1
|
Jiang S, Qiu S, Mu Y, Liu C, Han Y, Jiang J, Wang Y. Puerarin reduces susceptibility to ventricular arrhythmias and inhibits ferroptosis via Sirt1/Nrf2 signaling in high-fat-diet rats. Free Radic Biol Med 2025; 227:472-484. [PMID: 39647799 DOI: 10.1016/j.freeradbiomed.2024.12.005] [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: 09/08/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
Obesity is a significant risk factor for cardiac arrhythmias, and the ferroptosis is closely related to cardiac arrhythmias. This study aimed to investigate whether puerarin (Pue), a natural isoflavone, could reduce the susceptibility to ventricular arrhythmias (VAs) associated with obesity and inhibit ferroptosis, with a particular focus on the Sirt1/Nrf2 signaling pathway. Male rats were randomly divided into three groups: normal chow diet (NC), high-fat diet (HFD), and HFD with Pue treatment (100mg/kg, HFD + Pue). After 16 weeks, electrophysiological, structural, and molecular analysis were performed. Compared to the NC group, HFD rats exhibited prolonged QT interval and Tpeak-Tend interval, amplified transmural dispersion of ventricular repolarization, and increased susceptibility to VAs. Pue treatment significantly ameliorated these electrophysiological abnormalities and reduced VAs susceptibility. HFD rats showed cardiac hypertrophy, fibrosis, and inflammation, which were alleviated by Pue application. Cardiac lipid peroxidation, iron deposition, mitochondrial abnormality, and ferroptosis marker induction were observed in HFD rats. Further, treatment with Pue improved these alterations. Additionally, molecular docking analysis confirmed the interaction of Pue with Sirt1 and Nrf2. Furthermore, Pue treatment upregulated Sirt1 and Nrf2 expression in HFD rats, thereby reducing reactive oxygen species (ROS) generation and ferroptosis. Moreover, Pue protected cardiomyocytes against palmitic acid (PA)-induced injury by inhibiting ferroptosis via the Sirt1/Nrf2 pathway in H9c2 cells. Overall, our study shows for the first time that Pue reduces susceptibility to VAs and inhibits ferroptosis in HFD rats by modulating the Sirt1/Nrf2 signaling pathway, offering a potential therapeutic strategy for obesity-related cardiac arrhythmias.
Collapse
MESH Headings
- Animals
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- Sirtuin 1/metabolism
- Sirtuin 1/genetics
- Diet, High-Fat/adverse effects
- Ferroptosis/drug effects
- Isoflavones/pharmacology
- Rats
- Male
- Arrhythmias, Cardiac/drug therapy
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/prevention & control
- Signal Transduction/drug effects
- Rats, Sprague-Dawley
- Obesity/complications
- Obesity/drug therapy
- Obesity/pathology
- Molecular Docking Simulation
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
Collapse
Affiliation(s)
- Shuang Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Suhua Qiu
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yu Mu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Chilu Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China.
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China.
| |
Collapse
|
2
|
Xiao W, Han P, Song L, Yang J, Zhou L, Deng X, Ma ZJ, Lang Y, Zhao H, Zhao Y, Chen H, Zhang W, Huang H, Sun N. A Retrospective Study to Investigate the Relationship Between Body Mass Index and Hemodynamic Characteristics in Hypertensive Patients. J Clin Hypertens (Greenwich) 2025; 27:e14946. [PMID: 39654491 PMCID: PMC11773678 DOI: 10.1111/jch.14946] [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: 07/21/2024] [Revised: 10/13/2024] [Accepted: 11/06/2024] [Indexed: 01/29/2025]
Abstract
The relationship between hemodynamic parameters and body mass index (BMI) in the context of blood pressure regulation in hypertension is unclear. Here, we analyzed data from 1368 uncontrolled hypertensive patients, including key hemodynamic indicators such as heart rate (HR), cardiac index (CI), arterial stiffness (AS), systemic vascular resistance index (SVRI), and thoracic blood ratio (TBR). In this cohort, the average BMI across all hypertension patients was 26.859 ± 3.897 (kg/m2), with obese patients (BMI ≥ 28 kg/m2) averaging 31.01 ± 2.87 kg/m2 and non-obese (BMI<28 kg/m2) averaging 24.70 ± 2.28 kg/m2. Younger obese patients exhibited higher diastolic pressures than non-obese peers (p < 0.01). Hemodynamic analysis showed obese patients had increased HR and SVRI but lower CI and AS (p < 0.01). Hypertensive males aged under 60 with obesity displayed a more prominent peripheral vascular phenotype (p < 0.05) and volemic phenotype (p < 0.01) than non-obese males. Obese females aged under 60 showed a higher incidence of cardiac phenotype (p < 0.01). Across genders, obese hypertensive patients aged over 60 had a greater prevalence of volemic phenotype than non-obese patients (p < 0.05, p < 0.01, respectively). BMI inversely correlated with CI and positively with SVRI across age and gender categories (p < 0.01). Taken together, we find that patients with hypertension exhibit diverse hemodynamic profiles, and BMI significantly correlates with hemodynamic parameters such as SVRI and CI. Our research identifies BMI as a valuable target for personalized hypertension treatment.
Collapse
Affiliation(s)
- Wenqi Xiao
- Department of CardiologyBeijing Haidian HospitalBeijingChina
| | - Ping Han
- Department of CardiologyBeijing Haidian HospitalBeijingChina
| | - Liping Song
- Department of CardiologyBeijing Haidian HospitalBeijingChina
| | - Jingwen Yang
- Department of CardiologyBeijing Haidian HospitalBeijingChina
| | - Lin Zhou
- Department of CardiologyBeijing Haidian HospitalBeijingChina
| | - Xiangning Deng
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Zheng J. Ma
- SJTU‐Yale Joint Center for BiostatisticsShanghai Jiao Tong UniversityShanghaiChina
- Department of Research & DevelopmentBeijing Li‐Heng Medical Technologies, LtdBeijingChina
| | - Yukun Lang
- Qinglongqiao Community Service CenterBeijingChina
| | - Hai Zhao
- Shuangyushu Community Health Service CenterBeijingChina
| | - Yuzhuo Zhao
- Qingmao Community Health Service StationBeijingChina
| | - Hui Chen
- Wenquan Town Community Health Service CenterBeijingChina
| | - Wenjing Zhang
- Haidian Town Community Health Service CenterBeijingChina
| | - Huixian Huang
- Department of CardiologyBeijing Haidian HospitalBeijingChina
| | - Ningling Sun
- Department of CardiologyBeijing Haidian HospitalBeijingChina
- Department of HypertensionPeking University People's HospitalBeijingChina
| |
Collapse
|
3
|
Janssens JV, Raaijmakers AJA, Koutsifeli P, Weeks KL, Bell JR, Van Eyk JE, Curl CL, Mellor KM, Delbridge LMD. Mechanical loading reveals an intrinsic cardiomyocyte stiffness contribution to diastolic dysfunction in murine cardiometabolic disease. J Physiol 2024; 602:6705-6727. [PMID: 39629708 DOI: 10.1113/jp286437] [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: 04/18/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Cardiometabolic syndromes including diabetes and obesity are associated with occurrence of heart failure with diastolic dysfunction. There are no specific treatments for diastolic dysfunction, and therapies to manage symptoms have limited efficacy. Understanding of the cardiomyocyte origins of diastolic dysfunction is an important priority to identify new therapeutics. The investigative goal was to experimentally define in vitro stiffness properties of isolated cardiomyocytes derived from rodent hearts exhibiting diastolic dysfunction in vivo in response to dietary induction of cardiometabolic disease. Male mice fed a high fat/sugar diet (HFSD vs. control) exhibited diastolic dysfunction (echo E/e' Doppler ratio). Intact paced cardiomyocytes were functionally investigated in three conditions: non-loaded, loaded and stretched. Mean stiffness of HFSD cardiomyocytes was 70% higher than control. E/e' for the HFSD hearts was elevated by 35%. A significant relationship was identified between in vitro cardiomyocyte stiffness and in vivo dysfunction severity. With conversion from the non-loaded to loaded condition, the decrement in maximal sarcomere lengthening rate was more accentuated in HFSD cardiomyocytes (vs. control). With stretch, the Ca2+ transient decay time course was prolonged. With increased pacing, cardiomyocyte stiffness was elevated, yet diastolic Ca2+ elevation was attenuated. Our findings show unequivocally that cardiomyocyte mechanical dysfunction cannot be detected by analysis of non-loaded shortening. Collectively, these findings demonstrate that a component of cardiac diastolic dysfunction in cardiometabolic disease is derived from cardiomyocyte stiffness. Differential responses to load, stretch and pacing suggest that a previously undescribed alteration in myofilament-Ca2+ interaction contributes to intrinsic cardiomyocyte stiffness in cardiometabolic disease. KEY POINTS: Understanding cardiomyocyte stiffness components is an important priority for identifying new therapeutics for diastolic dysfunction, a key feature of cardiometabolic disease. In this study cardiac function was measured in vivo (echocardiography) for mice fed a high-fat/sugar diet (HFSD, ≥25 weeks). Performance of intact isolated cardiomyocytes derived from the same hearts was measured during pacing under non-loaded, loaded and stretched conditions in vitro. Calibrated cardiomyocyte stretches demonstrated that stiffness (stress/strain) was elevated in HFSD cardiomyocytes in vitro and correlated with diastolic dysfunction (E/e') in vivo. HFSD cardiomyocyte Ca2+ transient decay was prolonged in response to stretch. Stiffness was accentuated with pacing increase while the elevation in diastolic Ca2+ was attenuated. Data show unequivocally that cardiomyocyte mechanical dysfunction cannot be detected by analysis of non-loaded shortening. These findings suggest that stretch-dependent augmentation of the myofilament-Ca2+ response during diastole partially underlies elevated cardiomyocyte stiffness and diastolic dysfunction of hearts of animals with cardiometabolic disease.
Collapse
Affiliation(s)
- Johannes V Janssens
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Parisa Koutsifeli
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Kate L Weeks
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiometabolic Health (Baker), University of Melbourne, Melbourne, Australia
| | - James R Bell
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, La Trobe University, Melbourne, Australia
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Claire L Curl
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
| | - Kimberley M Mellor
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
- Auckland Bioengineering Institute, University of Auckland, New Zealand
- Department of Physiology, University of Auckland, New Zealand
| | - Lea M D Delbridge
- Department of Anatomy & Physiology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Janssens JV, Raaijmakers AJA, Koutsifeli P, Weeks KL, Bell JR, Van Eyk JE, Curl CL, Mellor KM, Delbridge LMD. Mechanical loading reveals an intrinsic cardiomyocyte stiffness contribution to diastolic dysfunction in murine cardiometabolic disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581448. [PMID: 38659933 PMCID: PMC11042179 DOI: 10.1101/2024.02.21.581448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cardiometabolic syndromes including diabetes and obesity are associated with occurrence of heart failure with diastolic dysfunction. There are no specific treatments for diastolic dysfunction, and therapies to manage symptoms have limited efficacy. Understanding of the cardiomyocyte origins of diastolic dysfunction is an important priority to identify new therapeutics. The investigative goal was to experimentally define in vitro stiffness properties of isolated cardiomyocytes derived from rodent hearts exhibiting diastolic dysfunction in vivo in response to dietary induction of cardiometabolic disease. Male mice fed a high fat/sugar diet (HFSD vs control) exhibited diastolic dysfunction (echo E/e' doppler ratio). Intact paced cardiomyocytes were functionally investigated in three conditions: non-loaded, loaded and stretched. Mean stiffness of HFSD cardiomyocytes was 70% higher than control. E/e' for the origin hearts was elevated by 35%. A significant relationship was identified between in vitro cardiomyocyte stiffness and in vivo dysfunction severity. With conversion from non-loaded to loaded condition, the decrement in maximal sarcomere lengthening rate was more accentuated in HFSD cardiomyocytes (vs control). With stretch, the Ca2+ transient decay time course was prolonged. With increased pacing, cardiomyocyte stiffness was elevated, yet diastolic Ca2+ elevation was attenuated. Our findings show unequivocally that cardiomyocyte mechanical dysfunction cannot be detected by analysis of non-loaded shortening. Collectively, these findings demonstrate that a component of cardiac diastolic dysfunction in cardiometabolic disease is derived from cardiomyocyte stiffness. Differential responses to load, stretch and pacing suggest that a previously undescribed alteration in myofilament-Ca2+ interaction contributes to intrinsic cardiomyocyte stiffness in cardiometabolic disease.
Collapse
|
5
|
Plawecki M, Gayrard N, Jeanson L, Chauvin A, Lajoix AD, Cristol JP, Jover B, Raynaud F. Cardiac remodeling associated with chronic kidney disease is enhanced in a rat model of metabolic syndrome: Preparation of mesenchymal transition. Mol Cell Biochem 2024; 479:29-39. [PMID: 36976428 DOI: 10.1007/s11010-023-04710-6] [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: 12/12/2022] [Accepted: 03/13/2023] [Indexed: 03/29/2023]
Abstract
Cardiac alteration due to chronic kidney disease is described by tissue fibrosis. This remodeling involves myofibroblasts of various origins, including epithelial or endothelial to mesenchymal transitions. In addition, obesity and insulin resistance together or separately seem to exacerbate cardiovascular risk in chronic kidney disease (CKD). The main objective of this study was to assess if pre-existing metabolic disease exacerbates CKD-induced cardiac alterations. In addition, we hypothesised that endothelial to mesenchymal transition participates in this enhancement of cardiac fibrosis. Rats fed cafeteria type diet for 6 months underwent a subtotal nephrectomy at 4 months. Cardiac fibrosis was evaluated by histology and qRT-PCR. Collagens and macrophages were quantified by immunohistochemistry. Endothelial to mesenchymal transitions were assessed by qRT-PCR (CD31, VE-cadherin, α-SMA, nestin) and also by CD31 immunofluorescence staining. Rats fed with cafeteria type regimen were obese, hypertensive and insulin resistant. Cardiac fibrosis was predominant in CKD rats and was highly majored by cafeteria regimen. Collagen-1 and nestin expressions were higher in CKD rats, independently of regimen. Interestingly, in rats with CKD and cafeteria diet we found an increase of CD31 and α-SMA co-staining with suggest an implication of endothelial to mesenchymal transition during heart fibrosis. We showed that rats already obese and insulin resistant had an enhanced cardiac alteration to a subsequent renal injury. Cardiac fibrosis process could be supported by a involvement of the endothelial to mesenchymal transition phenomenon.
Collapse
Affiliation(s)
- Maëlle Plawecki
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
- Laboratoire de Biochimie et d'hormonologie, CHU Lapeyronie, Montpellier, France
| | | | - Laura Jeanson
- BC2M, Université de Montpellier, Montpellier, France
| | - Anthony Chauvin
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | | | - Jean-Paul Cristol
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
- Laboratoire de Biochimie et d'hormonologie, CHU Lapeyronie, Montpellier, France
| | - Bernard Jover
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Fabrice Raynaud
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
6
|
Mao Y, Zhao K, Li P, Sheng Y. The emerging role of leptin in obesity-associated cardiac fibrosis: evidence and mechanism. Mol Cell Biochem 2022; 478:991-1011. [PMID: 36214893 DOI: 10.1007/s11010-022-04562-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022]
Abstract
Cardiac fibrosis is a hallmark of various cardiovascular diseases, which is quite commonly found in obesity, and may contribute to the increased incidence of heart failure arrhythmias, and sudden cardiac death in obese populations. As an endogenous regulator of adiposity metabolism, body mass, and energy balance, obesity, characterized by increased circulating levels of the adipocyte-derived hormone leptin, is a critical contributor to the pathogenesis of cardiac fibrosis. Although there are some gaps in our knowledge linking leptin and cardiac fibrosis, this review will focus on the interplay between leptin and major effectors involved in the pathogenesis underlying cardiac fibrosis at both cellular and molecular levels based on the current reports. The profibrotic effect of leptin is predominantly mediated by activated cardiac fibroblasts but may also involve cardiomyocytes, endothelial cells, and immune cells. Moreover, a series of molecular signals with a known profibrotic property is closely involved in leptin-induced fibrotic events. A more comprehensive understanding of the underlying mechanisms through which leptin contributes to the pathogenesis of cardiac fibrosis may open up a new avenue for the rapid emergence of a novel therapy for preventing or even reversing obesity-associated cardiac fibrosis.
Collapse
Affiliation(s)
- Yukang Mao
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China. .,Department of Cardiology, Jiangsu Province Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
7
|
Wingard MC, Dalal S, Shook PL, Ramirez P, Raza MU, Johnson P, Connelly BA, Thewke D, Singh M, Singh K. Deficiency of ataxia-telangiectasia mutated kinase attenuates Western-type diet-induced cardiac dysfunction in female mice. Physiol Rep 2022; 10:e15434. [PMID: 36117462 PMCID: PMC9483716 DOI: 10.14814/phy2.15434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
Chronic consumption of Western-type diet (WD) induces cardiac structural and functional abnormalities. Previously, we have shown that WD consumption in male ATM (ataxia-telangiectasia mutated kinase) deficient mice associates with accelerated body weight (BW) gain, cardiac systolic dysfunction with increased preload, and exacerbation of hypertrophy, apoptosis, and inflammation. This study investigated the role of ATM deficiency in WD-induced changes in functional and biochemical parameters of the heart in female mice. Six-week-old wild-type (WT) and ATM heterozygous knockout (hKO) female mice were placed on WD or NC (normal chow) for 14 weeks. BW gain, fat accumulation, and cardiac functional and biochemical parameters were measured 14 weeks post-WD. WD-induced subcutaneous and total fat contents normalized to body weight were higher in WT-WD versus hKO-WD. Heart function measured using echocardiography revealed decreased percent fractional shortening and ejection fraction, and increased LV end systolic diameter and volume in WT-WD versus WT-NC. These functional parameters remained unchanged in hKO-WD versus hKO-NC. Myocardial fibrosis, myocyte hypertrophy, and apoptosis were higher in WT-WD versus WT-NC. However, apoptosis was significantly lower and hypertrophy was significantly higher in hKO-WD versus WT-WD. MMP-9 and Bax expression, and Akt activation were higher in WT-WD versus WT-NC. PARP-1 (full-length) expression and mTOR activation were lower in WT-WD versus hKO-WD. Thus, ATM deficiency in female mice attenuates fat weight gain, preserves heart function, and associates with decreased cardiac cell apoptosis in response to WD.
Collapse
Affiliation(s)
- Mary C. Wingard
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Suman Dalal
- Department of Health SciencesEast Tennessee State UniversityJohnson CityTennesseeUSA
- Center of Excellence in Inflammation, Infectious Disease and ImmunityJohnson CityTennesseeUSA
| | - Paige L. Shook
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Paulina Ramirez
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Muhammad U. Raza
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Patrick Johnson
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Barbara A. Connelly
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
- Research and Development ServiceJames H Quillen Veterans Affairs Medical CenterMountain HomeTennesseeUSA
| | - Douglas P. Thewke
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Mahipal Singh
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
| | - Krishna Singh
- Department of Biomedical SciencesJames H Quillen College of Medicine, East Tennessee State UniversityJohnson CityTennesseeUSA
- Center of Excellence in Inflammation, Infectious Disease and ImmunityJohnson CityTennesseeUSA
- Research and Development ServiceJames H Quillen Veterans Affairs Medical CenterMountain HomeTennesseeUSA
| |
Collapse
|
8
|
Larson KF, Malik A, Brozovich FV. Aging and Heart Failure with Preserved Ejection Fraction. Compr Physiol 2022; 12:3813-3822. [PMID: 35950652 DOI: 10.1002/cphy.c210035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Heart failure is a clinical syndrome characterized by the inability of the cardiovascular system to provide adequate cardiac output at normal filling pressures. This results in a clinical syndrome characterized by dyspnea, edema, and decreased exertional tolerance. Heart failure with preserved ejection fraction (HFpEF) is an increasingly common disease, and the incidence of HFpEF increases with age. There are a variety of factors which contribute to the development of HFpEF, including the presence of hypertension, diabetes, obesity, and other pro-inflammatory states. These comorbid conditions result in changes at the biochemical and cell signaling level which ultimately lead to a disease with a great deal of phenotypic heterogeneity. In general, the physiologic dysfunction of HFpEF is characterized by vascular stiffness, increased cardiac filling pressures, pulmonary hypertension, and impaired volume management. The normal and abnormal processes associated with aging serve as an accelerant in this process, resulting in the hypothesis that HFpEF represents a form of presbycardia. In this article, we aim to review the processes importance of aging in the development of HFpEF by examining the disease and its causes from the biochemical to physiologic level. © 2022 American Physiological Society. Compr Physiol 12: 1-10, 2022.
Collapse
Affiliation(s)
- Kathryn F Larson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Awais Malik
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Frank V Brozovich
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Physiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Li Y, Li T, Zhou Z, Xiao Y. Emerging roles of Galectin-3 in diabetes and diabetes complications: A snapshot. Rev Endocr Metab Disord 2022; 23:569-577. [PMID: 35083706 PMCID: PMC9156459 DOI: 10.1007/s11154-021-09704-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Galectin-3 is a member of the galectin family, widely expressed in immune cells and plays a role mainly in inflammation, autoimmunity, apoptosis, and chemotaxis. We summarized the roles of Galectin-3 in diabetes and its complications, as well as the underlying mechanisms. Clinical research has determined that the circulating level of Galectin-3 is closely related to diabetes and its complications, thus it is promising to use Galectin-3 as a predictor and biomarker for those diseases. Galectin-3 also may be considered as an ideal therapeutic target, which has broad prospects in the prevention and treatment of diabetes and its complications, especially macrovascular and microvascular complications.
Collapse
Affiliation(s)
- Yanhua Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, No. 139, Renmin Rd, Changsha, 410011, China
- Department of Metabolism and Endocrinology, The Third Hospital of Changsha, 176, West Labour Road, Changsha, 410011, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Rd, Xi'an, 710032, China.
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, No. 139, Renmin Rd, Changsha, 410011, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, No. 139, Renmin Rd, Changsha, 410011, China.
| |
Collapse
|
10
|
Raggi F, Rossi C, Faita F, Distaso M, Kusmic C, Solini A. P2X7 Receptor and Heart Function in a Mouse Model of Systemic Inflammation Due to High Fat Diet. J Inflamm Res 2022; 15:2425-2439. [PMID: 35444452 PMCID: PMC9015053 DOI: 10.2147/jir.s356038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/09/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Low-grade inflammation contributes to heart failure in obesity or type 2 diabetes mellitus. The P2X7 receptor (P2X7R) is a key regulator of several pro-inflammatory responses in multiple tissues and organs; however, its involvement in the onset of heart dysfunction remains unclear. The study evaluated the role of P2X7R as a cardiac function regulator in C57BL/6J wild-type (WT) and P2X7R knockout (KO) mice by inducing systemic inflammation with high fat diet (HFD). Methods Specific parameters of systolic and diastolic function and heart morphology were measured in vivo before animal sacrifice by high-frequency ultrasonographic analysis. Gene and protein expression of cardiac biomarkers associated with inflammatory-oxidative pathways were evaluated by real-time PCR and Western Blotting. Results P2X7R-mediated up-regulation of the NLRP3-caspase-1 complex, increased expression of key oxidative stress (NOS-2, TNFα), and chemotactic (MCP-1) mediators were revealed in WT-HFD animals. In KO-HFD mice, such inflammatory-oxidative pathway was silent. Nevertheless, HFD induced in vivo a clear alteration of diastolic pattern (E/A: p < 0.03 vs WT-HFD) and a cardiac morphologic remodelling (left ventricular mass: p < 0.05 vs WT-HFD) only in P2X7R KO animals. Surprisingly, the transcriptional and protein expression of IL-1β and IL-6, usually regulated through P2X7R activation, were significantly higher in KO-HFD than in WT-HFD mice (both p < 0.05). Furthermore, an up-regulation of miR-214 and a down-regulation of miR-126 in heart of HFD-KO mice were observed, suggesting a link between such epigenetic dysregulation and cytokine overexpression as a potential pathophysiologic mechanism concurring to the progressive cardiac dysfunction. Conclusion These findings seem to suggest a cardioprotective role of P2X7R toward this tissue-specific inflammatory damage, likely through tissue homeostasis and organ functionality preservation.
Collapse
Affiliation(s)
- Francesco Raggi
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Chiara Rossi
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Francesco Faita
- Institute of Clinical Physiology, Italian National Research Council, Pisa, Italy
| | - Mariarosaria Distaso
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Claudia Kusmic
- Institute of Clinical Physiology, Italian National Research Council, Pisa, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
11
|
Remme CA. Sudden cardiac death in diabetes and obesity: mechanisms and therapeutic strategies. Can J Cardiol 2022; 38:418-426. [PMID: 35017043 DOI: 10.1016/j.cjca.2022.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/07/2023] Open
Abstract
Ventricular arrhythmias and sudden cardiac death (SCD) occur most frequently in the setting of coronary artery disease, cardiomyopathy and heart failure, but are also increasingly observed in individuals suffering from diabetes mellitus and obesity. The incidence of these metabolic disorders is rising in Western countries, but adequate prevention and treatment of arrhythmias and SCD in affected patients is limited due to our incomplete knowledge of the underlying disease mechanisms. Here, an overview is presented of the prevalence of electrophysiological disturbances, ventricular arrhythmias and SCD in the clinical setting of diabetes and obesity. Experimental studies are reviewed, which have identified disease pathways and associated modulatory factors, in addition to pro-arrhythmic mechanisms. Key processes are discussed, including mitochondrial dysfunction, oxidative stress, cardiac structural derangements, abnormal cardiac conduction, ion channel dysfunction, prolonged repolarization and dysregulation of intracellular sodium and calcium homeostasis. In addition, the recently identified pro-arrhythmic effects of dysregulated branched chain amino acid metabolism, a common feature in patients with metabolic disorders, are addressed. Finally, current management options are discussed, in addition to the potential development of novel preventive and therapeutic strategies based on recent insight gained from translational studies.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Vileigas DF, de Souza SLB, Corrêa CR, Silva CCVDA, de Campos DHS, Padovani CR, Cicogna AC. The effects of two types of Western diet on the induction of metabolic syndrome and cardiac remodeling in obese rats. J Nutr Biochem 2021; 92:108625. [PMID: 33705955 DOI: 10.1016/j.jnutbio.2021.108625] [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: 04/14/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022]
Abstract
Metabolic syndrome (MetS) include obesity as a critical feature and is strongly associated with risk of cardiovascular disease (CVD). Insights into mechanisms involved in the pathophysiology of these clinical manifestations are essential for the development of therapeutic strategies. Thus, Western diets (WD) have been widely employed in diet-induced obesity (DIO) model. However, there are variations in fat and sugar proportions of such diets, making comparisons challenging. We aimed to assess the impact of two types of the WD on metabolic status and cardiac remodeling, to achieve a DIO model that better mimics the human pathogenesis of MetS-induced CVD. Male Wistar rats were distributed into three groups: control diet, Western diet fat (WDF), and Western diet sugar (WDS) for 41 weeks. Metabolic and inflammatory parameters and cardiac changes were characterized. WDF and WDS feeding promoted higher serum triglycerides, glucose intolerance, and insulin resistance, while just WDF presented inflammation in adipose tissue. WDF-fed rats showed increased catalase activity and malondialdehyde (MDA) and carbonyl protein levels, suggesting cardiac oxidative stress, while WDS-fed rats only raised MDA. Both WD equally elevated protein expressions involved in lipid metabolism, but only WDF downregulated the glycolysis pathway. Furthermore, the mechanical myocardial function was impaired in obese rats, being more relevant in WDF. In conclusion, both WD effectively triggered MetS features, although inflammation was detected just on the WDF-fed animals. Moreover, the WDF promoted a more pronounced functional, metabolic, and oxidative cardiac disorder, suggesting to be an adequate model for studying CVD in the scenario of MetS.
Collapse
Affiliation(s)
- Danielle Fernandes Vileigas
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Camila Renata Corrêa
- Department of Patology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | | | | | - Carlos Roberto Padovani
- Department of Biostatistics, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
13
|
Wingard MC, Dalal S, Shook PL, Myers R, Connelly BA, Thewke DP, Singh M, Singh K. Deficiency of ataxia-telangiectasia mutated kinase modulates functional and biochemical parameters of the heart in response to Western-type diet. Am J Physiol Heart Circ Physiol 2021; 320:H2324-H2338. [PMID: 33929897 PMCID: PMC8289354 DOI: 10.1152/ajpheart.00990.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/31/2021] [Accepted: 04/14/2021] [Indexed: 02/08/2023]
Abstract
Ataxia-telangiectasia mutated (ATM) kinase deficiency exacerbates heart dysfunction late after myocardial infarction. Here, we hypothesized that ATM deficiency modulates Western-type diet (WD)-induced cardiac remodeling with an emphasis on functional and biochemical parameters of the heart. Weight gain was assessed in male wild-type (WT) and ATM heterozygous knockout (hKO) mice on weekly basis, whereas cardiac functional and biochemical parameters were measured 14 wk post-WD. hKO-WD mice exhibited rapid body weight gain at weeks 5, 6, 7, 8, and 10 versus WT-WD. WD decreased percent fractional shortening and ejection fraction, and increased end-systolic volumes and diameters to a similar extent in both genotypes. However, WD decreased stroke volume, cardiac output, peak velocity of early ventricular filling, and aortic ejection time and increased isovolumetric relaxation time (IVRT) and Tei index versus WT-NC (normal chow). Conversely, IVRT, isovolumetric contraction time, and Tei index were lower in hKO-WD versus hKO-NC and WT-WD. Myocyte apoptosis and hypertrophy were higher in hKO-WD versus WT-WD. WD increased fibrosis and expression of collagen-1α1, matrix metalloproteinase (MMP)-2, and MMP-9 in WT. WD enhanced AMPK activation, while decreasing mTOR activation in hKO. Akt and IKK-α/β activation, and Bax, PARP-1, and Glut-4 expression were higher in WT-WD versus WT-NC, whereas NF-κB activation and Glut-4 expression were lower in hKO-WD versus hKO-NC. Circulating concentrations of IL-12(p70), eotaxin, IFN-γ, macrophage inflammatory protein (MIP)-1α, and MIP-1β were higher in hKO-WD versus WT-WD. Thus, ATM deficiency accelerates weight gain, induces systolic dysfunction with increased preload, and associates with increased apoptosis, hypertrophy, and inflammation in response to WD.NEW & NOTEWORTHY Ataxia-telangiectasia mutated (ATM) kinase deficiency in humans associates with enhanced susceptibility to ischemic heart disease. Here, we provide evidence that ATM deficiency accelerates body weight gain and associates with increased cardiac preload, hypertrophy, and apoptosis in mice fed with Western-type diet (WD). Further investigations of the role of ATM deficiency in WD-induced alterations in function and biochemical parameters of the heart may provide clinically applicable information on treatment and/or nutritional counseling for patients with ATM deficiency.
Collapse
Affiliation(s)
- Mary C Wingard
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Suman Dalal
- Department of Health Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Paige L Shook
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Rachel Myers
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Barbara A Connelly
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- James H Quillen Veterans Affairs Medical Center, East Tennessee State University, Johnson City, Tennessee
| | - Douglas P Thewke
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Mahipal Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Krishna Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
- James H Quillen Veterans Affairs Medical Center, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
14
|
Mühlfeld C, Pfeiffer C, Schneider V, Bornemann M, Schipke J. Voluntary activity reverses spermidine-induced myocardial fibrosis and lipid accumulation in the obese male mouse. Histochem Cell Biol 2020; 155:75-88. [PMID: 33108533 PMCID: PMC7847856 DOI: 10.1007/s00418-020-01926-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2020] [Indexed: 02/06/2023]
Abstract
Obesity due to high calorie intake induces cardiac hypertrophy and dysfunction, thus contributing to cardiovascular morbidity and mortality. Recent studies in aging suggest that oral supplementation with the natural polyamine spermidine has a cardioprotective effect. Here, the hypothesis was tested that spermidine or voluntary activity alone or in combination protect the heart from adverse effects induced by obesity. Therefore, C57Bl/6 mice (n = 8–10 per group) were subjected to control or high fat diet (HFD) and were left untreated, or either received spermidine via drinking water or were voluntarily active or both. After 30 weeks, the mice were killed and the left ventricle of the hearts was processed for light and electron microscopy. Design-based stereology was used to estimate parameters of hypertrophy, fibrosis, and lipid accumulation. HFD induced cardiac hypertrophy as demonstrated by higher volumes of the left ventricle, cardiomyocytes, interstitium, myofibrils and cardiomyocyte mitochondria. These changes were not influenced by spermidine or voluntary activity. HFD also induced myocardial fibrosis and accumulation of lipid droplets within cardiomyocytes. These HFD effects were enhanced in spermidine treated animals but not in voluntarily active mice. This was even the case in voluntarily active mice that received spermidine. In conclusion, the data confirm the induction of left ventricular hypertrophy by high-fat diet and suggest that—under high fat diet—spermidine enhances cardiomyocyte lipid accumulation and interstitial fibrosis which is counteracted by voluntary activity.
Collapse
Affiliation(s)
- Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Clara Pfeiffer
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Vanessa Schneider
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Melanie Bornemann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Julia Schipke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
15
|
Oliveira-Junior SAD, Muzili NDA, Carvalho MRD, Ota GE, Morais CSD, Vieira LFDC, Ortiz MO, Campos DHS, Cezar MDM, Okoshi MP, Okoshi K, Cicogna AC, Martinez PF. AT1Receptor Blockade Improves Myocardial Functional Performance in Obesity. Arq Bras Cardiol 2020; 115:17-28. [PMID: 32401842 PMCID: PMC8384332 DOI: 10.36660/abc.20190131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
Fundamento A obesidade tem sido associada com ativação crônica do sistema renina-angiotensina-aldosterona e importantes alterações no desempenho cardíaco. Objetivo Avaliar a influência do bloqueio de receptores de angiotensina-II do tipo 1 (AT1) sobre a morfologia e desempenho cardíaco de ratos obesos por dieta Métodos Ratos Wistar (n=48) foram submetidos a dieta controle (2,9 kcal/g) ou hiperlipídica (3,6 kcal/g) durante 20 semanas. Após a 16ª semana, foram distribuídos em quatro grupos: Controle (CO), Obeso (OB), Controle Losartan (CL) e Obeso Losartan (OL). CL e OL receberam losartan (30 mg/kg/dia) na água durante quatro semanas. Posteriormente, foram analisadas composição corporal, pressão arterial sistólica (PAS) e ecocardiograma. A função de músculos papilares foi avaliada em situação basal com concentração de cálcio ([Ca2+]o) de 2,50 mM e após manobras inotrópicas: potencial pós-pausa (PPP), elevação da [Ca2+]o e durante estimulação beta-adrenérgica com isoproterenol. A análise dos resultados foi feita por meio de Two-Way ANOVA e teste de comparações apropriado. O nível de significância considerado foi de 5%. Resultados Embora a alteração da PAS não tenha se mantido ao final do experimento, a obesidade se associou com hipertrofia cardíaca e maior velocidade de encurtamento da parede posterior do ventrículo esquerdo.No estudo de músculos papilares em condição basal, CL mostrou menor velocidade máxima de variação negativa da tensão desenvolvida (-dT/dt) do que CO. O PPP de 60s promoveu menor -dT/dt e pico de tensão desenvolvida (TD) em OB e CL, comparados ao CO, e maior variação relativa de TD e velocidade máxima de variação positiva (+dT/dt) no OL em relação a CL e OB. Sob 1,5, 2,0 e 2,5mM de [Ca2+]o, o grupo OL exibiu maior -dT/dt do que CL. Conclusão Losartan melhora a função miocárdica de ratos com obesidade induzida por dieta. (Arq Bras Cardiol. 2020; 115(1):17-28)
Collapse
Affiliation(s)
| | | | | | - Gabriel Elias Ota
- Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brasil
| | | | | | | | | | | | - Marina Politi Okoshi
- Departamento de Medicina Interna, Universidade Estadual de São Paulo, Botucatu, SP, Brasil
| | - Katashi Okoshi
- Faculdade de Medicina, Universidade Estadual Paulista Júlio de Mesquita Filho, Botucatu, SP, Brasil
| | | | | |
Collapse
|
16
|
Boldt K, Rios JL, Joumaa V, Herzog W. Mechanical function of cardiac fibre bundles is partly protected by exercise in response to diet-induced obesity in rats. Appl Physiol Nutr Metab 2020; 46:46-54. [PMID: 32598858 DOI: 10.1139/apnm-2020-0275] [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: 12/17/2022]
Abstract
Decrements in contractile function resulting from obesity are thought to be major reasons for the link between obesity and cardiovascular disease, while exercise has been shown to improve cardiac muscle contractile function. The purpose of this study was to evaluate cardiac contractile properties following obesity induction and the potential protective effect of exercise. Twelve-week-old rats (n = 30) were organized into either a chow diet or a high-fat, high-sucrose (HFHS) diet group. Following 12 weeks of obesity induction the HFHS group animals were stratified and grouped into sedentary (HFHS+Sed) and exercise (HFHS+Ex) groups for an additional 12 weeks. Following 24 weeks of diet intervention, with 12 weeks of aerobic exercise (25 m/min, 30 min/day, 5 days/week) for the HFHS+Ex group, skinned cardiac fibre bundle testing was used to evaluate cardiac contractile properties. Body fat and mass were significantly greater in the HFHS-fed animals compared with the chow controls (p < 0.043). Hearts from rats in the HFHS+Sed group had significantly greater mass (p < 0.03), significantly slower maximum shortening velocity (p = 0.001), and tended to have lower calcium sensitivity (p = 0.077) and a lower proportion of α-myosin heavy chain composition (p = 0.074) than the sedentary chow animals. However, 12 weeks of moderate aerobic exercise partially prevented these decrements in contractile properties. Novelty Cardiac muscle from animals exposed to an obesogenic diet for 24 weeks had impaired contractile properties compared with controls. Obesity-induced impairment of contractile properties of the heart were partially prevented by a 12-week aerobic exercise regime.
Collapse
Affiliation(s)
- Kevin Boldt
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada.,Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jaqueline Lourdes Rios
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada.,Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Venus Joumaa
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada.,Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Walter Herzog
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada.,Human Performance Laboratory, University of Calgary, Calgary, AB T2N 1N4, Canada.,Biomechanics Laboratory, School of Sports, Federal University of Santa Catarina, SC 88040-900, Brazil
| |
Collapse
|
17
|
Vileigas DF, Marciano CLDC, Mota GAF, de Souza SLB, Sant’Ana PG, Okoshi K, Padovani CR, Cicogna AC. Temporal Measures in Cardiac Structure and Function During the Development of Obesity Induced by Different Types of Western Diet in a Rat Model. Nutrients 2019; 12:nu12010068. [PMID: 31888029 PMCID: PMC7019835 DOI: 10.3390/nu12010068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is recognized worldwide as a complex metabolic disorder that has reached epidemic proportions and is often associated with a high incidence of cardiovascular diseases. To study this pathology and evaluate cardiac function, several models of diet-induced obesity (DIO) have been developed. The Western diet (WD) is one of the most widely used models; however, variations in diet composition and time period of the experimental protocol make comparisons challenging. Thus, this study aimed to evaluate the effects of two different types of Western diet on cardiac remodeling in obese rats with sequential analyses during a long-term follow-up. Male Wistar rats were distributed into three groups fed with control diet (CD), Western diet fat (WDF), and Western diet sugar (WDS) for 41 weeks. The animal nutritional profile and cardiac histology were assessed at the 41st week. Cardiac structure and function were evaluated by echocardiogram at four different moments: 17, 25, 33, and 41 weeks. A noninvasive method was performed to assess systolic blood pressure at the 33rd and 41st week. The animals fed with WD (WDF and WDS) developed pronounced obesity with an average increase of 86.5% in adiposity index at the end of the experiment. WDF and WDS groups also presented hypertension. The echocardiographic data showed no structural differences among the three groups, but WDF animals presented decreased endocardial fractional shortening and ejection fraction at the 33rd and 41st week, suggesting altered systolic function. Moreover, WDF and WFS animals did not present hypertrophy and interstitial collagen accumulation in the left ventricle. In conclusion, both WD were effective in triggering severe obesity in rats; however, only the WDF induced mild cardiac dysfunction after long-term diet exposure. Further studies are needed to search for an appropriate DIO model with relevant cardiac remodeling.
Collapse
Affiliation(s)
- Danielle Fernandes Vileigas
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
| | - Cecília Lume de Carvalho Marciano
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
| | - Gustavo Augusto Ferreira Mota
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
| | - Paula Grippa Sant’Ana
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
| | - Katashi Okoshi
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
| | - Carlos Roberto Padovani
- Department of Biostatistics, Institute of Biosciences, São Paulo State University, Botucatu 18618970, Brazil;
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, Botucatu 18618687, Brazil; (D.F.V.); (C.L.d.C.M.); (G.A.F.M.); (S.L.B.d.S.); (P.G.S.); (K.O.)
- Correspondence: ; Tel.: +55-14-3880-1618
| |
Collapse
|
18
|
Bracht JR, Vieira‐Potter VJ, De Souza Santos R, Öz OK, Palmer BF, Clegg DJ. The role of estrogens in the adipose tissue milieu. Ann N Y Acad Sci 2019; 1461:127-143. [DOI: 10.1111/nyas.14281] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | | | | | - Orhan K. Öz
- Department of RadiologyUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Biff F. Palmer
- Department of MedicineUniversity of Texas Southwestern Medical Center Dallas Texas
| | - Deborah J. Clegg
- College of Nursing and Health ProfessionsDrexel University Philadelphia Pennsylvania
| |
Collapse
|
19
|
Espinoza-Derout J, Hasan KM, Shao XM, Jordan MC, Sims C, Lee DL, Sinha S, Simmons Z, Mtume N, Liu Y, Roos KP, Sinha-Hikim AP, Friedman TC. Chronic intermittent electronic cigarette exposure induces cardiac dysfunction and atherosclerosis in apolipoprotein-E knockout mice. Am J Physiol Heart Circ Physiol 2019; 317:H445-H459. [PMID: 31172811 DOI: 10.1152/ajpheart.00738.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Electronic cigarettes (e-cigarettes), also known as electronic nicotine delivery systems, are a popular alternative to conventional nicotine cigarettes, both among smokers and those who have never smoked. In spite of the widespread use of e-cigarettes and the proposed detrimental cardiac and atherosclerotic effects of nicotine, the effects of e-cigarettes on these systems are not known. In this study, we investigated the cardiovascular and cardiac effects of e-cigarettes with and without nicotine in apolipoprotein-E knockout (ApoE-/-) mice. We developed an e-cigarette exposure model that delivers nicotine in a manner similar to that of human e-cigarettes users. Using commercially available e-cigarettes, bluCig PLUS, ApoE-/- mice were exposed to saline, e-cigarette without nicotine [e-cigarette (0%)], and e-cigarette with 2.4% nicotine [e-cigarette (2.4%)] aerosol for 12 wk. Echocardiographic data show that mice treated with e-cigarette (2.4%) had decreased left ventricular fractional shortening and ejection fraction compared with e-cigarette (0%) and saline. Ventricular transcriptomic analysis revealed changes in genes associated with metabolism, circadian rhythm, and inflammation in e-cigarette (2.4%)-treated ApoE-/- mice. Transmission electron microscopy revealed that cardiomyocytes of mice treated with e-cigarette (2.4%) exhibited ultrastructural abnormalities indicative of cardiomyopathy. Additionally, we observed increased oxidative stress and mitochondrial DNA mutations in mice treated with e-cigarette (2.4%). ApoE-/- mice on e-cigarette (2.4%) had also increased atherosclerotic lesions compared with saline aerosol-treated mice. These results demonstrate adverse effects of e-cigarettes on cardiac function in mice.NEW & NOTEWORTHY The present study is the first to show that mice exposed to nicotine electronic cigarettes (e-cigarettes) have decreased cardiac fractional shortening and ejection fraction in comparison with controls. RNA-seq analysis reveals a proinflammatory phenotype induced by e-cigarettes with nicotine. We also found increased atherosclerosis in the aortic root of mice treated with e-cigarettes with nicotine. Our results show that e-cigarettes with nicotine lead to detrimental effects on the heart that should serve as a warning to e-cigarette users and agencies that regulate them.
Collapse
Affiliation(s)
- Jorge Espinoza-Derout
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Kamrul M Hasan
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Xuesi M Shao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Maria C Jordan
- David Geffen School of Medicine at University of California, Los Angeles, California
| | - Carl Sims
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Desean L Lee
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Satyesh Sinha
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Zena Simmons
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Norma Mtume
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Yanjun Liu
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Kenneth P Roos
- David Geffen School of Medicine at University of California, Los Angeles, California
| | - Amiya P Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Theodore C Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| |
Collapse
|
20
|
Oliveira‐Junior SA, Dal Pai M, Guizoni DM, Torres BP, Martinez PF, Campos DHS, Okoshi MP, Okoshi K, Padovani CR, Cicogna AC. Effects of AT1 receptor antagonism on interstitial and ultrastructural remodeling of heart in response to a hypercaloric diet. Physiol Rep 2019; 7:e13964. [PMID: 30592176 PMCID: PMC6308213 DOI: 10.14814/phy2.13964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/29/2022] Open
Abstract
Palatable hypercaloric feeding has been associated with angiotensin-II type 1 receptor (AT1R) stimulation and cardiac remodeling. This study analyzed whether AT1R antagonism attenuates cardiac remodeling in rats subjected to a palatable hypercaloric diet. Male Wistar-Kyoto rats were subjected to a commercial standard rat chow (CD) or a palatable hypercaloric diet (HD) for 35 weeks and then allocated into four groups: CD, CL, HD, and HL; L groups received losartan in drinking water (30 mg/kg/day) for 5 weeks. Body weight, adiposity, and glycemia were evaluated. The cardiovascular study included echocardiography, and myocardial morphometric and ultrastructural evaluation. Myocardial collagen isoforms Type I and III were analyzed by Western blot. Both HD and HL had higher adiposity than their respective controls. Cardiomyocyte cross-sectional-area (CD 285 ± 49; HD 344 ± 91; CL 327 ± 49; HL 303 ± 49 μm2 ) and interstitial collagen fractional area were significantly higher in HD than CD and unchanged by losartan. HD showed marked ultrastructural alterations such as myofilament loss, and severe mitochondrial swelling. CL presented higher Type I collagen expression when compared to CD and HL groups. The ultrastructural changes and type I collagen expression were attenuated by losartan in HL. Losartan attenuates systolic dysfunction and ultrastructural abnormalities without changing myocardial interstitial remodeling in rats subjected to a palatable hypercaloric diet.
Collapse
Affiliation(s)
- Silvio A. Oliveira‐Junior
- School of Physical TherapyFederal University of Mato Grosso do SulCampo GrandeMato Grosso do SulBrazil
| | - Maeli Dal Pai
- Botucatu Biosciences InstituteUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Daniele M. Guizoni
- Internal Medicine DepartmentBotucatu Medical SchoolUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Barbara P. Torres
- Botucatu Biosciences InstituteUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Paula F. Martinez
- School of Physical TherapyFederal University of Mato Grosso do SulCampo GrandeMato Grosso do SulBrazil
| | - Dijon H. S. Campos
- Internal Medicine DepartmentBotucatu Medical SchoolUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Marina P. Okoshi
- Internal Medicine DepartmentBotucatu Medical SchoolUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Katashi Okoshi
- Internal Medicine DepartmentBotucatu Medical SchoolUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Carlos R. Padovani
- Botucatu Biosciences InstituteUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| | - Antonio C. Cicogna
- Internal Medicine DepartmentBotucatu Medical SchoolUniv. Estadual PaulistaUNESPBotucatuSão PauloBrazil
| |
Collapse
|
21
|
Ferron AJT, Francisqueti FV, Minatel IO, Silva CCVDA, Bazan SGZ, Kitawara KAH, Garcia JL, Corrêa CR, Moreto F, Ferreira ALA. Association between Cardiac Remodeling and Metabolic Alteration in an Experimental Model of Obesity Induced by Western Diet. Nutrients 2018; 10:nu10111675. [PMID: 30400581 PMCID: PMC6266980 DOI: 10.3390/nu10111675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/02/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022] Open
Abstract
The high consumption of fat and sugar contributes to the development of obesity and co-morbidities, such as dyslipidemia, hypertension, and cardiovascular disease. The aim of this study was to evaluate the association between dyslipidemia and cardiac dysfunction induced by western diet consumption. Wistar rats were randomly divided into two experimental groups and fed ad libitum for 20 weeks with a control diet (Control, n = 12) or a high-sugar and high-fat diet (HSF, n = 12). The HSF group also received water + sucrose (25%). Evaluations included feed and caloric intake; body weight; plasma glucose; insulin; uric acid; HOMA-IR; lipid profile: [total cholesterol (T-chol), high-density lipoprotein (HDL), non-HDL Chol, triglycerides (TG)]; systolic blood pressure, and Doppler echocardiographic. Compared to the control group, animals that consumed the HSF diet presented higher weight gain, caloric intake, feed efficiency, insulin, HOMA-IR, and glucose levels, and lipid profile impairment (higher TG, T-chol, non-HDL chol and lower HDL). HSF diet was also associated with atrial-ventricular structural impairment and systolic-diastolic dysfunction. Positive correlation was also found among the following parameters: insulin versus estimated LV mass (r = 0.90, p = 0.001); non-HDL versus deceleration time (r = 0.46, p = 0.02); TG versus deceleration time (r = 0.50, p = 0.01). In summary, our results suggest cardiac remodeling lead by western diet is associated with metabolic parameters.
Collapse
Affiliation(s)
| | | | - Igor Otávio Minatel
- São Paulo State University (Unesp), Institute of Biosciences, Botucatu 18618-689, Brazil.
| | | | | | | | | | | | - Fernando Moreto
- São Paulo State University (Unesp), Medical School, Botucatu 18618-687, Brazil.
| | | |
Collapse
|
22
|
Quercetin Prevents Diastolic Dysfunction Induced by a High-Cholesterol Diet: Role of Oxidative Stress and Bioenergetics in Hyperglycemic Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7239123. [PMID: 29576853 PMCID: PMC5821945 DOI: 10.1155/2018/7239123] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/23/2017] [Indexed: 01/08/2023]
Abstract
Alterations in cardiac energy metabolism play a key role in the pathogenesis of diabetic cardiomyopathy. Hypercholesterolemia associated with bioenergetic impairment and oxidative stress has not been well characterized in the cardiac function under glycemic control deficiency conditions. This work aimed to determine the cardioprotective effects of quercetin (QUE) against the damage induced by a high-cholesterol (HC) diet in hyperglycemic rats, addressing intracellular antioxidant mechanisms and bioenergetics. Quercetin reduced HC-induced alterations in the lipid profile and glycemia in rats. In addition, QUE attenuated cardiac diastolic dysfunction (increased E:A ratio), prevented cardiac cholesterol accumulation, and reduced the increase in HC-induced myocyte density. Moreover, QUE reduced HC-induced oxidative stress by preventing the decrease in GSH/GSSG ratio, Nrf2 nuclear translocation, HO-1 expression, and antioxidant enzymatic activity. Quercetin also counteracted HC-induced bioenergetic impairment, preventing a reduction in ATP levels and alterations in PGC-1α, UCP2, and PPARγ expression. In conclusion, the mechanisms that support the cardioprotective effect of QUE in rats with HC might be mediated by the upregulation of antioxidant mechanisms and improved bioenergetics on the heart. Targeting bioenergetics with QUE can be used as a pharmacological approach to modulate structural and functional changes of the heart under hypercholesterolemic and hyperglycemic conditions.
Collapse
|
23
|
Dietary Fat, Sugar Consumption, and Cardiorespiratory Fitness in Patients With Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2017; 2:513-525. [PMID: 30062167 PMCID: PMC6058958 DOI: 10.1016/j.jacbts.2017.06.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/22/2017] [Accepted: 06/01/2017] [Indexed: 01/05/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is associated with obesity and, indirectly, with unhealthy diet. The role of dietary components in HFpEF is, however, largely unknown. In this study, the authors showed that in obese HFpEF patients, consumption of unsaturated fatty acids (UFA), was associated with better cardiorespiratory fitness, and UFA consumption correlated with better diastolic function and with greater fat-free mass. Similarly, mice fed with a high-fat diet rich in UFA and low in sugars had preserved myocardial function and reduced weight gain. Randomized clinical trials increasing dietary UFA consumption and reducing sugar consumption are warranted to confirm and expand our findings.
Collapse
Key Words
- CPX, cardiopulmonary exercise testing
- CRF, cardiorespiratory fitness
- CV, cardiovascular
- DT, deceleration time
- FFM, fat-free mass
- FM, fat mass
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- IQR, interquartile range
- MUFA, monounsaturated fatty acid
- PUFA, polyunsaturated fatty acid
- SFA, saturated fatty acid
- UFA, unsaturated fatty acid
- Vo2, oxygen consumption
- body composition
- diet
- heart failure with preserved ejection fraction
- obesity
- unsaturated fatty acids
Collapse
|
24
|
Vileigas DF, de Deus AF, da Silva DCT, de Tomasi LC, de Campos DHS, Adorni CS, de Oliveira SM, Sant'Ana PG, Okoshi K, Padovani CR, Cicogna AC. Saturated high-fat diet-induced obesity increases adenylate cyclase of myocardial β-adrenergic system and does not compromise cardiac function. Physiol Rep 2017; 4:4/17/e12914. [PMID: 27582064 PMCID: PMC5027348 DOI: 10.14814/phy2.12914] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/01/2016] [Indexed: 01/13/2023] Open
Abstract
Obesity is a worldwide pandemic associated with high incidence of cardiovascular disease. The mechanisms by which the obesity leads cardiac dysfunction are not fully elucidated and few studies have evaluated the relationship between obesity and proteins involved in myocardial β‐adrenergic (βA) system. The purpose of this study was to evaluate the cardiac function and βA pathway components in myocardium of obese rats. Male Wistar rats were distributed into two groups: control (n = 17; standard diet) and obese (n = 17; saturated high‐fat diet) fed for 33 weeks. Nutritional profile and comorbidities were assessed. Cardiac structure and function was evaluated by macroscopic postmortem, echocardiographic and isolated papillary muscle analyzes. Myocardial protein expression of β1‐ and β2‐adrenergic receptors, Gαs protein, adenylate cyclase (AC) and protein kinase A (PKA) was performed by Western blot. Cardiac cyclic adenosine monophosphate (cAMP) levels and PKA activity were assessed by ELISA. Obese rats showed increased adiposity index (P < 0.001) and several comorbidities as hypertension, glucose intolerance, insulin resistance, and dyslipidemia compared with control rats. Echocardiographic assessment revealed increased left atrium diameter (C: 4.98 ± 0.38 vs. Ob: 5.47 ± 0.53, P = 0.024) and posterior wall shortening velocity (C: 37.1 ± 3.6 vs. Ob: 41.8 ± 3.8, P = 0.007) in obese group. Papillary muscle evaluation indicated that baseline data and myocardial responsiveness to isoproterenol stimulation were similar between the groups. Protein expression of myocardial AC was higher in obese group than in the control (C: 1.00 ± 0.21 vs. Ob: 1.25 ± 0.10, P = 0.025), whereas the other components were unchanged. These results suggest that saturated high‐fat diet‐induced obesity was not effective in triggering cardiac dysfunction and impair the beta‐adrenergic signaling.
Collapse
Affiliation(s)
- Danielle F Vileigas
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Adriana F de Deus
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Danielle C T da Silva
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Loreta C de Tomasi
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Dijon H S de Campos
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Caroline S Adorni
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Scarlet M de Oliveira
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Paula G Sant'Ana
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Katashi Okoshi
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Carlos R Padovani
- Department of Biostatistics, Biosciences Institute São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| | - Antonio C Cicogna
- Department of Internal Medicine, Medical School São Paulo State University "Júlio de Mesquita Filho", Botucatu, São Paulo, Brazil
| |
Collapse
|
25
|
Proinflammatory Cytokines Are Soluble Mediators Linked with Ventricular Arrhythmias and Contractile Dysfunction in a Rat Model of Metabolic Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7682569. [PMID: 29201273 PMCID: PMC5671748 DOI: 10.1155/2017/7682569] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 08/27/2017] [Indexed: 12/15/2022]
Abstract
Metabolic syndrome (MS) increases cardiovascular risk and is associated with cardiac dysfunction and arrhythmias, although the precise mechanisms are still under study. Chronic inflammation in MS has emerged as a possible cause of adverse cardiac events. Male Wistar rats fed with 30% sucrose in drinking water and standard chow for 25–27 weeks were compared to a control group. The MS group showed increased weight, visceral fat, blood pressure, and serum triglycerides. The most important increases in serum cytokines included IL-1β (7-fold), TNF-α (84%), IL-6 (41%), and leptin (2-fold), the latter also showing increased gene expression in heart tissue (35-fold). Heart function ex vivo in MS group showed a decreased mechanical performance response to isoproterenol challenge (ISO). Importantly, MS hearts under ISO showed nearly twofold the incidence of ventricular fibrillation. Healthy rat cardiomyocytes exposed to MS group serum displayed impaired contractile function and Ca2+ handling during ISO treatment, showing slightly decreased cell shortening and Ca2+ transient amplitude (23%), slower cytosolic calcium removal (17%), and more frequent spontaneous Ca2+ release events (7.5-fold). As spontaneous Ca2+ releases provide a substrate for ventricular arrhythmias, our study highlights the possible role of serum proinflammatory mediators in the development of arrhythmic events during MS.
Collapse
|
26
|
Rietdorf K, MacQueen H. Investigating interactions between epicardial adipose tissue and cardiac myocytes: what can we learn from different approaches? Br J Pharmacol 2017; 174:3542-3560. [PMID: 27882550 PMCID: PMC5610165 DOI: 10.1111/bph.13678] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/14/2016] [Accepted: 11/18/2016] [Indexed: 01/08/2023] Open
Abstract
Heart disease is a major cause of morbidity and mortality throughout the world. Some cardiovascular conditions can be modulated by lifestyle factors such as increased exercise or a healthier diet, but many require surgical or pharmacological interventions for their management. More targeted and less invasive therapies would be beneficial. Recently, it has become apparent that epicardial adipose tissue plays an important role in normal and pathological cardiac function, and it is now the focus of considerable research. Epicardial adipose tissue can be studied by imaging of various kinds, and these approaches have yielded much useful information. However, at a molecular level, it is more difficult to study as it is relatively scarce in animal models and, for practical and ethical reasons, not always available in sufficient quantities from patients. What is needed is a robust model system in which the interactions between epicardial adipocytes and cardiac myocytes can be studied, and physiologically relevant manipulations performed. There are drawbacks to conventional culture methods, not least the difficulty of culturing both cardiac myocytes and adipocytes, each of which has special requirements. We discuss the benefits of a three-dimensional co-culture model in which in vivo interactions can be replicated. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Katja Rietdorf
- School of Life, Health and Chemical SciencesThe Open UniversityMilton KeynesUK
| | - Hilary MacQueen
- School of Life, Health and Chemical SciencesThe Open UniversityMilton KeynesUK
| |
Collapse
|
27
|
GSK-3 Inhibitors: Anti-Diabetic Treatment Associated with Cardiac Risk? : Editorial to: "The Impact of Chronic Glycogen Synthase Kinase-3 Inhibition on Remodeling of Normal and Pre-Diabetic Rat Hearts." by Barbara Huisamen et al. Cardiovasc Drugs Ther 2017; 30:233-5. [PMID: 27311575 DOI: 10.1007/s10557-016-6669-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
28
|
Affiliation(s)
- John S. Gottdiener
- From the Department of Medicine, Division of Cardiology, University of Maryland School of Medicine, Baltimore (J.S.G.); and Department of Medical and Clinical Psychology, Centre of Research on Psychology in Somatic Disease, Tilburg University, The Netherlands (W.J.K)
| | - Willem J. Kop
- From the Department of Medicine, Division of Cardiology, University of Maryland School of Medicine, Baltimore (J.S.G.); and Department of Medical and Clinical Psychology, Centre of Research on Psychology in Somatic Disease, Tilburg University, The Netherlands (W.J.K)
| |
Collapse
|
29
|
Carbone S, Lavie CJ, Arena R. Obesity and Heart Failure: Focus on the Obesity Paradox. Mayo Clin Proc 2017; 92:266-279. [PMID: 28109619 DOI: 10.1016/j.mayocp.2016.11.001] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/14/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022]
Abstract
The escalating prevalence of obesity has been linked to substantial increases in both metabolic and cardiovascular disease. Nevertheless, the direct effects of obesity on cardiovascular health and function require further exploration. In particular, the relationship between obesity and cardiac function has received intense scrutiny. Although obesity increases the risk for development of heart failure (HF), it appears to exert a protective effect in patients in whom HF has already been diagnosed (the "obesity paradox"). The protective effects of obesity in patients with previously diagnosed HF are the focus of particularly intense research. Several explanations have been proposed, but most studies are limited by the use of body mass index to classify obesity. Because body mass index does not distinguish between fat mass, fat-free mass, and lean mass, individuals with similar body mass indices may have vastly different body composition. This article discusses the roles of body composition, diet, cardiorespiratory fitness, and weight loss in the development of cardiac dysfunction and HF and the potential protective role that body composition compartments might play in improving HF prognosis. Based on an intensive literature search (Pubmed, Google Scholar) and critical review of the literature, we also discuss how a multidisciplinary approach including a nutritional intervention targeted to reduce systemic inflammation and lean mass-targeted exercise training could potentially exert beneficial effects for patients with HF.
Collapse
Affiliation(s)
- Salvatore Carbone
- Pauley Heart Center, Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, LA
| | - Ross Arena
- Department of Physical Therapy and Integrative Physiology Laboratory, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
30
|
Heinonen IHA, Boushel R, Kalliokoski KK. The Circulatory and Metabolic Responses to Hypoxia in Humans - With Special Reference to Adipose Tissue Physiology and Obesity. Front Endocrinol (Lausanne) 2016; 7:116. [PMID: 27621722 PMCID: PMC5002918 DOI: 10.3389/fendo.2016.00116] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/10/2016] [Indexed: 01/07/2023] Open
Abstract
Adipose tissue metabolism and circulation play an important role in human health. It is well-known that adipose tissue mass is increased in response to excess caloric intake leading to obesity and further to local hypoxia and inflammatory signaling. Acute exercise increases blood supply to adipose tissue and mobilization of fat stores for energy. However, acute exercise during systemic hypoxia reduces subcutaneous blood flow in healthy young subjects, but the response in overweight or obese subjects remains to be investigated. Emerging evidence also indicates that exercise training during hypoxic exposure may provide additive benefits with respect to many traditional cardiovascular risk factors as compared to exercise performed in normoxia, but unfavorable effects of hypoxia have also been documented. These topics will be covered in this brief review dealing with hypoxia and adipose tissue physiology.
Collapse
Affiliation(s)
- Ilkka H. A. Heinonen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Turku, Turku, Finland
- Division of Experimental Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Ilkka H. A. Heinonen,
| | - Robert Boushel
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|