1
|
Kacperska M, Mizera J, Pilecki M, Pomierny-Chamioło L. The impact of excessive maternal weight on the risk of neuropsychiatric disorders in offspring-a narrative review of clinical studies. Pharmacol Rep 2024; 76:452-462. [PMID: 38649593 PMCID: PMC11126479 DOI: 10.1007/s43440-024-00598-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
The global prevalence of overweight and obesity is a significant public health concern that also largely affects women of childbearing age. Human epidemiological studies indicate that prenatal exposure to excessive maternal weight or excessive gestational weight gain is linked to various neurodevelopmental disorders in children, including attention deficit hyperactivity disorder, autism spectrum disorder, internalizing and externalizing problems, schizophrenia, and cognitive/intellectual impairment. Considering that inadequate maternal body mass can induce serious disorders in offspring, it is important to increase efforts to prevent such outcomes. In this paper, we review human studies linking excessive maternal weight and the occurrence of mental disorders in children.
Collapse
Affiliation(s)
- Magdalena Kacperska
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Józef Mizera
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Maciej Pilecki
- Department of Psychiatry, Jagiellonian University Medical College, Kopernika 21a, 31-500, Kraków, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
2
|
Diniz MS, Grilo LF, Tocantins C, Falcão-Pires I, Pereira SP. Made in the Womb: Maternal Programming of Offspring Cardiovascular Function by an Obesogenic Womb. Metabolites 2023; 13:845. [PMID: 37512552 PMCID: PMC10386510 DOI: 10.3390/metabo13070845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/02/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Obesity incidence has been increasing at an alarming rate, especially in women of reproductive age. It is estimated that 50% of pregnancies occur in overweight or obese women. It has been described that maternal obesity (MO) predisposes the offspring to an increased risk of developing many chronic diseases in an early stage of life, including obesity, type 2 diabetes, and cardiovascular disease (CVD). CVD is the main cause of death worldwide among men and women, and it is manifested in a sex-divergent way. Maternal nutrition and MO during gestation could prompt CVD development in the offspring through adaptations of the offspring's cardiovascular system in the womb, including cardiac epigenetic and persistent metabolic programming of signaling pathways and modulation of mitochondrial metabolic function. Currently, despite diet supplementation, effective therapeutical solutions to prevent the deleterious cardiac offspring function programming by an obesogenic womb are lacking. In this review, we discuss the mechanisms by which an obesogenic intrauterine environment could program the offspring's cardiovascular metabolism in a sex-divergent way, with a special focus on cardiac mitochondrial function, and debate possible strategies to implement during MO pregnancy that could ameliorate, revert, or even prevent deleterious effects of MO on the offspring's cardiovascular system. The impact of maternal physical exercise during an obesogenic pregnancy, nutritional interventions, and supplementation on offspring's cardiac metabolism are discussed, highlighting changes that may be favorable to MO offspring's cardiovascular health, which might result in the attenuation or even prevention of the development of CVD in MO offspring. The objectives of this manuscript are to comprehensively examine the various aspects of MO during pregnancy and explore the underlying mechanisms that contribute to an increased CVD risk in the offspring. We review the current literature on MO and its impact on the offspring's cardiometabolic health. Furthermore, we discuss the potential long-term consequences for the offspring. Understanding the multifaceted effects of MO on the offspring's health is crucial for healthcare providers, researchers, and policymakers to develop effective strategies for prevention and intervention to improve care.
Collapse
Affiliation(s)
- Mariana S Diniz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Luís F Grilo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Carolina Tocantins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal
- 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 Sports, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
3
|
Cerf ME. Maternal and Child Health, Non-Communicable Diseases and Metabolites. Metabolites 2023; 13:756. [PMID: 37367913 DOI: 10.3390/metabo13060756] [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: 04/04/2023] [Revised: 06/02/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023] Open
Abstract
Mothers influence the health and disease trajectories of their children, particularly during the critical developmental windows of fetal and neonatal life reflecting the gestational-fetal and lactational-neonatal phases. As children grow and develop, they are exposed to various stimuli and insults, such as metabolites, that shape their physiology and metabolism to impact their health. Non-communicable diseases, such as diabetes, cardiovascular disease, cancer and mental illness, have high global prevalence and are increasing in incidence. Non-communicable diseases often overlap with maternal and child health. The maternal milieu shapes progeny outcomes, and some diseases, such as gestational diabetes and preeclampsia, have gestational origins. Metabolite aberrations occur from diets and physiological changes. Differential metabolite profiles can predict the onset of non-communicable diseases and therefore inform prevention and/or better treatment. In mothers and children, understanding the metabolite influence on health and disease can provide insights for maintaining maternal physiology and sustaining optimal progeny health over the life course. The role and interplay of metabolites on physiological systems and signaling pathways in shaping health and disease present opportunities for biomarker discovery and identifying novel therapeutic agents, particularly in the context of maternal and child health, and non-communicable diseases.
Collapse
Affiliation(s)
- Marlon E Cerf
- Grants, Innovation and Product Development, South African Medical Research Council, P.O. Box 19070, Tygerberg, Cape Town 7505, South Africa
- Biomedical Research and Innovation Platform, South African Medical Research Council, P.O. Box 19070, Tygerberg, Cape Town 7505, South Africa
| |
Collapse
|
4
|
Ahmed N, Kassis A, Malone J, Yang J, Zamzami E, Lin AH, Gordon SM, Gong M, Bardo M, Dalmasso C, Loria AS. Prenatal Morphine Exposure Increases Cardiovascular Disease Risk and Programs Neurogenic Hypertension in the Adult Offspring. Hypertension 2023; 80:1283-1296. [PMID: 37042247 PMCID: PMC10274123 DOI: 10.1161/hypertensionaha.122.20262] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/20/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND The opioid overdose and opioid use disorder epidemics are concomitant with increased metabolic and CVD risk. Although opioid use disorder causes adverse pregnancy outcomes, the offspring's cardiovascular health is understudied. We hypothesized that offspring exposed to in utero morphine exposure (IUME) would show increased CVD risk factors and endogenous opioid system dysregulation. METHODS Sprague Dawley dams were treated with saline (vehicle, n=10) or escalating doses of morphine (5-20 mg/kg per day, SC, n=10) during gestation. Cardiovascular and metabolic parameters were assessed in adult offspring. RESULTS Litter size and pups' birth weight were not different in response to IUME. Female and male IUME offspring showed reduced body length at birth (P<0.05) and body weight from weeks 1 to 3 of life (P<0.05), followed by a catch-up growth effect. By week 16, female and male IUME rats showed reduced tibia length (P<0.05) and fat mass. IUME increases the mean arterial pressure and the depressor response to mecamylamine (5 mg/kg per day, IP) induced by IUME were abolished by a chronic treatment with an alpha-adrenergic receptor blocker (prazosin; 1 mg/kg per day, IP). Although circulating levels of angiotensin peptides were similar between groups, IUME exacerbated maximal ex vivo Ang (angiotensin) II-induced vasoconstriction (P<0.05) and induced endothelial dysfunction in a sex-specific manner (P<0.05). Proenkephalin, an endogenous opioid peptide that lowers blood pressure and sympathetic-mediated vasoconstriction, showed reduced mRNA expression in the heart, aorta, and kidneys from morphine versus vehicle group (P<0.05). CONCLUSIONS Among the effects of IUME, neurogenic hypertension, vascular dysfunction, and metabolic dysfunction could be associated with the dysregulation of the endogenous opioid system.
Collapse
Affiliation(s)
- Nermin Ahmed
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Alana Kassis
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Jena Malone
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Jodie Yang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Esraa Zamzami
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - An-Hsuan Lin
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Scott M. Gordon
- SAHA Cardiovascular Center, University of Kentucky, Lexington, KY 40536
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Ming Gong
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Michael Bardo
- Department of Psychology, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536
| | - Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536
- SAHA Cardiovascular Center, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
5
|
Kim J, Kim J, Kwon YH. Leucine supplementation in maternal high-fat diet alleviated adiposity and glucose intolerance of adult mice offspring fed a postweaning high-fat diet. Lipids Health Dis 2023; 22:50. [PMID: 37061742 PMCID: PMC10105473 DOI: 10.1186/s12944-023-01812-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/31/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Combined maternal and postnatal high-fat (HF) diet intake predisposes offspring to metabolic dysregulation during adulthood. As the inhibitory effects of leucine consumption on obesity and metabolic disorders have been reported, the effects of maternal leucine supplementation on metabolic dysregulation in adult offspring were investigated. METHODS Female mice were exposed to a control (C) or HF diet, with or without leucine (L) supplementation (1.5%, w/v), 3 weeks before mating, during pregnancy, and during lactation (C, CL, HF, and HFL). Male offspring were exposed to an HF diet for 12 weeks after weaning (C/HF, CL/HF, HF/HF, and HFL/HF). Serum biochemical parameters were determined for both the dams and offspring. Oral glucose tolerance test and qRT-PCR analysis were used to investigate metabolic dysregulation in the offspring. RESULTS HFL dams exhibited higher relative adipose tissue weights than HF dams. Body weight, relative adipose tissue weight, and serum glucose levels were lower in the HFL/HF offspring than in the HF/HF offspring. Maternal leucine supplementation tended to alleviate glucose intolerance in the offspring of HF diet-fed dams. Additionally, mRNA levels of fibroblast growth factor 21 (FGF21), a hepatokine associated with glucose homeostasis, were higher in HFL/HF offspring than in HF/HF offspring and were negatively correlated with adiposity and serum glucose levels. The mRNA levels of genes encoding a FGF21 receptor complex, Fgf receptor 1 and klotho β, and its downstream targets, proliferator-activated receptor-γ co-activator 1α and sirtuin 1, were higher in adipose tissues of the HFL/HF offspring than in those of the HF/HF offspring. Serum lipid peroxide levels were lower in HFL dams than in HF dams and positively correlated with body and adipose tissue weights of offspring. CONCLUSIONS Leucine supplementation in HF diet-fed dams, but not in control diet-fed dams, resulted in an anti-obesity phenotype accompanied by glucose homeostasis in male offspring challenged with postnatal HF feeding. Activation of FGF21 signaling in the adipose tissue of offspring may be responsible for these beneficial effects of leucine.
Collapse
Affiliation(s)
- Juhae Kim
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Juyoung Kim
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
- Research Institute of Human Ecology, Seoul National University, Seoul, Korea.
| |
Collapse
|
6
|
Wang L, O'Kane AM, Zhang Y, Ren J. Maternal obesity and offspring health: Adapting metabolic changes through autophagy and mitophagy. Obes Rev 2023:e13567. [PMID: 37055041 DOI: 10.1111/obr.13567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/08/2022] [Accepted: 03/25/2023] [Indexed: 04/15/2023]
Abstract
Maternal obesity leads to obstetric complications and a high prevalence of metabolic anomalies in the offspring. Among various contributing factors for maternal obesity-evoked health sequelae, developmental programming is considered as one of the leading culprit factors for maternal obesity-associated chronic comorbidities. Although a unified theory is still lacking to systematically address multiple unfavorable postnatal health sequelae, a cadre of etiological machineries have been put forward, including lipotoxicity, inflammation, oxidative stress, autophagy/mitophagy defect, and cell death. Hereinto, autophagy and mitophagy play an essential housekeeping role in the clearance of long-lived, damaged, and unnecessary cell components to maintain and restore cellular homeostasis. Defective autophagy/mitophagy has been reported in maternal obesity and negatively impacts fetal development and postnatal health. This review will provide an update on metabolic disorders in fetal development and postnatal health issues evoked by maternal obesity and/or intrauterine overnutrition and discuss the possible contribution of autophagy/mitophagy in metabolic diseases. Moreover, relevant mechanisms and potential therapeutic strategies will be discussed in an effort to target autophagy/mitophagy and metabolic disturbances in maternal obesity.
Collapse
Affiliation(s)
- Litao Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Aislinn M O'Kane
- Department of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| |
Collapse
|
7
|
Embryonic Hyperglycemia Disrupts Myocardial Growth, Morphological Development, and Cellular Organization: An In Vivo Experimental Study. Life (Basel) 2023; 13:life13030768. [PMID: 36983924 PMCID: PMC10056749 DOI: 10.3390/life13030768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Hyperglycemia during gestation can disrupt fetal heart development and increase postnatal cardiovascular disease risk. It is therefore imperative to identify early biomarkers of hyperglycemia during gestation-induced fetal heart damage and elucidate the underlying molecular pathomechanisms. Clinical investigations of diabetic adults with heart dysfunction and transgenic mouse studies have revealed that overexpression or increased expression of TNNI3K, a heart-specific kinase that binds troponin cardiac I, may contribute to abnormal cardiac remodeling, ventricular hypertrophy, and heart failure. Optimal heart function also depends on the precise organization of contractile and excitable tissues conferred by intercellular occlusive, adherent, and communicating junctions. The current study evaluated changes in embryonic heart development and the expression levels of sarcomeric proteins (troponin I, desmin, and TNNI3K), junctional proteins, glucose transporter-1, and Ki-67 under fetal hyperglycemia. Stage 22HH Gallus domesticus embryos were randomly divided into two groups: a hyperglycemia (HG) group, in which individual embryos were injected with 30 mmol/L glucose solution every 24 h for 10 days, and a no-treatment (NT) control group, in which individual embryos were injected with physiological saline every 24 h for 10 days (stage 36HH). Embryonic blood glucose, height, and weight, as well as heart size, were measured periodically during treatment, followed by histopathological analysis and estimation of sarcomeric and junctional protein expression by western blotting and immunostaining. Hyperglycemic embryos demonstrated delayed heart maturation, with histopathological analysis revealing reduced left and right ventricular wall thickness (−39% and −35% vs. NT). Immunoexpression levels of TNNI3K and troponin 1 increased (by 37% and 39%, respectively), and desmin immunofluorescence reduced (by 23%). Embryo-fetal hyperglycemia may trigger an increase in the expression levels of TNNI3K and troponin I, as well as dysfunction of occlusive and adherent junctions, ultimately inducing abnormal cardiac remodeling.
Collapse
|
8
|
Zhang J, Zhang R, Chi J, Li Y, Bai W. Pre-pregnancy body mass index has greater influence on newborn weight and perinatal outcome than weight control during pregnancy in obese women. Arch Public Health 2023; 81:5. [PMID: 36639806 PMCID: PMC9838058 DOI: 10.1186/s13690-023-01025-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The pre-pregnancy weight and gestational weight gain (GWG) are closely related to perinatal maternal and infant complications, but the relationship between pre-pregnancy weight and GWG and the pattern of interaction have not been reported. This study investigated the influence of weight control during pregnancy on the perinatal maternal and infant outcomes. METHODS A total of 835 singleton pregnant women who were hospitalized between January 2018 and December 2018 were retrospectively included in this study and divided into two groups: the diet guidance (DG) group (n = 167) and the control group (n = 668). The pre-pregnancy body mass index (BMI), GWG, and perinatal outcomes of the women and infants were determined in two groups. RESULTS The dietary modification and reasonable exercise during pregnancy effectively reduced the GWG, and even some women with pre-pregnancy obesity achieved weight loss during pregnancy. The GWG in the DG group was significantly lower than in the control group, especially in the second and third trimesters. GWG was positively related to birth weight. The birth weight in the DG group was significantly lower than in the control group when their mothers had similar GWG. In women with pre-pregnancy obesity, GWG seemed to be negatively related to birth weight. However, after adjusting the mean BMI, the pre-pregnancy BMI and GWG were positively related to the birth weight. Compared with the control group, the incidences of dystocia, postpartum hemorrhage, macrosomia, small for gestational age infants and neonatal complications significantly reduced in the DG group, and the preterm birth rate was comparable between two groups. Some women with pre-pregnancy obesity lose weight during pregnancy, but there was no premature birth or small for gestational age infant. The incidences of macrosomia, postpartum hemorrhage, dystocia, cesarean section and gestational diabetes increased significantly with the increase of pre-pregnancy BMI. CONCLUSION For women with increased pre-pregnancy BMI, strict weight control is required to reduce obesity-related complications of the mother and infant. The weight control in the second and third trimesters is especially important and most likely to prevent GWG. Compared with GWG, pre-pregnancy BMI has greater influence on the birth weight and maternal and infant complications, and may even compromise the benefits of weight control during pregnancy. Thus, weight control is recommended before pregnancy.
Collapse
Affiliation(s)
- Jin Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Haidian, Beijing, 100038 China
| | - Rui Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Haidian, Beijing, 100038 China
| | - Jingjing Chi
- grid.24696.3f0000 0004 0369 153XDepartment of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Haidian, Beijing, 100038 China
| | - Ya Li
- grid.24696.3f0000 0004 0369 153XDepartment of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Haidian, Beijing, 100038 China
| | - Wenpei Bai
- grid.24696.3f0000 0004 0369 153XDepartment of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Haidian, Beijing, 100038 China
| |
Collapse
|
9
|
Wu L, Li N, Liu Y. Association Between Maternal Factors and Risk of Congenital Heart Disease in Offspring: A Systematic Review and Meta-Analysis. Matern Child Health J 2023; 27:29-48. [PMID: 36344649 PMCID: PMC9867685 DOI: 10.1007/s10995-022-03538-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION This study aimed to summarize the evidence describing the relationship between maternal factors during gestation and risk of congenital heart disease (CHD) in offspring. METHODS PubMed, EMBASE, and the Cochrane Library were searched for potentially relevant reports from inception to May 2021. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) calculated by the random-effects model were used to evaluate the association between maternal factors and CHD risk. RESULTS There was a significant association between CHD risk and obesity in pregnancy (OR 1.29, 95% CI 1.22-1.37; P < 0.001), smoking in pregnancy (OR 1.16, 95% CI 1.07-1.25; P < 0.001), maternal diabetes (OR 2.65, 95% CI 2.20-3.19; P < 0.001), and exposure of pregnant women to organic solvents (OR 1.82, 95% CI 1.23-2.70; P = 0.003). No correlations were revealed between CHD susceptibility and advanced maternal age (OR 1.04, 95% CI 0.96-1.12; P = 0.328), underweight (OR 1.02, 95% CI 0.96-1.08; P = 0.519), alcohol intake in pregnancy (OR 1.08, 95% CI 0.95-1.22; P = 0.251), coffee intake (OR 1.18, 95% CI 0.97-1.44; P = 0.105), and exposure to irradiation (OR 1.80, 95% CI 0.85-3.80; P = 0.125). DISCUSSION Maternal factors including maternal obesity, smoking in pregnancy, maternal diabetes and exposure to organic solvents might predispose the offspring to CHD risk.
Collapse
Affiliation(s)
- Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Na Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yong Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
10
|
Adebayo-Gege G, Alicha V, Omayone TO, Nzekwe SC, Irozuoke CA, Ojo OA, Ajayi AF. Anti-atherogenic and cardio-protective properties of sweet melon (Cucumis melo. L. Inodorus) seed extract on high fat diet induced obesity in male wistar rats. BMC Complement Med Ther 2022; 22:334. [PMID: 36539762 PMCID: PMC9764567 DOI: 10.1186/s12906-022-03793-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cucumis melon is a medicinal plant with multiple pharmacological properties such as anti-inflammatory, antioxidant, and diuretic effects. An increasing body of scientific evidence established the anti-diabetic/anti-obesity effects of Cucumis melo in humans, mice, and hamster models. However, there are no tangible reports on its ability to prevent cardiovascular complications following diet-induced obesity. The anti-atherogenic and cardioprotective effects of the Methanolic extract of Cucumis melo. L. Inodorus seeds on a high-fat diet (HFD)-induced obese rats was assessed in this study. METHODS: Forty male Wistar rats were randomly divided into five groups, (n = 8/group); i.e., Normal (N), HFD, HFD + 50 mg/kg b.w. of MCMs (Methanolic extract of Cucumis melon seeds), HFD + 100 mg/Kg b.w. of MCMs and HFD + 200 mg/kg b.w. of MCMs. The experimental animals were anaesthetized and sacrificed after 10 weeks, and blood samples and heart tissue were collected for further analysis. Using the Graph Pad Prism version 5.0, the results expressed as Mean ± SD was tested using the one-way ANOVA to show intergroup differences, followed by Bonferonni 's post hoc test. The level of significance was determined at P ≤ 0.05. RESULTS MCMs significantly (P < 0.05) reduced body weight, adiposity index, total fat mass, low-density lipoprotein cholesterol (LDL-c), and total cholesterol (TC) compared with the HFD obese groups MCMs caused a significant reduction in the body weight, total fat mass, adiposity index, low-density lipoprotein cholesterol (LDL-c), and total cholesterol (TC) when compared to the animals in HFD obese groups. Also, the Atherogenic index of plasma (AIP), Castelli index and, malondialdehyde (MDA) significantly (P < 0.05) decreased in MCMs treated groups compared to the HFD obese group. The catalase, protein, and HDL levels were significantly increased in MCMs treated groups compared to HFD-obese animals. Expression of nitric oxide in the form of nitrite in the heart tissue significantly increased in the MCMs treated compared to the HFD-obese rats, with the majority of the positive results recorded at 100 mg/Kg b.w. of MCMs. CONCLUSIONS MCMs have anti-atherogenic and Cardio-protective properties on High Fat Diet-Induced Obesity in Male rats via an antioxidant and nitric oxide-dependent mechanism. Further study is recommended to evaluate the molecular mechanisms to which these anti-atherogenic and cardio-protective actions can be attributed and exploit the GCMS result in the development of drug candidates.
Collapse
Affiliation(s)
- G Adebayo-Gege
- Department of Human Physiology, Faculty of Basic Medical Sciences, Baze University, Jabi, Nigeria
| | - V Alicha
- Department of Human Physiology, Faculty of Basic Medical Sciences, College of Medicine Bingham University, Jos, Nigeria
| | - T O Omayone
- Department of Human Physiology, Faculty of Basic Medical Sciences, College of Medicine, FUTA, Akure, Nigeria
| | - S C Nzekwe
- Department of Biochemistry, Faculty of Science, Adeleke University, Ede, Osun State, Nigeria
| | - C A Irozuoke
- Department of Anatomy, Faculty of Basic Medical Sciences, Baze University, Jabi, Nigeria
| | - O A Ojo
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, 232101, Nigeria
| | - A F Ajayi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria.
- Anchor BioMed Researh Institute, Ogbomoso, Oyo State, Nigeria.
| |
Collapse
|
11
|
Resistin Modulates the Functional Activity of Colostral Macrophages from Mothers with Obesity and Diabetes. Biomedicines 2022; 10:biomedicines10102332. [DOI: 10.3390/biomedicines10102332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Obesity and diabetes are major public health problems. Resistin is an adipokine that links the two diseases. There are few reports regarding colostrum cells and resistin from mothers with obesity and diabetes. Thus, this study aimed to determine the functional activity of macrophages present in the breast milk and colostrum of diabetic mothers with obesity and the effects of resistin on these cells. Methods: The women were divided according to BMI and glycemic status into normal weight non-diabetic, obese non-diabetic, normal weight type 2 diabetic, or obese type 2 diabetic groups. ELISA determined the resistin in colostrum. The cell subsets and apoptosis were determined by flow cytometry and the functional activity of cells by fluorescence microscopy. Results: The resistin levels were higher in the colostrum from diabetic mothers with obesity. The frequencies of CD14+ cells and cells expressing CD95+, independent of resistin treatment, were higher in the colostrum from diabetic mothers with obesity. The frequency of cells expressing CD14+CD95+ was higher in cells not treated with resistin in the colostrum from diabetic mothers with obesity. Apoptosis, irrespective of the presence of resistin, increased, whereas microbicidal activity decreased in cells from diabetic mothers with obesity. Conclusion: The data suggest that hyperglycemia associated with low-grade inflammation caused by obesity affects the percentage of cells expressing CD14+CD95+, death by apoptosis, and microbicidal indices; meanwhile, resistin restored the microbicidal activity of colostrum cells.
Collapse
|
12
|
Ma Z, Wang Y, Quan Y, Wang Z, Liu Y, Ding Z. Maternal obesity alters methylation level of cytosine in CpG island for epigenetic inheritance in fetal umbilical cord blood. Hum Genomics 2022; 16:34. [PMID: 36045397 PMCID: PMC9429776 DOI: 10.1186/s40246-022-00410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Over the past few decades, global maternal obesity prevalence has rapidly increased. This condition may induce long-lasting pathophysiological effects on either fetal or infant health that could be attributable to unknown unique changes in the umbilical blood composition. METHODS A total of 34 overweight/obese and 32 normal-weight pregnant women were recruited. Fifteen umbilical blood samples including 8 overweight/obese subjects and 7 normal weight women were sequenced using Targeted Bisulfite Sequencing technology to detect the average methylation level of cytosine and identify the differentially methylated region (DMR). GO and KEGG analyses were then employed to perform pathway enrichment analysis of DMR-related genes and promoters. Moreover, the mRNA levels of methylation-related genes histone deacetylases (HDACs) and DNA methyltransferases (DNMTs) were characterized in the samples obtained from these two groups. RESULTS Average methylated cytosine levels in both the CpG islands (CGI) and promoter significantly decreased in overweight/obese groups. A total of 1669 DMRs exhibited differences in their DNA methylation status between the overweight/obese and control groups. GO and KEGG analyses revealed that DMR-related genes and promoters were enriched in the metabolism, cancer and cardiomyopathy signaling pathways. Furthermore, the HDACs and DNMTs mRNA levels trended to decline in overweight/obese groups. CONCLUSIONS Decreased methylated cytosine levels in overweight/obese women induce the gene expression activity at a higher level than in the control group. DMRs between these two groups in the fetal blood may contribute to the changes in gene transcription that underlie the increased risk of metabolic disorders, cancers and cardiomyopathy in their offspring.
Collapse
Affiliation(s)
- Zhuoyao Ma
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China
| | - Yingjin Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Yanmei Quan
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China
| | - Zhijie Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| | - Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China.
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), Shanghai, 200025, China.
| |
Collapse
|
13
|
Mizera J, Kazek G, Pomierny B, Bystrowska B, Niedzielska-Andres E, Pomierny-Chamiolo L. Maternal High-Fat diet During Pregnancy and Lactation Disrupts NMDA Receptor Expression and Spatial Memory in the Offspring. Mol Neurobiol 2022; 59:5695-5721. [PMID: 35773600 DOI: 10.1007/s12035-022-02908-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 05/30/2022] [Indexed: 10/17/2022]
Abstract
The problem of an unbalanced diet, overly rich in fats, affects a significant proportion of the population, including women of childbearing age. Negative metabolic and endocrine outcomes for offspring associated with maternal high-fat diet during pregnancy and/or lactation are well documented in the literature. In this paper, we present our findings on the little-studied effects of this diet on NMDA receptors and cognitive functions in offspring. The subject of the study was the rat offspring born from dams fed a high-fat diet before mating and throughout pregnancy and lactation. Using a novel object location test, spatial memory impairment was detected in adolescent offspring as well as in young adult female offspring. The recognition memory of the adolescent and young adult offspring remained unaltered. We also found multiple alterations in the expression of the NMDA receptor subunits, NMDA receptor-associated scaffolding proteins, and selected microRNAs that regulate the activity of the NMDA receptor in the medial prefrontal cortex and the hippocampus of the offspring. Sex-dependent changes in glutamate levels were identified in extracellular fluid obtained from the medial prefrontal cortex and the hippocampus of the offspring. The obtained results indicate that a maternal high-fat diet during pregnancy and lactation can induce in the offspring memory disturbances accompanied by alterations in NMDA receptor expression.
Collapse
Affiliation(s)
- Jozef Mizera
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, PL, Poland
| | - Grzegorz Kazek
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, PL, Poland
| | - Bartosz Pomierny
- Department of Biochemical Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, PL, Poland
| | - Beata Bystrowska
- Department of Biochemical Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, PL, Poland
| | - Ewa Niedzielska-Andres
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, PL, Poland.
| | - Lucyna Pomierny-Chamiolo
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, PL, Poland.
| |
Collapse
|
14
|
Tocantins C, Diniz MS, Grilo LF, Pereira SP. The birth of cardiac disease: Mechanisms linking gestational diabetes mellitus and early onset of cardiovascular disease in offspring. WIREs Mech Dis 2022; 14:e1555. [PMID: 35304833 DOI: 10.1002/wsbm.1555] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/10/2022] [Accepted: 03/09/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is the biggest killer worldwide, composing a major economic burden for health care systems. Obesity and diabetes are dual epidemics on the rise and major risk factors predisposing for CVD. Increased obesity- and diabetes-related incidence is now observed among children, adolescents, and young adults. Gestational diabetes mellitus (GDM) is the most common metabolic pregnancy disorder, and its prevalence is rapidly increasing. During pregnancies complicated by GDM, the offspring are exposed to a compromised intrauterine environment characterized by hyperglycemic periods. Unfavorable in utero conditions at critical periods of fetal cardiac development can produce developmental adaptations that remodel the cardiovascular system in a way that can contribute to adult-onset of heart disease due to the programming during fetal life. Epidemiological studies have reported increased cardiovascular complications among GDM-descendants, highlighting the urgent need to investigate and understand the mechanisms modulated during fetal development of in utero GDM-exposed offspring that predispose an individual to increased CVD during life. In this manuscript, we overview previous studies in this area and gather evidence linking GDM and CVD development in the offspring, providing new insights on novel mechanisms contributing to offspring CVD programming by GDM, from the role of maternal-fetal interactions to their impact on fetal cardiovascular development, how the perpetuation of cardiac programming is maintained in postnatal life, and advance the intergenerational implications contributing to increased CVD premature origin. Understanding the perpetuation of CVD can be the first step to manage and reverse this leading cause of morbidity and mortality. This article is categorized under: Reproductive System Diseases > Molecular and Cellular Physiology Cardiovascular Diseases > Molecular and Cellular Physiology Metabolic Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Carolina Tocantins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana S Diniz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Luís F Grilo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,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
|
15
|
Costa TJ, De Oliveira JC, Giachini FR, Lima VV, Tostes RC, Bomfim GF. Programming of Vascular Dysfunction by Maternal Stress: Immune System Implications. Front Physiol 2022; 13:787617. [PMID: 35360231 PMCID: PMC8961444 DOI: 10.3389/fphys.2022.787617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
A growing body of evidence highlights that several insults during pregnancy impact the vascular function and immune response of the male and female offspring. Overactivation of the immune system negatively influences cardiovascular function and contributes to cardiovascular disease. In this review, we propose that modulation of the immune system is a potential link between prenatal stress and offspring vascular dysfunction. Glucocorticoids are key mediators of stress and modulate the inflammatory response. The potential mechanisms whereby prenatal stress negatively impacts vascular function in the offspring, including poor hypothalamic–pituitary–adrenal axis regulation of inflammatory response, activation of Th17 cells, renin–angiotensin–aldosterone system hyperactivation, reactive oxygen species imbalance, generation of neoantigens and TLR4 activation, are discussed. Alterations in the immune system by maternal stress during pregnancy have broad relevance for vascular dysfunction and immune-mediated diseases, such as cardiovascular disease.
Collapse
Affiliation(s)
- Tiago J. Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Júlio Cezar De Oliveira
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Victor Vitorino Lima
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Rita C. Tostes
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
| | - Gisele Facholi Bomfim
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
- *Correspondence: Gisele Facholi Bomfim,
| |
Collapse
|
16
|
Pantaleão LC, Inzani I, Furse S, Loche E, Hufnagel A, Ashmore T, Blackmore HL, Jenkins B, Carpenter AAM, Wilczynska A, Bushell M, Koulman A, Fernandez-Twinn DS, Ozanne SE. Maternal diet-induced obesity during pregnancy alters lipid supply to mouse E18.5 fetuses and changes the cardiac tissue lipidome in a sex-dependent manner. eLife 2022; 11:e69078. [PMID: 35025731 PMCID: PMC8794468 DOI: 10.7554/elife.69078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022] Open
Abstract
Maternal obesity during pregnancy has immediate and long-term detrimental effects on the offspring heart. In this study, we characterized the cardiac and circulatory lipid profiles in late gestation E18.5 fetuses of diet-induced obese pregnant mice and established the changes in lipid abundance and fetal cardiac transcriptomics. We used untargeted and targeted lipidomics and transcriptomics to define changes in the serum and cardiac lipid composition and fatty acid metabolism in male and female fetuses. From these analyses we observed: (1) maternal obesity affects the maternal and fetal serum lipidome distinctly; (2) female fetal heart lipidomes are more sensitive to maternal obesity than males; (3) changes in lipid supply might contribute to early expression of lipolytic genes in mouse hearts exposed to maternal obesity. These results highlight the existence of sexually dimorphic responses of the fetal heart to the same in utero obesogenic environment and identify lipids species that might mediate programming of cardiovascular health.
Collapse
Affiliation(s)
- Lucas C Pantaleão
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Isabella Inzani
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Samuel Furse
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment CentreCambridgeUnited Kingdom
| | - Elena Loche
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Antonia Hufnagel
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Thomas Ashmore
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Heather L Blackmore
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Benjamin Jenkins
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment CentreCambridgeUnited Kingdom
| | - Asha A M Carpenter
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Ania Wilczynska
- Cancer Research UK Beatson InstituteGlasgowUnited Kingdom
- Institute of Cancer Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Martin Bushell
- Cancer Research UK Beatson InstituteGlasgowUnited Kingdom
- Institute of Cancer Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Albert Koulman
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment CentreCambridgeUnited Kingdom
| | - Denise S Fernandez-Twinn
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| | - Susan E Ozanne
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke’s HospitalCambridgeUnited Kingdom
| |
Collapse
|
17
|
Maternal Fructose Intake Exacerbates Cardiac Remodeling in Offspring with Ventricular Pressure Overload. Nutrients 2021; 13:nu13093267. [PMID: 34579143 PMCID: PMC8467570 DOI: 10.3390/nu13093267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
Recent studies demonstrated that metabolic syndrome and cardiovascular diseases could be elicited by developmental programming, which is regulated by prenatal nutritional and environmental stress. In this study, we utilized a rat model to examine the effect of excessive maternal fructose intake during pregnancy and lactation on cardiac development and progression of pressure overload-induced cardiac hypertrophy in offspring. Transverse aortic constriction (TAC) was performed on 3-month-old male offspring to induce ventricular pressure overload. Four weeks post-TAC, echocardiographic assessment as well as histopathological and biochemical examinations were performed on the myocardium of the offspring. Echocardiographic and gross examinations showed that heart weight, interventricular septal thickness in diastole (IVD; d), and left ventricular posterior wall thickness in diastole (LVPW; d) were elevated in offspring with TAC and further increased by maternal fructose exposure (MFE). However, the left ventricular ejection function was not significantly affected. Myocardial histopathological examination revealed that the indices of fibrosis and oxidative stress were higher in offspring with MFE and TAC than those in animals receiving either treatment. Molecular examinations on the myocardium demonstrated an MFE-induced upregulation of p38-MAPK signaling. Next generation sequence (NGS) analysis indicated a modulation of the expression levels of several cardiac hypertrophy-associated genes, including GPR22, Myh7, Nppa, P2RX4, and Npy by MFE. Subsequent RT-PCR indicated that MFE regulated the expression levels of genes responsive to cardiac hypertrophy (i.e., Myh-7, ANP) and oxidative stress (i.e., GR, GPx, and NQO-1). In conclusion, MFE during pregnancy and lactation modulated myocardial gene expression, increased oxidative stress, and exacerbated ventricular pressure overload-induced cardiac remodeling in rat offspring.
Collapse
|
18
|
Sudeep HV, Ramanaiah I, Amritha R, Naveen P, Gouthamchandra K, Shyamprasad K. A Standardized Aframomum melegueta Seed Extract Regulates Browning of White Adipose Tissue in High-Fat Diet Model Mice. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211033744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Thermogenesis-mediated energy expenditure is a promising strategy to combat obesity. Aframomum melegueta commonly known as grains of paradise (GP) is a popular spice with medicinal attributes in promoting health. We have demonstrated the thermogenic effects of a standardized A melegueta seed extract (AMSE) containing not <10% 6-paradol in high fat diet-fed (HFD) mice. The 6-week oral ingestion of 20 and 40 mg/kg AMSE significantly limited the weight gain, improved the brown adipose tissue (BAT) activity in HFD mice. Interestingly, AMSE markedly induced the beige adipocytes in epididymal white adipose tissue (eWAT). AMSE treatment led to the upregulation of marker proteins i.e., uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor-gamma-coactivator 1-alpha (PGC-1α), and peroxisome proliferator-activated receptor gamma (PPARγ) in eWAT and BAT. Our findings add to the current understanding of the thermogenic potentials of GP seed extract and report that the extract can stimulate the browning of WATs in addition to enhanced BAT activity. AMSE requires clinical validation to be explored as a dietary supplement/functional ingredient with thermogenic effect in food and beverages.
Collapse
Affiliation(s)
| | - Illuri Ramanaiah
- R&D Center for Excellence, Vidya Herbs Pvt. Ltd., Karnataka, India
| | - Raj Amritha
- R&D Center for Excellence, Vidya Herbs Pvt. Ltd., Karnataka, India
| | | | | | | |
Collapse
|
19
|
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: 17] [Impact Index Per Article: 5.7] [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
|
20
|
Ren J, Wu NN, Wang S, Sowers JR, Zhang Y. Obesity cardiomyopathy: evidence, mechanisms, and therapeutic implications. Physiol Rev 2021; 101:1745-1807. [PMID: 33949876 PMCID: PMC8422427 DOI: 10.1152/physrev.00030.2020] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The prevalence of heart failure is on the rise and imposes a major health threat, in part, due to the rapidly increased prevalence of overweight and obesity. To this point, epidemiological, clinical, and experimental evidence supports the existence of a unique disease entity termed “obesity cardiomyopathy,” which develops independent of hypertension, coronary heart disease, and other heart diseases. Our contemporary review evaluates the evidence for this pathological condition, examines putative responsible mechanisms, and discusses therapeutic options for this disorder. Clinical findings have consolidated the presence of left ventricular dysfunction in obesity. Experimental investigations have uncovered pathophysiological changes in myocardial structure and function in genetically predisposed and diet-induced obesity. Indeed, contemporary evidence consolidates a wide array of cellular and molecular mechanisms underlying the etiology of obesity cardiomyopathy including adipose tissue dysfunction, systemic inflammation, metabolic disturbances (insulin resistance, abnormal glucose transport, spillover of free fatty acids, lipotoxicity, and amino acid derangement), altered intracellular especially mitochondrial Ca2+ homeostasis, oxidative stress, autophagy/mitophagy defect, myocardial fibrosis, dampened coronary flow reserve, coronary microvascular disease (microangiopathy), and endothelial impairment. Given the important role of obesity in the increased risk of heart failure, especially that with preserved systolic function and the recent rises in COVID-19-associated cardiovascular mortality, this review should provide compelling evidence for the presence of obesity cardiomyopathy, independent of various comorbid conditions, underlying mechanisms, and offer new insights into potential therapeutic approaches (pharmacological and lifestyle modification) for the clinical management of obesity cardiomyopathy.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuyi Wang
- School of Medicine, Shanghai University, Shanghai, China.,University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - James R Sowers
- Dalton Cardiovascular Research Center, Diabetes and Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
21
|
da Silva LO, da Silva Aragão R, Duarte Barros MDL, Nogueira Ferraz-Pereira K, Lins Pinheiro I, Galindo LCM. Maternal exposure to high-fat diet modifies anxiety-like/depression-like behaviors and compounds of Serotonergic System in offspring: A preclinical systematic review. Int J Dev Neurosci 2021; 81:371-385. [PMID: 33788300 DOI: 10.1002/jdn.10110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 12/18/2022] Open
Abstract
Maternal nutrition affects offspring physiology and behavior including susceptibility to mental health-related states. Perinatal high-fat diet (HFD) consumption has been associated with lower levels of serotonin as well as the development of anxiety-like and depression-like behaviors in offspring. The aim of this systematic review was to investigate the effects of maternal HFD during pregnancy and/or lactation on these behaviors and on some aspects of the serotonergic system. Criteria for eligibility included studies of offspring of rodents and non-human primates exposed to HFD at least during pregnancy and/or lactation, offspring that showed outcomes related to anxiety-like and depression-like behaviors and to the serotonergic system. The searches were realized in the LILACS, Web of Science, Scopus, and PubMed databases. The systematic review protocol was registered on the CAMARADES website. The internal validity was assessed by the SYRCLE risk of bias tool. The Kappa index was used for analyzing agreement among the reviewers. In addition, the PRISMA statement was used to report this systematic review. Sixteen articles were included in this review. Most of which studied HFD prior to mating and during pregnancy and lactation. All studies analyzed outcomes related to emotional behavior; three analyzed outcomes related to serotonin system compounds. Maternal consumption of HFD was found to be associated with an inconsistent pattern of the expression of TPH2 as well as reduced the immunoreactivity of 5-HT in the prefrontal cortex and increased 5-HT1A receptor expression in the dorsal raphe of offspring. An association between an HFD and alterations in emotional behavior was found in most of the studies selected.
Collapse
Affiliation(s)
- Luana Olegário da Silva
- Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil
| | - Raquel da Silva Aragão
- Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil.,Graduate Program in Nutrition, Universidade Federal de Pernambuco, Recife, Brazil.,Physical Education and Sport Sciences Nucleus, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil.,Unity of Studies in Nutrition and Phenotypic Plasticity, Department of Nutrition, Universidade Federal de Pernambuco, Recife, Brazil
| | | | - Kelli Nogueira Ferraz-Pereira
- Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil.,Unity of Studies in Nutrition and Phenotypic Plasticity, Department of Nutrition, Universidade Federal de Pernambuco, Recife, Brazil
| | - Isabeli Lins Pinheiro
- Physical Education and Sport Sciences Nucleus, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil.,Unity of Studies in Nutrition and Phenotypic Plasticity, Department of Nutrition, Universidade Federal de Pernambuco, Recife, Brazil
| | - Lígia Cristina Monteiro Galindo
- Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil.,Unity of Studies in Nutrition and Phenotypic Plasticity, Department of Nutrition, Universidade Federal de Pernambuco, Recife, Brazil.,Departament of Anatomy, Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
22
|
Giglio RV, Stoian AP, Haluzik M, Pafili K, Patti AM, Rizvi AA, Ciaccio M, Papanas N, Rizzo M. Novel molecular markers of cardiovascular disease risk in type 2 diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166148. [PMID: 33892081 DOI: 10.1016/j.bbadis.2021.166148] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/15/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
Diabetes represents the leading risk factor for the development of cardiovascular disease (CVD). Chronic hyperglycemia and/or acute post-prandial changes in blood glucose determine an increase in reactive oxygen species (ROS), which play a fundamental role in endothelial dysfunction and in the nuclear transport of pro-atherogenic transcription factors that activate the "inflammasome". In addition, the glycemic alteration favors the formation and stabilization of atherosclerotic plaque through the mechanism of non-enzymatic glycation of different molecules, with the establishment of the so-called "advanced glycosylation end products" (AGE). Laboratory information provided by the level of biomarkers could make a quantitative and qualitative contribution to the clinical process of screening, prediction, prevention, diagnosis, prognosis and monitoring of cardiovascular (CV) risk linked to diabetes. This review describes the importance of specific biomarkers, with particular focus on novel ones, for stratifying and management of diabetes CV risk.
Collapse
Affiliation(s)
- Rosaria Vincenza Giglio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Anca Pantea Stoian
- Faculty of General Medicine, Diabetes, Nutrition and Metabolic Diseases Department, Carol Davila University, Bucharest, Romania
| | - Martin Haluzik
- Centre for Experimental Medicine and Department of Diabetes, Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Kalliopi Pafili
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| | - Angelo Maria Patti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Ali Abbas Rizvi
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University, Atlanta, Georgia, USA; Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine Columbia, South Carolina, USA
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy; Department of Laboratory Medicine, University-Hospital, Palermo, Italy
| | - Nikolaos Papanas
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine Columbia, South Carolina, USA
| |
Collapse
|
23
|
Clarke GD, Li J, Kuo AH, Moody AJ, Nathanielsz PW. Cardiac magnetic resonance imaging: insights into developmental programming and its consequences for aging. J Dev Orig Health Dis 2021; 12:203-219. [PMID: 33349289 PMCID: PMC7987688 DOI: 10.1017/s2040174420001233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVD) are important consequences of adverse perinatal conditions such as fetal hypoxia and maternal malnutrition. Cardiac magnetic resonance imaging (CMR) can produce a wealth of physiological information related to the development of the heart. This review outlines the current state of CMR technologies and describes the physiological biomarkers that can be measured. These phenotypes include impaired ventricular and atrial function, maladaptive ventricular remodeling, and the proliferation of myocardial steatosis and fibrosis. The discussion outlines the applications of CMR to understanding the developmental pathways leading to impaired cardiac function. The use of CMR, both in animal models of developmental programming and in human studies, is described. Specific examples are given in a baboon model of intrauterine growth restriction (IUGR). CMR offers great potential as a tool for understanding the sequence of dysfunctional adaptations of developmental origin that can affect the human cardiovascular system.
Collapse
Affiliation(s)
- G D Clarke
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - J Li
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - A H Kuo
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - A J Moody
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - P W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
24
|
Yang Y, Kurian J, Schena G, Johnson J, Kubo H, Travers JG, Kang C, Lucchese AM, Eaton DM, Lv M, Li N, Leynes LG, Yu D, Yang F, McKinsey TA, Kishore R, Khan M, Mohsin S, Houser SR. Cardiac Remodeling During Pregnancy With Metabolic Syndrome: Prologue of Pathological Remodeling. Circulation 2021; 143:699-712. [PMID: 33587660 PMCID: PMC7888689 DOI: 10.1161/circulationaha.120.051264] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/30/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND The heart undergoes physiological hypertrophy during pregnancy in healthy individuals. Metabolic syndrome (MetS) is now prevalent in women of child-bearing age and might add risks of adverse cardiovascular events during pregnancy. The present study asks if cardiac remodeling during pregnancy in obese individuals with MetS is abnormal and whether this predisposes them to a higher risk for cardiovascular disorders. METHODS The idea that MetS induces pathological cardiac remodeling during pregnancy was studied in a long-term (15 weeks) Western diet-feeding animal model that recapitulated features of human MetS. Pregnant female mice with Western diet (45% kcal fat)-induced MetS were compared with pregnant and nonpregnant females fed a control diet (10% kcal fat). RESULTS Pregnant mice fed a Western diet had increased heart mass and exhibited key features of pathological hypertrophy, including fibrosis and upregulation of fetal genes associated with pathological hypertrophy. Hearts from pregnant animals with WD-induced MetS had a distinct gene expression profile that could underlie their pathological remodeling. Concurrently, pregnant female mice with MetS showed more severe cardiac hypertrophy and exacerbated cardiac dysfunction when challenged with angiotensin II/phenylephrine infusion after delivery. CONCLUSIONS These results suggest that preexisting MetS could disrupt physiological hypertrophy during pregnancy to produce pathological cardiac remodeling that could predispose the heart to chronic disorders.
Collapse
Affiliation(s)
- Yijun Yang
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Justin Kurian
- Center for Metabolic Disease and Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Giana Schena
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jaslyn Johnson
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hajime Kubo
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Joshua G. Travers
- Department of Medicine, Division of Cardiology, and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Chunya Kang
- Medical University of Lublin, Lublin, Poland
| | - Anna Maria Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Deborah M. Eaton
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Maoting Lv
- Second Ultrasound Department, Cangzhou Central Hospital, Hebei, China
| | - Na Li
- Second Department of Obstetrics, Cangzhou Central Hospital, Hebei, China
| | - Lorianna G. Leynes
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, PA, United States
| | - Fengzhen Yang
- Second Department of Obstetrics, Cangzhou Central Hospital, Hebei, China
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology, and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease and Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Steven R. Houser
- Independence Blue Cross Cardiovascular Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
25
|
Hussain T, Tan B, Murtaza G, Metwally E, Yang H, Kalhoro MS, Kalhoro DH, Chughtai MI, Yin Y. Role of Dietary Amino Acids and Nutrient Sensing System in Pregnancy Associated Disorders. Front Pharmacol 2020; 11:586979. [PMID: 33414718 PMCID: PMC7783402 DOI: 10.3389/fphar.2020.586979] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Defective implantation is related to pregnancy-associated disorders such as spontaneous miscarriage, intrauterine fetal growth restriction and others. Several factors proclaimed to be involved such as physiological, nutritional, environmental and managemental that leads to cause oxidative stress. Overloading of free radicals promotes oxidative stress, and the internal body system could not combat its ability to encounter the damaging effects and subsequently leading to pregnancy-related disorders. During pregnancy, essential amino acids display important role for optimum fetal growth and other necessary functions for continuing fruitful pregnancy. In this context, dietary amino acids have received much attention regarding the nutritional concerns during pregnancy. Arginine, glutamine, tryptophan and taurine play a crucial role in fetal growth, development and survival while ornithine and proline are important players for the regulation of gene expression, protein synthesis and angiogenesis. Moreover, amino acids also stimulate the mammalian target of rapamycin (mTOR) signaling pathway which plays a central role in the synthesis of proteins in placenta, uterus and fetus. This review article explores the significances of dietary amino acids in pregnancy development, regulation of nutrient-sensing pathways such as mTOR, peroxisome proliferator-activated receptors (PPARs), insulin/insulin-like growth factor signaling pathway (IIS) and 5' adenosine monophosphate-activated protein kinase (AMPK) which exhibit important role in reproduction and its related problems. In addition, the antioxidant function of dietary amino acids against oxidative stress triggering pregnancy disorders and their possible outcomes will also be enlightened. Dietary supplementation of amino acids during pregnancy could help mitigate reproductive disorders and thereby improving fertility in animals as well as humans.
Collapse
Affiliation(s)
- Tarique Hussain
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C,PIEAS), Faisalabad, Pakistan
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ghulam Murtaza
- Department of Animal Reproduction, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Sindh, Pakistan
| | - Elsayed Metwally
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Huansheng Yang
- Hunan International Joint laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Muhammad Saleem Kalhoro
- Department of Animal Products Technology, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Sindh, Pakistan
| | - Dildar Hussain Kalhoro
- Department of Veterinary Microbiology, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Sindh, Pakistan
| | - Muhammad Ismail Chughtai
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C,PIEAS), Faisalabad, Pakistan
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
26
|
Louwagie EJ, Larsen TD, Wachal AL, Gandy TCT, Eclov JA, Rideout TC, Kern KA, Cain JT, Anderson RH, Mdaki KS, Baack ML. Age and Sex Influence Mitochondria and Cardiac Health in Offspring Exposed to Maternal Glucolipotoxicity. iScience 2020; 23:101746. [PMID: 33225249 PMCID: PMC7666357 DOI: 10.1016/j.isci.2020.101746] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/29/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
Infants of diabetic mothers are at risk of cardiomyopathy at birth and myocardial infarction in adulthood, but prevention is hindered because mechanisms remain unknown. We previously showed that maternal glucolipotoxicity increases the risk of cardiomyopathy and mortality in newborn rats through fuel-mediated mitochondrial dysfunction. Here we demonstrate ongoing cardiometabolic consequences by cross-fostering and following echocardiography, cardiomyocyte bioenergetics, mitochondria-mediated turnover, and cell death following metabolic stress in aged adults. Like humans, cardiac function improves by weaning with no apparent differences in early adulthood but declines again in aged diabetes-exposed offspring. This is preceded by impaired oxidative phosphorylation, exaggerated age-related increase in mitochondrial number, and higher oxygen consumption. Prenatally exposed male cardiomyocytes have more mitolysosomes indicating high baseline turnover; when exposed to metabolic stress, mitophagy cannot increase and cardiomyocytes have faster mitochondrial membrane potential loss and mitochondria-mediated cell death. Details highlight age- and sex-specific roles of mitochondria in developmentally programmed adult heart disease. Fetal exposures disrupt mitochondria, bioenergetics, & cardiac function at birth First, bioenergetics & function improve until greater reliance on OXPHOS with age At 6MO, poor respiration incites biogenesis & mitophagy, and then functional decline Fetal exposures cause faster mitochondria-mediated cell death in aged adult hearts
Collapse
Affiliation(s)
- Eli J Louwagie
- University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA.,Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Tricia D Larsen
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Angela L Wachal
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Tyler C T Gandy
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Julie A Eclov
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, State University of New York, Buffalo, NY 14214, USA
| | - Katherine A Kern
- Department of Exercise and Nutrition Sciences, State University of New York, Buffalo, NY 14214, USA
| | - Jacob T Cain
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Ruthellen H Anderson
- University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA.,Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Kennedy S Mdaki
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Michelle L Baack
- University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA.,Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA.,Boekelheide Neonatal Intensive Care Unit, Sanford Children's Hospital, Sioux Falls, SD 57117, USA
| |
Collapse
|
27
|
Pei L, Xiao H, Lai F, Li Z, Li Z, Yue S, Chen H, Li Y, Cao X. Early postpartum dyslipidemia and its potential predictors during pregnancy in women with a history of gestational diabetes mellitus. Lipids Health Dis 2020; 19:220. [PMID: 33036614 PMCID: PMC7547505 DOI: 10.1186/s12944-020-01398-1] [Citation(s) in RCA: 11] [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: 08/02/2020] [Accepted: 10/01/2020] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND This study aimed to analyze the incidence of early postpartum dyslipidemia and its potential predictors in women with a history of gestational diabetes mellitus (GDM). METHODS This was a retrospective study. Five hundred eighty-nine women diagnosed with GDM were enrolled and followed up at 6-12 weeks after delivery. A 75 g oral glucose tolerance test (OGTT) and lipid levels were performed during mid-trimester and the early postpartum period. Participants were divided into the normal lipid group and dyslipidemia group according to postpartum lipid levels. Demographic and metabolic parameters were analyzed. Multiple logistic regression was performed to analyze the potential predictors for early postpartum dyslipidemia. A receiver operating characteristic curve (ROC) was calculated to determine the cut-off values. RESULTS A total of 38.5% of the 589 women developed dyslipidemia in early postpartum and 60% of them had normal glucose metabolism. Delivery age, systolic blood pressure (SBP), glycated hemoglobin (HbA1c) and low-density lipoprotein cholesterol (LDL-C) were independent predictors of early postpartum dyslipidemia in women with a history of GDM. The cut-offs of maternal age, SBP, HbA1c values, and LDL-C levels were 35 years, 123 mmHg, 5.1%, and 3.56 mmol/L, respectively. LDL-C achieved a balanced mix of high sensitivity (63.9%) and specificity (69.2%), with the highest area under the receiver operating characteristic curve (AUC) (0.696). When LDL-C was combined with age, SBP, and HbA1c, the AUC reached to 0.733. CONCLUSIONS A lipid metabolism evaluation should be recommended in women with a history of GDM after delivery, particularly those with a maternal age > 35 years, SBP > 123 mmHg before labor, HbA1c value > 5.1%, or LDL-C levels > 3.56 mmol/L in the second trimester of pregnancy.
Collapse
Affiliation(s)
- Ling Pei
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China
| | - Huangmeng Xiao
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China
| | - Fenghua Lai
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China
| | - Zeting Li
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China
| | - Zhuyu Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shufan Yue
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China
| | - Haitian Chen
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanbing Li
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China
| | - Xiaopei Cao
- Department of Endocrinology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd, Guangzhou, 510080, China.
| |
Collapse
|
28
|
Preston CC, Larsen TD, Eclov JA, Louwagie EJ, Gandy TCT, Faustino RS, Baack ML. Maternal High Fat Diet and Diabetes Disrupts Transcriptomic Pathways That Regulate Cardiac Metabolism and Cell Fate in Newborn Rat Hearts. Front Endocrinol (Lausanne) 2020; 11:570846. [PMID: 33042024 PMCID: PMC7527411 DOI: 10.3389/fendo.2020.570846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Children born to diabetic or obese mothers have a higher risk of heart disease at birth and later in life. Using chromatin immunoprecipitation sequencing, we previously demonstrated that late-gestation diabetes, maternal high fat (HF) diet, and the combination causes distinct fuel-mediated epigenetic reprogramming of rat cardiac tissue during fetal cardiogenesis. The objective of the present study was to investigate the overall transcriptional signature of newborn offspring exposed to maternal diabetes and maternal H diet. Methods: Microarray gene expression profiling of hearts from diabetes exposed, HF diet exposed, and combination exposed newborn rats was compared to controls. Functional annotation, pathway and network analysis of differentially expressed genes were performed in combination exposed and control newborn rat hearts. Further downstream metabolic assessments included measurement of total and phosphorylated AKT2 and GSK3β, as well as quantification of glycolytic capacity by extracellular flux analysis and glycogen staining. Results: Transcriptional analysis identified significant fuel-mediated changes in offspring cardiac gene expression. Specifically, functional pathways analysis identified two key signaling cascades that were functionally prioritized in combination exposed offspring hearts: (1) downregulation of fibroblast growth factor (FGF) activated PI3K/AKT pathway and (2) upregulation of peroxisome proliferator-activated receptor gamma coactivator alpha (PGC1α) mitochondrial biogenesis signaling. Functional metabolic and histochemical assays supported these transcriptome changes, corroborating diabetes- and diet-induced cardiac transcriptome remodeling and cardiac metabolism in offspring. Conclusion: This study provides the first data accounting for the compounding effects of maternal hyperglycemia and hyperlipidemia on the developmental cardiac transcriptome, and elucidates nuanced and novel features of maternal diabetes and diet on regulation of heart health.
Collapse
Affiliation(s)
- Claudia C. Preston
- Genetics and Genomics Group, Sanford Research, Sioux Falls, SD, United States
| | - Tricia D. Larsen
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD, United States
| | - Julie A. Eclov
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD, United States
| | - Eli J. Louwagie
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD, United States
| | - Tyler C. T. Gandy
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD, United States
| | - Randolph S. Faustino
- Genetics and Genomics Group, Sanford Research, Sioux Falls, SD, United States
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD, United States
| | - Michelle L. Baack
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD, United States
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD, United States
| |
Collapse
|
29
|
Najman JM, Wang W, Plotnikova M, Mamun AA, McIntyre D, Williams GM, Scott JG, Bor W, Clavarino AM. Poverty over the early life course and young adult cardio-metabolic risk. Int J Public Health 2020; 65:759-768. [PMID: 32666221 DOI: 10.1007/s00038-020-01423-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES There is little known about whether exposure to family poverty at specific periods of the early life course independently contributes to coronary heart disease risk beyond the contribution of concurrent poverty. METHODS Children were recruited in early pregnancy and additional survey data obtained during the pregnancy and at the 5-, 14- and 30-year follow-ups. Fasting blood samples were also obtained at the 30-year follow-up. Analyses are multinominal logistic regressions stratified by gender and with adjustments for confounding. RESULTS For male offspring, family poverty at different stages of the early life course was not associated with measures of cardio-metabolic risk. For females early life course, poverty predicted obesity, homeostatic model assessment of insulin resistance (HOMA-IR) and total cholesterol/high-density lipoprotein cholesterol (TC/HDL-C), as well as concurrent family poverty associated with obesity, HOMA-IR, TC/HDL-C, HDL-C and increased systolic and diastolic blood pressure. CONCLUSIONS Family poverty in the early life course independently predicts increased levels of cardio-metabolic risk of females. The primary finding, however, is that concurrent poverty is independently and strongly associated with increased cardio-metabolic risk levels in young adulthood.
Collapse
Affiliation(s)
- Jake M Najman
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, Australia.
- School of Social Science, The University of Queensland, St. Lucia, Australia.
| | - William Wang
- Faculty of Medicine, The University of Queensland, Herston, Australia
| | - Maria Plotnikova
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, Australia
| | - Abdullah A Mamun
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, Australia
| | - David McIntyre
- Mater Research Institute, Faculty of Medicine, The University of Queensland, South Brisbane, Australia
| | - Gail M Williams
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, Australia
| | - James G Scott
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, Australia
- Metro North Mental Health Service, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - William Bor
- Mater Child and Youth Mental Health Service, Mater Hospital, University of Queensland, Brisbane, Australia
| | - Alexandra M Clavarino
- School of Public Health, Faculty of Medicine, The University of Queensland, Herston, Australia
| |
Collapse
|
30
|
Puttabyatappa M, Sargis RM, Padmanabhan V. Developmental programming of insulin resistance: are androgens the culprits? J Endocrinol 2020; 245:R23-R48. [PMID: 32240982 PMCID: PMC7219571 DOI: 10.1530/joe-20-0044] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023]
Abstract
Insulin resistance is a common feature of many metabolic disorders. The dramatic rise in the incidence of insulin resistance over the past decade has enhanced focus on its developmental origins. Since various developmental insults ranging from maternal disease, stress, over/undernutrition, and exposure to environmental chemicals can all program the development of insulin resistance, common mechanisms may be involved. This review discusses the possibility that increases in maternal androgens associated with these various insults are key mediators in programming insulin resistance. Additionally, the intermediaries through which androgens misprogram tissue insulin sensitivity, such as changes in inflammatory, oxidative, and lipotoxic states, epigenetic, gut microbiome and insulin, as well as data gaps to be filled are also discussed.
Collapse
Affiliation(s)
| | - Robert M. Sargis
- Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | | |
Collapse
|
31
|
Maternal exposure to a high-fat diet showed unfavorable effects on the body weight, apoptosis and morphology of cardiac myocytes in offspring. Arch Gynecol Obstet 2020; 301:837-844. [PMID: 32114674 DOI: 10.1007/s00404-020-05470-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/13/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The study intends to explore the functions of maternal high-fat diet exposure on progeny weight and heart. METHODS Sprague-Dawley (SD) rats, fed on a high-fat diet, were used to establish a model of weight gain before and during pregnancy. The body and cardiac weight of neonatal, 1-month- and 3-month-old rats were measured. The morphology of myocardial cells was observed by hemotoxylin and eosin (H&E) staining. The expression of caspase-3, 8, 9 was measured by qRT-PCR and western blot. RESULTS Normal pregnant rats, fed on a high-fat diet throughout pregnancy, had a significant increase in body and cardiac weight of their neonates, and more fat deposition in myocardial cells and an increased expression of caspase-3, 8, 9, compared with that of the normal pregnant rats + normal diet group. These phenomena were relieved through later diet control. Pregnant rats, which fed on a high-fat diet throughout pregnancy, showed more adverse effects on neonatal body and cardiac weight, myocardial cell fat deposition, and the expression of caspase-3, 8, 9, compared with pregnant rats exposed to high-fat diet + normal diet and pregnant rats exposed to high-fat diet + normal diet + exercise. These phenomena cannot be fully restored via controlling later diet. CONCLUSIONS Our results stated that a proper diet before and during pregnancy was important for the cardiac health of offspring.
Collapse
|
32
|
Maternal Obesity Programs Offspring Development and Resveratrol Potentially Reprograms the Effects of Maternal Obesity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17051610. [PMID: 32131513 PMCID: PMC7084214 DOI: 10.3390/ijerph17051610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022]
Abstract
Maternal obesity during pregnancy is a now a public health burden that may be the culprit underlying the ever-increasing rates of adult obesity worldwide. Understanding the association between maternal obesity and adult offspring’s obesity would inform policy and practice regarding offspring health through available resources and interventions. This review first summarizes the programming effects of maternal obesity and discusses the possible underlying mechanisms. We then summarize the current evidence suggesting that maternal consumption of resveratrol is helpful in maternal obesity and alleviates its consequences. In conclusion, maternal obesity can program offspring development in an adverse way. Maternal resveratrol could be considered as a potential regimen in reprogramming adverse outcomes in the context of maternal obesity.
Collapse
|
33
|
Yang L, Zhang B, Wang X, Liu Z, Li J, Zhang S, Gu X, Jia M, Guo H, Feng N, Fan R, Xie M, Pei J, Chen L. P53/PANK1/miR-107 signalling pathway spans the gap between metabolic reprogramming and insulin resistance induced by high-fat diet. J Cell Mol Med 2020; 24:3611-3624. [PMID: 32048816 PMCID: PMC7131928 DOI: 10.1111/jcmm.15053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
High-fat diet (HFD) leads to obesity, type II diabetes mellitus (T2DM) and increases the coincidence of cardiovascular diseases and cancer. Insulin resistance (IR) is considered as the 'common soil' of those diseases. Furthermore, people on HFD showed restrained glycolysis and enhanced fatty acid oxidation, which is the so-called metabolic reprogramming. However, the relationship between metabolic reprogramming and IR induced by HFD is still unclear. Here, we demonstrate that PANK1 and miR-107 were up-regulated in the liver tissue of mice on HFD for 16 weeks and involved in metabolic reprogramming induced by palmitate acid (PA) incubation. Importantly, miR-107 within an intron of PANK1 gene facilitated IR by targeting caveolin-1 in AML12 cells upon PA incubation. Moreover, we identify that HFD enhanced P53 expression, and activation of P53 with nutlin-3a induced PANK1 and miR-107 expression simultaneously in transcriptional level, leading to metabolic reprogramming and IR, respectively. Consistently, inhibition of P53 with pifithrin-α hydrobromide ameliorated PA-induced metabolic reprogramming and IR. Thus, our results revealing a new mechanism by which P53 regulate metabolism. In addition, the results distinguished the different roles of PANK1 and its intron miR-107 in metabolic regulation, which will provide more accurate intervention targets for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Lu Yang
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Bin Zhang
- Department of Aerospace PhysiologyFourth Military Medical UniversityXi'anChina
| | - Xinju Wang
- Battalion 5 of CadetsFourth Military Medical UniversityXi'anChina
| | - Zhenhua Liu
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Juan Li
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Shumiao Zhang
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Xiaoming Gu
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Min Jia
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Haitao Guo
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Na Feng
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Rong Fan
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Manjiang Xie
- Department of Aerospace PhysiologyFourth Military Medical UniversityXi'anChina
| | - Jianming Pei
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
| | - Li Chen
- Department of PhysiologyNational Key Discipline of Cell BiologyFourth Military Medical UniversityXi'anChina
- Department of Aerospace PhysiologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
34
|
Zhao S, Zhong S, Wang F, Wang H, Xu D, Li G. Microcystin-LR exposure decreased the fetal weight of mice by disturbance of placental development and ROS-mediated endoplasmic reticulum stress in the placenta. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 256:113362. [PMID: 31672369 DOI: 10.1016/j.envpol.2019.113362] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/23/2019] [Accepted: 10/07/2019] [Indexed: 06/10/2023]
Abstract
The placenta is essential for sustaining the growth of the fetus. The aim of this study was to investigate the role of the placenta in MCLR-induced significant reduction in fetal weight, especially the changes in placental structure and function. Pregnant mice were intraperitoneally injected with MCLR (5 or 20 μg/kg) from gestational day (GD) 13 to GD17. The results showed MCLR reduced fetal weight and placenta weight. The histological specimens of the placentas were taken for light and electron microscopy studies. The internal space of blood vessels decreased obviously in the placental labyrinth layer of mice treated with MCLR. After the ultrastructural examination, the edema and intracytoplasmic vacuolization, dilation of the endoplasmic reticulum and corrugation of the nucleus were observed. In addition, maternal MCLR exposure caused a reduction of 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2) expression in placentae, a critical regulator of fetal development. Several genes of placental growth factors, such as Vegfα and Pgf and several genes of nutrient transport pumps, such as Glut1 and Pcft were depressed in placentas of MCLR-treated mice, however nutrient transporters Fatp1 and Snat4 were promoted. Moreover, significant increases in malondialdehyde (MDA) revealed the occurrence of oxidative stress caused by MCLR, which was also verified by remarkable decrease in the glutathione levels, total antioxidant capacity (T-AOC) as well as the activity of antioxidant enzymes. Real-time PCR and western blot analysis revealed that GRP78, CHOP, XBP-1, peIF2α and pIRE1 were remarkable increased in placentas of MCLR-treated mice, indicating that endoplasmic reticulum (ER) stress pathway was activated by MCLR. Furthermore, oxidative stress and ER stress consequently triggered apoptosis which contributed to the impairment of placental development. Collectively, these results suggest maternal MCLR exposure results in reduced fetal body weight, which might be associated with ROS-mediated endoplasmic reticulum stress and impairment in placental structure and function.
Collapse
Affiliation(s)
- Sujuan Zhao
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Shengzheng Zhong
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Fang Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Honghui Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Dexiang Xu
- School of Public Health, Anhui Medical University, Hefei, 230032, China.
| | - Guangyu Li
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
35
|
Wang XF, Li JD, Huo YL, Zhang YP, Fang ZQ, Wang HP, Peng W, Johnson AK, Xue B. Blockade of angiotensin-converting enzyme or tumor necrosis factor-α reverses maternal high-fat diet-induced sensitization of angiotensin II hypertension in male rat offspring. Am J Physiol Regul Integr Comp Physiol 2019; 318:R351-R359. [PMID: 31746626 DOI: 10.1152/ajpregu.00200.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Maternal high-fat diet (HFD) is associated with metabolic syndrome and cardiovascular diseases in adult offspring. Our previous study demonstrated that maternal HFD enhances pressor responses to ANG II or a proinflammatory cytokine (PIC), which is associated with increased expression of brain renin-angiotensin system (RAS) components and PICs in adult offspring. The present study further investigated whether inhibition of angiotensin-converting enzyme (ACE) or tumor necrosis factor-α (TNF-α) blocks sensitization of ANG II hypertension in offspring of HFD dams. All offspring were bred from dams with normal fat diet (NFD) or HFD starting two weeks before mating and maintained until weaning of the offspring. Then the weaned offspring were treated with an ACE inhibitor (captopril) or a TNF-α inhibitor (pentoxifylline) in the drinking water through the end of testing with a slow-pressor dose of ANG II. RT-PCR analyses of the lamina terminalis and paraventricular nucleus revealed upregulation of mRNA expression of several RAS components and PICs in male offspring of HFD dams when compared with age-matched offspring of NFD dams. The enhanced gene expression was attenuated by blockade of either RAS or PICs. Likewise, ANG II administration produced an augmented pressor response in offspring of HFD dams. This was abolished by either ACE or TNF-α inhibitor. Taken together, this study provides mechanistic evidence and a therapeutic strategy that systemic inhibition of the RAS and PICs can block maternal HFD-induced sensitization of ANG II hypertension, which is associated with attenuation of brain RAS and PIC expression in offspring.
Collapse
Affiliation(s)
- Xue-Fang Wang
- Life Science Research Center, Hebei North University, Zhangjiakou City, China
| | - Jian-Dong Li
- Life Science Research Center, Hebei North University, Zhangjiakou City, China
| | - Yan-Li Huo
- Life Science Research Center, Hebei North University, Zhangjiakou City, China
| | - Yu-Ping Zhang
- Life Science Research Center, Hebei North University, Zhangjiakou City, China
| | - Zhi-Qin Fang
- First Affiliated Hospital, Hebei North University, Zhangjiakou City, China
| | - Hai-Ping Wang
- Life Science Research Center, Hebei North University, Zhangjiakou City, China
| | - Wei Peng
- Life Science Research Center, Hebei North University, Zhangjiakou City, China
| | - Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| |
Collapse
|
36
|
Maternal Body Mass Index and Risk of Congenital Heart Defects in Infants: A Dose-Response Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1315796. [PMID: 31360700 PMCID: PMC6642764 DOI: 10.1155/2019/1315796] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 01/19/2023]
Abstract
Objective The exact shape of the dose-response relationship between maternal body mass index (BMI) and the risk of congenital heart defects (CHDs) in infants has not been clearly defined yet. This study aims to further clarify the relationship between maternal obesity and the risk of CHDs in infants by an overall and dose-response meta-analysis. Methods PubMed, Embase, and Web of Science databases were searched to identify all related studies. The studies were limited to human cohort or case-control studies in English language. Random-effect models and dose-response meta-analysis were used to synthesize the results. Heterogeneity, subgroup analysis, sensitivity analysis, and publication bias were also assessed. Results Nineteen studies with 2,416,546 participants were included in our meta-analysis. Compared with the mothers with normal weight, the pooled relative risks (RRs) of infants with CHDs were 1.08 (95% CI=1.03-1.13) in overweight and 1.23 (95% CI=1.17-1.29) in obese mothers. According to the findings from the linear meta-analysis, we observed an increased risk of infants with CHDs (RR=1.07, 95% CI=1.06-1.08) for each 5 kg/m2 increase in maternal BMI. A nonlinear relationship between maternal BMI and risk of infants with CHDs was also found (p=0.012). Conclusion The results from our meta-analysis indicate that increased maternal BMI is related to increased risk of CHDs in infants.
Collapse
|
37
|
Merle L, Person O, Bonnet P, Grégoire S, Soubeyre V, Grosmaitre X, Jarriault D. Maternal high fat high sugar diet disrupts olfactory behavior but not mucosa sensitivity in the offspring. Psychoneuroendocrinology 2019; 104:249-258. [PMID: 30904822 DOI: 10.1016/j.psyneuen.2019.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/24/2018] [Accepted: 02/06/2019] [Indexed: 12/18/2022]
Abstract
The influence of maternal diet on progeny's metabolic health has been thoroughly investigated, but the impact on sensory systems remains unexplored. Neurons of the olfactory system start to develop during the embryonic life and carry on their maturation after birth. Besides, these neurons are under metabolic influences, and it has recently been shown that adult mice exposed to an obesogenic or diabetogenic diet display reduced olfactory abilities. However, whether or not Folfactory function is affected by the perinatal nutritional environment is unknown. Here we investigated the effect of a high fat high sucrose (HFHS) maternal diet (46% of total energy brought by lipids, 26.6% by sucrose) on progeny's olfactory system in mice. In male offspring at weaning stage, maternal HFHS diet induced overweight and increased gonadal fat, associated with hyperleptinemia. The progeny of HFHS diet fed dams showed reduced sniffing behavior in the presence of low doses of phenylethanol (an attractive odorant for mice), compared to the progeny of standard diet fed dams. Furthermore, they exhibited increased time to retrieve a piece of breakfast cereals hidden beneath the bedding in a buried food test. Meanwhile, electroolfactogram recordings revealed no change in the sensitivity of olfactory mucosa. mRNA levels for elements of the olfactory transduction cascade were not affected either. Our results demonstrate that maternal HFHS diet during gestation and lactation strongly modulates olfactory perception in the offspring, without impairing odor detection by the olfactory epithelium. Maternal HFHS diet starting two months before gestation did not induce additional impairments in progeny.
Collapse
Affiliation(s)
- Laëtitia Merle
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France
| | - Ophélie Person
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France
| | - Pierre Bonnet
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France
| | - Stéphane Grégoire
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France
| | - Vanessa Soubeyre
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France
| | - Xavier Grosmaitre
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France
| | - David Jarriault
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 9E boulevard Jeanne d'Arc, F-21000 Dijon, France.
| |
Collapse
|
38
|
Upadhyay A, Anjum B, Godbole NM, Rajak S, Shukla P, Tiwari S, Sinha RA, Godbole MM. Time-restricted feeding reduces high-fat diet associated placental inflammation and limits adverse effects on fetal organ development. Biochem Biophys Res Commun 2019; 514:415-421. [PMID: 31053302 DOI: 10.1016/j.bbrc.2019.04.154] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 12/18/2022]
Abstract
Maternal nutrition has become a major public health concern over recent years and is a known predictor of adverse long-term metabolic derangement in offspring. Time-restricted feeding (TRF), wherein food consumption is restricted to the metabolically active phase of the day, is a dietary approach that improves metabolic parameters when consuming a high-fat diet (HFD). Here, we tested whether TRF could reduce maternal HFD associated inflammation and thereby mitigate defects in fetal organ developmental. Female rats were kept on following three dietary regimens; Ad libitum normal chow diet (NCD-AL), Ad libitum HFD (HFD-AL) and Time-restricted fed HFD (HFD-TRF) from 5 months prior to mating and continued throughout pregnancy. Rat dams were sacrificed at embryonic day 18.5 (ED18.5) and placental tissues from these rats were processed for the analysis of cellular apoptosis, inflammatory cytokines (TNFα and IL-6), oxidative stress, endoplasmic reticulum (ER) stress and autophagy. Furthermore, fetal hepatic triglyceride (TG) content and fetal lung maturation were assessed at ED18.5. Biochemical analysis revealed that HFD-TRF rat had significantly lower serum TG levels and body weight compared to HFD-AL rats. Additionally, TRF significantly blocked HFD-induced placental apoptosis and inflammation via minimizing cellular stress, and restoring autophagic flux. In addition, fetal hepatosteatosis and delayed fetal lung maturation induced by HFD was significantly ameliorated in HFD-TRF compared to HFD-AL. Collectively, our results suggest that reducing placental inflammation via TRF could prevent adverse fetal metabolic outcomes in pregnancies complicated by maternal obesity.
Collapse
Affiliation(s)
- Aditya Upadhyay
- Dept. of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India; Dr. A.P.J. Abdul Kalam Technical University Uttar Pradesh, Lucknow, India
| | - B Anjum
- Dept. of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India; Dept of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Nachiket M Godbole
- Dept. of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Sangam Rajak
- Dept of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Pooja Shukla
- Dept of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Swasti Tiwari
- Dept. of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rohit A Sinha
- Dept of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Madan M Godbole
- Dept. of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| |
Collapse
|
39
|
Bendimerad-Benmokhtar S, Bouanane S, Merzouk H, Baba Ahmed FZ, Bendaoud A. Effects of Nannochloropsis Fed on Serum and Tissue Lipids Metabolism in Obese Offspring of Overfed Dams. CURRENT NUTRITION & FOOD SCIENCE 2019. [DOI: 10.2174/1573401313666171004153311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background: The present work aims at determining the effects of maternal-diet-induced obesity on offspring metabolism. The short-term of a marine microalgae diet and its effects on lipids metabolism was investigated. </P><P> Method: Before gestation, some rats are fed control diet and others cafeteria diet. Moreover, two groups of dams were fed standard and cafeteria diets, and two other groups were fed the same diets but containing 10% of microalgae. This feeding started at gestation, and continued throughout parturition, lactation until their offspring's weaning age. </P><P> Results: Cafeteria diet was shown to increase the body weight and visceral obesity, with aberration in lipid metabolism. The results obtained show that the microalgae diet supplement induces a significant decrease in the maternal body weight and relative adipose tissue weight, plasma glucose and lipid levels, liver-triglyceride (TG) and adipose tissue-TG at parturition and at the end of lactation. Also, the addition of the microalgae in both males and female offspring fed dams at birth and weaning showed significant decrease in body weight, liver-TG whereas significant increase in TG-HDL. </P><P> Conclusion: In the end, it was noted that the incorporation of 10% of microalgae has a beneficial effect on body weight and lipid metabolism.
Collapse
Affiliation(s)
- Soraya Bendimerad-Benmokhtar
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University of Tlemcen 13000, Tlemcen, Algeria
| | - Samira Bouanane
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University of Tlemcen 13000, Tlemcen, Algeria
| | - Hafida Merzouk
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University of Tlemcen 13000, Tlemcen, Algeria
| | - Fatima Zohra Baba Ahmed
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University of Tlemcen 13000, Tlemcen, Algeria
| | - Asme Bendaoud
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University of Tlemcen 13000, Tlemcen, Algeria
| |
Collapse
|
40
|
Guida MC, Birse RT, Dall'Agnese A, Toto PC, Diop SB, Mai A, Adams PD, Puri PL, Bodmer R. Intergenerational inheritance of high fat diet-induced cardiac lipotoxicity in Drosophila. Nat Commun 2019; 10:193. [PMID: 30643137 PMCID: PMC6331650 DOI: 10.1038/s41467-018-08128-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/17/2018] [Indexed: 12/26/2022] Open
Abstract
Obesity is strongly correlated with lipotoxic cardiomyopathy, heart failure and thus mortality. The incidence of obesity has reached alarming proportions worldwide, and increasing evidence suggests that the parents' nutritional status may predispose their offspring to lipotoxic cardiomyopathy. However, to date, mechanisms underlying intergenerational heart disease risks have yet to be elucidated. Here we report that cardiac dysfunction induced by high-fat-diet (HFD) persists for two subsequent generations in Drosophila and is associated with reduced expression of two key metabolic regulators, adipose triglyceride lipase (ATGL/bmm) and transcriptional cofactor PGC-1. We provide evidence that targeted expression of ATGL/bmm in the offspring of HFD-fed parents protects them, and the subsequent generation, from cardio-lipotoxicity. Furthermore, we find that intergenerational inheritance of lipotoxic cardiomyopathy correlates with elevated systemic H3K27 trimethylation. Lowering H3K27 trimethylation genetically or pharmacologically in the offspring of HFD-fed parents prevents cardiac pathology. This suggests that metabolic homeostasis is epigenetically regulated across generations.
Collapse
Affiliation(s)
- Maria Clara Guida
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Ryan Tyge Birse
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- Biocompatibles Inc., 300 Four Falls Corporate Center, 300 Conshohocken State Road, West Conshohocken, PA, 19428-2998, USA
| | - Alessandra Dall'Agnese
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA, 02142, USA
| | - Paula Coutinho Toto
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Soda Balla Diop
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | | | - Peter D Adams
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- IRCCS Fondazione Santa Lucia, 00142, Rome, Italy
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
41
|
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T, Wang X. A Maternal High-Fat Diet Induces DNA Methylation Changes That Contribute to Glucose Intolerance in Offspring. Front Endocrinol (Lausanne) 2019; 10:871. [PMID: 31920981 PMCID: PMC6923194 DOI: 10.3389/fendo.2019.00871] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
Scope: Overnutrition in utero is a critical contributor to the susceptibility of diabetes by programming, although the exact mechanism is not clear. In this paper, we aimed to study the long-term effect of a maternal high-fat (HF) diet on offspring through epigenetic modifications. Procedures: Five-week-old female C57BL6/J mice were fed a HF diet or control diet for 4 weeks before mating and throughout gestation and lactation. At postnatal week 3, pups continued to consume a HF or switched to a control diet for 5 weeks, resulting in four groups of offspring differing by their maternal and postweaning diets. Results: The maternal HF diet combined with the offspring HF diet caused hyperglycemia and insulin resistance in male pups. Even after changing to the control diet, male pups exposed to the maternal HF diet still exhibited hyperglycemia and glucose intolerance. The livers of pups exposed to a maternal HF diet had a hypermethylated insulin receptor substrate 2 (Irs2) gene and a hypomethylated mitogen-activated protein kinase kinase 4 (Map2k4) gene. Correspondingly, the expression of the Irs2 gene decreased and that of Map2k4 increased in pups exposed to a maternal HF diet. Conclusion: Maternal overnutrition programs long-term epigenetic modifications, namely, Irs2 and Map2k4 gene methylation in the offspring liver, which in turn predisposes the offspring to diabetes later in life.
Collapse
|
42
|
Mata-Greenwood E, Huber HF, Li C, Nathanielsz PW. Role of pregnancy and obesity on vitamin D status, transport, and metabolism in baboons. Am J Physiol Endocrinol Metab 2019; 316:E63-E72. [PMID: 30398904 PMCID: PMC6417685 DOI: 10.1152/ajpendo.00208.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/25/2018] [Accepted: 10/12/2018] [Indexed: 12/17/2022]
Abstract
Human studies show that obesity is associated with vitamin D insufficiency, which contributes to obesity-related disorders. Our aim was to elucidate the regulation of vitamin D during pregnancy and obesity in a nonhuman primate species. We studied lean and obese nonpregnant and pregnant baboons. Plasma 25-hydroxy vitamin D (25-OH-D) and 1α,25-(OH)2-D metabolites were analyzed using ELISA. Vitamin D-related gene expression was studied in maternal kidney, liver, subcutaneous fat, and placental tissue using real-time PCR and immunoblotting. Pregnancy was associated with an increase in plasma bioactive vitamin D levels compared with nonpregnant baboons in both lean and obese groups. Pregnant baboons had lower renal 24-hydroxylase CYP24A1 protein and chromatin-bound vitamin D receptor (VDR) than nonpregnant baboons. In contrast, pregnancy upregulated the expression of CYP24A1 and VDR in subcutaneous adipose tissue. Obesity decreased vitamin D status in pregnant baboons (162 ± 17 vs. 235 ± 28 nM for 25-OH-D, 671 ± 12 vs. 710 ± 10 pM for 1α,25-(OH)2-D; obese vs. lean pregnant baboons, P < 0.05). Lower vitamin D status correlated with decreased maternal renal expression of the vitamin D transporter cubulin and the 1α-hydroxylase CYP27B1. Maternal obesity also induced placental downregulation of the transporter megalin (LRP2), CYP27B1, the 25-hydroxylase CYP2J2, and VDR. We conclude that baboons represent a novel species to evaluate vitamin D regulation. Both pregnancy and obesity altered vitamin D status. Obesity-induced downregulation of vitamin D transport and bioactivation genes are novel mechanisms of obesity-induced vitamin D regulation.
Collapse
Affiliation(s)
- Eugenia Mata-Greenwood
- Lawrence Longo Center for Perinatal Biology, School of Medicine, Loma Linda University , Loma Linda, California
| | - Hillary F Huber
- Department of Animal Science, University of Wyoming , Laramie Wyoming
| | - Cun Li
- Department of Animal Science, University of Wyoming , Laramie Wyoming
- Southwest National Primate Research Center, Texas Biomedical Research Institute , San Antonio, Texas
| | - Peter W Nathanielsz
- Department of Animal Science, University of Wyoming , Laramie Wyoming
- Southwest National Primate Research Center, Texas Biomedical Research Institute , San Antonio, Texas
| |
Collapse
|
43
|
Jevjdovic T, Dakic T, Kopanja S, Lakic I, Vujovic P, Jasnic N, Djordjevic J. Sex-Related Effects of Prenatal Stress on Region-Specific Expression of Monoamine Oxidase A and β Adrenergic Receptors in Rat Hearts. Arq Bras Cardiol 2018; 112:67-75. [PMID: 30569948 PMCID: PMC6317614 DOI: 10.5935/abc.20190001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/23/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Prenatal stress may increase risk of developing cardiovascular disorders in adulthood. The cardiotoxic effects of catecholamines are mediated via prolonged adrenergic receptor stimulation and increased oxidative stress upon their degradation by monoamine oxidase A (MAO-A). OBJECTIVES We investigated long-term effects of prenatal stress on β (1, 2, 3) adrenergic receptors and MAO-A gene expression in the hearts of adult rat offspring. METHODS Pregnant rats were exposed to unpredictable mild stress during the third week of gestation. RNA was isolated from left ventricular apex and base of adult offspring. Quantitative PCR was used to measure gene expression in collected ventricular tissue samples. The level of significance was set to p < 0.05. RESULTS β3 adrenergic receptor mRNA was undetectable in rat left ventricle. β1 adrenergic receptor was the predominantly expressed subtype at the apical and basal left ventricular myocardium in the control females. Male offspring from unstressed mothers displayed higher apical cardiac β1 than β2 adrenergic receptor mRNA levels. However, β1 and β2 adrenergic receptor mRNAs were similarly expressed at the ventricular basal myocardium in males. Unlike males, prenatally stressed females exhibited decreased β1 adrenergic receptor mRNA expression at the apical myocardium. Prenatal stress did not affect cardiac MAO-A gene expression. CONCLUSIONS Collectively, our results show that prenatal stress may have exerted region- and sex-specific β1 and β2 adrenergic receptor expression patterns within the left ventricle.
Collapse
Affiliation(s)
- Tanja Jevjdovic
- Faculty of Biology - University of Belgrade, Belgrado - Sérvia
| | - Tamara Dakic
- Faculty of Biology - University of Belgrade, Belgrado - Sérvia
| | - Sonja Kopanja
- Department of Pediatrics and Adolescent Medicine - Medical University of Vienna, Viena - Áustria
| | - Iva Lakic
- Faculty of Biology - University of Belgrade, Belgrado - Sérvia
| | - Predrag Vujovic
- Faculty of Biology - University of Belgrade, Belgrado - Sérvia
| | - Nebojsa Jasnic
- Faculty of Biology - University of Belgrade, Belgrado - Sérvia
| | | |
Collapse
|
44
|
Abstract
The causes of essential hypertension remain an enigma. Interactions between genetic and external factors are generally recognized to act as aetiological mechanisms that trigger the pathogenesis of high blood pressure. However, the questions of which genes and factors are involved, and when and where such interactions occur, remain unresolved. Emerging evidence indicates that the hypertensive response to pressor stimuli, like many other physiological and behavioural adaptations, can become sensitized to particular stimuli. Studies in animal models show that, similarly to other response systems controlled by the brain, hypertensive response sensitization (HTRS) is mediated by neuroplasticity. The brain circuitry involved in HTRS controls the sympathetic nervous system. This Review outlines evidence supporting the phenomenon of HTRS and describes the range of physiological and psychosocial stressors that can produce a sensitized hypertensive state. Also discussed are the cellular and molecular changes in the brain neural network controlling sympathetic tone involved in long-term storage of information relating to stressors, which could serve to maintain a sensitized state. Finally, this Review concludes with a discussion of why a sensitized hypertensive response might previously have been beneficial and increased biological fitness under some environmental conditions and why today it has become a health-related liability.
Collapse
Affiliation(s)
- Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA.
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, USA.
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA.
- The François M. Abboud Cardiovascular Center, Iowa City, IA, USA.
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- The François M. Abboud Cardiovascular Center, Iowa City, IA, USA
| |
Collapse
|
45
|
Sallam NA, Palmgren VAC, Singh RD, John CM, Thompson JA. Programming of Vascular Dysfunction in the Intrauterine Milieu of Diabetic Pregnancies. Int J Mol Sci 2018; 19:E3665. [PMID: 30463313 PMCID: PMC6275067 DOI: 10.3390/ijms19113665] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023] Open
Abstract
With the rising global tide of obesity, gestational diabetes mellitus (GDM) burgeoned into one of the most common antenatal disorders worldwide. Macrosomic babies born to diabetic mothers are more likely to develop risk factors for cardiovascular disease (CVD) before they reach adulthood. Rodent studies in offspring born to hyperglycemic pregnancies show vascular dysfunction characterized by impaired nitric oxide (NO)-mediated vasodilation and increased production of contractile prostanoids by cyclooxygenase 2 (COX-2). Vascular dysfunction is a key pathogenic event in the progression of diabetes-related vascular disease, primarily attributable to glucotoxicity. Therefore, glucose-induced vascular injury may stem directly from the hyperglycemic intrauterine environment of GDM pregnancy, as evinced by studies showing endothelial activation and inflammation at birth or in childhood in offspring born to GDM mothers. This review discusses potential mechanisms by which intrauterine hyperglycemia programs dysfunction in the developing vasculature.
Collapse
Affiliation(s)
- Nada A Sallam
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
- Children's Hospital Research Institute; University of Calgary, Calgary, AB T2N 4N1, Canada.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Victoria A C Palmgren
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
| | - Radha D Singh
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
- Children's Hospital Research Institute; University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Cini M John
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
| | - Jennifer A Thompson
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
- Children's Hospital Research Institute; University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
46
|
Cox LA, Olivier M, Spradling-Reeves K, Karere GM, Comuzzie AG, VandeBerg JL. Nonhuman Primates and Translational Research-Cardiovascular Disease. ILAR J 2018; 58:235-250. [PMID: 28985395 DOI: 10.1093/ilar/ilx025] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the United States. Human epidemiological studies provide challenges for understanding mechanisms that regulate initiation and progression of CVD due to variation in lifestyle, diet, and other environmental factors. Studies describing metabolic and physiologic aspects of CVD, and those investigating genetic and epigenetic mechanisms influencing CVD initiation and progression, have been conducted in multiple Old World nonhuman primate (NHP) species. Major advantages of NHPs as models for understanding CVD are their genetic, metabolic, and physiologic similarities with humans, and the ability to control diet, environment, and breeding. These NHP species are also genetically and phenotypically heterogeneous, providing opportunities to study gene by environment interactions that are not feasible in inbred animal models. Each Old World NHP species included in this review brings unique strengths as models to better understand human CVD. All develop CVD without genetic manipulation providing multiple models to discover genetic variants that influence CVD risk. In addition, as each of these NHP species age, their age-related comorbidities such as dyslipidemia and diabetes are accelerated proportionally 3 to 4 times faster than in humans.In this review, we discuss current CVD-related research in NHPs focusing on selected aspects of CVD for which nonprimate model organism studies have left gaps in our understanding of human disease. We include studies on current knowledge of genetics, epigenetics, calorie restriction, maternal calorie restriction and offspring health, maternal obesity and offspring health, nonalcoholic steatohepatitis and steatosis, Chagas disease, microbiome, stem cells, and prevention of CVD.
Collapse
Affiliation(s)
- Laura A Cox
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Michael Olivier
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | | | - Genesio M Karere
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - John L VandeBerg
- South Texas Diabetes and Obesity Center, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, Texas
| |
Collapse
|
47
|
Cerf ME. High Fat Programming and Cardiovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E86. [PMID: 30428585 PMCID: PMC6262472 DOI: 10.3390/medicina54050086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
Abstract
Programming is triggered through events during critical developmental phases that alter offspring health outcomes. High fat programming is defined as the maintenance on a high fat diet during fetal and/or early postnatal life that induces metabolic and physiological alterations that compromise health. The maternal nutritional status, including the dietary fatty acid composition, during gestation and/or lactation, are key determinants of fetal and postnatal development. A maternal high fat diet and obesity during gestation compromises the maternal metabolic state and, through high fat programming, presents an unfavorable intrauterine milieu for fetal growth and development thereby conferring adverse cardiac outcomes to offspring. Stressors on the heart, such as a maternal high fat diet and obesity, alter the expression of cardiac-specific factors that alter cardiac structure and function. The proper nutritional balance, including the fatty acid balance, particularly during developmental windows, are critical for maintaining cardiac structure, preserving cardiac function and enhancing the cardiac response to metabolic challenges.
Collapse
Affiliation(s)
- Marlon E Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg 7505, South Africa.
| |
Collapse
|
48
|
Wang Q, Zhu C, Sun M, Maimaiti R, Ford SP, Nathanielsz PW, Ren J, Guo W. Maternal obesity impairs fetal cardiomyocyte contractile function in sheep. FASEB J 2018; 33:2587-2598. [PMID: 30289749 DOI: 10.1096/fj.201800988r] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity is a major public health problem worldwide. In the United States, one-third of women of reproductive age are obese. Human studies show that maternal obesity (MO) predisposes offspring to cardiovascular disease. However, the underlying mechanisms remain unclear. Given the similarities between pregnancy in sheep and humans, we studied sheep to examine the impact of MO on fetal cardiomyocyte contractility at term. We observed that MO impaired cardiomyocyte contractility by reducing peak shortening and shortening/relengthening velocity, prolonging time to relengthening. MO disrupted Ca2+ homeostasis in fetal cardiomyocytes, increasing intracellular Ca2+ and inducing cellular Ca2+ insensitivity. The Ca2+-release channel was impaired, but Ca2+ uptake was unaffected by MO. The upstream kinases that phosphorylate the Ca2+-release channel-ryanodine receptor-2, PKA, and calmodulin-dependent protein kinase II-were activated in MO fetuses. Contractile dysfunction was associated with an increased ratio of myosin heavy chain (MHC)-β to MHC-α and upregulated cardiac troponin (cTn)-T and tropomyosin, as well as cTn-I phosphorylation. In summary, this is the first characterization of the effects of MO on fetal cardiomyocyte contractility. Our findings indicate that MO impairs fetal cardiomyocyte contractility through altered intracellular Ca2+ handling, overloading fetal cardiomyocyte intracellular Ca2+ and aberrant myofilament protein composition. These mechanisms may contribute to developmental programming by MO of offspring cardiac function and predisposition to later life cardiovascular disease in the offspring.-Wang, Q., Zhu, C., Sun, M., Maimaiti, R., Ford, S. P., Nathanielsz, P. W., Ren, J., Guo, W. Maternal obesity impairs fetal cardiomyocyte contractile function in sheep.
Collapse
Affiliation(s)
- Qiurong Wang
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Chaoqun Zhu
- Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Mingming Sun
- Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Rexiati Maimaiti
- Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Stephen P Ford
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Peter W Nathanielsz
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming, USA
| | - Wei Guo
- Center for the Study of Fetal Programming, University of Wyoming, Laramie, Wyoming, USA.,Animal Science Department, University of Wyoming, Laramie, Wyoming, USA; and.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
49
|
Beeson JH, Blackmore HL, Carr SK, Dearden L, Duque-Guimarães DE, Kusinski LC, Pantaleão LC, Pinnock AG, Aiken CE, Giussani DA, Fernandez-Twinn DS, Ozanne SE. Maternal exercise intervention in obese pregnancy improves the cardiovascular health of the adult male offspring. Mol Metab 2018; 16:35-44. [PMID: 30293577 PMCID: PMC6157615 DOI: 10.1016/j.molmet.2018.06.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Obesity during pregnancy is associated with an elevated risk of cardiovascular disease in the offspring. With increased numbers of women entering pregnancy overweight or obese, there is a requirement for targeted interventions to reduce disease risk in future generations. Using an established murine model of maternal obesity during pregnancy, we investigated if a treadmill exercise intervention in the mother could improve offspring cardiac health and explored potential underlying mechanisms. METHODS A 20-minute treadmill exercise intervention protocol was performed 5 days a week in diet-induced obese female C57BL/6 mice 1 week prior to, and up to E17 of pregnancy. All male offspring were weaned onto a control diet and studied at 8 weeks of age when their cardiovascular physiology was assessed by in vivo echocardiography and non-invasive tail cuff plethysmography. Cardiomyocyte cell area, re-expression of fetal genes and the expression of calcium handling and sympathetic activation proteins were determined. RESULTS At 8 weeks, there was no difference in bodyweight or fat mass between groups. Offspring of obese dams developed pathologic cardiac hypertrophy, hypertension and cardiac dysfunction characterized by reduced ejection fraction (p < 0.001). Maternal exercise prevented cardiac hypertrophy and dysfunction but failed to prevent hypertension. These offspring of exercised dams also had enhanced (p < 0.001) levels of calcium handling proteins and a sympathetic-activated inotropic response. CONCLUSIONS Exercise in obese pregnancy was beneficial to offspring cardiac function and structure but did not influence hypertension suggesting they are programmed by separate mechanistic pathways. These data suggest combination interventions in obese pregnancies will be required to improve all aspects of the cardiovascular health of the next generation.
Collapse
Affiliation(s)
- Jessica H Beeson
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Heather L Blackmore
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Sarah K Carr
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Laura Dearden
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Daniella E Duque-Guimarães
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Laura C Kusinski
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Lucas C Pantaleão
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Adele G Pinnock
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Catherine E Aiken
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK; Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital and NIHR Cambridge Comprehensive Biomedical Research Centre, Box 223, Cambridge, CB2 0SW, UK.
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, CB2 3EG, UK.
| | - Denise S Fernandez-Twinn
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Susan E Ozanne
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Level 4, Box 289, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
50
|
Ganguly A, Devaskar SU. High-fat diet affects pregestational adiposity and glucose tolerance perturbing gestational placental macronutrient transporters culminating in an obese offspring in wild-type and glucose transporter isoform 3 heterozygous null mice. J Nutr Biochem 2018; 62:192-201. [PMID: 30308381 DOI: 10.1016/j.jnutbio.2018.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 07/12/2018] [Accepted: 09/10/2018] [Indexed: 12/15/2022]
Abstract
We examined the effect of a high-fat diet (HFD) vs. control diet (CD) upon pregestational and gestational wild-type (wt) and glucose transporter (glut)3 heterozygous (glut3+/-) female mice and observed an increase in pregestational body weights, white adiposity (wt > glut3+/-), circulating cholesterol, and high-density lipoproteins, with glucose intolerance in both genotypes. The HFD-exposed offspring displayed reduced birth weight with catch up to CD-fed in wt vs. an increased birth weight persisting as such at weaning by day 21 in glut3+/- mice. To decipher the mechanism behind this genotype-specific difference in the HFD offspring's phenotype, we first examined placental macronutrient transporters and noted HFD-induced increase in CD36 in wt with no change in other FATPs, sodium-coupled neutral amino acid transporters and system L amino acid transporter in both genotypes. In contrast, while placental Glut1 increased in both the genotypes, only Glut3 increased in the glut3+/- genotype in response to HFD. Hence, we next assessed glut3+/- embryonic (ES) cells under differing stressors of low glucose, hypoxia and inhibition of oxidative phosphorylation. Reduced Glut3-mediated glucose uptake in glut3+/- vs. wt ES cells culminated in deficient growth. We conclude that maternal HFD affects the in utero growth potential of the offspring by altering placental CD36 and Glut1 concentrations. In contrast, a differential effect on placental Glut3 concentrations between glut3+/- and wt genotypes is evident, with an increase occurring in the glut3+/- genotype alone. Deficient Glut3 in ES cells interferes with glucose uptake, cell survival and growth being further exaggerated with low glucose, hypoxia and inhibition of oxidative phosphorylation.
Collapse
Affiliation(s)
- Amit Ganguly
- Department of Pediatrics, Division of Neonatology & Developmental Biology and Neonatal Research Center of the UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095
| | - Sherin U Devaskar
- Department of Pediatrics, Division of Neonatology & Developmental Biology and Neonatal Research Center of the UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095.
| |
Collapse
|