevalence of obesity, and the limited success in its prevention points to the need for a better understanding of the diverse exposures that associate with, or protect against, this condition, and the mechanisms driving its pathogenesis. The objectives of this review were to 1. evaluate the evidence on the maternal metabolic exposome in the programming of offspring growth/obesity and 2. identify and discuss the mechanisms underlying the programming of obesity. A systematic review was conducted following PRISMA guidelines to capture articles that investigated early life metabolic exposures and offspring weight and/or obesity outcomes. Scientific databases were searched using pre-determined indexed search terms, and risk of bias assessments were conducted to determine study quality. A final total of 76 articles were obtained and extracted data from human and animal studies were visualised using GOfER diagrams. Multiple early life exposures, including maternal obesity, diabetes and adverse nutrition, increase the risk of high weight at birth and postnatally, and excess adipose accumulation in human and animal offspring. The main mechanisms through which the metabolic exposome programmes offspring growth and obesity risk include epigenetic modifications, altered placental function, altered composition of the gut microbiome and breast milk, and metabolic inflammation, with downstream effects on development of the central appetite system, adipose tissues and liver. Understanding early life risks and protectors, and the mechanisms through which the exposome modifies health trajectories, is critical for developing and applying early interventions to prevent offspring obesity later in life.
Collapse
Affiliation(s)
- Jamie Strain
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Mohamed Serhan
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, AB, Canada; Department of Physiology, University of Alberta, Edmonton, AB, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
10
|
Tarevnic R, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. Maternal swimming mitigates liver damage caused by paternal obesity. Nutrition 2021; 86:111168. [PMID: 33601122 DOI: 10.1016/j.nut.2021.111168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 01/06/2021] [Accepted: 01/16/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Parents' lifestyle and nutrition can program offspring obesity in adulthood. We hypothesized that maternal swimming has beneficial effects on the adversity caused by paternal obesity on offspring. METHODS Twelve-week-old male C57 BL/6 J mice (fed a high-fat diet, obese father [ObFa], or control diet, lean father [LFa]) were mated with female mice fed only the control diet. Mothers were trained (TMo) or untrained (UMo): swimming for 6 wk before and the first 2 wk of gestation. Pups were fed only the control diet. RESULTS Fathers showed different body mass (BM) at copulation, but not the mothers. The ObFa had 20% higher BM than the LFa. Twelve-week-old ObFa/UMo offspring showed a higher BM gain than the LFa/UMo and ObFa/TMo. There was BM sexual dimorphism in the LFa/UMo (female mice +24% than male mice). There was hyperglycemia and hyperinsulinemia in the ObFa/UMo, but low glycemia and insulin levels were seen in the ObFa/TMo. There was augmented liver steatosis in the ObFa/UMo compared with the LFa/UMo, and the ObFa/TMo compared with the LFa/TMo, but reduced steatosis in the ObFa/TMo compared with the ObFa/UMo. In addition, lipogenic markers were more expressed and beta-oxidation markers less expressed in the ObFa/UMo compared with the LFa/UMo, but the opposite was observed in the ObFa/TMo compared with the ObFa/UMo. Proinflammatory markers were higher in the liver of the ObFa/UMo compared with the LFa/UMo and lower in the ObFa/TMo compared with the ObFa/UMo. CONCLUSIONS Obese fathers produced offspring that were overweight and had altered fasting glycemia and insulin sensitivity, leading to higher liver lipogenesis and inflammation, as well as lower beta-oxidation. The swimming mother mitigated these adverse effects in mice offspring.
Collapse
Affiliation(s)
- Renata Tarevnic
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ornellas
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Zhang J, Cao L, Tan Y, Zheng Y, Gui Y. N-acetylcysteine protects neonatal mice from ventricular hypertrophy induced by maternal obesity in a sex-specific manner. Biomed Pharmacother 2021; 133:110989. [PMID: 33378994 DOI: 10.1016/j.biopha.2020.110989] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Maternal obesity induces adverse cardiac programming in offspring, and effective interventions are needed to prevent cardiovascular ill-health. Herein we hypothesized that exposure to maternal obesogenic diet-induced obesity in mice results in left ventricular remodelling and hypertrophy in early childhood, and that maternal N-acetylcysteine (NAC) treatment alleviates these effects in a sex-dependent manner. METHODS AND RESULTS The maternal obesity was induced in mice by the consumption of a Western diet accompanied by a 20 % sucrose solution. To determine the effect of NAC on the cardiac outcomes induced by maternal obesity, obese dams were continuously exposed to the obesogenic diet, with or without the oral NAC treatment during pregnancy. Left ventricular remodelling and hypertrophy occurred as early as 7 days after birth in the male offspring of obese dams (O-OB) compared with controls (O-CO). An over-expression of key genes and markers related to cardiac fibrosis accompanied by more disorganized myofibrils was observed in the hearts of neonatal male O-OB mice. When we next evaluated the level of oxidative stress in the hearts of neonatal mice, the activity of enzymatic antioxidants declined and expression of NOX enzyme complex was up-regulated in O-OB offspring hearts, but was normal in the offspring of NAC treated mice (O-OB/NAC). Maternal obesity also activated cardiac Akt and mammalian target of rapamycin (mTOR) signalling in offspring, and NAC treatment restored offspring cardiac Akt-mTOR signalling to normal irrespective of sex. NAC treatment did not prevent cardiomyocyte hypertrophy but did alleviate increased heart weight, interventricular septal thickness, and collagen content in male O-OB/NAC pups. CONCLUSIONS Collectively, our results indicated that NAC blunted cardiac fibrosis and related ventricular hypertrophy of male neonatal offspring in the setting of maternal obesity, potentially acting by reducing oxidative stress. The present study provides a basis for investigating the role of NAC in nutrition-related cardiac programming.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Animal Nutritional Physiological Phenomena
- Animals
- Animals, Newborn
- Antioxidants/pharmacology
- Disease Models, Animal
- Female
- Fibrosis
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Male
- Maternal Nutritional Physiological Phenomena
- Mice, Inbred C57BL
- Obesity, Maternal/complications
- Obesity, Maternal/physiopathology
- Oxidative Stress/drug effects
- Pregnancy
- Prenatal Exposure Delayed Effects
- Sex Factors
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Jialing Zhang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China; MOH Key Laboratory of Neonatal Diseases at Children's Hospital, Fudan University, Shanghai, China
| | - Li Cao
- Ultrasound Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yanfeng Tan
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Yuanzheng Zheng
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China; MOH Key Laboratory of Neonatal Diseases at Children's Hospital, Fudan University, Shanghai, China
| | - Yonghao Gui
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China; MOH Key Laboratory of Neonatal Diseases at Children's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Alves JM, Luo S, Chow T, Herting M, Xiang AH, Page KA. Sex differences in the association between prenatal exposure to maternal obesity and hippocampal volume in children. Brain Behav 2020; 10:e01522. [PMID: 31903710 PMCID: PMC7010582 DOI: 10.1002/brb3.1522] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Animal studies have shown that male but not female offspring exposed to maternal obesity have abnormal hippocampal development. Similar sex differences were observed in animal models of developmental programming by prenatal stress or maternal diabetes. We aimed to translate this work into humans by examining sex-specific effects of exposure to maternal obesity on hippocampal volume in children. METHODS Eighty-eight children (37 boys and 51 girls) aged 7-11 years completed the study. Maternal prepregnancy body mass index (BMI) was obtained from electronic medical records. A high-resolution anatomical scan was performed using a 3-Tesla magnetic resonance imaging (MRI) scanner. Total hippocampal volume and hippocampal subfield volumes were analyzed using FreeSurfer 6.0. Linear regression was used to investigate sex differences in relationships between maternal prepregnancy BMI and child hippocampal volume. RESULTS Maternal prepregnancy BMI ranged from 19.0 to 50.4 kg/m2 . We observed a significant interaction between maternal prepregnancy BMI and sex on total hippocampal volume (p < .001) such that boys (r = -.39, p = .018) but not girls (r = .11, p = .45) had a significant negative relationship between maternal prepregnancy BMI and total hippocampal volume. This relationship in boys remained significant after adjusting for child and maternal covariates (β = -126.98, p = .012). The sex interactions with prepregnancy BMI were consistently observed in hippocampal subfields CA1 (p = .008), CA2/3 (p = .016), CA4 (p = .002), dentate gyrus (p < .001), and subiculum (p < .001). CONCLUSIONS Our results support findings in animal models and suggest that boys may be more vulnerable to the adverse effects of exposure to maternal obesity on hippocampal development than girls.
Collapse
Affiliation(s)
- Jasmin M. Alves
- Division of EndocrinologyDepartment of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Diabetes and Obesity Research InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Shan Luo
- Division of EndocrinologyDepartment of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Diabetes and Obesity Research InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Ting Chow
- Department of Research and EvaluationPasadenaCAUSA
| | - Megan Herting
- Department of Preventive MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | | | - Kathleen A. Page
- Division of EndocrinologyDepartment of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Diabetes and Obesity Research InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| |
Collapse
|
13
|
Sex-specific programming effects of parental obesity in pre-implantation embryonic development. Int J Obes (Lond) 2019; 44:1185-1190. [PMID: 31776435 DOI: 10.1038/s41366-019-0494-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/25/2019] [Accepted: 11/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Obesity is a global rising problem with epidemiological dimension. Obese parents can have programming effects on their offspring leading to obesity and associated diseases in later life. This constitutes a vicious circle. Epidemiological data and studies in rodents demonstrated differential programming effects in male and female offspring, but the timing of their developmental origin is not known. METHODS This study investigated if sex-specific programming effects of parental obesity can already be detected in the pre-implantation period. Diet-induced obese male or female mice were mated with normal-weight partners and blastocysts were recovered. RESULTS Gene expression profiling revealed sex-specific responses of the blastocyst transcriptome to maternal and paternal obesity. The changes in the transcriptome of male blastocysts were more pronounced than those of female blastocysts, with a stronger impact of paternal than of maternal obesity. The sperm of obese mice revealed an increased abundance of several miRNAs compared with lean mice. CONCLUSIONS Our study indicates that sex-specific programming effects of parental obesity already start in the pre-implantation period and reveals specific alterations of the sperm miRNA profile as mechanistic link to programming effects of paternal obesity.
Collapse
|
14
|
Zhang JK, Miao J, Chen ZQ, Duan SZ, Zhang X, Ji WJ, Niu JM, Yuan F, Zhou X, Li YM, Zhang Z. β3-Adrenergic Activation Improves Maternal and Offspring Perinatal Outcomes in Diet-Induced Prepregnancy Obesity in Mice. Obesity (Silver Spring) 2019; 27:1482-1493. [PMID: 31328894 DOI: 10.1002/oby.22561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/20/2019] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Prepregnancy obesity is an epidemic disorder that seriously threatens both maternal and offspring health. This study investigated the effects of β3-adrenergic receptor (β3-AR) activation on the perinatal outcomes in a diet-induced prepregnancy obese (PPO) murine model. METHODS Four-week-old female C57BL/6 mice were fed high-fat diet or chow diet for 16 weeks to yield PPO mice and chow-fed (CF) lean mice, respectively. After successful mating with CF males, the PPO and CF mice were both randomly divided into vehicle control- or CL316,243 (a highly selective β3-AR agonist)-treated groups. On gestational day 7, subcutaneous infusion of CL316,243 or saline vehicle (1 mg/kg/d) was provided using osmotic pumps. The perinatal outcomes, adipose tissue morphology, and metabolic and inflammatory markers were examined. RESULTS Chronic β3-AR agonist infusion induced brown adipose tissue activation and white adipose tissue browning and countered obesity-induced alterations in lipid profiles, insulin resistance, and systemic and local inflammatory states. Moreover, β3-AR activation was associated with improved placental perfusion and offspring outcomes. CONCLUSIONS Our results provide proof-of-principle evidence that pharmacological β3-AR activation may be of therapeutic potential in preventing prepregnancy-obesity-associated adverse maternal and offspring perinatal outcomes.
Collapse
Affiliation(s)
- Jun-Kai Zhang
- Logistics University of the Chinese People's Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Jun Miao
- Logistics University of the Chinese People's Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Zu-Qin Chen
- Logistics University of the Chinese People's Armed Police Force, Tianjin, China
- Department of MRI, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Si-Zhang Duan
- Logistics University of the Chinese People's Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Xin Zhang
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Wen-Jie Ji
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Jian-Min Niu
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Fei Yuan
- Department of MRI, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu-Ming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Characteristic Medical Center of the Chinese People's Armed Police Forces, Tianjin, China
| | - Zhuoli Zhang
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
15
|
Badran M, Yassin BA, Lin DTS, Kobor MS, Ayas N, Laher I. Gestational intermittent hypoxia induces endothelial dysfunction, reduces perivascular adiponectin and causes epigenetic changes in adult male offspring. J Physiol 2019; 597:5349-5364. [PMID: 31441069 DOI: 10.1113/jp277936] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Obstructive sleep apnoea (OSA) is characterized by intermittent hypoxia, which causes oxidative stress and inflammation and increases the risk of cardiovascular disease. OSA during pregnancy causes adverse maternal and fetal outcomes. The effects of pre-existing OSA in pregnant women on cardiometabolic outcomes in the offspring are unknown. We evaluated basic metabolic parameters, as well as aortic vascular and perivascular adipose tissue (PVAT) function in response to adiponectin, and examined DNA methylation of adiponectin gene promoter in PVAT in 16-week-old adult offspring exposed to gestational intermittent hypoxia (GIH). GIH decreased body weights at week 1 in both male and female offspring, and caused subsequent increases in body weight and food consumption in male offspring only. Adult female offspring had normal levels of lipids, glucose and insulin, with no endothelial dysfunction. Adult male offspring exhibited dyslipidaemia, insulin resistance and hyperleptinaemia. Decreased endothelial-dependent vasodilatation, loss of anti-contractile activity of PVAT and low circulating PVAT adiponectin levels, as well as increased pro-inflammatory gene expression and DNA methylation of adiponectin gene promoter, occurred in adult male offspring. Our results suggest that male offspring of women with OSA could be at risk of developing cardiometabolic disease during adulthood. ABSTRACT Perturbations during pregnancy can program the offspring to develop cardiometabolic diseases later in life. Obstructive sleep apnoea (OSA) is a chronic condition that frequently affects pregnancies and leads to adverse fetal outcomes. We assessed the offspring of female mice experiencing gestational intermittent hypoxia (GIH), a hallmark of OSA, for changes in metabolic profiles, aortic nitric oxide (NO)-dependent relaxations, perivascular adipose tissue (PVAT) anti-contractile activities and the responses to adiponectin, and DNA methylation of the adiponectin gene promoter in PVAT tissue. Pregnant mouse dams were exposed to intermittent hypoxic cycles ( F I O 2 21-12%) for 18 days. GIH resulted in lower body weights of pups at week 1, followed by significant weight gain by week 16 of age in male but not female offspring. Plasma lipids, leptin and insulin resistance were higher in GIH male adult offspring. Endothelium-dependent relaxation in response to ACh and the anti-contractile activity of PVAT in the abdominal aorta was reduced in GIH adult male offspring. Incubation of arteries from GIH adult male offspring with adiponectin restored the anti-contractile activity of PVAT. Both circulating and PVAT tissue homogenate levels of adiponectin, as well as gene expression of adiponectin in PVAT, were lower in GIH male offspring, along with an increased gene expression of inflammatory cytokines. Pyrosequencing of adiponectin gene promoter in PVAT showed increased DNA methylation in GIH male offspring. Our results indicate that GIH leads to vascular disease in adult male offspring through PVAT dysfunction, which was associated with low adiponectin levels and epigenetic modifications on the adiponectin gene promoter.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Bisher Abu Yassin
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - David Tse Shen Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Najib Ayas
- Divisions of Critical Care and Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Sleep Disorders Program, UBC Hospital, Vancouver, BC, Canada.,Division of Critical Care Medicine, Providence Healthcare, Vancouver, BC, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Normalisation of circulating adiponectin levels in obese pregnant mice prevents cardiac dysfunction in adult offspring. Int J Obes (Lond) 2019; 44:488-499. [PMID: 31076636 DOI: 10.1038/s41366-019-0374-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/06/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND/OBJECTIVES Adiponectin concentrations are low in obese pregnant women. Restoring normal adiponectin concentrations by infusion in obese pregnant mice prevents placental dysfunction, foetal overgrowth and metabolic syndrome in the offspring. We hypothesised that normalising maternal adiponectin in obese late pregnant dams prevents cardiac dysfunction in the adult offspring. SUBJECTS/METHODS Pregnant female mice with diet-induced obesity were infused with adiponectin (0.62 μg g-1 day-1, n = 24) or saline (n = 22) over days 14.5-18.5 of pregnancy (term = day 19.5). Control dams ate standard chow and received saline (n = 22). Offspring were studied at 3 and 6 months of age. RESULTS Maternal obesity impaired ventricular diastolic function, increased cardiomyocyte cross-sectional area and upregulated cardiac brain natriuretic peptide (Nppb) and α-skeletal actin (Acta1) gene expression in adult male offspring, compared to control offspring. In adult female offspring, maternal obesity increased Nppb expression, decreased end-diastolic volume and caused age-dependent diastolic dysfunction but not cardiomyocyte hypertrophy. Maternal obesity also activated cardiac Akt and mechanistic target of rapamycin (mTOR) signalling in male, but not in female, offspring and inhibited cardiac extracellular signal-regulated kinase 1/2 (ERK1/2) in both sexes. Normalising maternal circulating adiponectin concentrations by infusing obese dams with adiponectin prevented offspring diastolic dysfunction and ventricular dilation and normalised cardiac Akt-mTOR signalling irrespective of sex. Maternal adiponectin infusion also reduced cardiac Nppb expression and increased ERK1/2 signalling in offspring of obese dams. Adiponectin infusion did not prevent cardiomyocyte hypertrophy but reduced ventricular wall thickness in male offspring and increased collagen content in female offspring of obese dams, compared to controls. CONCLUSIONS Low maternal adiponectin levels in obese mice in late pregnancy are mechanistically linked to in utero programming of cardiac dysfunction in their offspring. Interventions enhancing endogenous adiponectin secretion or signalling in obese pregnant women could prevent the development of cardiac dysfunction in their children.
Collapse
|
17
|
Smoothy J, Larcombe AN, Chivers EK, Matthews VB, Gorman S. Maternal high fat diet compromises survival and modulates lung development of offspring, and impairs lung function of dams (female mice). Respir Res 2019; 20:21. [PMID: 30700289 PMCID: PMC6354360 DOI: 10.1186/s12931-019-0976-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/03/2019] [Indexed: 12/25/2022] Open
Abstract
Background Epidemiological studies have identified strong relationships between maternal obesity and offspring respiratory dysfunction; however, the causal direction is not known. We tested whether maternal obesity alters respiratory function of offspring in early life. Methods Female C57Bl/6 J mice were fed a high or low fat diet prior to and during two rounds of mating and resulting pregnancies with offspring lung function assessed at 2 weeks of age. The lung function of dams was measured at 33 weeks of age. Results A high fat diet caused significant weight gain prior to conception with dams exhibiting elevated fasting glucose, and glucose intolerance. The number of surviving litters was significantly less for dams fed a high fat diet, and surviving offspring weighed more, were longer and had larger lung volumes than those born to dams fed a low fat diet. The larger lung volumes significantly correlated in a linear fashion with body length. Pups born from the second pregnancy had reduced tissue elastance compared to pups born from the first pregnancy, regardless of the dam’s diet. As there was reduced offspring survival born to dams fed a high fat diet, the statistical power of lung function measures of offspring was limited. There were signs of increased inflammation in the bronchoalveolar lavage fluid of dams (but not offspring) fed a high fat diet, with more tumour necrosis factor-α, interleukin(IL)-5, IL-33 and leptin detected. Dams that were fed a high fat diet and became pregnant twice had reduced fasting glucose immediately prior to the second mating, and lower levels of IL-33 and leptin in bronchoalveolar lavage fluid. Conclusions While maternal high fat diet compromised litter survival, it also promoted somatic and lung growth (increased lung volume) in the offspring. Further studies are required to examine downstream effects of this enhanced lung volume on respiratory function in disease settings.
Collapse
Affiliation(s)
- Jordan Smoothy
- Telethon Kids Institute, University of Western Australia, Northern Entrance Perth Children's Hospital, 15 Hospital Ave, Nedlands, Western Australia, 6009, Australia
| | - Alexander N Larcombe
- Telethon Kids Institute, University of Western Australia, Northern Entrance Perth Children's Hospital, 15 Hospital Ave, Nedlands, Western Australia, 6009, Australia.,School of Public Health, Curtin University, Perth, Western Australia, 6845, Australia
| | - Emily K Chivers
- Telethon Kids Institute, University of Western Australia, Northern Entrance Perth Children's Hospital, 15 Hospital Ave, Nedlands, Western Australia, 6009, Australia
| | - Vance B Matthews
- School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Shelley Gorman
- Telethon Kids Institute, University of Western Australia, Northern Entrance Perth Children's Hospital, 15 Hospital Ave, Nedlands, Western Australia, 6009, Australia.
| |
Collapse
|
18
|
Beneficial effects of maternal swimming during pregnancy on offspring metabolism when the father is obese. J Dev Orig Health Dis 2018; 10:502-506. [PMID: 30560765 DOI: 10.1017/s2040174418001046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We aimed to evaluate the impact of maternal exercise training on the offspring metabolism and body size caused by father obesity. C57BL/6 male 4-week-old mice were fed a high-fat diet (HF father) or control diet (C father), while equal age female mice were fed only a C diet and were separated into two groups: trained (T mother) and non-trained (NT mother), and at 12 weeks of age mice were mated. A continuous swimming protocol was applied for 10 weeks (before and during gestation), and offspring were followed since weaning until sacrifice (at 12 weeks of age). HF father, compared to C father, showed obesity, elevated total cholesterol (TC) and triglycerides (TG), and glucose intolerance. Both sexes HF/NT offspring showed hyperglycemia, glucose intolerance and high levels of TC and TG, without obesity. However, HF/T offspring showed data close to C/NT, demonstrating the beneficial effect of maternal exercise in the offspring of obese fathers.
Collapse
|
19
|
Talbot CPJ, Dolinsky VW. Sex differences in the developmental origins of cardiometabolic disease following exposure to maternal obesity and gestational diabetes 1. Appl Physiol Nutr Metab 2018; 44:687-695. [PMID: 30500266 DOI: 10.1139/apnm-2018-0667] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Over the past 30 years, the worldwide prevalence of obesity has nearly doubled. In addition, more and more women in their child-bearing years are overweight or obese, which increases the risk of gestational diabetes mellitus (GDM). It is increasingly accepted by the scientific community that early life exposure to environmental stress influences the long-term health of an individual, which has been termed the Developmental Origins of Health and Disease theory. Evidence from human cohorts and epidemiological and animal studies has shown that maternal obesity and GDM condition the offspring for cardiometabolic disease development. These effects are most likely regulated by epigenetic mechanisms; however, biological sex is an important factor in defining the risk of the development of several metabolic health disorders. The aim of this review is to describe the current evidence from human cohort and animal model studies that implicates sex differences in the developmental origins of cardiometabolic disease following exposure to maternal obesity and GDM. In addition, this review addresses the potential mechanisms involved in these sex differences. In many studies, sex is ignored as an important variable in disease development; however, the results presented in this review highlight important differences between sexes in the developmental programming of biological responses to exposures during the fetal stage. This knowledge will ultimately help in the development of effective therapeutic strategies for the treatment of cardiometabolic diseases that exhibit sexual dimorphism.
Collapse
Affiliation(s)
- Charlotte Pauline Joëlle Talbot
- a Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.,b Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.,c Manitoba Developmental Origins of Chronic Diseases in Children Network (DEVOTION), University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Vernon Wayne Dolinsky
- a Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.,b Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.,c Manitoba Developmental Origins of Chronic Diseases in Children Network (DEVOTION), University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
20
|
Gender-divergent expression of lipid and bile acid metabolism-related genes in adult mice offspring of dams fed a high-fat diet. J Biosci 2018. [DOI: 10.1007/s12038-018-9750-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Ornellas F, Carapeto PV, Mandarim‐de‐Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
22
|
Ornellas F, Carapeto PV, Mandarim-de-Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J Pediatr (Rio J) 2017; 93:551-559. [PMID: 28822233 DOI: 10.1016/j.jped.2017.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. SOURCES A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. SUMMARY OF FINDINGS Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. CONCLUSIONS Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Priscila V Carapeto
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil.
| | - Marcia B Aguila
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
23
|
Bronson SL, Chan JC, Bale TL. Sex-Specific Neurodevelopmental Programming by Placental Insulin Receptors on Stress Reactivity and Sensorimotor Gating. Biol Psychiatry 2017; 82:127-138. [PMID: 28168960 PMCID: PMC5483189 DOI: 10.1016/j.biopsych.2016.12.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/22/2016] [Accepted: 12/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diabetes, obesity, and overweight are prevalent pregnancy complications that predispose offspring to neurodevelopmental disorders, including autism, attention-deficit/hyperactivity disorder, and schizophrenia. Although male individuals are three to four times more likely than female individuals to develop these disorders, the mechanisms driving the sex specificity of disease vulnerability remain unclear. Because defective placental insulin receptor (InsR) signaling is a hallmark of pregnancy metabolic dysfunction, we hypothesized that it may be an important contributor and novel mechanistic link to sex-specific neurodevelopmental changes underlying disease risk. METHODS We used Cre/loxP transgenic mice to conditionally target InsRs in fetally derived placental trophoblasts. Adult offspring were evaluated for effects of placental trophoblast-specific InsR deficiency on stress sensitivity, cognitive function, sensorimotor gating, and prefrontal cortical transcriptional reprogramming. To evaluate molecular mechanisms driving sex-specific outcomes, we assessed genome-wide expression profiles in the placenta and fetal brain. RESULTS Male, but not female, mice with placental trophoblast-specific InsR deficiency showed a significantly increased hypothalamic-pituitary-adrenal axis stress response and impaired sensorimotor gating, phenotypic effects that were associated with dysregulated nucleotide metabolic processes in the male prefrontal cortex. Within the placenta, InsR deficiency elicited changes in gene expression, predominantly in male mice, reflecting potential shifts in vasculature, amino acid transport, serotonin homeostasis, and mitochondrial function. These placental disruptions were associated with altered gene expression profiles in the male fetal brain and suggested delayed cortical development. CONCLUSIONS Together, these data demonstrate the novel role of placental InsRs in sex-specific neurodevelopment and reveal a potential mechanism for neurodevelopmental disorder risk in pregnancies complicated by maternal metabolic disorders, including diabetes and obesity.
Collapse
Affiliation(s)
- Stefanie L Bronson
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer C Chan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
24
|
Father’s obesity programs the adipose tissue in the offspring via the local renin–angiotensin system and MAPKs pathways, especially in adult male mice. Eur J Nutr 2017; 57:1901-1912. [DOI: 10.1007/s00394-017-1473-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/16/2017] [Indexed: 12/31/2022]
|
25
|
High-fat diet induced changes in lumbar vertebra of the male rat offsprings. Acta Histochem 2016; 118:711-721. [PMID: 27577321 DOI: 10.1016/j.acthis.2016.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/04/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022]
Abstract
In obesity, bone marrow adiposity increases and proinflammatory cytokines excretion activates RANK/RANKL/OPG system, which leads to increased bone resorption. The aim of this study was to analyze trabecular and cortical bone parameters in animals exposed to the high-fat diet in utero and after lactation. Skeletal organ of interest was the fifth lumbar vertebra, which is not exposed to biomechanical loading in rats. Further aims were to determine TNF-α and IL-6 serum concentrations, and the intensity of the TNF-α immunohistochemical staining in the bone marrow. Ten female Sprague Dawley rats, nine weeks old, were randomly divided in two groups and fed either standard laboratory chow or food rich in saturated fatty acids during five weeks, and then mated with genetically similar male subjects. After birth and lactation male offsprings from both groups were divided in four subgroups depending on the diet they were fed until twenty-two weeks of age. The highest cholesterol and triglyceride concentration were found in both groups of offsprings fed with high-fat diet. The lowest trabecular bone volume, lowest trabecular number and highest trabecular separation were found in offsprings fed with high-fat diet of mothers on standard laboratory chow. The same group of offsprings was also characterized by the highest intensity of TNF-α immunostaining in the bone marrow and the highest TNF-α serum concentration, which suggest that this proinflammatory cytokine has interfered with bone metabolism, possibly by stimulation of bone resorption, which led to inadequate trabecular bone development and bone modeling of the fifth lumbar vertebra.
Collapse
|
26
|
Hokke S, Puelles VG, Armitage JA, Fong K, Bertram JF, Cullen-McEwen LA. Maternal Fat Feeding Augments Offspring Nephron Endowment in Mice. PLoS One 2016; 11:e0161578. [PMID: 27547968 PMCID: PMC4993378 DOI: 10.1371/journal.pone.0161578] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022] Open
Abstract
Increasing consumption of a high fat 'Western' diet has led to a growing number of pregnancies complicated by maternal obesity. Maternal overnutrition and obesity have health implications for offspring, yet little is known about their effects on offspring kidney development and renal function. Female C57Bl6 mice were fed a high fat diet (HFD, 21% fat) or matched normal fat diet (NFD, 6% fat) for 6 weeks prior to pregnancy and throughout gestation and lactation. HFD dams were overweight and glucose intolerant prior to mating but not in late gestation. Offspring of NFD and HFD dams had similar body weights at embryonic day (E)15.5, E18.5 and at postnatal day (PN)21. HFD offspring had normal ureteric tree development and nephron number at E15.5. However, using unbiased stereology, kidneys of HFD offspring were found to have 20-25% more nephrons than offspring of NFD dams at E18.5 and PN21. Offspring of HFD dams with body weight and glucose profiles similar to NFD dams prior to pregnancy also had an elevated nephron endowment. At 9 months of age, adult offspring of HFD dams displayed mild fasting hyperglycaemia but similar body weights to NFD offspring. Renal function and morphology, measured by transcutaneous clearance of FITC-sinistrin and stereology respectively, were normal. This study demonstrates that maternal fat feeding augments offspring nephron endowment with no long-term consequences for offspring renal health. Future studies assessing the effects of a chronic stressor on adult mice with augmented nephron number are warranted, as are studies investigating the molecular mechanisms that result in high nephron endowment.
Collapse
Affiliation(s)
- Stacey Hokke
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Victor G. Puelles
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - James A. Armitage
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
- School of Medicine (Optometry), Deakin University, Waurn Ponds, Victoria, Australia
| | - Karen Fong
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - John F. Bertram
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Luise A. Cullen-McEwen
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Lecoutre S, Deracinois B, Laborie C, Eberlé D, Guinez C, Panchenko PE, Lesage J, Vieau D, Junien C, Gabory A, Breton C. Depot- and sex-specific effects of maternal obesity in offspring's adipose tissue. J Endocrinol 2016; 230:39-53. [PMID: 27122310 DOI: 10.1530/joe-16-0037] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/27/2016] [Indexed: 12/19/2022]
Abstract
According to the Developmental Origin of Health and Disease (DOHaD) concept, alterations of nutrient supply in the fetus or neonate result in long-term programming of individual body weight (BW) setpoint. In particular, maternal obesity, excessive nutrition, and accelerated growth in neonates have been shown to sensitize offspring to obesity. The white adipose tissue may represent a prime target of metabolic programming induced by maternal obesity. In order to unravel the underlying mechanisms, we have developed a rat model of maternal obesity using a high-fat (HF) diet (containing 60% lipids) before and during gestation and lactation. At birth, newborns from obese dams (called HF) were normotrophs. However, HF neonates exhibited a rapid weight gain during lactation, a key period of adipose tissue development in rodents. In males, increased BW at weaning (+30%) persists until 3months of age. Nine-month-old HF male offspring was normoglycemic but showed mild glucose intolerance, hyperinsulinemia, and hypercorticosteronemia. Despite no difference in BW and energy intake, HF adult male offspring was predisposed to fat accumulation showing increased visceral (gonadal and perirenal) depots weights and hyperleptinemia. However, only perirenal adipose tissue depot exhibited marked adipocyte hypertrophy and hyperplasia with elevated lipogenic (i.e. sterol-regulated element binding protein 1 (Srebp1), fatty acid synthase (Fas), and leptin) and diminished adipogenic (i.e. peroxisome proliferator-activated receptor gamma (Pparγ), 11β-hydroxysteroid dehydrogenase type 1 (11β-Hds1)) mRNA levels. By contrast, very few metabolic variations were observed in HF female offspring. Thus, maternal obesity and accelerated growth during lactation program offspring for higher adiposity via transcriptional alterations of visceral adipose tissue in a depot- and sex-specific manner.
Collapse
Affiliation(s)
- Simon Lecoutre
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Barbara Deracinois
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Christine Laborie
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Delphine Eberlé
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Céline Guinez
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Polina E Panchenko
- INRAUMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| | - Jean Lesage
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Didier Vieau
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| | - Claudine Junien
- INRAUMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France UVSQUniversité Versailles-Saint-Quentin-en-Yvelines, Versailles, France
| | - Anne Gabory
- INRAUMR1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France
| | - Christophe Breton
- Univ. LilleEA4489, Équipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F59000 Lille, France
| |
Collapse
|
28
|
Janoschek R, Bae-Gartz I, Vohlen C, Alcázar MAA, Dinger K, Appel S, Dötsch J, Hucklenbruch-Rother E. Dietary intervention in obese dams protects male offspring from WAT induction of TRPV4, adiposity, and hyperinsulinemia. Obesity (Silver Spring) 2016; 24:1266-73. [PMID: 27106804 DOI: 10.1002/oby.21486] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE One major risk factor for childhood overweight is maternal obesity. The underlying molecular mechanisms are ill-defined, and effective prevention strategies are missing. METHODS Diet-induced obese mouse dams were changed to standard chow during pregnancy and lactation as an intervention against predisposition for obesity and metabolic sequelea in the offspring. Expression of adipokines and TRPV4, a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis, in offspring's white adipose tissue (WAT) was assessed. RESULTS Pathological effects on offspring's body weight, fat content, and serum insulin were fully reversed in intervention offspring on postnatal day 21. In WAT, a sixfold increase of Trpv4 mRNA expression in offspring consuming high-fat-containing diet was found, which was completely blunted in the intervention group. Simultaneously, WAT adipokine, interleukin-6, and peroxisome proliferator-activated receptor-γ mRNA and UCP1 protein expression were largely returned to control levels in intervention offspring. CONCLUSIONS Improvement of maternal nutrition offers a powerful strategy to improve offspring's metabolic health. Targeting TRPV4-linked aspects of WAT metabolic function during early development might be a promising approach to prevent long-term adverse metabolic effects of maternal high-fat nutrition.
Collapse
Affiliation(s)
- Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | | | - Katharina Dinger
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | | |
Collapse
|
29
|
Bringhenti I, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. The insulin-signaling pathway of the pancreatic islet is impaired in adult mice offspring of mothers fed a high-fat diet. Nutrition 2016; 32:1138-43. [PMID: 27155954 DOI: 10.1016/j.nut.2016.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 02/02/2016] [Accepted: 03/01/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Mothers fed a high-fat (HF) diet can cause different adverse alterations in their offspring. The study aimed to verify the pancreatic islet structure and insulin-signaling pathway in adulthood of offspring of mothers fed a HF diet during the pregnancy. METHODS Female mice (mothers) were randomly assigned to receive either standard chow (Mo-SC) or a HF diet (Mo-HF) ad libitum. After 2 mo on the experimental diets, 3-mo-old female mice were mated with male C57 BL/6 mice that were fed a SC diet. The male offspring was evaluated at 6 mo old. RESULTS At 6 mo of age, Mo-HF offspring had an increment in body mass and adiposity, hypercholesterolemia, and hypertriacylglycerolemia, higher levels of insulin, and leptin with a concomitant decrease in adiponectin levels. In the islet, we observed an alteration in the structure characterized by the migration of some alpha cells from the edge to the core of the islet in association with an increase in the masses of the islet, beta cell, and alpha cell, featuring a pancreatic islet remodeling. Additionally, the Mo-HF offspring demonstrated a decrease in IRS1, PI3 k p-Akt, Pd-1, and Glut2 protein expressions compared to Mo-SC offspring. However, an increase was observed in FOXO1 and insulin protein expressions in Mo-HF offspring compared to Mo-SC offspring. CONCLUSION The present study demonstrated that a maternal HF diet is responsible for remodeling the islet structure coupled with an adverse carbohydrate metabolism and impairment of the insulin-signaling pathway in adult male mice offspring.
Collapse
Affiliation(s)
- Isabele Bringhenti
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ornellas
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
30
|
Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring. Physiol Behav 2015; 153:47-55. [PMID: 26485293 DOI: 10.1016/j.physbeh.2015.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/07/2015] [Accepted: 10/16/2015] [Indexed: 11/29/2022]
Abstract
We aimed to evaluate the effects of maternal and/or paternal obesity on offspring body mass, leptin signaling, appetite-regulating neurotransmitters and local inflammatory markers. C57BL/6 mice received standard chow (SC, lean groups) or high-fat diet (HF, obese groups) starting from one month of age. At three months, HF mice became obese relative to SC mice. They were then mated as follows: lean mother and lean father, lean mother and obese father, obese mother and lean father, and obese mother and obese father. The offspring received the SC diet from weaning until three months of age, when they were sacrificed. In the offspring, paternal obesity did not lead to changes in the Janus kinase (JAK)/signal transducer and activation of the transcription (STAT) pathway or feeding behavior but did induce hypothalamic inflammation. On the other hand, maternal obesity resulted in increased weight gain, hyperleptinemia, decreased leptin OBRb receptor expression, JAK/STAT pathway impairment, and increased SOCS3 signaling in the offspring. In addition, maternal obesity elevated inflammatory markers and altered NPY and POMC expression in the hypothalamus. Interestingly, combined parental obesity exacerbated the deleterious outcomes compared to single-parent obesity. In conclusion, while maternal obesity is known to program metabolic changes and obesity in offspring, the current study demonstrated that obese fathers induce hypothalamus inflammation in offspring, which may contribute to the development of metabolic syndromes in adulthood.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
31
|
Little appetite for obesity: meta-analysis of the effects of maternal obesogenic diets on offspring food intake and body mass in rodents. Int J Obes (Lond) 2015; 39:1669-78. [PMID: 26293233 DOI: 10.1038/ijo.2015.160] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 07/15/2015] [Accepted: 08/02/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND There is increasing recognition that maternal effects contribute to variation in individual food intake and metabolism. For example, many experimental studies on model animals have reported the effect of a maternal obesogenic diet during pregnancy on the appetite of offspring. However, the consistency of effects and the causes of variation among studies remain poorly understood. METHODS After a systematic search for relevant publications, we selected 53 studies on rats and mice for a meta-analysis. We extracted and analysed data on the differences in food intake and body weight between offspring of dams fed obesogenic diets and dams fed standard diets during gestation. We used meta-regression to study predictors of the strength and direction of the effect sizes. RESULTS We found that experimental offspring tended to eat more than control offspring but this difference was small and not statistically significant (0.198, 95% highest posterior density (HPD)=-0.118-0.627). However, offspring from dams on obesogenic diets were significantly heavier than offspring of control dams (0.591, 95% HPD=0.052-1.056). Meta-regression analysis revealed no significant influences of tested predictor variables (for example, use of choice vs no-choice maternal diet, offspring sex) on differences in offspring appetite. Dietary manipulations that extended into lactation had the largest effect on body weight. Subgroup analysis revealed that high protein to non-protein ratio of the maternal diet may promote increased body weight in experimental offspring in comparison with control offspring; low protein content in the maternal chow can have opposite effect. CONCLUSIONS Exposure to maternal obesogenic diets in early life is not likely to result in a substantial change in offspring appetite. Nevertheless, we found an effect on offspring body weight, consistent with permanent alterations of offspring metabolism in response to maternal diet. Additionally, it appears that protein content of the obesogenic diet and timing of manipulation modulate the effects on offspring body weight in later life.
Collapse
|
32
|
Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Programming of obesity and comorbidities in the progeny: lessons from a model of diet-induced obese parents. PLoS One 2015; 10:e0124737. [PMID: 25880318 PMCID: PMC4399989 DOI: 10.1371/journal.pone.0124737] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/03/2015] [Indexed: 01/07/2023] Open
Abstract
Aim To determine the impact of paternal obesity, maternal obesity or the combination of two obese parents on markers of adult offspring metabolism, with a focus on body mass (BM), lipid and carbohydrate, components of lipogenesis and beta-oxidation in the liver, sex dimorphism in the offspring that received a SC diet during the postnatal period. Materials and Methods Male and female C57BL/6 mice were fed a high-fat diet (HF; 49% lipids) or standard chow (SC; 17% lipids) for 8 weeks before mating until lactation. The offspring were labeled according to sex, maternal diet (first letters), paternal diet (second letters), and received a SCdiet until 12-weeks of age when they were sacrificed. BM, eating behavior, glucose tolerance, plasma analysis, gene and protein expression of the components of lipogenesis and beta-oxidation in the liver of offspring were evaluated. Results HF diet-fed mothers and fathers were overweight, hyperglycemic and glucose intolerant and had a deteriorating lipid profile. The adult male and female offspring of HF-mothers were overweight, with an increased adiposity index, hyperphagic, had an impaired glucose metabolism, increased total cholesterol and triacylglycerol levels, increased lipogenesis concomitant with decreased beta-oxidation resulting in liver steatosis. The male and female offspring of HF-father had impaired glucose metabolism, exacerbated lipogenesis without influencing beta-oxidation and enhanced hepatic steatosis. These findings are independent of BM. Male and female offspring of a mother and father that received a HF diet demonstrated these effects most prominently in adult life. Conclusion Paternal obesity leads to alterations in glucose metabolism, increase in components of lipogenesis and liver steatosis. In contrast, maternal obesity leads to overweight and changes in the metabolic profile and liver resulting from activation of hepatic lipogenesis with impaired beta-oxidation. When both parents are obese, the effects observed in the male and female offspring are exacerbated.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
33
|
Adult offspring of high-fat diet-fed dams can have normal glucose tolerance and body composition. J Dev Orig Health Dis 2015; 5:229-39. [PMID: 24901663 PMCID: PMC4098028 DOI: 10.1017/s2040174414000154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Maternal high-fat diet consumption and obesity have been shown to program long-term obesity and lead to impaired glucose tolerance in offspring. Many rodent studies, however, use non-purified, cereal-based diets as the control for purified high-fat diets. In this study, primiparous ICR mice were fed purified control diet (10–11 kcal% from fat of lard or butter origin) and lard (45 or 60 kcal% fat) or butter (32 or 60 kcal% fat)-based high-fat diets for 4 weeks before mating, throughout pregnancy, and for 2 weeks of nursing. Before mating, female mice fed the 32 and 60% butter-based high-fat diets exhibited impaired glucose tolerance but those females fed the lard-based diets showed normal glucose disposal following a glucose challenge. High-fat diet consumption by female mice of all groups decreased lean to fat mass ratios during the 4th week of diet treatment compared with those mice consuming the 10–11% fat diets. All females were bred to male mice and pregnancy and offspring outcomes were monitored. The body weight of pups born to 45% lard-fed dams was significantly increased before weaning, but only female offspring born to 32% butter-fed dams exhibited long-term body weight increases. Offspring glucose tolerance and body composition were measured for at least 1 year. Minimal, if any, differences were observed in the offspring parameters. These results suggest that many variables should be considered when designing future high-fat diet feeding and maternal obesity studies in mice.
Collapse
|
34
|
Bringhenti I, Ornellas F, Martins MA, Mandarim-de-Lacerda CA, Aguila MB. Early hepatic insult in the offspring of obese maternal mice. Nutr Res 2014; 35:136-45. [PMID: 25582085 DOI: 10.1016/j.nutres.2014.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/25/2014] [Accepted: 11/27/2014] [Indexed: 12/17/2022]
Abstract
We hypothesized that the maternal obesity initiates metabolic disorders associated with oxidative stress in the liver of offspring since early life. Mouse's mothers were assigned into 2 groups according to the diet offered (n = 10 per group): standard chow (SC) or high-fat diet (HF). The results revealed that HF offspring had an increase in body mass at day 10 (+25%, P < .05) and in glucose levels (+25%, P < .0001). Hepatic triacylglycerol was increased in HF offspring at day 1 and day 10 compared with SC offspring (+30%, P < .01 and +40%, P < .01) as was hepatic steatosis (+110%, P < .001; +145%, P < .0001). Fatty acid synthase was increased in HF offspring at day 1 (+450%, P < .01) and peroxisome proliferator activator receptor-γ was elevated at day 1 and day 10 (+140%, P < .01; +2741%, P < .01). Peroxisome proliferator activator receptor-α was diminished in HF offspring at day 10 compared with SC offspring (-100%, P < .01). Moreover, carnitine palmitoyl-CoA transferase-1 was decreased in HF offspring at day 1 and day 10 (-80%, P < .01; -60%, P < .05). In the HF offspring (compared with the SC offspring), the catalase and the superoxide dismutase were significantly lower in both days 1 and 10 (P < .05). In 10-day-old offspring, glutathione peroxidase 1 and glutathione reductase were lower in HF offspring than in SC offspring (P < .0001). Our findings suggest that the maternal obesity in mice induces an early oxidative dysfunction coupled with hepatic steatosis and might contribute to progressive liver injury later in life.
Collapse
Affiliation(s)
- Isabele Bringhenti
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ornellas
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcela Anjos Martins
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Graus-Nunes F, Dalla Corte Frantz E, Lannes WR, da Silva Menezes MC, Mandarim-de-Lacerda CA, Souza-Mello V. Pregestational maternal obesity impairs endocrine pancreas in male F1 and F2 progeny. Nutrition 2014; 31:380-7. [PMID: 25441581 DOI: 10.1016/j.nut.2014.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 08/04/2014] [Accepted: 08/10/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the effects of maternal obesity on pancreas structure and carbohydrate metabolism in early adult life, focusing on the F1 and F2 generations after F0 maternal pregestational, gestation, and lactation high-fat diet (HF). METHODS C57 BL/6 female mice (F0) were fed standard chow (SC) or an HF diet for 8 wk before mating and during the gestation and lactation periods to provide the F1 generation (F1-SC and F1-HF). At 3 mo old, F1 females were mated to produce the F2 generation (F2-SC and F2-HF). The male offspring from all groups were evaluated at 3 mo old. RESULTS F0-HF and F1-HF dams were overweight before gestation and had a higher body mass gain and energy intake during gestation, although only F0-HF dams presented pregestational hyperglycemia. The F1-HF offspring had higher body mass, energy intake, fasting glucose levels, and were glucose intolerant compared with F1-SC offspring. These parameters were not significantly altered in F2-HF offspring. Both F1-HF and F2-HF offspring showed hyperinsulinemia, hyperleptinemia, decreased adiponectin levels, increased pancreatic mass, and islet volume density with elevated α- and β-cell mass, hypertrophied islet characterized by an altered distribution of α- and β-cells and weak pancreatic-duodenal homeobox (Pdx)1 immunoreactivity. CONCLUSIONS Maternal HF diet consumed during the preconception period and throughout the gestation and lactation periods in mice promotes metabolism and pancreatic programming in F1 and F2 male offspring, implying intergenerational effects.
Collapse
Affiliation(s)
- Francielle Graus-Nunes
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Eliete Dalla Corte Frantz
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Wilian Rodrigues Lannes
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Mariel Caroline da Silva Menezes
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.
| |
Collapse
|
36
|
Dahlhoff M, Pfister S, Blutke A, Rozman J, Klingenspor M, Deutsch MJ, Rathkolb B, Fink B, Gimpfl M, Hrabě de Angelis M, Roscher AA, Wolf E, Ensenauer R. Peri-conceptional obesogenic exposure induces sex-specific programming of disease susceptibilities in adult mouse offspring. Biochim Biophys Acta Mol Basis Dis 2013; 1842:304-17. [PMID: 24275555 DOI: 10.1016/j.bbadis.2013.11.021] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 10/20/2013] [Accepted: 11/19/2013] [Indexed: 12/23/2022]
Abstract
Vulnerability of the fetus upon maternal obesity can potentially occur during all developmental phases. We aimed at elaborating longer-term health outcomes of fetal overnutrition during the earliest stages of development. We utilized Naval Medical Research Institute (NMRI) mice to induce pre-conceptional and gestational obesity and followed offspring outcomes in the absence of any postnatal obesogenic influences. Male adult offspring developed overweight, insulin resistance, hyperleptinemia, hyperuricemia and hepatic steatosis; all these features were not observed in females. Instead, they showed impaired fasting glucose and a reduced fat mass and adipocyte size. Influences of the interaction of maternal diet∗sex concerned offspring genes involved in fatty liver disease, lipid droplet size regulation and fat mass expansion. These data suggest that a peri-conceptional obesogenic exposure is sufficient to shape offspring gene expression patterns and health outcomes in a sex- and organ-specific manner, indicating varying developmental vulnerabilities between sexes towards metabolic disease in response to maternal overnutrition.
Collapse
Key Words
- ANOVA
- ATP citrate lyase
- AUC
- Acaca
- Acetyl-Coenzyme A carboxylase 1
- Acly
- Actb
- Analysis of variance
- Area under the curve
- B cell leukemia/lymphoma 2
- BW
- Bax
- Bcl2
- Bcl2-associated X protein
- Berardinelli–Seip congenital lipodystrophy 2 (also known as seipin)
- Beta-actin
- Body weight
- Bscl2
- CD
- CET
- CT
- Carbon dioxide production
- Carnitine palmitoyltransferase 1
- Cd36
- Cd36 antigen
- Cell death-inducing DNA fragmentation factor, alpha subunit-like effector A
- Central European Time
- Cidea
- Computed tomography
- Control diet
- Cpt1
- Day post coitum
- EEC
- European Economic Commission
- Exposure to maternal control diet
- Exposure to maternal high-fat, high-calorie diet
- FA
- Fabp4
- Fasn
- Fatty acid
- Fatty acid binding protein 4
- Fatty acid synthase
- GR
- GTT
- Glucocorticoid receptor
- Glucose tolerance test
- H&E
- HFD
- HMW
- HOMA-IR
- HP
- Hairy and enhancer of split 1
- Heat production
- Hematoxylin–eosin
- Hes1
- High-fat, high-calorie diet
- High-molecular-weight
- Homeostatic model assessment of insulin resistance
- Lep
- Leptin
- MD
- MDA
- MRI
- Magnetic resonance imaging
- Maintenance diet
- Malic enzyme 1
- Malondialdehyde
- Me1
- Mesoderm-specific transcript/imprinted paternally expressed gene 1 (also known as Peg1)
- Mest
- N
- NAFLD
- NEFA
- NMRI
- NRL
- Naval Medical Research Institute
- Nitrogen
- Non-alcoholic fatty liver disease
- Non-esterified fatty acid
- Nose–rump-length
- Nr1h3
- Nr3c1
- Nuclear receptor subfamily 1, group H, member 3 (also known as Lxra, liver X receptor alpha)
- Nuclear receptor subfamily 3, group C, member 1 (also known as Gr, glucocorticoid receptor)
- Obesity
- Offspring
- Oxygen consumption
- PFA
- Paraformaldehyde
- Patatin-like phospholipase domain-containing protein 2 (also known as Atgl, adipose triglyceride lipase)
- Peptidylprolyl isomerase A
- Peri-conceptional
- Perilipin 2
- Peroxisome proliferator activated receptor alpha
- Peroxisome proliferator activated receptor gamma
- Plin2
- Pnpla2
- Ppara
- Pparg
- Ppia
- Pregnancy
- Programming
- RER
- ROI
- Region of interest
- Respiratory exchange ratio
- S.e.m.
- Scd2
- Secreted frizzled-related sequence protein 5
- Sex-specificity
- Sfrp5
- Srebf1
- Standard error of the mean
- Stearoyl-Coenzyme A desaturase 2
- Sterol regulatory element binding transcription factor 1
- TBARS
- Thiobarbituric acid-reactive substances
- Ube2d2
- Ubiquitin-conjugating enzyme E2D 2
- VCO(2)
- VO(2)
- dpc
- mat-CD
- mat-HFD
Collapse
Affiliation(s)
- M Dahlhoff
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377 Munich, Germany.
| | - S Pfister
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337 Munich, Germany.
| | - A Blutke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Veterinärstrasse 13, 80539 Munich, Germany.
| | - J Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany; Molecular Nutritional Medicine, Else-Kröner Fresenius Center, Technische Universität München, Gregor-Mendel-Strasse 2, 85350 Freising-Weihenstephan, Germany.
| | - M Klingenspor
- Molecular Nutritional Medicine, Else-Kröner Fresenius Center, Technische Universität München, Gregor-Mendel-Strasse 2, 85350 Freising-Weihenstephan, Germany.
| | - M J Deutsch
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337 Munich, Germany.
| | - B Rathkolb
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377 Munich, Germany; German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany.
| | - B Fink
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337 Munich, Germany.
| | - M Gimpfl
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337 Munich, Germany.
| | - M Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Wissenschaftszentrum Weihenstephan, Technische Universität München, Alte Akademie 8, 85354 Freising, Germany; Member of German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany.
| | - A A Roscher
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337 Munich, Germany.
| | - E Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377 Munich, Germany.
| | - R Ensenauer
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337 Munich, Germany.
| |
Collapse
|