1
|
Ren J, Zhou L, Li S, Zhang Q, Xiao X. The roles of the gut microbiota, metabolites, and epigenetics in the effects of maternal exercise on offspring metabolism. Am J Physiol Endocrinol Metab 2024; 327:E760-E772. [PMID: 39535269 DOI: 10.1152/ajpendo.00200.2024] [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: 05/28/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Metabolic diseases, including obesity, dyslipidemia, and type 2 diabetes, have become severe challenges worldwide. The Developmental Origins of Health and Disease (DOHaD) hypothesis suggests that an adverse intrauterine environment can increase the risk of metabolic disorders in offspring. Studies have demonstrated that maternal exercise is an effective intervention for improving the offspring metabolic health. However, the pathways through which exercise works are unclear. It has been reported that the gut microbiota mediates the effect of maternal exercise on offspring metabolism, and epigenetic modifications have also been proposed to be important molecular mechanisms. Microbial metabolites can influence epigenetics by providing substrates for DNA or histone modifications, binding to G-protein-coupled receptors to affect downstream pathways, or regulating the activity of epigenetic modifying enzymes. This review aims to summarize the intergenerational effect of maternal exercise and proposes that gut microbiota-metabolites-epigenetic regulation is an important mechanism by which maternal exercise improves offspring metabolism, which may yield novel targets for the early prevention and intervention of metabolic diseases.
Collapse
Affiliation(s)
- Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Pontes CNR, Bessa ADSMD, Macedo LM, Ferreira-Junior MD, Cavalcante KVN, Campos HM, Cruz-Leite VRM, Neves ÂR, Gomes RM, Ghedini PC, Biancardi MF, Mendes EP, Borges CL, Pedrino GR, Castro CH. Angiotensin-(1-7) Treatment Early in Life Prevents Cardiac Hypertrophy in Adult Hypertensive Rats. J Cardiovasc Pharmacol 2024; 83:457-465. [PMID: 39536129 DOI: 10.1097/fjc.0000000000001530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/30/2023] [Indexed: 03/20/2024]
Abstract
ABSTRACT Angiotensin (Ang)-(1-7) is a cardioprotective peptide of the renin-angiotensin system. Prepuberty has been considered as a later susceptible window of development, and stressful factors in this life phase can induce chronic diseases in adulthood. We aimed to investigate whether the treatment with Ang-(1-7) during the prepuberty could attenuate the development of hypertension and cardiac injury in adult spontaneously hypertensive rats (SHRs). SHRs were treated with Ang-(1-7) (24 μg/kg/h) from age 4 to 7 weeks. Systolic blood pressure was measured by tail-cuff plethysmography up to 17th week. Thereafter, echocardiography was performed, and the rats were euthanized for the collection of tissues and blood. Ang-(1-7) did not change the systolic blood pressure but reduced the septal and posterior wall thickness, and cardiomyocyte hypertrophy and fibrosis in SHR. In addition, Ang-(1-7) reduced the gene expression of atrial natriuretic peptide and brain natriuretic peptide, increased the metalloproteinase 9 expression, and reduced the extracellular signal-regulated kinases 1/2 phosphorylation. Ang-(1-7) also prevented the reduction of Mas receptor but did not change the protein expression of angiotensin-converting enzyme, angiotensin-converting enzyme 2, AT1, and AT2. The treatment with Ang-(1-7) decreased the malondialdehyde (MDA) levels and increased superoxide dismutase-1 and catalase activities and protein expression of catalase. Our findings demonstrate that the treatment of SHR with Ang-(1-7) for 3 weeks early in life promotes beneficial effects in the heart later in life, even without altering blood pressure, through mechanisms involving the reduction of oxidative stress and ERK1/2 phosphorylation. In addition, this study supports the prepuberty as an important programming window.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ângela Ribeiro Neves
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Rodrigo Mello Gomes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | | | | | - Clayton Luiz Borges
- Department of Biochemistry and Molecular Biology, Federal University of Goiás, Goiânia, Brazil
| | | | - Carlos Henrique Castro
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics
| |
Collapse
|
3
|
Moholdt T, Stanford KI. Exercised breastmilk: a kick-start to prevent childhood obesity? Trends Endocrinol Metab 2024; 35:23-30. [PMID: 37735048 PMCID: PMC11005327 DOI: 10.1016/j.tem.2023.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023]
Abstract
Exercise has systemic health benefits through effects on multiple tissues, with intertissue communication. Recent studies indicate that exercise may improve breastmilk composition and thereby reduce the intergenerational transmission of obesity. Even if breastmilk is considered optimal infant nutrition, there is evidence for variations in its composition between mothers who are normal weight, those with obesity, and those who are physically active. Nutrition early in life is important for later-life susceptibility to obesity and other metabolic diseases, and maternal exercise may provide protection against the development of metabolic disease. Here we summarize recent research on the influence of maternal obesity on breastmilk composition and discuss the potential role of exercise-induced adaptations to breastmilk as a kick-start to prevent childhood obesity.
Collapse
Affiliation(s)
- Trine Moholdt
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynaecology and Obstetrics, St. Olav's Hospital, Trondheim, Norway.
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
4
|
Tando Y, Matsui Y. Inheritance of environment-induced phenotypic changes through epigenetic mechanisms. ENVIRONMENTAL EPIGENETICS 2023; 9:dvad008. [PMID: 38094661 PMCID: PMC10719065 DOI: 10.1093/eep/dvad008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 03/08/2024]
Abstract
Growing evidence suggests that epigenetic changes through various parental environmental factors alter the phenotypes of descendants in various organisms. Environmental factors, including exposure to chemicals, stress and abnormal nutrition, affect the epigenome in parental germ cells by different epigenetic mechanisms, such as DNA methylation, histone modification as well as small RNAs via metabolites. Some current remaining questions are the causal relationship between environment-induced epigenetic changes in germ cells and altered phenotypes of descendants, and the molecular basis of how the abnormal epigenetic changes escape reprogramming in germ cells. In this review, we introduce representative examples of intergenerational and transgenerational inheritance of phenotypic changes through parental environmental factors and the accompanied epigenetic and metabolic changes, with a focus on animal species. We also discuss the molecular mechanisms of epigenomic inheritance and their possible biological significance.
Collapse
Affiliation(s)
- Yukiko Tando
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Yasuhisa Matsui
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
5
|
Ruebel ML, Borengasser SJ, Zhong Y, Kang P, Faske J, Shankar K. Maternal Exercise Prior to and during Gestation Induces Sex-Specific Alterations in the Mouse Placenta. Int J Mol Sci 2023; 24:16441. [PMID: 38003633 PMCID: PMC10671464 DOI: 10.3390/ijms242216441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
While exercise (EX) during pregnancy is beneficial for both mother and child, little is known about the mechanisms by which maternal exercise mediates changes in utero. Six-week-old female C57BL/6 mice were divided into two groups: with (exercise, EX; N = 7) or without (sedentary, SED; N = 8) access to voluntary running wheels. EX was provided via 24 h access to wheels for 10 weeks prior to conception until late pregnancy (18.5 days post coitum). Sex-stratified placentas and fetal livers were collected. Microarray analysis of SED and EX placentas revealed that EX affected gene transcript expression of 283 and 661 transcripts in male and female placentas, respectively (±1.4-fold, p < 0.05). Gene Set Enrichment and Ingenuity Pathway Analyses of male placentas showed that EX led to inhibition of signaling pathways, biological functions, and down-regulation of transcripts related to lipid and steroid metabolism, while EX in female placentas led to activation of pathways, biological functions, and gene expression related to muscle growth, brain, vascular development, and growth factors. Overall, our results suggest that the effects of maternal EX on the placenta and presumably on the offspring are sexually dimorphic.
Collapse
Affiliation(s)
- Meghan L. Ruebel
- Microbiome and Metabolism Research Unit, USDA-ARS, Southeast Area, Little Rock, AR 72202, USA;
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Sarah J. Borengasser
- Tobacco Settlement Endowment Trust Health Promotion Research Center, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics—Endocrinology & Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ying Zhong
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Ping Kang
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
| | - Jennifer Faske
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (Y.Z.); (J.F.)
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Elías-López AL, Vázquez-Mena O, Sferruzzi-Perri AN. Mitochondrial dysfunction in the offspring of obese mothers and it's transmission through damaged oocyte mitochondria: Integration of mechanisms. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166802. [PMID: 37414229 DOI: 10.1016/j.bbadis.2023.166802] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/23/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
In vivo and in vitro studies demonstrate that mitochondria in the oocyte, are susceptible to damage by suboptimal pre/pregnancy conditions, such as obesity. These suboptimal conditions have been shown to induce mitochondrial dysfunction (MD) in multiple tissues of the offspring, suggesting that mitochondria of oocytes that pass from mother to offspring, can carry information that can programme mitochondrial and metabolic dysfunction of the next generation. They also suggest that transmission of MD could increase the risk of obesity and other metabolic diseases in the population inter- and trans-generationally. In this review, we examined whether MD observed in offspring tissues of high energetic demand, is the result of the transmission of damaged mitochondria from the oocytes of obese mothers to the offspring. The contribution of genome-independent mechanisms (namely mitophagy) in this transmission were also explored. Finally, potential interventions aimed at improving oocyte/embryo health were investigated, to see if they may provide an opportunity to halter the generational effects of MD.
Collapse
Affiliation(s)
- A L Elías-López
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", México.
| | | | - A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK.
| |
Collapse
|
7
|
Gupta MK, Peng H, Li Y, Xu CJ. The role of DNA methylation in personalized medicine for immune-related diseases. Pharmacol Ther 2023; 250:108508. [PMID: 37567513 DOI: 10.1016/j.pharmthera.2023.108508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Epigenetics functions as a bridge between host genetic & environmental factors, aiding in human health and diseases. Many immune-related diseases, including infectious and allergic diseases, have been linked to epigenetic mechanisms, particularly DNA methylation. In this review, we summarized an updated overview of DNA methylation and its importance in personalized medicine, and demonstrated that DNA methylation has excellent potential for disease prevention, diagnosis, and treatment in a personalized manner. The future implications and limitations of the DNA methylation study have also been well-discussed.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - He Peng
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; Department of Internal Medicine and Radboud Institute for Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany; Department of Internal Medicine and Radboud Institute for Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
8
|
Khasanova A, Henagan TM. Exercise Is Medicine: How Do We Implement It? Nutrients 2023; 15:3164. [PMID: 37513581 PMCID: PMC10385293 DOI: 10.3390/nu15143164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Exercise is well known to have beneficial effects on various disease states. In this paper, we broadly describe the fundamental concepts that are shared among various disease states, including obesity, type 2 diabetes (T2D), cardiovascular disease (CVD), heart failure (HF), cancer, and psychological well-being, and the beneficial effects of exercise training within these concepts. We highlight issues involved in implementing exercise recommendations and describe the potential impacts and challenges to medical professionals and patients. Problems are identified and discussed with respect to the future roles of professionals in the current built environment with its limited infrastructure to support current physical activity recommendations.
Collapse
Affiliation(s)
- Aliya Khasanova
- Department of Family Medicine, Baton Rouge General Family Health Center, Baton Rouge, LA 70806, USA
- Department of Family Medicine, Baton Rouge General Hospital, Baton Rouge, LA 70808, USA
| | - Tara M Henagan
- Department of Family Medicine, Baton Rouge General Family Health Center, Baton Rouge, LA 70806, USA
- Department of Family Medicine, Baton Rouge General Hospital, Baton Rouge, LA 70808, USA
| |
Collapse
|
9
|
Agarwal M, Hoffman J, Ngo Tenlep SY, Santarossa S, Pearson KJ, Sitarik AR, Cassidy-Bushrow AE, Petriello MC. Maternal polychlorinated biphenyl 126 (PCB 126) exposure modulates offspring gut microbiota irrespective of diet and exercise. Reprod Toxicol 2023; 118:108384. [PMID: 37061048 PMCID: PMC10257154 DOI: 10.1016/j.reprotox.2023.108384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
The gut microbiota plays an important role throughout the lifespan in maintaining host health, and several factors can modulate microbiota composition including diet, exercise, and environmental exposures. Maternal microbiota is transferred to offspring during early life; thus, environmental exposures before gestation may also modulate offspring microbiota. Here we aimed to investigate the effects of maternal exposure to dioxin-like polychlorinated biphenyls (PCBs) on the microbiota of aged offspring and to determine if lifestyle factors, including maternal exercise or offspring high-fat feeding alter these associations. To test this, dams were exposed to PCB 126 (0.5 μmole/kg body weight) or vehicle oil by oral gavage during preconception, gestation, and during lactation. Half of each group was allowed access to running wheels for ≥ 7 days before and during pregnancy and up through day 14 of lactation. Female offspring born from the 4 maternal groups (PCB exposure or not, with/without exercise) were subsequently placed either on regular diet or switched to a high-fat diet during adulthood. Microbiota composition was quantified in female offspring at 49 weeks of age by 16 S rRNA sequencing. Maternal exposure to PCB 126 resulted in significantly reduced richness and diversity in offspring microbiota regardless of diet or exercise. Overall compositional differences were largely driven by offspring diet, but alterations in specific taxa due to maternal PCB 126 exposure, included the depletion of Verrucomicrobiaceae and Akkermansia muciniphila, and an increase in Anaeroplasma. Perturbation of microbiota due to PCB 126 may predispose offspring to a variety of chronic diseases later in adulthood.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA
| | - Jessie Hoffman
- Department of Human Nutrition, Winthrop University, Rock Hill, SC 29733, USA
| | - Sara Y Ngo Tenlep
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, USA
| | - Sara Santarossa
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI 48202, USA
| | - Kevin J Pearson
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, USA
| | - Alexandra R Sitarik
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI 48202, USA
| | | | - Michael C Petriello
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
10
|
Purcell AR, Glastras SJ. Maternal Weight Management to Prevent the Developmental Programming of MAFLD in Offspring of Obese Mothers. Nutrients 2023; 15:2155. [PMID: 37432265 DOI: 10.3390/nu15092155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 07/12/2023] Open
Abstract
The global surge of obesity amongst women of reproductive age has raised concerns surrounding the health consequences for their offspring as there is a formidable link between an obesogenic maternal environment and the developmental programming of metabolic dysfunction in the offspring. Specifically, the offspring of mothers with obesity have a three-fold higher risk of developing metabolic-associated fatty liver disease (MAFLD) compared to the offspring of healthy-weight mothers. Given the burgeoning burden of obesity and its comorbidities, it is essential to focus research efforts on methods to alleviate the intergenerational onset of obesity and MAFLD. This review summarizes the current research surrounding the developmental programming of MAFLD in the offspring of mothers with obesity and examines the potential for weight interventions to prevent such metabolic dysfunction in the offspring. It focuses on the benefits of pre-pregnancy interventional strategies, including dietary and exercise intervention, to ameliorate adverse liver health outcomes in the offspring. The utility and translation of these interventions for humans may be difficult for prospective mothers with obesity, thus the use of pre-pregnancy therapeutic weight loss aids, such as glucagon-like peptide-1 receptor agonists, is also discussed.
Collapse
Affiliation(s)
- Amanda Renae Purcell
- Kolling Institute of Medical Research, Sydney 2065, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Sarah Jean Glastras
- Kolling Institute of Medical Research, Sydney 2065, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, Sydney 2065, Australia
| |
Collapse
|
11
|
Hernández-Saavedra D, Markunas C, Takahashi H, Baer LA, Harris JE, Hirshman MF, Ilkayeva O, Newgard CB, Stanford KI, Goodyear LJ. Maternal Exercise and Paternal Exercise Induce Distinct Metabolite Signatures in Offspring Tissues. Diabetes 2022; 71:2094-2105. [PMID: 35838316 PMCID: PMC9501651 DOI: 10.2337/db22-0341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/12/2022] [Indexed: 01/19/2023]
Abstract
That maternal and paternal exercise improve the metabolic health of adult offspring is well established. Tissue and serum metabolites play a fundamental role in the health of an organism, but how parental exercise affects offspring tissue and serum metabolites has not yet been investigated. Here, male and female breeders were fed a high-fat diet and housed with or without running wheels before breeding (males) and before and during gestation (females). Offspring were sedentary and chow fed, with parents as follows: sedentary (Sed), maternal exercise (MatEx), paternal exercise (PatEx), or maternal+paternal exercise (Mat+PatEx). Adult offspring from all parental exercise groups had similar improvement in glucose tolerance and hepatic glucose production. Targeted metabolomics was performed in offspring serum, liver, and triceps muscle. Offspring from MatEx, PatEx, and Mat+PatEx each had a unique tissue metabolite signature, but Mat+PatEx offspring had an additive phenotype relative to MatEx or PatEx alone in a subset of liver and muscle metabolites. Tissue metabolites consistently indicated that the metabolites altered with parental exercise contribute to enhanced fatty acid oxidation. These data identify distinct tissue-specific adaptations and mechanisms for parental exercise-induced improvement in offspring metabolic health. Further mining of this data set could aid the development of novel therapeutic targets to combat metabolic diseases.
Collapse
Affiliation(s)
- Diego Hernández-Saavedra
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL
| | - Christina Markunas
- Departments of Pharmacology and Cancer Biology and Medicine, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Durham, NC
| | - Hirokazu Takahashi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Lisa A. Baer
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Johan E. Harris
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Michael F. Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Olga Ilkayeva
- Departments of Pharmacology and Cancer Biology and Medicine, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Durham, NC
| | - Christopher B. Newgard
- Departments of Pharmacology and Cancer Biology and Medicine, Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Durham, NC
| | - Kristin I. Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Laurie J. Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Maternal training during lactation modifies breast milk fatty acid composition and male offspring glucose homeostasis in rat. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159223. [PMID: 35987325 DOI: 10.1016/j.bbalip.2022.159223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022]
Abstract
The perinatal exposome can modify offspring metabolism and health later in life. Within this concept, maternal exercise during gestation has been reported modifying offspring glucose sensing and homeostasis, while the impact of such exercise during lactation is little-known. We thus aimed at evaluating short- and long-term effects of it on offspring pancreatic function, assuming a link with changes in breast milk composition. Fifteen-week-old primiparous female Wistar rats exercised during lactation at a constant submaximal intensity (TR) or remained sedentary (CT). Male offspring were studied at weaning and at 7 months of age for growth, pancreas weight, glycemia and insulin responses. Milk protein content was determined by the bicinchoninic acid assay (BCA colorimetric method), and lipid content and fatty acid composition by gas chromatography. Mature milk from TR rats contained significantly less saturated (-7 %) and more monounsaturated (+18 %) and polyunsaturated (PUFA +12 %) fatty acids compared to CT rats, with no difference in total lipid and protein concentrations. In offspring from TR vs CT mothers, fasting glycemia was lower, pancreas weight was higher with a lower insulin content (-37 %) at weaning. Such outcomes were correlated with milk PUFA levels and indices of desaturase or elongase activities. These effects were no longer present at 7 months, whereas a more efficient muscle insulin sensitivity was observed. Maternal training during lactation led to a specific milk phenotype that was associated with a short-term impact on glucose homeostasis and pancreatic function of the male offspring.
Collapse
|
13
|
Effects of maternal controlled exercise on offspring adiposity and glucose tolerance. J Dev Orig Health Dis 2022; 13:455-462. [PMID: 34503602 PMCID: PMC8907328 DOI: 10.1017/s2040174421000489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
While metabolic disorders such as obesity and diabetes are costly and deadly to the current population, they are also extremely detrimental to the next generation. Much of the current literature focuses on the negative impact of poor maternal choices on offspring disease, while there is little work examining maternal behaviors that may improve offspring health. Research has shown that voluntary maternal exercise in mouse models improves metabolic function in offspring. In this study, we hypothesized that controlled maternal exercise in a mouse model will effect positive change on offspring obesity and glucose homeostasis. Female mice were separated into three groups: home cage, sedentary, and exercise. The sedentary home cage group was not removed from the home cage, while the sedentary wheel group was removed from the cage and placed in an immobile wheel apparatus. The exercise group was removed from the home cage and run on the same wheel apparatus but with the motor activated at 5-10 m/min for 1 h/d prior to and during pregnancy. Offspring were subjected to oral glucose tolerance testing and body composition analysis. There was no significant difference in offspring glucose tolerance or body composition as a consequence of the maternal exercise intervention compared to the sedentary wheel group. There were no marked negative consequences of the maternal controlled exercise intervention. Further research should clarify the potential advantages of the controlled exercise model and improve experimental techniques to facilitate translation of this research to human applications.
Collapse
|
14
|
Chaves A, Weyrauch LA, Zheng D, Biagioni EM, Krassovskaia PM, Davidson BL, Broskey NT, Boyle KE, May LE, Houmard JA. Influence of Maternal Exercise on Glucose and Lipid Metabolism in Offspring Stem Cells: ENHANCED by Mom. J Clin Endocrinol Metab 2022; 107:e3353-e3365. [PMID: 35511592 PMCID: PMC12102725 DOI: 10.1210/clinem/dgac270] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 02/06/2023]
Abstract
CONTEXT Recent preclinical data suggest exercise during pregnancy can improve the metabolic phenotype not only of the mother, but of the developing offspring as well. However, investigations in human offspring are lacking. OBJECTIVE To characterize the effect of maternal aerobic exercise on the metabolic phenotype of the offspring's mesenchymal stem cells (MSCs). DESIGN Randomized controlled trial. SETTING Clinical research facility. PATIENTS Healthy female adults between 18 and 35 years of age and ≤ 16 weeks' gestation. INTERVENTION Mothers were randomized into 1 of 2 groups: aerobic exercise (AE, n = 10) or nonexercise control (CTRL, n = 10). The AE group completed 150 minutes of weekly moderate-intensity exercise, according to American College of Sports Medicine guidelines, during pregnancy, whereas controls attended stretching sessions. MAIN OUTCOME MEASURES Following delivery, MSCs were isolated from the umbilical cord of the offspring and metabolic tracer and immunoblotting experiments were completed in the undifferentiated (D0) or myogenically differentiated (D21) state. RESULTS AE-MSCs at D0 had an elevated fold-change over basal in insulin-stimulated glycogen synthesis and reduced nonoxidized glucose metabolite (NOGM) production (P ≤ 0.05). At D21, AE-MSCs had a significant elevation in glucose partitioning toward oxidation (oxidation/NOGM ratio) compared with CTRL (P ≤ 0.05). Immunoblot analysis revealed elevated complex I expression in the AE-MSCs at D21 (P ≤ 0.05). Basal and palmitate-stimulated lipid metabolism was similar between groups at D0 and D21. CONCLUSIONS These data provide evidence of a programmed metabolic phenotype in human offspring with maternal AE during pregnancy.
Collapse
Affiliation(s)
- Alec Chaves
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Luke A Weyrauch
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Donghai Zheng
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Ericka M Biagioni
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Polina M Krassovskaia
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Breanna L Davidson
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Nicholas T Broskey
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Kristen E Boyle
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Linda E May
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Joseph A Houmard
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
15
|
Rodrigo N, Saad S, Pollock C, Glastras SJ. Diet Modification before or during Pregnancy on Maternal and Foetal Outcomes in Rodent Models of Maternal Obesity. Nutrients 2022; 14:2154. [PMID: 35631295 PMCID: PMC9146671 DOI: 10.3390/nu14102154] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/10/2022] Open
Abstract
The obesity epidemic has serious implications for women of reproductive age; its rising incidence is associated not just with health implications for the mother but also has transgenerational ramifications for the offspring. Increased incidence of diabetes, cardiovascular disease, obesity, and kidney disease are seen in both the mothers and the offspring. Animal models, such as rodent studies, are fundamental to studying maternal obesity and its impact on maternal and offspring health, as human studies lack rigorous controlled experimental design. Furthermore, the short and prolific reproductive potential of rodents enables examination across multiple generations and facilitates the exploration of interventional strategies to mitigate the impact of maternal obesity, both before and during pregnancy. Given that obesity is a major public health concern, it is important to obtain a greater understanding of its pathophysiology and interaction with reproductive health, placental physiology, and foetal development. This narrative review focuses on the known effects of maternal obesity on the mother and the offspring, and the benefits of interventional strategies, including dietary intervention, before or during pregnancy on maternal and foetal outcomes. It further examines the contribution of rodent models of maternal obesity to elucidating pathophysiological pathways of disease development, as well as methods to reduce the impact of obesity on the mothers and the developing foetus. The translation of these findings into the human experience will also be discussed.
Collapse
Affiliation(s)
- Natassia Rodrigo
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, Sydney 2065, Australia;
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Sonia Saad
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Carol Pollock
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
- Department of Renal Medicine, Royal North Shore Hospital, Sydney 2065, Australia
| | - Sarah J. Glastras
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, Sydney 2065, Australia;
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
16
|
Recent Experimental Studies of Maternal Obesity, Diabetes during Pregnancy and the Developmental Origins of Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms23084467. [PMID: 35457285 PMCID: PMC9027277 DOI: 10.3390/ijms23084467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/14/2022] Open
Abstract
Globally, cardiovascular disease remains the leading cause of death. Most concerning is the rise in cardiovascular risk factors including obesity, diabetes and hypertension among youth, which increases the likelihood of the development of earlier and more severe cardiovascular disease. While lifestyle factors are involved in these trends, an increasing body of evidence implicates environmental exposures in early life on health outcomes in adulthood. Maternal obesity and diabetes during pregnancy, which have increased dramatically in recent years, also have profound effects on fetal growth and development. Mounting evidence is emerging that maternal obesity and diabetes during pregnancy have lifelong effects on cardiovascular risk factors and heart disease development. However, the mechanisms responsible for these observations are unknown. In this review, we summarize the findings of recent experimental studies, showing that maternal obesity and diabetes during pregnancy affect energy metabolism and heart disease development in the offspring, with a focus on the mechanisms involved. We also evaluate early proof-of-concept studies for interventions that could mitigate maternal obesity and gestational diabetes-induced cardiovascular disease risk in the offspring.
Collapse
|
17
|
Stevanović-Silva J, Beleza J, Coxito P, Costa RC, Ascensão A, Magalhães J. Fit mothers for a healthy future: Breaking the intergenerational cycle of non-alcoholic fatty liver disease with maternal exercise. Eur J Clin Invest 2022; 52:e13596. [PMID: 34120338 DOI: 10.1111/eci.13596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022]
Abstract
UNLABELLED SPECIAL ISSUE: 'FOIEGRAS-Bioenergetic Remodelling in the Pathophysiology and Treatment of Non-Alcoholic Fatty Liver Disease'. BACKGROUND Non-alcoholic fatty liver disease (NAFLD) emerges as significant health burden worldwide. Lifestyle changes, unhealthy dietary habits and physical inactivity, can trigger NAFLD development. Persisting on these habits during pregnancy affects in utero environment and prompts a specific metabolic response in foetus resulting in offspring metabolic maladjustments potentially critical for developing NAFLD later in life. The increasing prevalence of NAFLD, particularly in children, has shifted the research focus towards preventive and therapeutic strategies. Yet, designing effective approaches that can break the NAFLD intergenerational cycle becomes even more complicated. Regular physical exercise (PE) is a powerful non-pharmacological strategy known to counteract deleterious metabolic outcomes. In this narrative review, we aimed to briefly describe NAFLD pathogenesis focusing on maternal nutritional challenge and foetal programming, and to provide potential mechanisms behind the putative intergenerational effect of PE against metabolic diseases, including liver diseases. METHODS Following detailed electronic database search, recent existing evidence about NAFLD development, intergenerational programming and gestational exercise effects was critically analysed and discussed. RESULTS PE during pregnancy could have a great potential to counteract intergenerational transmission of metabolic burden. The interplay between different PE roles-metabolic, endocrine and epigenetic-could offer a more stable in utero environment to the foetus, thus rescuing offspring vulnerability to metabolic disturbances. CONCLUSIONS The better understanding of maternal PE beneficial consequences on offspring metabolism could reinforce the importance of PE during pregnancy as an indispensable strategy in improving offspring health.
Collapse
Affiliation(s)
- Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
18
|
Maternal exercise and high-fat diet affect hypothalamic neural projections in rat offspring in a sex-specific manner. J Nutr Biochem 2022; 103:108958. [DOI: 10.1016/j.jnutbio.2022.108958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/20/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022]
|
19
|
Freitas-Dias R, Lima TI, Costa-Junior JM, Gonçalves LM, Araujo HN, Paula FMM, Santos GJ, Branco RCS, Ou K, Kaestner KH, Silveira LR, Oliveira CAM, Boschero AC, Zoppi CC, Carneiro EM. Offspring from trained male mice inherit improved muscle mitochondrial function through PPAR co-repressor modulation. Life Sci 2021; 291:120239. [PMID: 34942163 DOI: 10.1016/j.lfs.2021.120239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 10/19/2022]
Abstract
Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.
Collapse
Affiliation(s)
- Ricardo Freitas-Dias
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Physical Therapy, Laboratory of Exercise Physiology, University of Pernambuco, Petrolina, PE, Brazil
| | - Tanes I Lima
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jose Maria Costa-Junior
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luciana M Gonçalves
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Flavia M M Paula
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Gustavo J Santos
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianopolis, SC, Brazil
| | - Renato Chaves Souto Branco
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Kristy Ou
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Camila A M Oliveira
- Department of Biosciences, Federal University of Sao Paulo, Santos, SP, Brazil
| | - Antonio C Boschero
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Claudio C Zoppi
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Everardo M Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
20
|
Wu G, Zhang X, Gao F. The epigenetic landscape of exercise in cardiac health and disease. JOURNAL OF SPORT AND HEALTH SCIENCE 2021; 10:648-659. [PMID: 33333247 PMCID: PMC8724625 DOI: 10.1016/j.jshs.2020.12.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/16/2020] [Accepted: 11/16/2020] [Indexed: 05/02/2023]
Abstract
With the rising incidence of cardiovascular diseases, the concomitant mortality and morbidity impose huge burdens on quality of life and societal costs. It is generally accepted that physical inactivity is one of the major risk factors for cardiac disease and that exercise benefits the heart in both physiological and pathologic conditions. However, the molecular mechanisms governing the cardioprotective effects exerted by exercise remain incompletely understood. Most recently, an increasing number of studies indicate the involvement of epigenetic modifications in the promotion of cardiac health and prevention of cardiac disease. Exercise and other lifestyle factors extensively induce epigenetic modifications, including DNA/RNA methylation, histone post-translational modifications, and non-coding RNAs in multiple tissues, which may contribute to their positive effects in human health and diseases. In addition, several studies have shown that maternal or paternal exercise prevents age-associated or high-fat diet-induced metabolic dysfunction in the offspring, reinforcing the importance of epigenetics in mediating the beneficial effects of exercise. It has been shown that exercise can directly modify cardiac epigenetics to promote cardiac health and protect the heart against various pathological processes, or it can modify epigenetics in other tissues, which reduces the risk of cardiac disease and affords cardioprotection through exerkines. An in-depth understanding of the epigenetic landscape of cardioprotective response to exercise will provide new therapeutic targets for cardiac diseases. This review, therefore, aimed to acquaint the cardiac community with the rapidly advancing and evolving field of exercise and epigenetics.
Collapse
Affiliation(s)
- Guiling Wu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
21
|
Beleza J, Stevanović-Silva J, Coxito P, Costa RC, Ascensão A, Torrella JR, Magalhães J. Building-up fit muscles for the future: Transgenerational programming of skeletal muscle through physical exercise. Eur J Clin Invest 2021; 51:e13515. [PMID: 33580562 DOI: 10.1111/eci.13515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/19/2022]
Abstract
'Special issue - In Utero and Early Life Programming of Aging and Disease'. Skeletal muscle (SM) adaptations to physical exercise (PE) have been extensively studied due, not only to the relevance of its in situ plasticity, but also to the SM endocrine-like effects in noncontractile tissues, such as brain, liver or adipocytes. Regular PE has been considered a pleiotropic nonpharmacological strategy to prevent and counteract the deleterious consequences of several metabolic, cardiovascular, oncological and neurodegenerative disorders. Additionally, PE performed by parents seems to have a direct impact in the offspring through the transgenerational programming of different tissues, such as SM. In fact, SM offspring programming mechanisms seems to be orchestrated, at least in part, by epigenetic machinery conditioning transcriptional or post-transcriptional processes. Ultimately, PE performed in the early in life is also a critical window of opportunity to positively modulate the juvenile and adult phenotype. Parental PE has a positive impact in several health-related offspring outcomes, such as SM metabolism, differentiation, morphology and ultimately in offspring exercise volition and endurance. Also, early-life PE counteracts conceptional-related adverse effects and induces long-lasting healthy benefits throughout adulthood. Additionally, epigenetics mechanisms seem to play a key role in the PE-induced SM adaptations. Despite the undoubtedly positive role of parental and early-life PE on SM phenotype, a strong research effort is still needed to better understand the mechanisms that positively regulate PE-induced SM programming.
Collapse
Affiliation(s)
- Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Joan Ramon Torrella
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| |
Collapse
|
22
|
Song L, Yan J, Wang N, Wei X, Luo X, Meng K, Sun B. Prenatal exercise reverses high-fat-diet-induced placental alterations and alters male fetal hypothalamus during late gestation in rats†. Biol Reprod 2021; 102:705-716. [PMID: 31742332 DOI: 10.1093/biolre/ioz213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/14/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Maternal high-fat (HF) diet negatively affects maternal metabolism and placental function. This study aimed to determine whether gestational exercise prevents the effect of HF diet on placental amino acid transporter expression and nutrient-sensing signaling and the fetal response. Pregnant Sprague-Dawley rats were either fed with a CHOW (13.5% fat) or HF (60% fat) diet during gestation and further divided into two subgroups: voluntary exercised and sedentary. Placentae were collected on gestational day (GD) 14 and GD20, and male placentae were used in this study. We found that gestational exercise ameliorated the detrimental effects of HF diet on dams' adiposity, plasma leptin, and insulin concentrations. Maternal exercise did not influence fetoplacental growth but affected male fetal hypothalamic Leprb, Stat3, Insr, Agrp, and Pomc expressions on GD20. Maternal HF diet decreased placental labyrinth thickness and increased system A amino acid transporter SNAT2 expression, while these changes were normalized by exercise. The activation of placental mechanistic target of rapamycin complex 1/4E-BP1 and LepRb/STAT3 signaling might contribute to the increased placental SNAT2 expression in HF-fed dams, which were reversed by exercise on GD20. These data highlight that gestational exercise reverses HF-diet-induced placental alterations during late gestation without influencing fetal growth. However, maternal exercise altered fetal hypothalamic gene expression, which may affect long-term offspring health.
Collapse
Affiliation(s)
- Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Nan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kai Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
23
|
Krassovskaia PM, Chaves AB, Houmard JA, Broskey NT. Exercise during Pregnancy: Developmental Programming Effects and Future Directions in Humans. Int J Sports Med 2021; 43:107-118. [PMID: 34344043 DOI: 10.1055/a-1524-2278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Epidemiological studies show that low birth weight is associated with mortality from cardiovascular disease in adulthood, indicating that chronic diseases could be influenced by hormonal or metabolic insults encountered in utero. This concept, now known as the Developmental Origins of Health and Disease hypothesis, postulates that the intrauterine environment may alter the structure and function of the organs of the fetus as well as the expression of genes that impart an increased vulnerability to chronic diseases later in life. Lifestyle interventions initiated during the prenatal period are crucial as there is the potential to attenuate progression towards chronic diseases. However, how lifestyle interventions such as physical activity directly affect human offspring metabolism and the potential mechanisms involved in regulating metabolic balance at the cellular level are not known. The purpose of this review is to highlight the effects of exercise during pregnancy on offspring metabolic health and emphasize gaps in the current human literature and suggestions for future research.
Collapse
Affiliation(s)
- Polina M Krassovskaia
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| | - Alec B Chaves
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| | - Joseph A Houmard
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| | - Nicholas T Broskey
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| |
Collapse
|
24
|
Shrestha A, Prowak M, Berlandi-Short VM, Garay J, Ramalingam L. Maternal Obesity: A Focus on Maternal Interventions to Improve Health of Offspring. Front Cardiovasc Med 2021; 8:696812. [PMID: 34368253 PMCID: PMC8333710 DOI: 10.3389/fcvm.2021.696812] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity has many implications for offspring health that persist throughout their lifespan that include obesity and cardiovascular complications. Several different factors contribute to obesity and they encompass interplay between genetics and environment. In the prenatal period, untreated obesity establishes a foundation for a myriad of symptoms and negative delivery experiences, including gestational hypertensive disorders, gestational diabetes, macrosomia, and labor complications. However, data across human and animal studies show promise that nutritional interventions and physical activity may rescue much of the adverse effects of obesity on offspring metabolic health. Further, these maternal interventions improve the health of the offspring by reducing weight gain, cardiovascular disorders, and improving glucose tolerance. Mechanisms from animal studies have also been proposed to elucidate the signaling pathways that regulate inflammation, lipid metabolism, and oxidative capacity of the tissue, ultimately providing potential specific courses of treatment. This review aims to pinpoint the risks of maternal obesity and provide plausible intervention strategies. We delve into recent research involving both animal and human studies with maternal interventions. With the increasing concerning of obesity rates witnessed in the United States, it is imperative to acknowledge the long-term effects posed on future generations and specifically modify maternal nutrition and care to mitigate these adverse outcomes.
Collapse
Affiliation(s)
- Akriti Shrestha
- Department of Nutrition and Food Studies, Syracuse University, Syracuse, NY, United States
| | - Madison Prowak
- Department of Nutrition and Food Studies, Syracuse University, Syracuse, NY, United States
| | | | - Jessica Garay
- Department of Nutrition and Food Studies, Syracuse University, Syracuse, NY, United States
| | - Latha Ramalingam
- Department of Nutrition and Food Studies, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
25
|
Kusuyama J, Alves-Wagner AB, Conlin RH, Makarewicz NS, Albertson BG, Prince NB, Kobayashi S, Kozuka C, Møller M, Bjerre M, Fuglsang J, Miele E, Middelbeek RJW, Xiudong Y, Xia Y, Garneau L, Bhattacharjee J, Aguer C, Patti ME, Hirshman MF, Jessen N, Hatta T, Ovesen PG, Adamo KB, Nozik-Grayck E, Goodyear LJ. Placental superoxide dismutase 3 mediates benefits of maternal exercise on offspring health. Cell Metab 2021; 33:939-956.e8. [PMID: 33770509 PMCID: PMC8103776 DOI: 10.1016/j.cmet.2021.03.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/14/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022]
Abstract
Poor maternal diet increases the risk of obesity and type 2 diabetes in offspring, adding to the ever-increasing prevalence of these diseases. In contrast, we find that maternal exercise improves the metabolic health of offspring, and here, we demonstrate that this occurs through a vitamin D receptor-mediated increase in placental superoxide dismutase 3 (SOD3) expression and secretion. SOD3 activates an AMPK/TET signaling axis in fetal offspring liver, resulting in DNA demethylation at the promoters of glucose metabolic genes, enhancing liver function, and improving glucose tolerance. In humans, SOD3 is upregulated in serum and placenta from physically active pregnant women. The discovery of maternal exercise-induced cross talk between placenta-derived SOD3 and offspring liver provides a central mechanism for improved offspring metabolic health. These findings may lead to novel therapeutic approaches to limit the transmission of metabolic disease to the next generation.
Collapse
Affiliation(s)
- Joji Kusuyama
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Miyagi, Japan.
| | - Ana Barbara Alves-Wagner
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Royce H Conlin
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nathan S Makarewicz
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Brent G Albertson
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Noah B Prince
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Shio Kobayashi
- Section of Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Chisayo Kozuka
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; YCI Laboratory for Metabolic Epigenetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Magnus Møller
- Department of Gynecology and Obstetrics, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Fuglsang
- Department of Gynecology and Obstetrics, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| | - Emily Miele
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Yang Xiudong
- Graduate School of Biomedical Sciences, University of Texas at Houston, Houston, TX, USA
| | - Yang Xia
- Graduate School of Biomedical Sciences, University of Texas at Houston, Houston, TX, USA
| | - Léa Garneau
- Institut du Savoir Montfort, recherche, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Jayonta Bhattacharjee
- School of Human Kinetics, Faculty of Health Science University of Ottawa, Ottawa, Canada
| | - Céline Aguer
- Institut du Savoir Montfort, recherche, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada; School of Human Kinetics, Faculty of Health Science University of Ottawa, Ottawa, Canada; Interdisciplinary School of Health Sciences, Faculty of Health Science University of Ottawa, Ottawa, Canada
| | - Mary Elizabeth Patti
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Ishikawa, Japan
| | - Per Glud Ovesen
- Department of Gynecology and Obstetrics, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| | - Kristi B Adamo
- School of Human Kinetics, Faculty of Health Science University of Ottawa, Ottawa, Canada
| | - Eva Nozik-Grayck
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Reynolds L, Vsevolozhskaya O, Rice B, Chavan N, Dugan A, Maddox H, Preston J, DeHoff L, O'Brien J, Pearson K. Physical activity during pregnancy alters gene expression in neonatal tissue. Physiol Int 2021. [DOI: 10.1556/2060.2021.00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
AbstractReducing the risk of developing chronic disease, such as obesity and type 2 diabetes, is an important component of successful aging. Offspring born to mothers who exercise during pregnancy have improved body composition and metabolic profiles. However, mechanisms to explain this phenomenon are lacking.PurposeThis study examined whether maternal step counts were correlated with neonatal gene expression markers related to glucose metabolism and adipogenesis.MethodsPhysical activity levels were assessed in women with male neonates via Fitbit Flex® during the second and third trimester of pregnancy. The dartos and epidermal/dermal layers of the foreskin were collected following circumcision in full-term, singleton, neonates (n = 12 dartos and n = 14 dermal). Tissue was homogenized, RNA isolated, and a NanoString code set was run to quantify a panel of genes related to glucose metabolism and adipogenesis.ResultsTwelve genes were correlated to steps per day with a P-value of <0.05. After adjusting for multiple comparisons, six genes remained significantly correlated to steps per day (False Discovery Rate-corrected P-value < 0.10). Notably, glucose transporter 1, adiponectin receptor 1, and CCAAT/enhancer-binding protein alpha and beta were positively correlated with steps per day, while peroxisome proliferator-activated receptor alpha and peroxisome proliferator-activated receptor gamma coactivator 1- alpha were negatively correlated with steps per day.ConclusionMaternal physical activity is associated with offspring gene expression markers of adipogenesis, insulin sensitivity and glucose uptake. Future studies should aim to mechanistically examine whether these markers are driving increased adiposity in offspring born to sedentary mothers.
Collapse
Affiliation(s)
- L.J. Reynolds
- 1Department of Human Movement Sciences, College of Education and Professional Studies, Old Dominion University, Norfolk, VA, USA
| | - O.A. Vsevolozhskaya
- 2Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - B.B. Rice
- 3Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - N.R. Chavan
- 4Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - A.J. Dugan
- 2Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - H.F. Maddox
- 3Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - J.D. Preston
- 3Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
- 5School of Medicine, Emory University, Atlanta, GA, USA
| | - L.B. DeHoff
- 3Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - J.M. O'Brien
- 4Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - K.J. Pearson
- 3Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
27
|
August PM, Rodrigues KDS, Klein CP, Dos Santos BG, Matté C. Influence of gestational exercise practice and litter size reduction on maternal care. Neurosci Lett 2021; 741:135454. [PMID: 33166634 DOI: 10.1016/j.neulet.2020.135454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 01/21/2023]
Abstract
Mother-pup interactions are extremely important to offspring survival and growth. The goal of this study was to evaluate the influence of prenatal and neonatal interventions on maternal care, analyzing the effect of maternal exercise, as a healthy intervention, and also the litter size reduction, a model that has been widely used to study early overfeeding in rats. Female Wistar rats were divided into 1) sedentary, and 2) swimming exercise for four weeks, starting one week before mating (5 days/week, 30 min/session). One day after birth, the litter was culled to 8 pups (normal) or 3 pups (small) per dam, yielding control and overfed subgroups for each maternal group, respectively. From postnatal days 2-9 the litter was observed 5 periods a day, to evaluate maternal behavior. Litter reduction caused important alterations in maternal behavior, reducing the total time out of the nest and increasing the frequency of maternal care and lactation in several observation periods, justifying the increased pup's weight gain already demonstrated by this animal model. The practice of maternal exercise did not prevent, but cause the less intensive frequency of non-maternal behavior and lactation in arched-back position, induced by the reduction of litter size. These data demonstrated that small litter size altered maternal behavior, and gestational exercise does not influence significantly these changes.
Collapse
Affiliation(s)
- Pauline Maciel August
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Karoline Dos Santos Rodrigues
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Caroline Peres Klein
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bernardo Gindri Dos Santos
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cristiane Matté
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Ciências Biológicas: Fisiologia, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
28
|
Sofiabadi M, Zarbaf R, Abdolahpour A, Koushki Jahromi M, Peymani A, Khosravi N. Effect of exercise training before mating on mRNA expression of breast cancer-related genes in offspring in rats. Sci Sports 2020; 35:312.e1-312.e9. [DOI: 10.1016/j.scispo.2020.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
29
|
Kusuyama J, Alves-Wagner AB, Makarewicz NS, Goodyear LJ. Effects of maternal and paternal exercise on offspring metabolism. Nat Metab 2020; 2:858-872. [PMID: 32929233 PMCID: PMC7643050 DOI: 10.1038/s42255-020-00274-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
Maternal and paternal obesity and type 2 diabetes are recognized risk factors for the development of metabolic dysfunction in offspring, even when the offspring follow a healthful lifestyle. Multiple studies have demonstrated that regular physical activity in mothers and fathers has striking beneficial effects on offspring health, including preventing the development of metabolic disease in rodent offspring as they age. Here, we review the benefits of maternal and paternal exercise in combating the development of metabolic dysfunction in adult offspring, focusing on offspring glucose homeostasis and adaptations to metabolic tissues. We discuss recent findings regarding the roles of the placenta and sperm in mediating the effects of parental exercise on offspring metabolic health, as well as the mechanisms hypothesized to underlie these beneficial changes. Given the worldwide epidemics of obesity and type 2 diabetes, if these findings translate to humans, regular exercise during the reproductive years might limit the vicious cycles in which increased metabolic risk propagates across generations.
Collapse
Affiliation(s)
- Joji Kusuyama
- Integrative Physiology and Metabolism Section, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Ana Barbara Alves-Wagner
- Integrative Physiology and Metabolism Section, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nathan S Makarewicz
- Integrative Physiology and Metabolism Section, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Laurie J Goodyear
- Integrative Physiology and Metabolism Section, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Axsom JE, Libonati JR. Impact of parental exercise on epigenetic modifications inherited by offspring: A systematic review. Physiol Rep 2020; 7:e14287. [PMID: 31758667 PMCID: PMC6874781 DOI: 10.14814/phy2.14287] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 01/07/2023] Open
Abstract
Performing regular exercise is associated with numerous health benefits including a reduction in all‐cause mortality. The mechanisms associated with exercise‐induced health improvements are wide ranging and benefit virtually every organ system in the body. Of significance, recent evidence has suggested that some of these protective benefits may also be passed to offspring through multiple generations via alterations in gamete presentation, changes to the in‐utero and offspring rearing environments, and epigenetic modifications. The purpose of this review was to systematically examine the current literature for evidence of exercise‐induced epigenetic modifications in offspring. A systematic search yielded four papers that met inclusion criteria. Parental exercise interventions were associated with differential DNA methylation patterns in offspring. These shifts in methylation patterns were consistent with concurrent changes in offspring mRNA levels, protein expression, and functional measures. Many of the observed changes were related to metabolic pathways. Hence, the evidence suggests that exercise‐induced epigenetic changes can be observed in offspring and may play a pivotal role among the multifactorial intergenerational‐health impact of exercise. A proposed mechanism for the wide‐ranging health benefits of exercise is epigenetic changes and there is potential for epigenetic changes to be passed on to offspring through intergenerational inheritance.![]()
Collapse
Affiliation(s)
- Jessie E Axsom
- Department of Nursing Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph R Libonati
- Department of Nursing Science, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Harris JE, Pinckard KM, Wright KR, Baer LA, Arts PJ, Abay E, Shettigar VK, Lehnig AC, Robertson B, Madaris K, Canova TJ, Sims C, Goodyear LJ, Andres A, Ziolo MT, Bode L, Stanford KI. Exercise-induced 3'-sialyllactose in breast milk is a critical mediator to improve metabolic health and cardiac function in mouse offspring. Nat Metab 2020; 2:678-687. [PMID: 32694823 PMCID: PMC7438265 DOI: 10.1038/s42255-020-0223-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 05/27/2020] [Indexed: 01/22/2023]
Abstract
Poor maternal environments, such as under- or overnutrition, can increase the risk for the development of obesity, type 2 diabetes and cardiovascular disease in offspring1-9. Recent studies in animal models have shown that maternal exercise before and during pregnancy abolishes the age-related development of impaired glucose metabolism10-15, decreased cardiovascular function16 and increased adiposity11,15; however, the underlying mechanisms for maternal exercise to improve offspring's health have not been identified. In the present study, we identify an exercise-induced increase in the oligosaccharide 3'-sialyllactose (3'-SL) in milk in humans and mice, and show that the beneficial effects of maternal exercise on mouse offspring's metabolic health and cardiac function are mediated by 3'-SL. In global 3'-SL knockout mice (3'-SL-/-), maternal exercise training failed to improve offspring metabolic health or cardiac function in mice. There was no beneficial effect of maternal exercise on wild-type offspring who consumed milk from exercise-trained 3'-SL-/- dams, whereas supplementing 3'-SL during lactation to wild-type mice improved metabolic health and cardiac function in offspring during adulthood. Importantly, supplementation of 3'-SL negated the detrimental effects of a high-fat diet on body composition and metabolism. The present study reveals a critical role for the oligosaccharide 3'-SL in milk to mediate the effects of maternal exercise on offspring's health. 3'-SL supplementation is a potential therapeutic approach to combat the development of obesity, type 2 diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Johan E Harris
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kelsey M Pinckard
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Katherine R Wright
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Peter J Arts
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eaman Abay
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Vikram K Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Adam C Lehnig
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bianca Robertson
- Department of Pediatrics and Larsson-Rosenquist-Foundation Mother-Milk-Infant Center of Research Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Kendra Madaris
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tyler J Canova
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Clark Sims
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aline Andres
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist-Foundation Mother-Milk-Infant Center of Research Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
32
|
Moreira JBN, Wisløff U. Post-exercise breast milk: the new polypill? Nat Metab 2020; 2:653-654. [PMID: 32694822 DOI: 10.1038/s42255-020-0234-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jose B N Moreira
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- School of Human Movement and Nutrition Science, University of Queensland, Queensland, Australia.
| |
Collapse
|
33
|
Falcão-Tebas F, Marin EC, Kuang J, Bishop DJ, McConell GK. Maternal exercise attenuates the lower skeletal muscle glucose uptake and insulin secretion caused by paternal obesity in female adult rat offspring. J Physiol 2020; 598:4251-4270. [PMID: 32539156 PMCID: PMC7586952 DOI: 10.1113/jp279582] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Paternal obesity negatively influences metabolic outcomes in adult rat offspring. Maternal voluntary physical activity has previously been reported to improve glucose metabolism in adult rat offspring sired by healthy fathers. Here, we investigated whether a structured programme of maternal exercise training before and during gestation can attenuate the negative impacts that paternal obesity has on insulin sensitivity and secretion in female adult offspring. Exercise before and during pregnancy normalised the lower insulin sensitivity in skeletal muscle and the lower insulin secretion observed in female offspring sired by obese fathers. This paper presents a feasible, low-cost and translatable intervention strategy that can be applied perinatally to support multifactor interventions to break the cycle of metabolic dysfunction caused by paternal obesity. ABSTRACT We investigated whether maternal exercise before and during gestation could attenuate the negative metabolic effects of paternal high-fat diet-induced obesity in female adult rat offspring. Fathers consumed a normal chow or high-fat diet before mating. Mothers exercised on a treadmill before and during gestation or remained sedentary. In adulthood, female offspring were assessed using intraperitoneal insulin and glucose tolerance tests (IPITT and IPGTT, respectively), pancreatic morphology, ex vivo skeletal muscle insulin-stimulated glucose uptake and mitochondrial respiratory function. Paternal obesity impaired whole-body and skeletal muscle insulin sensitivity and insulin secretion in adult offspring. Maternal exercise attenuated the lower insulin-stimulated glucose uptake in offspring sired by obese fathers but distal insulin signalling components (p-AKT Thr308 and Ser473, p-TBC1D4 Thr642 and GLUT4) remained unchanged (P > 0.05). Maternal exercise increased citrate synthase activity only in offspring sired by obese fathers. Maternal exercise also reversed the lower insulin secretion in vivo observed in offspring of obese fathers, probably due to an attenuation of the decrease in pancreatic beta cell mass. In summary, maternal exercise before and during pregnancy in rats attenuated skeletal muscle insulin resistance and attenuated the decrease in pancreatic beta cell mass and insulin secretion observed in the female offspring of obese fathers.
Collapse
Affiliation(s)
- Filippe Falcão-Tebas
- Institute for Health and Sport (IHES), Victoria University, Melbourne, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Evelyn C Marin
- Institute for Health and Sport (IHES), Victoria University, Melbourne, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| | - Jujiao Kuang
- Institute for Health and Sport (IHES), Victoria University, Melbourne, Australia
| | - David J Bishop
- Institute for Health and Sport (IHES), Victoria University, Melbourne, Australia
| | - Glenn K McConell
- Institute for Health and Sport (IHES), Victoria University, Melbourne, Australia.,College of Health and Biomedicine, Victoria University, Melbourne, Australia
| |
Collapse
|
34
|
Protective effect of maternal exercise against amyloid-β neurotoxicity in the male rat offspring's cerebellum. J Dev Orig Health Dis 2020; 11:521-532. [PMID: 32631472 DOI: 10.1017/s2040174420000562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Developmental Origins of Health and Disease (DOHaD) states that intrauterine maternal environment influences postnatal life by programming offspring's metabolism. Intrauterine milieu induced by exercise during pregnancy promotes long-lasting benefits to the offspring's health and seems to offer some resistance against chronic diseases in adult life. Alzheimer's disease is a public health concern with limited treatment options. In the present study, we assessed the potential of maternal exercise during pregnancy in long-term programming of young adult male rat offspring's cerebellar metabolism in conferring neuroprotection against amyloid-β (Aβ) neurotoxicity. Female Wistar rats were submitted to a swimming protocol 1 week prior mating and throughout pregnancy (five sessions/a week lasting 30 min). Aβ oligomers were infused bilaterally in the brain ventricles of 60-day-old male offspring. Fourteen days after surgery, we measured parameters related to redox state, mitochondrial function, and the immunocontent of proteins related to synaptic function. We found that maternal exercise during pregnancy attenuated several parameters in the offspring's male rat cerebellum, such as the reactive species rise, the increase of inducible nitric oxide synthase immunocontent and tau phosphorylation induced by Aβ oligomers, increased mitochondrial fission indicated by dynamin-related protein 1 (DRP1), and protein oxidation identified by carbonylation. Strikingly, we find that maternal exercise promotes changes in the rat offspring's cerebellum that are still evident in young adult life. These favorable neurochemical changes in offspring's cerebellum induced by maternal exercise may contribute to a protective phenotype against Aβ-induced neurotoxicity in young adult male rat offspring.
Collapse
|
35
|
Castro-Rodríguez DC, Rodríguez-González GL, Menjivar M, Zambrano E. Maternal interventions to prevent adverse fetal programming outcomes due to maternal malnutrition: Evidence in animal models. Placenta 2020; 102:49-54. [PMID: 33218579 DOI: 10.1016/j.placenta.2020.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/22/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022]
Abstract
Animal studies indicate that suboptimal conditions during pregnancy adversely impact both maternal health and offspring phenotype, predisposing offspring to development of later-life diseases including obesity, diabetes, cardiovascular diseases, and behavioral and reproductive dysfunction. Effective interventions during pregnancy and/or lactation are needed to improve both maternal and offspring health. This review addresses the relationship between adverse perinatal insults and its negative impact on offspring development and presents some maternal intervention studies in animal models, such as maternal nutrition (diet modification, antioxidants, omega-3-6 (n-3-6), probiotics) or physical activity, which can prevent or alleviate negative outcomes in both mother and offspring.
Collapse
Affiliation(s)
- Diana C Castro-Rodríguez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico; CONACyT-Cátedras, Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Guadalupe L Rodríguez-González
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Marta Menjivar
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico; Unidad Académica de Ciencias y Tecnología, Universidad Nacional Autónoma de México-Yucatán, Yucatán, Mexico
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico.
| |
Collapse
|
36
|
Abstract
It shows that detrimental exposures and conditions in mothers can lead to the development of obesity and type 2 diabetes in offspring. This can lead to a vicious cycle of metabolic dysfunction, where rising rates of obesity, pre-diabetes, and diabetes in individuals of reproductive age, propagating risks to subsequent generations. It is well established that regular exercise has important health benefits for people with obesity and type 2 diabetes. Recently, increasing studies aim to examine the effects of maternal exercise on metabolic health in offspring. This review aims to demonstrate the evidence linking maternal exercise during critical periods of development and its implications for glucose metabolism in offspring, including intervention timing, sexual dimorphism, different exercise type, and intensity. Then we further examine the potential role of epigenetic modifications in this process.
Collapse
|
37
|
Zheng J, Alves-Wagner AB, Stanford KI, Prince NB, So K, Mul JD, Dirice E, Hirshman MF, Kulkarni RN, Goodyear LJ. Maternal and paternal exercise regulate offspring metabolic health and beta cell phenotype. BMJ Open Diabetes Res Care 2020; 8:8/1/e000890. [PMID: 32111717 PMCID: PMC7050345 DOI: 10.1136/bmjdrc-2019-000890] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/20/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Poor maternal and paternal environments increase the risk for obesity and diabetes in offspring, whereas maternal and paternal exercise in mice can improve offspring metabolic health. We determined the effects of combined maternal and paternal exercise on offspring health and the effects of parental exercise on offspring pancreas phenotype, a major tissue regulating glucose homeostasis. RESEARCH DESIGN AND METHODS Breeders were high fat fed and housed±running wheels before breeding (males) and before and during gestation (females). Offspring groups were: both parents sedentary (Sed); maternal exercise only (Mat Ex); paternal exercise only (Pat Ex); and maternal+paternal exercise (Mat+Pat Ex). Offspring were sedentary, chow fed, and studied at weaning, 12, 20 and 52 weeks. RESULTS While there was no effect of parental exercise on glucose tolerance at younger ages, at 52 weeks, offspring of Mat Ex, Pat Ex and Mat+Pat Ex displayed lower glycemia and improved glucose tolerance. The greatest effects were in offspring from parents that both exercised (Mat+Pat Ex). Offspring from Mat Ex, Pat Ex, and Mat+Pat Ex had decreased beta cell size, whereas islet size and beta cell mass only decreased in Mat+Pat Ex offspring. CONCLUSIONS Maternal and paternal exercise have additive effects to improve glucose tolerance in offspring as they age, accompanied by changes in the offspring endocrine pancreas. These findings have important implications for the prevention and treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Jia Zheng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Ana Barbara Alves-Wagner
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kristin I Stanford
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Noah B Prince
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Kawai So
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Joram D Mul
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Ercument Dirice
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Rohit N Kulkarni
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Stimulation of the ACE2/Ang-(1-7)/Mas axis in hypertensive pregnant rats attenuates cardiovascular dysfunction in adult male offspring. Hypertens Res 2019; 42:1883-1893. [PMID: 31506648 DOI: 10.1038/s41440-019-0321-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/31/2022]
Abstract
The aim of this study was to investigate whether treatment with diminazene aceturate (DIZE), a putative ACE2 activator, or with angiotensin-(1-7) during pregnancy could attenuate the development of cardiovascular dysfunction in the adult offspring of spontaneously hypertensive rats (SHRs). For this, pregnant SHRs received DIZE or Ang-(1-7) throughout gestation. The systolic blood pressure (SBP) was measured in the male offspring from the 6th to16th weeks of age by tail-cuff plethysmography. Thereafter, the left ventricular contractile function and coronary reactivity were evaluated by the Langendorff technique. Samples of the left ventricles (LVs) and kidneys were collected for histology and western blot assay in another batch of adult rat offspring. Maternal treatment with DIZE or Ang-(1-7) during pregnancy attenuated the increase in SBP in adult offspring. In addition, both DIZE and Ang-(1-7) treatments reduced the cardiomyocyte diameter and fibrosis deposition in the LV, and treatment with Ang-(1-7) also reduced the fibrosis deposition in the kidneys. Maternal treatment with DIZE, as well as Ang-(1-7), improved the coronary vasodilation induced by bradykinin in isolated hearts from adult offspring. However, no difference was observed in the contractile function of the LVs of these animals. The expression levels of AT1 and Mas receptors, ACE, ACE2, SOD, and catalase in the LV were not modified by maternal treatment with Ang-(1-7), but this treatment elicited a reduction in AT2 expression. These data show that treatment with DIZE or Ang-(1-7) during gestation promoted beneficial effects of attenuating hypertension and cardiac remodeling in adult offspring.
Collapse
|
39
|
McGee SL, Hargreaves M. Epigenetics and Exercise. Trends Endocrinol Metab 2019; 30:636-645. [PMID: 31279665 DOI: 10.1016/j.tem.2019.06.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 01/12/2023]
Abstract
Epigenetics can be defined as 'the structural adaptation of chromosomal regions so as to register, signal, or perpetuate altered activity states.' Increased transcription of key regulatory, metabolic, and myogenic genes is an early response to exercise and is important in mediating subsequent adaptations in skeletal muscle. DNA hypomethylation and histone hyperacetylation are emerging as important crucial events for increased transcription. The complex interactions between multiple epigenetic modifications and their regulation by metabolic changes and signaling events during exercise, with implications for enhanced understanding of the acute and chronic adaptations to exercise, are questions for further investigation.
Collapse
Affiliation(s)
- Sean L McGee
- Metabolic Research Unit, School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong Waurn Ponds, VIC 3216, Australia.
| | - Mark Hargreaves
- Department of Physiology, The University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
40
|
Musial B, Fernandez‐Twinn DS, Duque‐Guimaraes D, Carr SK, Fowden AL, Ozanne SE, Sferruzzi‐Perri AN. Exercise alters the molecular pathways of insulin signaling and lipid handling in maternal tissues of obese pregnant mice. Physiol Rep 2019; 7:e14202. [PMID: 31466137 PMCID: PMC6715452 DOI: 10.14814/phy2.14202] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 02/02/2023] Open
Abstract
Obesity during gestation adversely affects maternal and infant health both during pregnancy and for long afterwards. However, recent work suggests that a period of maternal exercise during pregnancy can improve metabolic health of the obese mother and her offspring. This study aimed to identify the physiological and molecular impact of exercise on the obese mother during pregnancy that may lead to improved metabolic outcomes. To achieve this, a 20-min treadmill exercise intervention was performed 5 days a week in diet-induced obese female mice from 1 week before and up to day 17 of pregnancy. Biometric, biochemical and molecular analyses of maternal tissues and/or plasma were performed on day 19 of pregnancy. We found exercise prevented some of the adverse changes in insulin signaling and lipid metabolic pathways seen in the liver, skeletal muscle and white adipose tissue of sedentary-obese pregnant dams (p110β, p110α, AKT, SREBP). Exercise also induced changes in the insulin and lipid signaling pathways in obese dams that were different from those observed in control and sedentary-obese dams. The changes induced by obesity and exercise were tissue-specific and related to alterations in tissue lipid, protein and glycogen content and plasma insulin, leptin and triglyceride concentrations. We conclude that the beneficial effects of exercise on metabolic outcomes in obese mothers may be related to specific molecular signatures in metabolically active maternal tissues during pregnancy. These findings highlight potential metabolic targets for therapeutic intervention and the importance of lifestyle in reducing the burden of the current obesity epidemic on healthcare systems.
Collapse
Affiliation(s)
- Barbara Musial
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| | - Denise S. Fernandez‐Twinn
- MRC Metabolic Disease UnitUniversity of Cambridge Metabolic Research Laboratories, Wellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Daniella Duque‐Guimaraes
- MRC Metabolic Disease UnitUniversity of Cambridge Metabolic Research Laboratories, Wellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Sarah K. Carr
- MRC Metabolic Disease UnitUniversity of Cambridge Metabolic Research Laboratories, Wellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Abigail L. Fowden
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| | - Susan E. Ozanne
- MRC Metabolic Disease UnitUniversity of Cambridge Metabolic Research Laboratories, Wellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Amanda N. Sferruzzi‐Perri
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
41
|
Chatmethakul T, Roghair RD. Risk of hypertension following perinatal adversity: IUGR and prematurity. J Endocrinol 2019; 242:T21-T32. [PMID: 30657741 PMCID: PMC6594910 DOI: 10.1530/joe-18-0687] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
Consistent with the paradigm shifting observations of David Barker and colleagues that revealed a powerful relationship between decreased weight through 2 years of age and adult disease, intrauterine growth restriction (IUGR) and preterm birth are independent risk factors for the development of subsequent hypertension. Animal models have been indispensable in defining the mechanisms responsible for these associations and the potential targets for therapeutic intervention. Among the modifiable risk factors, micronutrient deficiency, physical immobility, exaggerated stress hormone exposure and deficient trophic hormone production are leading candidates for targeted therapies. With the strong inverse relationship seen between gestational age at delivery and the risk of hypertension in adulthood trumping all other major cardiovascular risk factors, improvements in neonatal care are required. Unfortunately, therapeutic breakthroughs have not kept pace with rapidly improving perinatal survival, and groundbreaking bench-to-bedside studies are urgently needed to mitigate and ultimately prevent the tsunami of prematurity-related adult cardiovascular disease that may be on the horizon. This review highlights our current understanding of the developmental origins of hypertension and draws attention to the importance of increasing the availability of lactation consultants, nutritionists, pharmacists and physical therapists as critical allies in the battle that IUGR or premature infants are waging not just for survival but also for their future cardiometabolic health.
Collapse
Affiliation(s)
- Trassanee Chatmethakul
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Robert D Roghair
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
42
|
Maternally expressed gene 3 in metabolic programming. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194396. [PMID: 31271897 DOI: 10.1016/j.bbagrm.2019.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/20/2019] [Indexed: 01/10/2023]
Abstract
Maternally Expressed Gene 3 (MEG3) is a long noncoding RNA (lncRNA) that coordinates a diverse array of cellular processes requiring epigenetic regulation of genes and interactions with key signaling proteins and by acting as a competitive endogenous (ce)RNA. Epigenetic modifications driven by in utero nutrition affect MEG3 expression and its role in the development of multiple metabolic disorders. This review examines how epigenetic modification of MEG3 expression can confer adaptedness to different metabolic environments. To this end, we discuss how nutritional status that leads to an increase of MEG3 expression can protect against cancer and metabolic dysfunctions, while interventions that promote MEG3 downregulation minimize the pleiotropic costs associated with its expression. Lastly, we identify research directions that would further shed light on the role of MEG3 in metabolic regulation and in functional imprinted gene networks. This article is part of a Special Issue entitled: ncRNA in control of gene expression edited by Kotb Abdelmohsen.
Collapse
|
43
|
Siti F, Dubouchaud H, Hininger I, Quiclet C, Vial G, Galinier A, Casteilla L, Fontaine E, Batandier C, Couturier K. Maternal exercise before and during gestation modifies liver and muscle mitochondria in rat offspring. ACTA ACUST UNITED AC 2019; 222:jeb.194969. [PMID: 31019067 DOI: 10.1242/jeb.194969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 04/05/2019] [Indexed: 01/09/2023]
Abstract
It is now well established that the intrauterine environment is of major importance for offspring health during later life. Endurance training during pregnancy is associated with positive metabolic adjustments and beneficial effects on the balance between pro-oxidants and antioxidants (redox state) in the offspring. Our hypothesis was that these changes could rely on mitochondrial adaptations in the offspring due to modifications of the fetal environment induced by maternal endurance training. Therefore, we compared the liver and skeletal muscle mitochondrial function and the redox status of young rats whose mothers underwent moderate endurance training (treadmill running) before and during gestation (T) with those of young rats from untrained mothers (C). Our results show a significant reduction in the spontaneous H2O2 release by liver and muscle mitochondria in the T versus C offspring (P<0.05). These changes were accompanied by alterations in oxygen consumption. Moreover, the percentage of short-chain fatty acids increased significantly in liver mitochondria from T offspring. This may lead to improvements in the fluidity and the flexibility of the membrane. In plasma, glutathione peroxidase activity and protein oxidation were significantly higher in T offspring than in C offspring (P<0.05). Such changes in plasma could represent an adaptive signal transmitted from mothers to their offspring. We thus demonstrated for the first time, to our knowledge, that it is possible to act on bioenergetic function including alterations of mitochondrial function in offspring by modifying maternal physical activity before and during pregnancy. These changes could be crucial for the future health of the offspring.
Collapse
Affiliation(s)
- Farida Siti
- Université Grenoble Alpes, INSERM, LBFA, 38058 Grenoble, France.,Université Grenoble Alpes, UFR STAPS, SFR Sport Exercice Motricité, 38058 Grenoble, France.,Department of Medical Pharmacy, Universitas Indonesia, 10430 Jakarta, Indonesia
| | - Hervé Dubouchaud
- Université Grenoble Alpes, INSERM, LBFA, 38058 Grenoble, France.,Université Grenoble Alpes, UFR STAPS, SFR Sport Exercice Motricité, 38058 Grenoble, France
| | | | - Charline Quiclet
- Université Grenoble Alpes, INSERM, LBFA, 38058 Grenoble, France.,Université Grenoble Alpes, UFR STAPS, SFR Sport Exercice Motricité, 38058 Grenoble, France
| | - Guillaume Vial
- Université Grenoble Alpes, INSERM, HP2, 38000 Grenoble, France
| | - Anne Galinier
- Université de Toulouse, STROMALab, CNRS: ERL5311, EFS: INP-ENVT, INSERM: U-1031, UPS, 31100 Toulouse, France
| | - Louis Casteilla
- Université de Toulouse, STROMALab, CNRS: ERL5311, EFS: INP-ENVT, INSERM: U-1031, UPS, 31100 Toulouse, France
| | - Eric Fontaine
- Université Grenoble Alpes, INSERM, LBFA, 38058 Grenoble, France.,Centre Hospitalier Universitaire Grenoble Alpes, 38000 Grenoble, France
| | | | - Karine Couturier
- Université Grenoble Alpes, INSERM, LBFA, 38058 Grenoble, France .,Université Grenoble Alpes, UFR STAPS, SFR Sport Exercice Motricité, 38058 Grenoble, France
| |
Collapse
|
44
|
Rodríguez-González GL, Castro-Rodríguez DC, Zambrano E. Pregnancy and Lactation: A Window of Opportunity to Improve Individual Health. Methods Mol Biol 2018; 1735:115-144. [PMID: 29380310 DOI: 10.1007/978-1-4939-7614-0_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Human and animal studies indicate that obesity during pregnancy adversely impacts both maternal health and offspring phenotype predisposing them to chronic diseases later in life including obesity, dyslipidemia, type 2 diabetes mellitus, and hypertension. Effective interventions during human pregnancy and/or lactation are needed to improve both maternal and offspring health. This review addresses the relationship between adverse perinatal insults and its negative impact on offspring development and presents some maternal intervention studies such as diet modification, probiotic consumption, or maternal exercise, to prevent or alleviate the negative outcomes in both the mother and her child.
Collapse
Affiliation(s)
- Guadalupe L Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana C Castro-Rodríguez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
45
|
Klein CP, Dos Santos Rodrigues K, Hözer RM, de Sá Couto-Pereira N, Saccomori AB, Dal Magro BM, Crestani MS, Hoppe JB, Salbego CG, Dalmaz C, Matté C. Swimming exercise before and during pregnancy: Promising preventive approach to impact offspring´s health. Int J Dev Neurosci 2018; 71:83-93. [PMID: 30172896 DOI: 10.1016/j.ijdevneu.2018.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/25/2018] [Accepted: 08/27/2018] [Indexed: 02/08/2023] Open
Abstract
Several environmental factors affect child development, such as the intrauterine environment during the embryonic and fetal development and early postnatal environment provided by maternal behavior. Although mechanistic effects of maternal exercise on offspring health improvement are not yet completely understood, the number of reports published demonstrating the positive influence of maternal exercise have increase. Herein, we addressed issues related to early postnatal environment provided by maternal behavior and early developmental physical landmarks, sensorimotor reflexes, and motor movements ontogeny. In brief, adult female rats underwent involuntary swimming exercise, in a moderated intensity, one week before mating and throughout pregnancy, 30 min a day, 5 days a week. Maternal exercised dams have unchanged gestational outcomes compared to sedentary dams. We found no differences concerning the frequency of pup-directed behavior displayed by dams. However, sedentary dams displayed a poorer pattern of maternal care quality during dark cycle than exercised dams. Physical landmarks and sensorimotor reflexes development of female and male littermates did not differ between maternal groups. Developmental motor parameters such as immobility, lateral head movements, head elevation, pivoting, rearing with forelimb support and crawling frequencies did not differ between groups. Pups born to exercised dams presented higher frequency of walking and rearing on the hind legs. These data suggest that female and male littermates of exercised group present a high frequency of exploratory behavior over sedentary littermates. Taken together, the present findings reinforce that maternal exercise throughout pregnancy represent a window of opportunity to improve offspring's postnatal health.
Collapse
Affiliation(s)
- Caroline Peres Klein
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Karoline Dos Santos Rodrigues
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Régis Mateus Hözer
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natividade de Sá Couto-Pereira
- Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - André Brum Saccomori
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bárbara Mariño Dal Magro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mariana Scortegagna Crestani
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Juliana Bender Hoppe
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Christianne Gazzana Salbego
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carla Dalmaz
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cristiane Matté
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Fisiologia, Instituto de Ciências Básicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
46
|
Murphy MO, Herald JB, Leachman J, Villasante Tezanos A, Cohn DM, Loria AS. A model of neglect during postnatal life heightens obesity-induced hypertension and is linked to a greater metabolic compromise in female mice. Int J Obes (Lond) 2018; 42:1354-1365. [PMID: 29535450 PMCID: PMC6054818 DOI: 10.1038/s41366-018-0035-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 01/05/2018] [Accepted: 01/14/2018] [Indexed: 12/12/2022]
Abstract
.: Exposure to early life stress (ELS) is associated with behavioral-related alterations, increases in body mass index and higher systolic blood pressure in humans. Postnatal maternal separation and early weaning (MSEW) is a mouse model of neglect characterized by a long-term dysregulation of the neuroendocrine system. OBJECTIVES Given the contribution of adrenal-derived hormones to the development of obesity, we hypothesized that exposure to MSEW could contribute to the worsening of cardiometabolic function in response to chronic high-fat diet (HF) feeding by promoting adipose tissue expansion and insulin resistance. SUBJECTS MSEW was performed in C57BL/6 mice from postnatal days 2-16 and weaned at postnatal day 17. Undisturbed litters weaned at postnatal day 21 served as the control (C) group. At the weaning day, mice were placed on a low-fat diet (LF) or HF for 16 weeks. RESULTS When fed a LF, male and female mice exposed to MSEW display similar body weight but increased fat mass compared to controls. However, when fed a HF, only female MSEW mice display increased body weight, fat mass, and adipocyte hypertrophy compared with controls. Also, female MSEW mice display evidence of an early onset of cardiometabolic risk factors, including hyperinsulinemia, glucose intolerance, and hypercholesterolemia. Yet, both male and female MSEW mice fed a HF show increased blood pressure compared with controls. CONCLUSIONS This study shows that MSEW promotes a sex-specific dysregulation of the adipose tissue expansion and glucose homeostasis that precedes the development of obesity-induced hypertension.
Collapse
Affiliation(s)
- Margaret O Murphy
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Joseph B Herald
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Jacqueline Leachman
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Dianne M Cohn
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
47
|
Momma H, Sawada SS, Sloan RA, Gando Y, Kawakami R, Terada S, Miyachi M, Kinugawa C, Okamoto T, Tsukamoto K, Huang C, Nagatomi R, Blair SN. Importance of Achieving a "Fit" Cardiorespiratory Fitness Level for Several Years on the Incidence of Type 2 Diabetes Mellitus: A Japanese Cohort Study. J Epidemiol 2018; 28:230-236. [PMID: 29176273 PMCID: PMC5911673 DOI: 10.2188/jea.je20160199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background The “Physical Activity Reference for Health Promotion 2013” provides “fit” reference values for cardiorespiratory fitness (CRF) for good health. The importance of achieving a fit CRF level for several years on the subsequent prevention of type 2 diabetes mellitus (T2DM) remains to be clarified. Methods This cohort study was conducted in 2,235 nondiabetic males aged 21 to 59 years, enrolled in April 1986 through March 1987. We calculated the ratio of the area under the curve (AUCratio) for actual measured values and the AUC for the reference values of CRF in each individual during an 8-year measurement period before the baseline. According to whether they met a fit CRF level or not, participants were categorized into groups based on the AUCratio (FitAUC or UnfitAUC) and initial CRF (Fitinitial or Unfitinitial), respectively. T2DM was evaluated on health checkups until March 2010. Results During the follow-up period, 400 men developed T2DM. After adjustment for confounders, as compared with those in the FitAUC group, the hazard ratio (HR) for those in the UnfitAUC group was 1.33 (95% confidence interval [CI], 1.06–1.65). A combined analysis with the categories of initial value and AUCratio showed that, compared with the Fitinitial and FitAUC group, the HRs of Fitinitial and UnfitAUC, Unfitinitial and FitAUC, and Unfitinitial and UnfitAUC groups were 1.41 (95% CI, 0.99–2.00), 1.18 (95% CI, 0.81–1.70), and 1.40 (95% CI, 1.08–1.83), respectively. Conclusion Achievement of a fit CRF level established in the Japan physical activity guideline for several years was associated with lower subsequent risk of T2DM.
Collapse
Affiliation(s)
- Haruki Momma
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering.,Department of Health Promotion and Exercise, National Institutes of Biomedical Innovation, Health and Nutrition
| | - Susumu S Sawada
- Department of Health Promotion and Exercise, National Institutes of Biomedical Innovation, Health and Nutrition
| | - Robert A Sloan
- Department of Psychosomatic Internal Medicine, Graduate Medical and Dental School, Kagoshima University
| | - Yuko Gando
- Department of Health Promotion and Exercise, National Institutes of Biomedical Innovation, Health and Nutrition
| | | | - Shin Terada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo
| | - Motohiko Miyachi
- Department of Health Promotion and Exercise, National Institutes of Biomedical Innovation, Health and Nutrition
| | | | | | | | - Cong Huang
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering
| | - Ryoichi Nagatomi
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering
| | - Steven N Blair
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina.,Department of Exercise Science, Arnold School of Public Health, University of South Carolina
| |
Collapse
|
48
|
Harris JE, Baer LA, Stanford KI. Maternal Exercise Improves the Metabolic Health of Adult Offspring. Trends Endocrinol Metab 2018; 29:164-177. [PMID: 29402734 PMCID: PMC5826804 DOI: 10.1016/j.tem.2018.01.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 12/22/2022]
Abstract
The intrauterine environment can modulate the course of development and confer an enduring effect on offspring health. The effects of maternal diet to impair offspring metabolic health are well established, but the effects of maternal exercise on offspring metabolic health have been less defined. Because physical exercise is a treatment for obesity and type 2 diabetes (T2D), maternal exercise is an appealing intervention to positively influence the intrauterine environment and improve the metabolic health of offspring. Recent research has provided insights into the effects of maternal exercise on the metabolic health of adult offspring, which is the focus of this review.
Collapse
Affiliation(s)
- Johan E Harris
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Lisa A Baer
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
49
|
Therapies for gestational diabetes and their implications for maternal and offspring health: Evidence from human and animal studies. Pharmacol Res 2018; 130:52-73. [PMID: 29421161 DOI: 10.1016/j.phrs.2018.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/05/2018] [Accepted: 02/01/2018] [Indexed: 01/21/2023]
Abstract
Obesity prior to and during pregnancy is associated with an increased risk of complications during pregnancy. One of the most common complications of pregnancy is gestational diabetes mellitus (GDM), a condition characterized by hyperglycemia and insulin resistance that is diagnosed in the third trimester of pregnancy. GDM predisposes both mothers and their children to increased obesity and cardiometabolic disorders, namely type 2 diabetes and cardiovascular disease. Current treatments include lifestyle changes and insulin injections, but oral anti-diabetic drugs such as metformin and glyburide are increasingly prescribed as they do not require injections. However, the long-term implications of therapies for diabetes during pregnancy on mothers and their offspring are not fully understood. In this review, we describe current treatments for GDM, including the first line lifestyle interventions such as exercise as well as insulin, glyburides and metformin. We also review selected natural health products that are sometimes used by individuals during pregnancy that could also be an effective therapeutic in pregnancies characterized by obesity or GDM. We focus on both the short- and long-term effects of treatments on the health of mothers and their offspring. We review the current literature from clinical research and animal studies.
Collapse
|
50
|
Preston JD, Reynolds LJ, Pearson KJ. Developmental Origins of Health Span and Life Span: A Mini-Review. Gerontology 2018; 64:237-245. [PMID: 29324453 DOI: 10.1159/000485506] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A vast body of research has demonstrated that disease susceptibility and offspring health can be influenced by perinatal factors, which include both paternal and maternal behavior and environment. Offspring disease risk has the potential to affect the health span and life span of offspring. KEY FINDINGS Various maternal factors, such as environmental toxicant exposure, diet, stress, exercise, age at conception, and longevity have the potential to influence age-associated diseases such as cardiovascular disease, obesity, diabetes, and cancer risk in offspring. Paternal factors such as diet, age at conception, and longevity can potentially impact offspring health span and life span-reducing traits as well. PRACTICAL IMPLICATIONS Continued research could go a long way toward defining mechanisms of the developmental origins of life span and health span, and eventually establishing regimens to avoid negative developmental influences and to encourage positive interventions to potentially increase life span and improve health span in offspring.
Collapse
Affiliation(s)
- Joshua D Preston
- Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | | |
Collapse
|