1
|
Li Y, Han Q, Liu Y, Yin J, Ma J. Role of the histone deacetylase family in lipid metabolism: Structural specificity and functional diversity. Pharmacol Res 2024; 210:107493. [PMID: 39491635 DOI: 10.1016/j.phrs.2024.107493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/23/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Lipids play crucial roles in signal transduction. Lipid metabolism is associated with several transcriptional regulators, including peroxisome proliferator activated receptor γ, sterol regulatory element-binding protein 1, and acetyl-CoA carboxylase. In recent years, increasing evidence has suggested that members of the histone deacetylase (HDAC) family play key roles in lipid metabolism. However, the mechanisms by which each member of this family regulates lipid metabolism remain unclear. This review discusses the latest research on the roles played by HDACs in fat metabolism. The role of HDACs in obesity, diabetes, and atherosclerosis has also been discussed. In addition, the interaction of HDACs with the gut microbiome and circadian rhythm has been reviewed, and the future development trend in HDACs has been predicted, which may potentiate therapeutic application of targeted HDACs in related metabolic diseases.
Collapse
Affiliation(s)
- Yunxia Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China
| | - Qi Han
- College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China
| | - Yuxin Liu
- College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China.
| | - Jie Ma
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| |
Collapse
|
2
|
Alexandre-Santos B, Reis GDS, Medeiros GR, Stockler-Pinto MB, Oliveira NSC, Miranda-Alves L, Nóbrega ACLD, Magliano DC, Frantz EDC. Bisphenol S exposure induces cardiac remodeling and aggravates high-fat diet-induced cardiac hypertrophy in mice. ENVIRONMENTAL RESEARCH 2024; 261:119781. [PMID: 39142458 DOI: 10.1016/j.envres.2024.119781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Bisphenol S (BPS) is widely used in the manufacture products and increase the risk of cardiovascular diseases. The effect of the association between obesity and BPS on cardiac outcomes is still unknown. Male C57BL/6 mice were divided into standard chow diet (SC; 15 kJ/g), standard chow diet + BPS (SCB), high-fat diet (HF; 21 kJ/g), and high-fat diet + BPS (HFB). Over 12 weeks, the groups were exposed to BPS through drinking water (dose: 25 μg/kg/day) and/or a HF diet. We evaluated: body mass (BM), total cholesterol, systolic blood pressure (SBP), left ventricle (LV) mass, and cardiac remodeling. In the SCB group, BM, total cholesterol, and SBP increase were augmented in relation to the SC group. In the HF and HFB groups, these parameters were higher than in the SC and SCB groups. Cardiac hypertrophy was evidenced by augmented LV mass and wall thickness, and ANP protein expression in all groups in comparison to the SC group. Only the HFB group had a thicker LV wall than SCB and HF groups, and increased cardiomyocyte area when compared with SC and SCB groups. Concerning cardiac fibrosis, SCB, HF, and HFB groups presented higher interstitial collagen area, TGFβ, and α-SMA protein expression than the SC group. Perivascular collagen area was increased only in the HF and HFB groups than SC group. Higher IL-6, TNFα, and CD11c protein expression in all groups than the SC group evidenced inflammation. All groups had elevated CD36 and PPARα protein expression in relation to the SC group, but only HF and HFB groups promoted cardiac steatosis with increased perilipin 5 protein expression than the SC group. BPS exposure alone promoted cardiac remodeling with pathological concentric hypertrophy, fibrosis, and inflammation. Diet-induced remodeling is aggravated when associated with BPS, with marked hypertrophy, alongside fibrosis, inflammation, and lipid accumulation.
Collapse
Affiliation(s)
- Beatriz Alexandre-Santos
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Guilherme Dos Santos Reis
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Gabriela Rodrigues Medeiros
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Milena Barcza Stockler-Pinto
- Research Center on Nutrigenetics and Nutrigenomics, Faculty of Nutrition, Fluminense Federal University, Niteroi, RJ, Brazil
| | | | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology, Institute of Biomedical Science, Federal University of Rio de Janeiro, RJ, Brazil
| | | | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Laboratory of Experimental Endocrinology, Institute of Biomedical Science, Federal University of Rio de Janeiro, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
3
|
Frigoli GF, Quadreli DH, Santos DPD, Costa IRD, Ferreira ARO, Peres MNC, Ribeiro MVG, Ceravolo GS, Mathias PC, Palma-Rigo K, Fernandes GSA. Low protein uptake during peripuberty impairs the testis, epididymis, and spermatozoa in pubertal and adult Wistar rats. J Dev Orig Health Dis 2024; 15:e23. [PMID: 39444313 DOI: 10.1017/s2040174424000308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Protein malnutrition during critical periods poses significant risks to reproductive health. Thus, this study aims to evaluate the immediate and delayed effects of a 30-day low-protein diet on the postnatal development of the male reproductive system. For so, male rats were fed a protein-deficient diet from postnatal day 30-60, followed by evaluations of testis, epididymis, and spermatozoa both at the end of the diet and after a 60-day recovery period. Testicular and epididymal weight was lowered in pubertal animals. Several histological alterations were found in the testis, such as acidophilic cells and vacuoles in the seminiferous epithelium, and sperm production was compromised. In the epididymis, the luminal compartment was diminished, and the stroma was enlarged both in the caput and cauda; in the cauda, the epithelial compartment was enlarged; the transit time of spermatozoa through this organ was diminished. Testosterone production was lowered. Spermatozoa's motility, mitochondrial activation, and acrosomal integrity were impaired, and several alterations in morphology were observed. After the recovery period, testicular and epididymal weight was restored. Tissue remodulation was observed in the epididymis, but the spermatozoa's transit time in this organ was not altered. Sperm and testosterone production, spermatozoa motility, mitochondrial activation, and acrosomal integrity were also restored. However, testicular histological alterations and spermatic morphological abnormalities were maintained in protein-restricted animals. Protein restriction during peripuberty impairs the reproductive maturation of pubertal Wistar rats, impairing testicular and epididymal function, with lasting effects even after dietary correction.
Collapse
Affiliation(s)
- Giovanna Fachetti Frigoli
- Laboratory of Toxicology and Metabolic Disturbs of Reproduction, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| | - Débora Hipólito Quadreli
- Laboratory of Toxicology and Metabolic Disturbs of Reproduction, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| | - Dayane Priscila Dos Santos
- Laboratory of Toxicology and Metabolic Disturbs of Reproduction, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| | - Ivana Regina da Costa
- Laboratory of Toxicology and Metabolic Disturbs of Reproduction, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Cell Biology and Genetics, Center of Biological Sciences, State University of Maringá, Maringá, Brazil
| | - Maria Natália Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Cell Biology and Genetics, Center of Biological Sciences, State University of Maringá, Maringá, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Cell Biology and Genetics, Center of Biological Sciences, State University of Maringá, Maringá, Brazil
| | - Graziela Scalianti Ceravolo
- Laboratory of Vascular Pharmacology, Department of Physiological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| | - Paulo Cezar Mathias
- Laboratory of Secretion Cell Biology, Department of Cell Biology and Genetics, Center of Biological Sciences, State University of Maringá, Maringá, Brazil
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Cell Biology and Genetics, Center of Biological Sciences, State University of Maringá, Maringá, Brazil
| | - Glaura Scantamburlo Alves Fernandes
- Laboratory of Toxicology and Metabolic Disturbs of Reproduction, General Biology Department, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| |
Collapse
|
4
|
Gonzalez K, Merlin AC, Roye E, Ju B, Lee Y, Chicco AJ, Chung E. Voluntary Wheel Running Reduces Cardiometabolic Risks in Female Offspring Exposed to Lifelong High-Fat, High-Sucrose Diet. Med Sci Sports Exerc 2024; 56:1378-1389. [PMID: 38595204 PMCID: PMC11250925 DOI: 10.1249/mss.0000000000003443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
PURPOSE Maternal and postnatal overnutrition has been linked to an increased risk of cardiometabolic diseases in offspring. This study investigated the impact of adult-onset voluntary wheel running to counteract cardiometabolic risks in female offspring exposed to a life-long high-fat, high-sucrose (HFHS) diet. METHODS Dams were fed either an HFHS or a low-fat, low-sucrose (LFLS) diet starting from 8 wk before pregnancy and continuing throughout gestation and lactation. Offspring followed their mothers' diets. At 15 wk of age, they were divided into sedentary (Sed) or voluntary wheel running (Ex) groups, resulting in four groups: LFLS/Sed ( n = 10), LFLS/Ex ( n = 5), HFHS/Sed ( n = 6), HFHS/Ex ( n = 5). Cardiac function was assessed at 25 wk, with tissue collection at 26 wk for mitochondrial respiratory function and protein analysis. Data were analyzed using two-way ANOVA. RESULTS Although maternal HFHS diet did not affect the offspring's body weight at weaning, continuous HFHS feeding postweaning resulted in increased body weight and adiposity, irrespective of the exercise regimen. HFHS/Sed offspring showed increased left ventricular wall thickness and elevated expression of enzymes involved in fatty acid transport (CD36, FABP3), lipogenesis (DGAT), glucose transport (GLUT4), oxidative stress (protein carbonyls, nitrotyrosine), and early senescence markers (p16, p21). Their cardiac mitochondria displayed lower oxidative phosphorylation (OXPHOS) efficiency and reduced expression of OXPHOS complexes and fatty acid metabolism enzymes (ACSL5, CPT1B). However, HFHS/Ex offspring mitigated these effects, aligning more with LFLS/Sed offspring. CONCLUSIONS Adult-onset voluntary wheel running effectively counteracts the detrimental cardiac effects of a lifelong HFHS diet, improving mitochondrial efficiency, reducing oxidative stress, and preventing early senescence. This underscores the significant role of physical activity in mitigating diet-induced cardiometabolic risks.
Collapse
Affiliation(s)
- Kassandra Gonzalez
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| | - Andrea Chiñas Merlin
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
- Biomedical Engineering, Tecnologico de Monterrey, Campus Monterrey, MEXICO
| | - Erin Roye
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| | - Beomsoo Ju
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, University of West Florida, Pensacola, FL
| | - Youngil Lee
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, University of West Florida, Pensacola, FL
| | - Adam J. Chicco
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Eunhee Chung
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, TX
| |
Collapse
|
5
|
Huang C, Liu Z, Chen M, Zhang H, Mo R, Chen R, Liu Y, Wang S, Xue Q. Up-regulation of BRD4 contributes to gestational diabetes mellitus-induced cardiac hypertrophy in offspring by promoting mitochondria dysfunction in sex-independent manner. Biochem Pharmacol 2024; 226:116387. [PMID: 38944397 DOI: 10.1016/j.bcp.2024.116387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/27/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024]
Abstract
Gestational diabetes mellitus (GDM) is associated with cardiovascular disease in postnatal life. The current study tested the hypothesis that GDM caused the cardiac hypertrophy in fetal (ED18.5), postnatal day 7 (PD7), postnatal day 21 (PD21) and postnatal day 90 (PD90) offspring by upregulation of BRD4 and mitochondrial dysfunction. Pregnant mice were divided into control and GDM groups. Hearts were isolated from ED18.5, PD7, PD21 and PD90. GDM increased the body weight (BW) and heart weight (HW) in ED18.5 and PD7, but not PD21 and PD90 offspring. However, HW/BW ratio was increased in all ages of GDM offspring compared to control group. Electron microscopy showed disorganized myofibrils, mitochondrial swelling, vacuolization, and cristae disorder in GDM offspring. GDM resulted in myocardial hypertrophy in offspring, which persisted from fetus to adult in a sex-independent manner. Echocardiography analysis revealed that GDM caused diastolic dysfunction, but had no effect on systolic function. Meanwhile, myocardial BRD4 was significantly upregulated in GDM offspring and BRD4 inhibition by JQ1 alleviated GDM-induced myocardial hypertrophy in offspring. Co-immunoprecipitation showed that BRD4 interacted with DRP1 and there was an increase of BRD4 and DRP1 interaction in GDM offspring. Furthermore, GDM caused the accumulation of damaged mitochondria in hearts from all ages of offspring, including mitochondrial fusion fission imbalance (upregulation of DRP1, and downregulation of MFN1, MFN2 and OPA1) and myocardial mitochondrial ROS accumulation, which was reversed by JQ1. These results suggested that the upregulation of BRD4 is involved in GDM-induced myocardial hypertrophy in the offspring through promoting mitochondrial damage in a gender-independent manner.
Collapse
Affiliation(s)
- Cailing Huang
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Zimo Liu
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Mei Chen
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Haichuan Zhang
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Ruyao Mo
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Renshan Chen
- Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, Guangdong, China
| | - Yinghua Liu
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shixiang Wang
- Department of Cardiology, the third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Qin Xue
- Department of Pharmacology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
6
|
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: 8] [Impact Index Per Article: 4.0] [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
|
7
|
Espírito-Santo DA, Cordeiro GS, Oliveira TWS, Santos LS, Silva RT, Costa CAS, Boaventura GT, Barreto-Medeiros JM. Exposure to a high-fat diet during intrauterine life and post-birth causes cardiac histomorphometric changes in rats: A systematic review. Life Sci 2022; 303:120658. [PMID: 35662554 DOI: 10.1016/j.lfs.2022.120658] [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: 03/07/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Abstract
Cardiac histomorphometric changes are conditions present as an adaptive response to increased cardiovascular demand, such as in obesity or the consumption of a high-fat diet. Epidemiologic studies show an increase in maternal obese individuals, with repercussions on offspring cardiovascular health. OBJECTIVE The goal of this study was to systematically review studies that evaluated cardiac histomorphometric changes in rodents exposed to a high-fat diet. DATA SOURCE PubMed, Embase, Science Direct, Web of Science and Lilacs. DATA EXTRACTION Animal species, percentage of dietary fat, period and time of exposure and main cardiac change results were extracted. DATA ANALYSIS A total of 1687 studies were found, and 20 met the inclusion criteria for this systematic review. A maternal high-fat diet was started 3 to 4 weeks before mating in most (70%) of the studies. Nutritional manipulation of offspring was initiated during pregnancy and maintained until the end of lactation in most (45%) of the studies. The fat percentage of high-fat diets ranged between 20% and 62%. The studies showed increases in cardiomyocytes, left ventricle size, and whole heart hypertrophy. Some studies showed increased thickness of the middle intima layer of the aorta and atherosclerosis. Studies that maintained a high-fat diet after the lactation period also showed an increase in cardiac hypertrophy. CONCLUSION Maternal exposure to a hyperlipidic diet in the fetal stages of cardiac development causes cardiac hypertrophy in offspring. The high variation in the dietary fat and the difference in the time and period of exposure of the offspring to the high - fat diet suggest the high degree of sensitivity of the cardiac structure.
Collapse
Affiliation(s)
- Djane A Espírito-Santo
- Department of Nutrition, Graduate Program of Food Nutrition and Health, Federal University of Bahia, Brazil.
| | - Gabriele S Cordeiro
- Department of Nutrition, Graduate Program of Food Nutrition and Health, Federal University of Bahia, Brazil
| | | | | | - Rafael T Silva
- Department of Nutrition, Graduate Program of Food Nutrition and Health, Federal University of Bahia, Brazil
| | - Carlos A S Costa
- Health Sciences Center, Federal University of Recôncavo Baiano, Santo Antonio de Jesus, Bahia, Brazil
| | - Gilson T Boaventura
- Department of Nutrition, Graduate Program of Food Nutrition and Health, Federal University of Bahia, Brazil
| | - Jairza M Barreto-Medeiros
- Department of Nutrition, Graduate Program of Food Nutrition and Health, Federal University of Bahia, Brazil
| |
Collapse
|
8
|
Ferreira ARO, Ribeiro MVG, Peres MNC, Piovan S, Gonçalves GD, Saavedra LPJ, Martins JNDL, Junior MDF, Cavalcante KVN, Lopes GKG, Carneiro M, Almeida DL, Gomes RM, Comar JF, Armitage JA, Mathias PCDF, Palma-Rigo K. Protein Restriction in the Peri-Pubertal Period Induces Autonomic Dysfunction and Cardiac and Vascular Structural Changes in Adult Rats. Front Physiol 2022; 13:840179. [PMID: 35574445 PMCID: PMC9095958 DOI: 10.3389/fphys.2022.840179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/22/2022] [Indexed: 11/30/2022] Open
Abstract
Perturbations to nutrition during critical periods are associated with changes in embryonic, fetal or postnatal developmental patterns that may render the offspring more likely to develop cardiovascular disease in later life. The aim of this study was to evaluate whether autonomic nervous system imbalance underpins in the long-term hypertension induced by dietary protein restriction during peri-pubertal period. Male Wistar rats were assigned to groups fed with a low protein (4% protein, LP) or control diet (20.5% protein; NP) during peri-puberty, from post-natal day (PN) 30 until PN60, and then all were returned to a normal protein diet until evaluation of cardiovascular and autonomic function at PN120. LP rats showed long-term increased mean arterial pressure (p = 0.002) and sympathetic arousal; increased power of the low frequency (LF) band of the arterial pressure spectral (p = 0.080) compared with NP animals. The depressor response to the ganglion blocker hexamethonium was increased in LP compared with control animals (p = 0.006). Pulse interval variability showed an increase in the LF band and LF/HF ratio (p = 0.062 and p = 0.048) in LP animals. The cardiac response to atenolol and/or methylatropine and the baroreflex sensitivity were similar between groups. LP animals showed ventricular hypertrophy (p = 0.044) and increased interstitial fibrosis (p = 0.028) compared with controls. Reduced protein carbonyls (PC) (p = 0.030) and catalase activity (p = 0.001) were observed in hearts from LP animals compared with control. In the brainstem, the levels of PC (p = 0.002) and the activity of superoxide dismutase and catalase (p = 0.044 and p = 0.012) were reduced in LP animals, while the levels of GSH and total glutathione were higher (p = 0.039 and p = 0.038) compared with NP animals. Protein restriction during peri-pubertal period leads to hypertension later in life accompanied by sustained sympathetic arousal, which may be associated with a disorganization of brain and cardiac redox state and structural cardiac alteration.
Collapse
Affiliation(s)
- Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Maria Natalia Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Silvano Piovan
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Géssica Dutra Gonçalves
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Juliana Nunes de Lima Martins
- Laboratory of Liver Metabolism and Radioisotopes, Department of Biochemistry, State University of Maringa, Maringa, Brazil
| | - Marcos Divino Ferreira Junior
- Laboratory of Endocrine Physiology and Metabolism, Department of Physiological Sciences, Federal University of Goias, Goiania, Brazil
| | - Keilah Valeria Naves Cavalcante
- Laboratory of Endocrine Physiology and Metabolism, Department of Physiological Sciences, Federal University of Goias, Goiania, Brazil
| | - Gabriel kian Guimarães Lopes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Mariane Carneiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Rodrigo Mello Gomes
- Laboratory of Endocrine Physiology and Metabolism, Department of Physiological Sciences, Federal University of Goias, Goiania, Brazil
| | - Jurandir Fernando Comar
- Laboratory of Liver Metabolism and Radioisotopes, Department of Biochemistry, State University of Maringa, Maringa, Brazil
| | | | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
- Adventist College of Parana, Ivatuba, Brazil
- *Correspondence: Kesia Palma-Rigo,
| |
Collapse
|
9
|
Hufnagel A, Dearden L, Fernandez-Twinn DS, Ozanne SE. Programming of cardiometabolic health: the role of maternal and fetal hyperinsulinaemia. J Endocrinol 2022; 253:R47-R63. [PMID: 35258482 PMCID: PMC9066586 DOI: 10.1530/joe-21-0332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022]
Abstract
Obesity and gestational diabetes during pregnancy have multiple short- and long-term consequences for both mother and child. One common feature of pregnancies complicated by maternal obesity and gestational diabetes is maternal hyperinsulinaemia, which has effects on the mother and her adaptation to pregnancy. Even though insulin does not cross the placenta insulin can act on the placenta as well affecting placental growth, angiogenesis and lipid metabolism. Obese and gestational diabetic pregnancies are often characterised by maternal hyperglycaemia resulting in exposure of the fetus to high levels of glucose, which freely crosses the placenta. This leads to stimulation of fetal ß-cells and insulin secretion in the fetus. Fetal hyperglycaemia/hyperinsulinaemia has been shown to cause multiple complications in fetal development, such as altered growth trajectories, impaired neuronal and cardiac development and early exhaustion of the pancreas. These changes could increase the susceptibility of the offspring to develop cardiometabolic diseases later in life. In this review, we aim to summarize and review the mechanisms by which maternal and fetal hyperinsulinaemia impact on (i) maternal health during pregnancy; (ii) placental and fetal development; (iii) offspring energy homeostasis and long-term cardiometabolic health; (iv) how interventions can alleviate these effects.
Collapse
Affiliation(s)
- Antonia Hufnagel
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Laura Dearden
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Level 4, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| |
Collapse
|
10
|
Ferreira LA, Ferreira-Junior MD, Amaral KDJV, Cavalcante KVN, Pontes CNR, Cristin L, Ribeiro DS, dos Santos BG, Xavier CH, Mathias PCDF, Andersen ML, Pedrino GR, de Castro CH, Mazaro-Costa R, Gomes RM. Maternal postnatal early overfeeding induces sex-related cardiac dysfunction and alters sexually hormones levels in young offspring. J Nutr Biochem 2022; 103:108969. [DOI: 10.1016/j.jnutbio.2022.108969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/08/2021] [Accepted: 01/31/2022] [Indexed: 12/18/2022]
|
11
|
Environmental Alterations during Embryonic Development: Studying the Impact of Stressors on Pluripotent Stem Cell-Derived Cardiomyocytes. Genes (Basel) 2021; 12:genes12101564. [PMID: 34680959 PMCID: PMC8536136 DOI: 10.3390/genes12101564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022] Open
Abstract
Non-communicable diseases (NCDs) sauch as diabetes, obesity and cardiovascular diseases are rising rapidly in all countries world-wide. Environmental maternal factors (e.g., diet, oxidative stress, drugs and many others), maternal illnesses and other stressors can predispose the newborn to develop diseases during different stages of life. The connection between environmental factors and NCDs was formulated by David Barker and colleagues as the Developmental Origins of Health and Disease (DOHaD) hypothesis. In this review, we describe the DOHaD concept and the effects of several environmental stressors on the health of the progeny, providing both animal and human evidence. We focus on cardiovascular diseases which represent the leading cause of death worldwide. The purpose of this review is to discuss how in vitro studies with pluripotent stem cells (PSCs), such as embryonic and induced pluripotent stem cells (ESC, iPSC), can underpin the research on non-genetic heart conditions. The PSCs could provide a tool to recapitulate aspects of embryonic development “in a dish”, studying the effects of environmental exposure during cardiomyocyte (CM) differentiation and maturation, establishing a link to molecular mechanism and epigenetics.
Collapse
|
12
|
Zhang J, Cao L, Tan Y, Zheng Y, Gui Y. N-acetylcysteine protects neonatal mice from ventricular hypertrophy induced by maternal obesity in a sex-specific manner. Biomed Pharmacother 2021; 133:110989. [PMID: 33378994 DOI: 10.1016/j.biopha.2020.110989] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Maternal obesity induces adverse cardiac programming in offspring, and effective interventions are needed to prevent cardiovascular ill-health. Herein we hypothesized that exposure to maternal obesogenic diet-induced obesity in mice results in left ventricular remodelling and hypertrophy in early childhood, and that maternal N-acetylcysteine (NAC) treatment alleviates these effects in a sex-dependent manner. METHODS AND RESULTS The maternal obesity was induced in mice by the consumption of a Western diet accompanied by a 20 % sucrose solution. To determine the effect of NAC on the cardiac outcomes induced by maternal obesity, obese dams were continuously exposed to the obesogenic diet, with or without the oral NAC treatment during pregnancy. Left ventricular remodelling and hypertrophy occurred as early as 7 days after birth in the male offspring of obese dams (O-OB) compared with controls (O-CO). An over-expression of key genes and markers related to cardiac fibrosis accompanied by more disorganized myofibrils was observed in the hearts of neonatal male O-OB mice. When we next evaluated the level of oxidative stress in the hearts of neonatal mice, the activity of enzymatic antioxidants declined and expression of NOX enzyme complex was up-regulated in O-OB offspring hearts, but was normal in the offspring of NAC treated mice (O-OB/NAC). Maternal obesity also activated cardiac Akt and mammalian target of rapamycin (mTOR) signalling in offspring, and NAC treatment restored offspring cardiac Akt-mTOR signalling to normal irrespective of sex. NAC treatment did not prevent cardiomyocyte hypertrophy but did alleviate increased heart weight, interventricular septal thickness, and collagen content in male O-OB/NAC pups. CONCLUSIONS Collectively, our results indicated that NAC blunted cardiac fibrosis and related ventricular hypertrophy of male neonatal offspring in the setting of maternal obesity, potentially acting by reducing oxidative stress. The present study provides a basis for investigating the role of NAC in nutrition-related cardiac programming.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Animal Nutritional Physiological Phenomena
- Animals
- Animals, Newborn
- Antioxidants/pharmacology
- Disease Models, Animal
- Female
- Fibrosis
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Male
- Maternal Nutritional Physiological Phenomena
- Mice, Inbred C57BL
- Obesity, Maternal/complications
- Obesity, Maternal/physiopathology
- Oxidative Stress/drug effects
- Pregnancy
- Prenatal Exposure Delayed Effects
- Sex Factors
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Jialing Zhang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China; MOH Key Laboratory of Neonatal Diseases at Children's Hospital, Fudan University, Shanghai, China
| | - Li Cao
- Ultrasound Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yanfeng Tan
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Yuanzheng Zheng
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China; MOH Key Laboratory of Neonatal Diseases at Children's Hospital, Fudan University, Shanghai, China
| | - Yonghao Gui
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, China; MOH Key Laboratory of Neonatal Diseases at Children's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Morales-Scholz MG, Swinton C, Murphy RM, Kowalski GM, Bruce CR, Howlett KF, Shaw CS. Autophagy is not involved in lipid accumulation and the development of insulin resistance in skeletal muscle. Biochem Biophys Res Commun 2021; 534:533-539. [PMID: 33261883 DOI: 10.1016/j.bbrc.2020.11.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To investigate the effect of high fat diet-induced insulin resistance on autophagy markers in the liver and skeletal muscle of mice in the fasted state and following an oral glucose bolus. METHODS Forty C57BL/6J male mice were fed either a high fat, high sucrose (HFSD, n = 20) or standard chow control (CON, n = 20) diet for 16 weeks. Upon trial completion, mice were gavaged with water or glucose and skeletal muscle and liver were collected 15 min post gavage. Protein abundance and gene expression of autophagy markers and activation of related signalling pathways were assessed. RESULTS Compared to CON, the HFSD intervention increased LC3B-II and p62/SQSTM1 protein abundance in the liver which is indicative of elevated autophagosome content via reduced clearance. These changes coincided with inhibitory autophagy signalling through elevated p-mTOR S2448 and p-ULK1S758. HFSD did not alter autophagy markers in skeletal muscle. Administration of an oral glucose bolus had no effect on autophagy markers or upstream signalling responses in either tissue regardless of diet. CONCLUSION HFSD induces tissue-specific autophagy impairments, with autophagosome accumulation indicating reduced lysosomal clearance in the liver. In contrast, autophagy markers were unchanged in skeletal muscle, indicating that autophagy is not involved in the development of skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- María G Morales-Scholz
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia
| | - Courtney Swinton
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, VIC, Australia
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia
| | - Clinton R Bruce
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia
| | - Christopher S Shaw
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, 3216, VIC, Australia.
| |
Collapse
|
14
|
Xue Q, Chen F, Zhang H, Liu Y, Chen P, Patterson AJ, Luo J. Maternal high-fat diet alters angiotensin II receptors and causes changes in fetal and neonatal rats†. Biol Reprod 2020; 100:1193-1203. [PMID: 30596890 DOI: 10.1093/biolre/ioy262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/07/2018] [Accepted: 12/24/2018] [Indexed: 01/01/2023] Open
Abstract
Maternal high-fat diet (HFD) during pregnancy is linked to cardiovascular diseases in postnatal life. The current study tested the hypothesis that maternal HFD causes myocardial changes through angiotensin II receptor (AGTR) expression modulation in fetal and neonatal rat hearts. The control group of pregnant rats was fed a normal diet and the treatment group of pregnant rats was on a HFD (60% kcal fat). Hearts were isolated from embryonic day 21 fetuses (E21) and postnatal day 7 pups (PD7). Maternal HFD decreased the body weight of the offspring in both E21 and PD7. The ratio of heart weight to body weight was increased in E21, but not PD7, when compared to the control group. Transmission electron microscopy revealed disorganized myofibrils and effacement of mitochondria cristae in the treatment group. Maternal HFD decreased S-phase and increased G1-phase of the cellular cycle for fetal and neonatal cardiac cells. Molecular markers of cardiac hypertrophy, such as Nppa and Myh7, were found to be increased in the treatment group. There was an associated increase in Agtr2 mRNA and protein, whereas Agtr1a mRNA and AGTR1 protein were decreased in HFD fetal and neonatal hearts. Furthermore, maternal HFD decreased glucocorticoid receptors (GRs) binding to glucocorticoid response elements at the Agtr1a and Agtr2 promoter, which correlated with downregulation of GR in fetal and neonatal hearts. These findings suggest that maternal HFD may promote premature termination of fetal and neonatal cardiomyocyte proliferation and compensatory hypertrophy through intrauterine modulation of AGTR1 and AGTR2 expression via GR dependent mechanism.
Collapse
Affiliation(s)
- Qin Xue
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Fangyuan Chen
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Haichuan Zhang
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yinghua Liu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Pinxian Chen
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Andrew J Patterson
- University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Jiandong Luo
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| |
Collapse
|
15
|
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: 5] [Impact Index Per Article: 1.0] [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
|
16
|
Krzeczkowski JE, Boylan K, Arbuckle TE, Muckle G, Poliakova N, Séguin JR, Favotto LA, Savoy C, Amani B, Mortaji N, Van Lieshout RJ. Maternal Pregnancy Diet Quality Is Directly Associated with Autonomic Nervous System Function in 6-Month-Old Offspring. J Nutr 2020; 150:267-275. [PMID: 31573610 DOI: 10.1093/jn/nxz228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/15/2019] [Accepted: 08/28/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Many pregnant women are consuming diets of poor overall quality. Although many studies have linked poor prenatal diet quality to an increased risk of specific diseases in offspring, it is not known if exposure to poor prenatal diet affects core neurophysiological regulatory systems in offspring known to lie upstream of multiple diseases. OBJECTIVE We aimed to examine the association between prenatal diet quality and autonomic nervous system (ANS) function in infants at 6 mo of age. METHODS Data from 400 women (aged >18 y, with uncomplicated pregnancies) and their infants participating in the Maternal-Infant Research on Environmental Chemicals-Infant Development cohort were used to investigate links between prenatal diet quality and infant ANS function at 6 mo of age. Prenatal diet quality was assessed using the Healthy Eating Index (2010), calculated from a validated FFQ completed by women during the first trimester. Infant ANS function was measured using 2 assessments of heart rate variability (HRV) including root mean square of successive differences (RMSSD) and SD of N-N intervals (SDNN). Associations were analyzed before and after adjustment for socioeconomic status, maternal depression symptoms, maternal cardiometabolic dysfunction, breastfeeding, and prenatal smoking. RESULTS Poorer prenatal diet quality was associated with lower infant HRV assessed using RMSSD (B: 0.07; 95% CI: 0.01, 0.13; R2 = 0.013) and SDNN (B: 0.18; 95% CI: 0.02, 0.35; R2 = 0.011). These associations remained significant after adjustment for confounding variables [RMSSD: B: 0.09; 95% CI: 0.003, 0.18; squared semipartial correlation (sp2) = 0.14 and SDNN B: 0.24; 95% CI: 0.0, 0.49; sp2 = 0.13]. CONCLUSIONS In a large cohort study, poorer prenatal diet quality was associated with lower offspring HRV, a marker of decreased capacity of the ANS to respond adaptively to challenge. Therefore, poor prenatal diet may play a significant role in the programming of multiple organ systems and could increase general susceptibility to disease in offspring.
Collapse
Affiliation(s)
- John E Krzeczkowski
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Khrista Boylan
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Tye E Arbuckle
- Population Studies Division, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Gina Muckle
- School of Psychology, Université Laval, Quebec City, Quebec, Canada.,Research Centre of CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Natalia Poliakova
- Population Health and Optimal Health Practices Research Branch, CHU de Québec Research Centre, Quebec City, Quebec, Canada
| | - Jean R Séguin
- Department of Psychiatry and Addiction, CHU Ste-Justine Research Centre, Montreal, Quebec, Canada
| | - Lindsay A Favotto
- Department of Health Research Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Calan Savoy
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Bahar Amani
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Neda Mortaji
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Ryan J Van Lieshout
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada.,Department of Health Research Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
17
|
Blin G, Liand M, Mauduit C, Chehade H, Benahmed M, Simeoni U, Siddeek B. Maternal Exposure to High-Fat Diet Induces Long-Term Derepressive Chromatin Marks in the Heart. Nutrients 2020; 12:E181. [PMID: 31936461 PMCID: PMC7019950 DOI: 10.3390/nu12010181] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/30/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Heart diseases are a leading cause of death. While the link between early exposure to nutritional excess and heart disease risk is clear, the molecular mechanisms involved are poorly understood. In the developmental programming field, increasing evidence is pointing out the critical role of epigenetic mechanisms. Among them, polycomb repressive complex 2 (PRC2) and DNA methylation play a critical role in heart development and pathogenesis. In this context, we aimed at evaluating the role of these epigenetic marks in the long-term cardiac alterations induced by early dietary challenge. Using a model of rats exposed to maternal high-fat diet during gestation and lactation, we evaluated cardiac alterations at adulthood. Expression levels of PRC2 components, its histone marks di- and trimethylated histone H3 (H3K27me2/3), associated histone mark (ubiquitinated histone H2A, H2AK119ub1) and target genes were measured by Western blot. Global DNA methylation level and DNA methyl transferase 3B (DNMT3B) protein levels were measured. Maternal high-fat diet decreased H3K27me3, H2Ak119ub1 and DNA methylation levels, down-regulated the enhancer of zeste homolog 2 (EZH2), and DNMT3B expression. The levels of the target genes, isl lim homeobox 1 (Isl1), six homeobox 1 (Six1) and mads box transcription enhancer factor 2, polypeptide C (Mef2c), involved in cardiac pathogenesis were up regulated. Overall, our data suggest that the programming of cardiac alterations by maternal exposure to high-fat diet involves the derepression of pro-fibrotic and pro-hypertrophic genes through the induction of EZH2 and DNMT3B deficiency.
Collapse
Affiliation(s)
- Guillaume Blin
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Marjorie Liand
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Claire Mauduit
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, 06204 Nice, France; (C.M.); (M.B.)
| | - Hassib Chehade
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Mohamed Benahmed
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, 06204 Nice, France; (C.M.); (M.B.)
| | - Umberto Simeoni
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Benazir Siddeek
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| |
Collapse
|
18
|
Mabasa L, Samodien E, Sangweni NF, Pheiffer C, Louw J, Johnson R. In Utero One-Carbon Metabolism Interplay and Metabolic Syndrome in Cardiovascular Disease Risk Reduction. Mol Nutr Food Res 2019; 64:e1900377. [PMID: 31408914 DOI: 10.1002/mnfr.201900377] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/13/2019] [Indexed: 12/16/2022]
Abstract
The maternal obesogenic environment plays a role in programing the susceptibility of the fetus to postnatal non-alcoholic fatty liver disease (NAFLD), a risk factor for cardiovascular disease (CVD). NAFLD is a multisystem disease that is characterized by hepatic fat accumulation due in part to dysregulated energy metabolism network through epigenetic mechanisms such as DNA methylation. DNA methylation affects fetal programing and disease risk via regulation of gene transcription; it is affected by methyl donor nutrients such as vitamin B12 , methionine, folic acid, vitamin B6 , and choline. Although several studies have documented the role of several maternal methyl donor nutrients on obesity-induced NAFLD in offspring, currently, data are lacking on its impact on CVD risk as an endpoint. The aim of this paper is to use current knowledge to construct a postulation for the potential role of a comprehensive gestational methyl donor nutrients supplementary approach on the susceptibility of offspring to developing metabolic-syndrome-related cardiovascular complications.
Collapse
Affiliation(s)
- Lawrence Mabasa
- South African Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Ebrahim Samodien
- South African Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Nonhlakanipho F Sangweni
- South African Medical Research Council, Tygerberg, Cape Town, South Africa.,Stellenbosch University, Tygerberg, South Africa
| | - Carmen Pheiffer
- South African Medical Research Council, Tygerberg, Cape Town, South Africa.,Stellenbosch University, Tygerberg, South Africa
| | - Johan Louw
- South African Medical Research Council, Tygerberg, Cape Town, South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Rabia Johnson
- South African Medical Research Council, Tygerberg, Cape Town, South Africa.,Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
19
|
Bagchi RA, Weeks KL. Histone deacetylases in cardiovascular and metabolic diseases. J Mol Cell Cardiol 2019; 130:151-159. [PMID: 30978343 DOI: 10.1016/j.yjmcc.2019.04.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) regulate gene transcription by catalyzing the removal of acetyl groups from key lysine residues in nucleosomal histones and via the recruitment of other epigenetic regulators to DNA promoter/enhancer regions. Over the past two decades, HDACs have been implicated in multiple processes pertinent to cardiovascular and metabolic diseases, including cardiac hypertrophy and remodeling, fibrosis, calcium handling, inflammation and energy metabolism. The development of small molecule HDAC inhibitors and genetically modified loss- and gain-of-function mouse models has allowed interrogation of the roles of specific HDAC isoforms in these processes. Isoform-selective HDAC inhibitors may prove to be powerful therapeutic agents for the treatment of cardiovascular diseases, obesity and diabetes.
Collapse
Affiliation(s)
- Rushita A Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
20
|
Long-term impact of maternal high-fat diet on offspring cardiac health: role of micro-RNA biogenesis. Cell Death Discov 2019; 5:71. [PMID: 30854230 PMCID: PMC6397280 DOI: 10.1038/s41420-019-0153-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/25/2022] Open
Abstract
Heart failure is a worldwide leading cause of death. Diet and obesity are particularly of high concern in heart disease etiology. Gravely, altered nutrition during developmental windows of vulnerability can have long-term impact on heart health; however, the underlying mechanisms are poorly understood. In the understanding of the initiation of chronic diseases related to developmental exposure to environmental challenges, deregulations in epigenetic mechanisms including micro-RNAs have been proposed as key events. In this context, we aimed at delineating the role of micro-RNAs in the programming of cardiac alterations induced by early developmental exposure to nutritional imbalance. To reach our aim, we developed a human relevant model of developmental exposure to nutritional imbalance by maternally exposing rat to high-fat diet during gestation and lactation. In this model, offspring exposed to maternal high-fat diet developed cardiac hypertrophy and increased extracellular matrix depot compared to those exposed to chow diet. Microarray approach performed on cardiac tissue allowed the identification of a micro-RNA subset which was down-regulated in high-fat diet-exposed animals and which were predicted to regulate transforming growth factor-beta (TGFβ)-mediated remodeling. As indicated by in vitro approaches and gene expression measurement in the heart of our animals, decrease in DiGeorge critical region 8 (DGCR8) expression, involved in micro-RNA biogenesis, seems to be a critical point in the alterations of the micro-RNA profile and the TGFβ-mediated remodeling induced by maternal exposure to high-fat diet. Finally, increasing DGCR8 activity and/or expression through hemin treatment in vitro revealed its potential in the rescue of the pro-fibrotic phenotype in cardiomyocytes driven by DGCR8 decrease. These findings suggest that cardiac alterations induced by maternal exposure to high-fat diet is related to abnormalities in TGFβ pathway and associated with down-regulated micro-RNA processing. Our study highlighted DGCR8 as a potential therapeutic target for heart diseases related to early exposure to dietary challenge.
Collapse
|