1
|
de Oliveira Silva T, Lunardon G, Lino CA, de Almeida Silva A, Zhang S, Irigoyen MCC, Lu YW, Mably JD, Barreto-Chaves MLM, Wang DZ, Diniz GP. Senescent cell depletion alleviates obesity-related metabolic and cardiac disorders. Mol Metab 2025; 91:102065. [PMID: 39557194 PMCID: PMC11636344 DOI: 10.1016/j.molmet.2024.102065] [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: 07/24/2024] [Revised: 10/06/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Obesity is a major contributor to metabolic and cardiovascular disease. Although senescent cells have been shown to accumulate in adipose tissue, the role of senescence in obesity-induced metabolic disorders and in cardiac dysfunction is not yet clear; therefore, the therapeutic potential of managing senescence in obesity-related metabolic and cardiac disorders remains to be fully defined. OBJECTIVE We investigated the beneficial effects of a senolytic cocktail (dasatinib and quercetin) on senescence and its influence on obesity-related parameters. METHODS AND RESULTS We found that the increase in body weight and adiposity, glucose intolerance, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic disorders which were induced by an obesogenic diet were alleviated by senolytic cocktail treatment in mice. Treatment with senolytic compounds eliminated senescent cells, counteracting the activation of the senescence program and DNA damage in white adipose tissue (WAT) observed with an obesogenic diet. Moreover, the senolytic cocktail prevented the brown adipose tissue (BAT) whitening and increased the expression of the thermogenic gene profile in BAT and pWAT. In the hearts of obese mice, senolytic combination abolished myocardial maladaptation, reducing the senescence-associated secretory phenotype (SASP) and DNA damage, repressing cardiac hypertrophy, and improving diastolic dysfunction. Additionally, we showed that treatment with the senolytic cocktail corrected gene expression programs associated with fatty acid metabolism, oxidative phosphorylation, the P53 pathway, and DNA repair, which were all downregulated in obese mice. CONCLUSIONS Collectively, these data suggest that a senolytic cocktail can prevent the activation of the senescence program in the heart and WAT and activate the thermogenic program in BAT. Our results suggest that targeting senescent cells may be a novel therapeutic strategy for alleviating obesity-related metabolic and cardiac disorders.
Collapse
Affiliation(s)
- Tábatha de Oliveira Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Guilherme Lunardon
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Amanda de Almeida Silva
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Shiju Zhang
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | | | - Yao Wei Lu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John D Mably
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | | | - Da-Zhi Wang
- Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Gabriela P Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
2
|
Renton MC, McGee SL, Howlett KF. The role of protein kinase D (PKD) in obesity: Lessons from the heart and other tissues. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119814. [PMID: 39128598 DOI: 10.1016/j.bbamcr.2024.119814] [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: 03/28/2023] [Revised: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Obesity causes a range of tissue dysfunctions that increases the risk for morbidity and mortality. Protein kinase D (PKD) represents a family of stress-activated intracellular signalling proteins that regulate essential processes such as cell proliferation and differentiation, cell survival, and exocytosis. Evidence suggests that PKD regulates the cellular adaptations to the obese environment in metabolically important tissues and drives the development of a variety of diseases. This review explores the role that PKD plays in tissue dysfunction in obesity, with special consideration of the development of obesity-mediated cardiomyopathy, a distinct cardiovascular disease that occurs in the absence of common comorbidities and leads to eventual heart failure and death. The downstream mechanisms mediated by PKD that could contribute to dysfunctions observed in the heart and other metabolically important tissues in obesity, and the predicted cell types involved are discussed to suggest potential targets for the development of therapeutics against obesity-related disease.
Collapse
Affiliation(s)
- Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia; The Fralin Biomedical Research Institute at Virginia Tech Carilion, Centre for Vascular and Heart Research, Roanoke, VA, USA.
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia.
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia.
| |
Collapse
|
3
|
Vieira AKG, Bernardo AF, Neves FA, Soares VM, Guedes RM, Soares PN, Lisboa PC, Cortez E, Moura EG, da Silva BG, Garcia-Souza EP, Moura AS. Impact of early postnatal overnutrition on cardiac mitochondrial dysfunction in adult mice with ischemia/reperfusion. Nutr Metab Cardiovasc Dis 2024:S0939-4753(24)00356-9. [PMID: 39433457 DOI: 10.1016/j.numecd.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND AND AIMS Nutritional imbalance at the beginning of life, a critical window period, leads to the development of obesity, overweight, dyslipidemia, diabetes, and cardiovascular disease in adulthood. In this study, the effects and associations of overnutrition during lactation on energy metabolism and oxidative stress in cardiomyocytes of adult male Swiss mice were examined. METHODS AND RESULTS Animals were divided into two groups (control and overfed) subjected to baseline and ischemia/reperfusion conditions, forming four groups: control baseline (CBL), control ischemia/reperfusion (CIR), overfed baseline (OBL), and overfed ischemia/reperfusion (OIR). The hearts were analyzed for hemodynamics using the Langendorff technique, mitochondrial energy metabolism using the Oroboros apparatus, ATP production, oxidative stress, and SIRT1, pSTAT3 and STAT3 protein content by Western blotting. Hemodynamic abnormalities in the cardiovascular system were associated with mitochondrial dysfunction, as demonstrated by impaired carbohydrate and fatty acid oxidation capacity, decreased mitochondrial coupling in the OG, and reduced ATP production in the OIR group. Alteration in pSTAT3 and SIRT1 proteins expression in overfed mice reinforce energy metabolism impairment. Lipid and/or protein degradation is altered in the heart of OG, suggesting increased oxidative stress. CONCLUSION Overnutrition during lactation associated with heart ischemia leads to molecular cardiac alterations in STAT3 and SIRT1 proteins, compromising energy metabolism via reduced mitochondrial oxidation capacity, ATP production and increased lipid peroxidation.
Collapse
Affiliation(s)
- Anatalia K G Vieira
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Amélia F Bernardo
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiana A Neves
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivian M Soares
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roberta M Guedes
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia N Soares
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia C Lisboa
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Erika Cortez
- Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Egberto G Moura
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna G da Silva
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Erica P Garcia-Souza
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anibal S Moura
- Department of Physiological Sciences, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, 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
|
Xu B, Lv L, Chen X, Li X, Zhao X, Yang H, Feng W, Jiang X, Li J. Temporal relationships between BMI and obesity-related predictors of cardiometabolic and breast cancer risk in a longitudinal cohort. Sci Rep 2023; 13:12361. [PMID: 37524743 PMCID: PMC10390576 DOI: 10.1038/s41598-023-39387-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Prospective inter-relationships among biomarkers were unexplored, which may provide mechanistic insights into diseases. We investigated the longitudinal associations of BMI change with trajectories of biomarkers related to cardiometabolic or breast cancer risk. A longitudinal study was conducted among 444 healthy women between 2019 to 2021. Cross‑lagged path analysis was used to examine the temporal relationships among BMI, cardiometabolic risk score (CRS), and obesity‑related proteins score (OPS) of breast cancer. Linear mixed-effect models were applied to investigate associations of time-varying BMI with biomarker-based risk score trajectories. Baseline BMI was associated with subsequent change of breast cancer predictors (P = 0.03), and baseline CRS were positively associated with OPS change (P < 0.001) but not vice versa. After fully adjustment of confounders, we found a 0.058 (95%CI = 0.009-0.107, P = 0.020) units increase of CRS and a 1.021 (95%CI = 0.041-1.995, P = 0.040) units increase of OPS as BMI increased 1 kg/m2 per year in postmenopausal women. OPS increased 0.784 (95%CI = 0.053-1.512, P = 0.035) units as CRS increased 1 unit per year. However, among premenopausal women, BMI only significantly affected CRS (β = 0.057, 95%CI = 0.007 to 0.107, P = 0.025). No significant change of OPS with time-varying CRS was found. Higher increase rates of BMI were associated with worse trajectories of biomarker-based risk of cardiometabolic and breast cancer. The longitudinal impact of CRS on OPS is unidirectional. Recommendations such as weight control for the reduction of cardiometabolic risk factors may benefit breast cancer prevention, especially in postmenopausal women.
Collapse
Affiliation(s)
- Bin Xu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Liang Lv
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xingyue Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Huifang Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wanting Feng
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xia Jiang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, 16#, Section 3, Renmin Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
6
|
Abstract
Hypertrophic cardiomyopathy is one of the most frequently diagnosed primary conditions of the heart muscle. It is considered to be inherited, caused by genetic mutations encoding for sarcomere proteins. The marked heterogeneity in clinical manifestations and natural course of the disease, even among family members sharing the same genetic mutation, has raised the question of non-genetic environmental factors contributing to the phenotype. Obesity has been associated with worse cardiovascular outcomes in the general population. Its prevalence is increased in hypertrophic cardiomyopathy, and the two conditions share some similar pathophysiological and clinical characteristics. In this review, we aim to summarise the effects of obesity in the cardiac phenotype, the symptoms and management in patients with hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
| | - Konstantinos Savvatis
- Barts Health NHS Trust, London, UK; William Harvey Research Institute, Queen Mary University London, UK; Institute for Cardiovascular Science, University College London, London, UK
| |
Collapse
|
7
|
Huang S, Li Y, Shi K, Wang J, Jiang L, Gao Y, Yan WF, Yang ZG. Impact of Metabolic Syndrome on Left Ventricular Deformation and Myocardial Energetic Efficiency Compared Between Women and Men: An MRI Study. J Magn Reson Imaging 2022; 57:1743-1751. [PMID: 36196628 DOI: 10.1002/jmri.28455] [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: 08/05/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Metabolic and hemodynamic alterations in metabolic syndrome (MetS) can cause a reduced myocardial energetic efficiency (MEE). Indexed MEE (MEEi), as a simple estimate of MEE, is emerging as a novel and useful imaging parameter. PURPOSE To investigate the impact of MetS on MEE and systolic myocardial strain and to assess any sex difference. STUDY TYPE Retrospective. POPULATION A total of 161 patients with MetS (female: n = 82, 52.2 ± 11.7 years; male: n = 79, 51.8 ± 10.6 years) and 77 healthy subjects (female: n = 46, 52.7 ± 8.2 years; male: n = 31, 54.1 ± 11.2 years). Patients with left ventricular (LV) ejection fraction <50% were excluded. FIELD STRENGTH/SEQUENCE A 3.0 T; balanced steady-state free precession sequence. ASSESSMENT LV volumes and mass (LVM) and global longitudinal strain (GLS) were obtained by MRI. Stroke volume (SV) divided by HR was used as a surrogate measure of MEE and normalized to LVM (MEEi). STATISTICAL TESTS Student's t-test or Mann-Whitney U-test; Multivariable linear regression (coefficient of determination, R2 ). P < 0.05 was considered statistically significant. RESULTS For both males and females, MEEi and GLS were lower in MetS patients than in the normal controls. Among MetS patients, men had significantly higher LVM (59.7 ± 13.4 g/m2 vs. 48.8 ± 11.3 g/m2 ) and significantly lower MEEi (0.68 ± 0.23 mL/g/s vs. 0.84 ± 0.23 mL/g/s) and GLS (-11.7% ± 2.8% vs. -13.9% ± 2.7%) than women. After adjustment for clinical variables, male gender (β = -0.291) was found to be inversely correlated with MEEi. Multivariable analysis showed that MEEi (β = 0.454) were independently associated with GLS (adjusted R2 = 0.454) after adjustment for clinical and other MRI parameters. DATA CONCLUSION MEEi was significantly impaired in MetS without overt systolic dysfunction. There was a sex difference regarding the cardiac alterations in MetS, with men having significantly lower MEEi and GLS and significantly higher LVM than women. Further, MEEi was independently associated with GLS. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Shan Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ke Shi
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Jiang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Gao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Feng Yan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Qin X, Zhang Y, Zheng Q. Metabolic Inflexibility as a Pathogenic Basis for Atrial Fibrillation. Int J Mol Sci 2022; 23:ijms23158291. [PMID: 35955426 PMCID: PMC9368187 DOI: 10.3390/ijms23158291] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained arrhythmia, is closely intertwined with metabolic abnormalities. Recently, a metabolic paradox in AF pathogenesis has been suggested: under different forms of pathogenesis, the metabolic balance shifts either towards (e.g., obesity and diabetes) or away from (e.g., aging, heart failure, and hypertension) fatty acid oxidation, yet they all increase the risk of AF. This has raised the urgent need for a general consensus regarding the metabolic changes that predispose patients to AF. “Metabolic flexibility” aptly describes switches between substrates (fatty acids, glucose, amino acids, and ketones) in response to various energy stresses depending on availability and requirements. AF, characterized by irregular high-frequency excitation and the contraction of the atria, is an energy challenge and triggers a metabolic switch from preferential fatty acid utilization to glucose metabolism to increase the efficiency of ATP produced in relation to oxygen consumed. Therefore, the heart needs metabolic flexibility. In this review, we will briefly discuss (1) the current understanding of cardiac metabolic flexibility with an emphasis on the specificity of atrial metabolic characteristics; (2) metabolic heterogeneity among AF pathogenesis and metabolic inflexibility as a common pathological basis for AF; and (3) the substrate-metabolism mechanism underlying metabolic inflexibility in AF pathogenesis.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Yudi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- Correspondence: or
| |
Collapse
|
9
|
Zhao J, Li W, Wang J, Hu Z, Huang Y, Zhang Y, Zhang L. Evaluation of left ventricular function in obese patients with obstructive sleep apnea by three-dimensional speckle tracking echocardiography. Int J Cardiovasc Imaging 2022; 38:2311-2322. [DOI: 10.1007/s10554-022-02660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/23/2022] [Indexed: 11/05/2022]
|
10
|
Daneii P, Neshat S, Mirnasiry MS, Moghimi Z, Dehghan Niri F, Farid A, Shekarchizadeh M, Heshmat-Ghahdarijani K. Lipids and diastolic dysfunction: Recent evidence and findings. Nutr Metab Cardiovasc Dis 2022; 32:1343-1352. [PMID: 35428541 DOI: 10.1016/j.numecd.2022.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 11/25/2022]
Abstract
AIM Diastolic dysfunction is the decreased flexibility of the left ventricle due to the impaired ability of the myocardium to relax and plays an important role in the pathogenesis of heart failure. Lipid metabolism is a well-known contributor to cardiac conditions, including ventricular function. In this article, we aimed to review the literature addressing the connections between lipids, their storage, and metabolism with left ventricular diastolic dysfunction. DATA SYNTHESIS We searched Google scholar, Pubmed, Embase and Researchgate for our keywords: "Diastolic function", "Fat" and "Lipid profile". Initially, 250 articles were selected by title and 84 of them were chosen as most relevant and directly reviewed. CONCLUSIONS Alterations of lipid metabolism in cardiac muscle and cardiac lipid content can occur in many conditions, including consumption of a high-fat diet, obesity, metabolic syndrome, and non-alcoholic fatty liver disease (NAFLD). These conditions induce alterations in myocardial lipid metabolism, increase myocardial fat content and epicardial fat thickness and increase inflammation and oxidative stress which ultimately lead to cardiac lipotoxicity and diastolic dysfunction. The effects of lipids on diastolic function can differ based on gender. Lipid profile and metabolism are as important in the pathogenesis of diastolic dysfunction as they are in other cardiovascular disorders. A more careful look at cardiac lipid metabolism in molecular, histological and gross levels results in more precise understanding of its role in myocardial function and leads to development of potential treatments for diastolic dysfunction.
Collapse
Affiliation(s)
- Padideh Daneii
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sina Neshat
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | | - Zahra Moghimi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | | - Armita Farid
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Masood Shekarchizadeh
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Science, Iran
| | - Kiyan Heshmat-Ghahdarijani
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
11
|
Jiang M, Meng Z, Cheng Z, Zhan K, Ma X, Yang T, Huang Y, Yan Q, Gong X, Zhao G. Effects of Buffalo Milk and Cow Milk on Lipid Metabolism in Obese Mice Induced by High Fat. Front Nutr 2022; 9:841800. [PMID: 35558744 PMCID: PMC9089190 DOI: 10.3389/fnut.2022.841800] [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: 12/22/2021] [Accepted: 03/30/2022] [Indexed: 12/25/2022] Open
Abstract
The aim of this study was to evaluate the effects of buffalo milk and cow milk on lipid metabolism in obese mice. Milk composition analysis showed fat, protein, and total solid content in buffalo milk was higher than cow milk, while the lactose content of buffalo milk was lower than cow milk. After milk metabolite extraction and LC-MS/MS analysis, differential metabolites were mainly enriched in “linoleic acid metabolism pathways,” “pentose and glucuronate interconversion pathways,” and “metabolism of xenobiotics by cytochrome P450 pathways.” We fed three groups of C57BL/6J mice (n = 6 per group) for 5 weeks: (1) high-fat diet group (HFD group); (2) high-fat diet + buffalo milk group (HBM group); and (3) high-fat diet + cow milk group (HCM group). Our results showed that body weight of mice was significantly decreased in HBM and HCM groups from 1 to 4 weeks compared with the HFD group. The mRNA expression of ACAA2, ACACB, and SLC27A5 genes involved in the lipid metabolism in liver tissue were significantly elevated in HCM group, relatively to HFD and HBM group. In addition, the adipocyte number, size and lipid accumulation in the liver were significantly decreased in HCM group compared with the HFD group by H&E staining and oil red O staining, but was not change in HBM group. The mRNA levels of TNF-α and IL-1β inflammatory genes were significantly increased in HBM group, relatively to HFD and HCM group, which is consistent with results from inflammatory cell infiltration and tissue disruption by colon tissue sections. In conclusion, dietary supplementation of cow milk has beneficial effects on loss of weight and lipid metabolism in obese mice.
Collapse
Affiliation(s)
- Maocheng Jiang
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Zitong Meng
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Zhiqiang Cheng
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Kang Zhan
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Xiaoyu Ma
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Tianyu Yang
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Yinghao Huang
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Qi Yan
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Xiaoxiao Gong
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China
| | - Guoqi Zhao
- College of Animal Science and Technology, Institute of Animal Culture Collection and Application, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Ministry of Education of China, Yangzhou, China
| |
Collapse
|
12
|
Haidar A, Taegtmeyer H. Strategies for Imaging Metabolic Remodeling of the Heart in Obesity and Heart Failure. Curr Cardiol Rep 2022; 24:327-335. [PMID: 35107704 PMCID: PMC9074778 DOI: 10.1007/s11886-022-01650-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Define early myocardial metabolic changes among patients with obesity and heart failure, and to describe noninvasive methods and their applications for imaging cardiac metabolic remodeling. RECENT FINDINGS Metabolic remodeling precedes, triggers, and sustains functional and structural remodeling in the stressed heart. Alterations in cardiac metabolism can be assessed by using a variety of molecular probes. The glucose tracer analog, 18F-FDG, and the labeled tracer 11C-palmitate are still the most commonly used tracers to assess glucose and fatty acid metabolism, respectively. The development of new tracer analogs and imaging agents, including those targeting the peroxisome proliferator-activated receptor (PPAR), provides new opportunities for imaging metabolic activities at a molecular level. While the use of cardiac magnetic resonance spectroscopy in the clinical setting is limited to the assessment of intramyocardial and epicardial fat, new technical improvements are likely to increase its usage in the setting of heart failure. Noninvasive imaging methods are an effective tool for the serial assessment of alterations in cardiac metabolism, either during disease progression, or in response to treatment.
Collapse
Affiliation(s)
- Amier Haidar
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Dhore-Patil A, Thannoun T, Samson R, Le Jemtel TH. Diabetes Mellitus and Heart Failure With Preserved Ejection Fraction: Role of Obesity. Front Physiol 2022; 12:785879. [PMID: 35242044 PMCID: PMC8886215 DOI: 10.3389/fphys.2021.785879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022] Open
Abstract
Heart failure with preserved ejection fraction is a growing epidemic and accounts for half of all patients with heart failure. Increasing prevalence, morbidity, and clinical inertia have spurred a rethinking of the pathophysiology of heart failure with preserved ejection fraction. Unlike heart failure with reduced ejection fraction, heart failure with preserved ejection fraction has distinct clinical phenotypes. The obese-diabetic phenotype is the most often encountered phenotype in clinical practice and shares the greatest burden of morbidity and mortality. Left ventricular remodeling plays a major role in its pathophysiology. Understanding the interplay of obesity, diabetes mellitus, and inflammation in the pathophysiology of left ventricular remodeling may help in the discovery of new therapeutic targets to improve clinical outcomes in heart failure with preserved ejection fraction. Anti-diabetic agents like glucagon-like-peptide 1 analogs and sodium-glucose co-transporter 2 are promising therapeutic modalities for the obese-diabetic phenotype of heart failure with preserved ejection fraction and aggressive weight loss via lifestyle or bariatric surgery is still key to reverse adverse left ventricular remodeling. This review focuses on the obese-diabetic phenotype of heart failure with preserved ejection fraction highlighting the interaction between obesity, diabetes, and coronary microvascular dysfunction in the development and progression of left ventricular remodeling. Recent therapeutic advances are reviewed.
Collapse
Affiliation(s)
- Aneesh Dhore-Patil
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States.,Tulane University Heart and Vascular Institute, New Orleans, LA, United States
| | - Tariq Thannoun
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States.,Tulane University Heart and Vascular Institute, New Orleans, LA, United States
| | - Rohan Samson
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States.,Tulane University Heart and Vascular Institute, New Orleans, LA, United States
| | - Thierry H Le Jemtel
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States.,Tulane University Heart and Vascular Institute, New Orleans, LA, United States
| |
Collapse
|
14
|
Telomere Length, Apoptotic, and Inflammatory Genes: Novel Biomarkers of Gastrointestinal Tract Pathology and Meat Quality Traits in Chickens under Chronic Stress ( Gallus gallus domesticus). Animals (Basel) 2021; 11:ani11113276. [PMID: 34828008 PMCID: PMC8614256 DOI: 10.3390/ani11113276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/16/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The assessment of poultry’s gastrointestinal (GI) tract and meat quality traits are crucial for sustainable poultry production in the tropics. The search for well-conserved and more reliable biomarkers for the GI tract and meat traits has faced many challenges. In this study, we observed the effect of corticosterone (CORT) and age on body weight, buffy coat telomere length, GI tract, and meat quality traits. The critical evaluation of the GI tract and meat traits in this study revealed that telomere length, mitochondria, and acute phase protein genes were altered by chronic stress and were associated with the traits. This study informed us of the potential of telomere length, mitochondria, and acute phase protein genes in the assessment of GI tract pathological conditions and meat quality in the poultry sector for sustainable production. Abstract This study was designed to examine the potentials of telomere length, mitochondria, and acute phase protein genes as novel biomarkers of gastrointestinal (GI) tract pathologies and meat quality traits. Chickens were fed a diet containing corticosterone (CORT) for 4 weeks and records on body weight, telomere length, GI tract and muscle histopathological test, meat quality traits, mitochondria, and acute phase protein genes were obtained at weeks 4 and 6 of age. The body weight of CORT-fed chickens was significantly suppressed (p < 0.05). CORT significantly altered the GI tract and meat quality traits. The interaction effect of CORT and age on body weight, duodenum and ileum crypt depth, pH, and meat color was significant (p < 0.05). CORT significantly (p < 0.05) shortened buffy coat telomere length. UCP3 and COX6A1 were diversely and significantly expressed in the muscle, liver, and heart of the CORT-fed chicken. Significant expression of SAAL1 and CRP in the liver and hypothalamus of the CORT-fed chickens was observed at week 4 and 6. Therefore, telomere lengths, mitochondria, and acute phase protein genes could be used as novel biomarkers for GI tract pathologies and meat quality traits.
Collapse
|
15
|
Piché ME, Clavel MA, Auclair A, Rodríguez-Flores M, O'Connor K, Garceau P, Rakowski H, Poirier P. Early benefits of bariatric surgery on subclinical cardiac function: Contribution of visceral fat mobilization. Metabolism 2021; 119:154773. [PMID: 33838144 DOI: 10.1016/j.metabol.2021.154773] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/10/2021] [Accepted: 04/01/2021] [Indexed: 01/01/2023]
Abstract
AIMS We explored the early effects of bariatric surgery on subclinical myocardial function in individuals with severe obesity and preserved left ventricular (LV) ejection fraction. METHODS Thirty-eight patients with severe obesity [body mass index (BMI) ≥35 kg/m2] and preserved LV ejection fraction (≥50%) who underwent bariatric surgery (biliopancreatic diversion with duodenal switch [BPD-DS]) (Surgery group), 19 patients with severe obesity managed with usual care (Medical group), and 18 age and sex-matched non-obese controls (non-obese group) were included. Left ventricular global longitudinal strain (LV GLS) was evaluated with echocardiography speckle tracking imaging. Abnormal myocardial function was defined as LV GLS <18%. RESULTS Age of the participants was 42 ± 11 years with a BMI of 48 ± 8 kg/m2 (mean ± standard deviation); 82% were female. The percentage of total weight loss at 6 months after bariatric surgery was 26.3 ± 5.2%. Proportions of hypertension (61 vs. 30%, P = 0.0005), dyslipidemia (42 vs. 5%, P = 0.0001) and type 2 diabetes (40 vs. 13%, P = 0.002) were reduced postoperatively. Before surgery, patients with obesity displayed abnormal subclinical myocardial function vs. non-obese controls (LV GLS, 16.3 ± 2.5 vs. 19.6 ± 1.7%, P < 0.001). Six months after bariatric surgery, the subclinical myocardial function was comparable to non-obese (LV GLS, 18.2 ± 1.9 vs. 19.6 ± 1.7%, surgery vs. non-obese, P = NS). On the contrary, half of individuals with obesity managed medically worsened their myocardial function during the follow-up (P = 0.002). Improvement in subclinical myocardial function following bariatric surgery was associated with changes in abdominal visceral fat (r = 0.43, P < 0.05) and inflammatory markers (r = 0.45, P < 0.01), whereas no significant association was found with weight loss or change in insulin sensitivity (HOMA-IR) (P > 0.05). In a multivariate model, losing visceral fat mass was independently associated with improved subclinical myocardial function. CONCLUSIONS Bariatric surgery was associated with significant improvement in the metabolic profile and in subclinical myocardial function. Early improvement in subclinical myocardial function following bariatric surgery was related to a greater mobilization of visceral fat depot, linked to global fat dysfunction and cardiometabolic morbidity.
Collapse
Affiliation(s)
- Marie-Eve Piché
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Canada; Faculty of Medicine, Laval University, Québec, Canada
| | - Marie-Annick Clavel
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Canada; Faculty of Medicine, Laval University, Québec, Canada
| | - Audrey Auclair
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Canada
| | - Marcela Rodríguez-Flores
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Canada; Instituto Nacional de Ciencias Médicas y Nutrición, Mexico City, Mexico
| | - Kim O'Connor
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Canada; Faculty of Medicine, Laval University, Québec, Canada
| | - Patrick Garceau
- Faculty of Medicine, Montreal University, Montréal, Canada; Institut de Cardiologie de Montréal, Montréal, Canada
| | | | - Paul Poirier
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Canada; Faculty of Pharmacy, Laval University, Québec, Canada.
| |
Collapse
|
16
|
Sangweni NF, Mosa RA, Dludla PV, Kappo AP, Opoku AR, Muller CJF, Johnson R. The triterpene, methyl-3β-hydroxylanosta-9,24-dien-21-oate (RA3), attenuates high glucose-induced oxidative damage and apoptosis by improving energy metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153546. [PMID: 33799221 DOI: 10.1016/j.phymed.2021.153546] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/11/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Hyperglycemia-induced cardiovascular dysfunction has been linked to oxidative stress and accelerated apoptosis in the diabetic myocardium. While there is currently no treatment for diabetic cardiomyopathy (DCM), studies suggest that the combinational use of anti-hyperglycemic agents and triterpenes could be effective in alleviating DCM. HYPOTHESIS To investigate the therapeutic effect of methyl-3β-hydroxylanosta-9,24-dien-21-oate (RA3), in the absence or presence of the anti-diabetic drug, metformin (MET), against hyperglycemia-induced cardiac injury using an in vitro H9c2 cell model. METHODS To mimic a hyperglycemic state, H9c2 cells were exposed to high glucose (HG, 33 mM) for 24 h. Thereafter, the cells were treated with RA3 (1 μM), MET (1 μM) and the combination of MET (1 μM) plus RA3 (1 μM) for 24 h, to assess the treatments therapeutic effect. RESULTS Biochemical analysis revealed that RA3, with or without MET, improves glucose uptake via insulin-dependent (IRS-1/PI3K/Akt signaling) and independent (AMPK) pathways whilst ameliorating the activity of antioxidant enzymes in the H9c2 cells. Mechanistically, RA3 was able to alleviate HG-stimulated oxidative stress through the inhibition of reactive oxygen species (ROS) and lipid peroxidation as well as the reduced expression of the PKC/NF-кB cascade through decreased intracellular lipid content. Subsequently, RA3 was able to mitigate HG-induced apoptosis by decreasing the activity of caspase 3/7 and DNA fragmentation in the cardiomyoblasts. CONCLUSION RA3, in the absence or presence of MET, demonstrated potent therapeutic properties against hyperglycemia-mediated cardiac damage and could be a suitable candidate in the prevention of DCM.
Collapse
Affiliation(s)
- Nonhlakanipho F Sangweni
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg 7505, South Africa; Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| | - Rebamang A Mosa
- Department of Biochemistry, Genetics and Microbiology (BGM), Division of Biochemistry, University of Pretoria, Hatfield 0028, South Africa
| | - Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg 7505, South Africa; Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy.
| | - Abidemi P Kappo
- Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park 2006, South Africa
| | - Andy R Opoku
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg 7505, South Africa; Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa.
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg 7505, South Africa; Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.
| |
Collapse
|
17
|
Mancusi C, Midtbø H, De Luca N, Halland H, de Simone G, Gerdts E. Association of Myocardial Energetic Efficiency with Circumferential and Longitudinal Left Ventricular Myocardial Function in Subjects with Increased Body Mass Index (the FATCOR Study). J Clin Med 2021; 10:jcm10081581. [PMID: 33918093 PMCID: PMC8069856 DOI: 10.3390/jcm10081581] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Lower myocardial mechanic-energetic efficiency (MEEi), expressed as stroke volume/heart rate ratio (SV/HR) in mL/s/g of the left ventricular (LV) mass, is associated with the incidence of heart failure in subjects with cardiometabolic disorders. We explored the association of MEEi with LV systolic circumferential and longitudinal myocardial function in 480 subjects with increased body mass index (BMI) without known cardiovascular disease (mean age 47 ± 9 years, 61% women, 63% obese, 74% with hypertension) participating in the fat-associated cardiovascular dysfunction (FATCOR) study. Insulin resistance was assessed by the homeostasis model assessment insulin-resistance index (HOMA-IR). SV was calculated by Doppler echocardiography. The LV systolic circumferential myocardial function was evaluated by midwall fractional shortening (MFS) and longitudinal function by global longitudinal strain (GLS). Patients were grouped into MEEi quartiles. The lowest MEEi quartile (<0.41 mL/s per g) was considered low MEEi. The association of MEEi with MFS and GLS were tested in multivariable linear regression analyses. Patients with low MEEi were more frequently men, with obesity and hypertension, dyslipidemia and higher HOMA-IR index (all p for trend <0.05). In multivariable analyses, lower MEEi was associated with lower LV myocardial function by MFS and GLS independent of higher LV mass and clinical variables, including older age, male sex, presence of hypertension and a higher triglycerides level (all p < 0.05). In conclusion, in subjects with increased BMI without known cardiovascular disease participating in the FATCOR study, reduced MEEi was associated with lower LV myocardial function both in the circumferential and longitudinal direction, independent of cardiometabolic factors.
Collapse
Affiliation(s)
- Costantino Mancusi
- Hypertension Research Center, Department of Advanced Biomedical Sciences, University Federico II of Naples, 80131 Naples, Italy; (N.D.L.); (G.d.S.)
- Correspondence: ; Tel.: +39-081-746-211
| | - Helga Midtbø
- Department of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway; (H.M.); (H.H.); (E.G.)
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Nicola De Luca
- Hypertension Research Center, Department of Advanced Biomedical Sciences, University Federico II of Naples, 80131 Naples, Italy; (N.D.L.); (G.d.S.)
| | - Hilde Halland
- Department of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway; (H.M.); (H.H.); (E.G.)
| | - Giovanni de Simone
- Hypertension Research Center, Department of Advanced Biomedical Sciences, University Federico II of Naples, 80131 Naples, Italy; (N.D.L.); (G.d.S.)
| | - Eva Gerdts
- Department of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway; (H.M.); (H.H.); (E.G.)
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
18
|
Paiman EHM, de Mutsert R, Widya RL, Rosendaal FR, Jukema JW, Lamb HJ. The role of insulin resistance in the relation of visceral, abdominal subcutaneous and total body fat to cardiovascular function. Nutr Metab Cardiovasc Dis 2020; 30:2230-2241. [PMID: 32912791 DOI: 10.1016/j.numecd.2020.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/30/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS The separate cardiovascular effects of type 2 diabetes and adiposity remain to be examined. This study aimed to investigate the role of insulin resistance in the relations of visceral (VAT), abdominal subcutaneous (aSAT) adipose tissue and total body fat (TBF) to cardiovascular remodeling. METHODS AND RESULTS In this cross-sectional analysis of the population-based Netherlands Epidemiology of Obesity study, 914 middle-aged individuals (46% men) were included. Participants underwent magnetic resonance imaging. Standardized linear regression coefficients (95%CI) were calculated, adjusted for potential confounding factors. All fat depots and insulin resistance (HOMA-IR), separate from VAT and TBF, were associated with lower mitral early and late peak filling rate ratios (E/A): -0.04 (-0.09;0.01) per SD (54 cm2) VAT; -0.05 (-0.10;0.00) per SD (94 cm2) aSAT; -0.09 (-0.16;-0.02) per SD (8%) TBF; -0.11 (-0.17;-0.05) per 10-fold increase in HOMA-IR, whereas VAT and TBF were differently associated with left ventricular (LV) end-diastolic volume: -8.9 (-11.7;-6.1) mL per SD VAT; +5.4 (1.1;9.7) mL per SD TBF. After adding HOMA-IR to the model to evaluate the mediating role of insulin resistance, change in E/A was -0.02 (-0.07;0.04) per SD VAT; -0.03 (-0.08;0.02) per SD aSAT; -0.06 (-0.13;0.01) per SD TBF, and change in LV end-diastolic volume was -7.0 (-9.7;-4.3) mL per SD VAT. In women, adiposity but not HOMA-IR was related to higher aortic arch pulse wave velocity. CONCLUSION Insulin resistance was associated with reduced diastolic function, separately from VAT and TBF, and partly mediated the associations between adiposity depots and lower diastolic function.
Collapse
Affiliation(s)
- Elisabeth H M Paiman
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ralph L Widya
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
19
|
Song Y, Qi Z, Zhang Y, Wei J, Liao X, Li R, Dong C, Zhu L, Yang Z, Cai Z. Effects of exposure to ambient fine particulate matter on the heart of diet-induced obesity mouse model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 732:139304. [PMID: 32438171 DOI: 10.1016/j.scitotenv.2020.139304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 06/11/2023]
Abstract
Exposure to fine particulate matter (PM2.5) is associated with decreased cardiac function, especially in high risk populations such as obese ones. In this study, impacts of PM2.5 exposure on cardiac function were investigated by using the diet-induced obesity mice model. Mice were fed with normal diet or high-fat diet (HFD) for four weeks and then exposed to phosphate-buffered solution or Taiyuan winter PM2.5 (0.25 mg/kg body/day) through intratracheal instillation for another four weeks. Among physiological indices recorded, heart rate and blood pressure were increased after PM2.5 exposure in the heart of the obese mice. Metabolomics and lipidomics were applied to explore molecular alterations in response to the co-treatment of PM2.5 and HFD. Our results demonstrated both direct impacts on cardiac function and indirect effects resulted from the injury of other organs. Inflammation of lung and hypothalamus may be responsible for the elevation of phenylalanine metabolism in serum and its downstream products: epinephrine and norepinephrine, the catecholamines involves in regulating cardiac system. In intracardiac system, the co-treatment led to imbalance of energy metabolism, in addition to oxidative stress and inflammation. In contrast to the upregulation of glucose and fatty acids uptake and CoA synthesis, levels of ATP, acetyl-CoA and the intermediates in glycolysis pathway decreased in the heart. The results indicated that energy metabolism disorder was possibly one of the important contributing factors to the more severe adverse effects of the combined treatment of HFD and PM2.5.
Collapse
Affiliation(s)
- Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zenghua Qi
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Yanhao Zhang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Juntong Wei
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoliang Liao
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Lin Zhu
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
20
|
Cardiac Transcriptome Analysis Reveals Nr4a1 Mediated Glucose Metabolism Dysregulation in Response to High-Fat Diet. Genes (Basel) 2020; 11:genes11070720. [PMID: 32610475 PMCID: PMC7397175 DOI: 10.3390/genes11070720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/23/2022] Open
Abstract
Obesity is associated with an increased risk of developing cardiovascular disease (CVD), with limited alterations in cardiac genomic characteristics known. Cardiac transcriptome analysis was conducted to profile gene signatures in high-fat diet (HFD)-induced obese mice. A total of 184 differentially expressed genes (DEGs) were identified between groups. Based on the gene ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs, the critical role of closely interlocked glucose metabolism was determined in HFD-induced cardiac remodeling DEGs, including Nr4a1, Fgf21, Slc2a3, Pck1, Gck, Hmgcs2, and Bpgm. Subsequently, the expression levels of these DEGs were evaluated in both the myocardium and palmitic acid (PA)-stimulated H9c2 cardiomyocytes using qPCR. Nr4a1 was highlighted according to its overexpression resulting from the HFD. Additionally, inhibition of Nr4a1 by siRNA reversed the PA-induced altered expression of glucose metabolism-related DEGs and hexokinase 2 (HK2), the rate-limiting enzyme in glycolysis, thus indicating that Nr4a1 could modulate glucose metabolism homeostasis by regulating the expression of key enzymes in glycolysis, which may subsequently influence cardiac function in obesity. Overall, we provide a comprehensive understanding of the myocardium transcript molecular framework influenced by HFD and propose Nr4a1 as a key glucose metabolism target in obesity-induced CVD.
Collapse
|
21
|
Abstract
This review addresses the interplay between obesity, type 2 diabetes mellitus, and cardiovascular diseases. It is proposed that obesity, generally defined by an excess of body fat causing prejudice to health, can no longer be evaluated solely by the body mass index (expressed in kg/m2) because it represents a heterogeneous entity. For instance, several cardiometabolic imaging studies have shown that some individuals who have a normal weight or who are overweight are at high risk if they have an excess of visceral adipose tissue-a condition often accompanied by accumulation of fat in normally lean tissues (ectopic fat deposition in liver, heart, skeletal muscle, etc). On the other hand, individuals who are overweight or obese can nevertheless be at much lower risk than expected when faced with excess energy intake if they have the ability to expand their subcutaneous adipose tissue mass, particularly in the gluteal-femoral area. Hence, excessive amounts of visceral adipose tissue and of ectopic fat largely define the cardiovascular disease risk of overweight and moderate obesity. There is also a rapidly expanding subgroup of patients characterized by a high accumulation of body fat (severe obesity). Severe obesity is characterized by specific additional cardiovascular health issues that should receive attention. Because of the difficulties of normalizing body fat content in patients with severe obesity, more aggressive treatments have been studied in this subgroup of individuals such as obesity surgery, also referred to as metabolic surgery. On the basis of the above, we propose that we should refer to obesities rather than obesity.
Collapse
Affiliation(s)
- Marie-Eve Piché
- From the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.,Department of Medicine, Faculty of Medicine (M.-E.P.), Université Laval, Québec, QC, Canada
| | - André Tchernof
- From the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.,School of Nutrition (A.T.), Université Laval, Québec, QC, Canada
| | - Jean-Pierre Després
- From the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.,Vitam - Centre de recherche en santé durable, CIUSSS - Capitale-Nationale (J.-P.D.), Université Laval, Québec, QC, Canada.,Department of Kinesiology, Faculty of Medicine (J.-P.D.), Université Laval, Québec, QC, Canada
| |
Collapse
|
22
|
Paternal Resistance Training Induced Modifications in the Left Ventricle Proteome Independent of Offspring Diet. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5603580. [PMID: 32454941 PMCID: PMC7218999 DOI: 10.1155/2020/5603580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/18/2019] [Indexed: 01/13/2023]
Abstract
Ancestral obesogenic exposure is able to trigger harmful effects in the offspring left ventricle (LV) which could lead to cardiovascular diseases. However, the impact of the father's lifestyle on the offspring LV is largely unexplored. The aim of this study was to investigate the effects of 8 weeks of paternal resistance training (RT) on the offspring left ventricle (LV) proteome exposed to control or high-fat (HF) diet. Wistar rats were randomly divided into two groups: sedentary fathers and trained fathers (8 weeks, 3 times per week with weights secured to the animals' tails). The offspring were obtained by mating with sedentary females. Upon weaning, male offspring were divided into 4 groups (5 animals per group): offspring from sedentary fathers, exposed to control diet (SFO-C); offspring from trained fathers, exposed to control diet (TFO-C); offspring from sedentary fathers, exposed to high-fat diet (SFO-HF); and offspring from trained fathers, exposed to high-fat diet (TFO-HF). The LC-MS/MS analysis revealed 537 regulated proteins among groups. Offspring exposure to HF diet caused reduction in the abundance levels of proteins related to cell component organization, metabolic processes, and transport. Proteins related to antioxidant activity, transport, and transcription regulation were increased in TFO-C and TFO-HF as compared with the SFO-C and SFO-HF groups. Paternal RT demonstrated to be an important intervention capable of inducing significant effects on the LV proteome regardless of offspring diet due to the increase of proteins involved into LV homeostasis maintenance. This study contributes to a better understanding of the molecular aspects involved in transgenerational inheritance.
Collapse
|
23
|
Zaric B, Obradovic M, Trpkovic A, Banach M, Mikhailidis DP, Isenovic ER. Endothelial Dysfunction in Dyslipidaemia: Molecular Mechanisms and Clinical Implications. Curr Med Chem 2020; 27:1021-1040. [DOI: 10.2174/0929867326666190903112146] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022]
Abstract
The endothelium consists of a monolayer of Endothelial Cells (ECs) which form
the inner cellular lining of veins, arteries, capillaries and lymphatic vessels. ECs interact with
the blood and lymph. The endothelium fulfils functions such as vasodilatation, regulation of
adhesion, infiltration of leukocytes, inhibition of platelet adhesion, vessel remodeling and
lipoprotein metabolism. ECs synthesize and release compounds such as Nitric Oxide (NO),
metabolites of arachidonic acid, Reactive Oxygen Species (ROS) and enzymes that degrade
the extracellular matrix. Endothelial dysfunction represents a phenotype prone to atherogenesis
and may be used as a marker of atherosclerotic risk. Such dysfunction includes impaired
synthesis and availability of NO and an imbalance in the relative contribution of endothelialderived
relaxing factors and contracting factors such as endothelin-1 and angiotensin. This
dysfunction appears before the earliest anatomic evidence of atherosclerosis and could be an
important initial step in further development of atherosclerosis. Endothelial dysfunction was
historically treated with vitamin C supplementation and L-arginine supplementation. Short
term improvement of the expression of adhesion molecule and endothelial function during
antioxidant therapy has been observed. Statins are used in the treatment of hyperlipidaemia, a
risk factor for cardiovascular disease. Future studies should focus on identifying the mechanisms
involved in the beneficial effects of statins on the endothelium. This may help develop
drugs specifically aimed at endothelial dysfunction.
Collapse
Affiliation(s)
- Bozidarka Zaric
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Milan Obradovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland
| | - Dimitri P. Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, United Kingdom
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| |
Collapse
|
24
|
Chen B, Shi X, Cui Y, Hou A, Zhao P. A Review of PCSK9 Inhibitors and their Effects on Cardiovascular Diseases. Curr Top Med Chem 2019; 19:1790-1817. [PMID: 31400268 DOI: 10.2174/1568026619666190809094203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/07/2019] [Accepted: 07/25/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiovascular diseases remain the leading cause of morbidity and mortality in the world, with elevated Low-Density Lipoprotein-Cholesterol (LDL-C) levels as the major risk factor. Lower levels of LDL-C can effectively reduce the risk of cardiovascular diseases. Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in regulating the degradation of hepatic LDL receptors that remove LDL-C from the circulation. PCSK9 inhibitors are a new class of agents that are becoming increasingly important in the treatment to reduce LDL-C levels. Two PCSK9 inhibitors, alirocumab and evolocumab, have been approved to treat hypercholesterolemia and are available in the United States and the European Union. Through the inhibition of PCSK9 and increased recycling of LDL receptors, serum LDL-C levels can be significantly reduced. OBJECTIVE This review will describe the chemistry, pharmacokinetics, and pharmacodynamics of PCSK9 inhibitors and their clinical effects.
Collapse
Affiliation(s)
- Bo Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 20092, China
| | - Xin Shi
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 20092, China
| | - Yanping Cui
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 20092, China
| | - Aiping Hou
- Department of Pediatric, Shidong Hospital, Shanghai 20092, China
| | - Pengjun Zhao
- Department of Pediatric, Shidong Hospital, Shanghai 20092, China
| |
Collapse
|
25
|
Filardi T, Ghinassi B, Di Baldassarre A, Tanzilli G, Morano S, Lenzi A, Basili S, Crescioli C. Cardiomyopathy Associated with Diabetes: The Central Role of the Cardiomyocyte. Int J Mol Sci 2019; 20:ijms20133299. [PMID: 31284374 PMCID: PMC6651183 DOI: 10.3390/ijms20133299] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
Abstract
The term diabetic cardiomyopathy (DCM) labels an abnormal cardiac structure and performance due to intrinsic heart muscle malfunction, independently of other vascular co-morbidity. DCM, accounting for 50%–80% of deaths in diabetic patients, represents a worldwide problem for human health and related economics. Optimal glycemic control is not sufficient to prevent DCM, which derives from heart remodeling and geometrical changes, with both consequences of critical events initially occurring at the cardiomyocyte level. Cardiac cells, under hyperglycemia, very early undergo metabolic abnormalities and contribute to T helper (Th)-driven inflammatory perturbation, behaving as immunoactive units capable of releasing critical biomediators, such as cytokines and chemokines. This paper aims to focus onto the role of cardiomyocytes, no longer considered as “passive” targets but as “active” units participating in the inflammatory dialogue between local and systemic counterparts underlying DCM development and maintenance. Some of the main biomolecular/metabolic/inflammatory processes triggered within cardiac cells by high glucose are overviewed; particular attention is addressed to early inflammatory cytokines and chemokines, representing potential therapeutic targets for a prompt early intervention when no signs or symptoms of DCM are manifesting yet. DCM clinical management still represents a challenge and further translational investigations, including studies at female/male cell level, are warranted.
Collapse
Affiliation(s)
- Tiziana Filardi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Susanna Morano
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", Piazza L. de Bosis 6, 00135 Rome, Italy.
| |
Collapse
|
26
|
Ortega-Loubon C, Fernández-Molina M, Singh G, Correa R. Obesity and its cardiovascular effects. Diabetes Metab Res Rev 2019; 35:e3135. [PMID: 30715772 DOI: 10.1002/dmrr.3135] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/21/2022]
Abstract
Obesity is described in terms of body fat percentage or body mass index (BMI), despite the fact that these measures do not give full insight about the body fat distribution. It is presently a consistently growing universal challenge since it has tripled in the last 10 years, killing approximately 28 million people each year. In this review, we aim to clarify the different results of obesity on the working and physiology of the cardiovascular system and to reveal changes in the obesity "paradox"-a variety of cardiovascular outcomes in typical/overweight people. Central fat build-up in ordinary/overweight populaces has been related to expanded occurrences of myocardial infarction, heart failure, or all-cause mortality when contrasted with the obese populace. These discoveries are additionally clarified as the abundance and prolonged vulnerability to free fatty acids (FFAs) in obesity. This has been believed to cause the myocardial substrate to move from glucose to FFAs digestion, which causes lipid gathering in cardiomyocytes, spilling over to other lean tissues, and prompting a general atherogenic impact. This cardiomyocyte lipid aggregation has been demonstrated to cause insulin resistance and cardiovascular hypertrophy, and to lessen the heart functions in general. There is a proof backing the fact that fat tissue is not only an energy reservoir, it also coordinates hormones and proinflammatory cytokines and deals with the energy transition of the body by putting away abundant lipids in diverse tissues.
Collapse
Affiliation(s)
- Christian Ortega-Loubon
- Department of Pediatric Cardiac Surgery, Universidad Autonoma de Barcelona, Barcelona, Spain
| | | | - Gauri Singh
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Ricardo Correa
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| |
Collapse
|
27
|
Le Jemtel TH, Samson R, Ayinapudi K, Singh T, Oparil S. Epicardial Adipose Tissue and Cardiovascular Disease. Curr Hypertens Rep 2019; 21:36. [PMID: 30953236 DOI: 10.1007/s11906-019-0939-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Epicardial adipose tissue has been associated with the development/progression of cardiovascular disease. We appraise the strength of the association between epicardial adipose tissue and development/progression of cardiovascular diseases like coronary artery disease, atrial fibrillation, and heart failure with preserved ejection fraction. RECENT FINDINGS Cross-sectional clinical and translational correlative studies have established an association between epicardial adipose tissue and progression of coronary artery disease. Recent studies question this association and underline the need for longitudinal studies. Epicardial adipose tissue also plays a definite role in the pathobiology of atrial fibrillation and its recurrence after ablation. In contrast to an early paradigm, epicardial adipose tissue does not appear to play a key role in the pathogenesis of heart failure with preserved ejection fraction in obese patients. The association of epicardial adipose tissue with atrial fibrillation is robust. In contrast, the association of epicardial adipose tissue with coronary artery disease and heart failure with preserved ejection fraction is tenuous. Additional research, including longitudinal studies, is needed to confirm or refute these proposed associations.
Collapse
Affiliation(s)
- Thierry H Le Jemtel
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine; Tulane University Heart and Vascular Institute, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA.
| | - Rohan Samson
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine; Tulane University Heart and Vascular Institute, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA
| | - Karnika Ayinapudi
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine; Tulane University Heart and Vascular Institute, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA
| | - Twinkle Singh
- Section of Cardiology, Department of Medicine, Tulane University School of Medicine; Tulane University Heart and Vascular Institute, 1430 Tulane Avenue, SL-48, New Orleans, LA, 70112, USA
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
28
|
Granéli C, Hicks R, Brolén G, Synnergren J, Sartipy P. Diabetic Cardiomyopathy Modelling Using Induced Pluripotent Stem Cell Derived Cardiomyocytes: Recent Advances and Emerging Models. Stem Cell Rev Rep 2019; 15:13-22. [PMID: 30343468 PMCID: PMC6513824 DOI: 10.1007/s12015-018-9858-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The global burden of diabetes has drastically increased over the past decades and in 2017 approximately 4 million deaths were caused by diabetes and cardiovascular complications. Diabetic cardiomyopathy is a common complication of diabetes with early manifestations of diastolic dysfunction and left ventricular hypertrophy with subsequent progression to systolic dysfunction and ultimately heart failure. An in vitro model accurately recapitulating key processes of diabetic cardiomyopathy would provide a useful tool for investigations of underlying disease mechanisms to further our understanding of the disease and thereby potentially advance treatment strategies for patients. With their proliferative capacity and differentiation potential, human induced pluripotent stem cells (iPSCs) represent an appealing cell source for such a model system and cardiomyocytes derived from induced pluripotent stem cells have been used to establish other cardiovascular related disease models. Here we review recently made advances and discuss challenges still to be overcome with regard to diabetic cardiomyopathy models, with a special focus on iPSC-based systems. Recent publications as well as preliminary data presented here demonstrate the feasibility of generating cardiomyocytes with a diabetic phenotype, displaying insulin resistance, impaired calcium handling and hypertrophy. However, capturing the full metabolic- and functional phenotype of the diabetic cardiomyocyte remains to be accomplished.
Collapse
Affiliation(s)
- Cecilia Granéli
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28, Skövde, Sweden.
- Discovery Sciences, IMED Biotech Unit, AstraZeneca Gothenburg, SE-431 50, Mölndal, Sweden.
| | - Ryan Hicks
- Discovery Sciences, IMED Biotech Unit, AstraZeneca Gothenburg, SE-431 50, Mölndal, Sweden
| | - Gabriella Brolén
- Discovery Sciences, IMED Biotech Unit, AstraZeneca Gothenburg, SE-431 50, Mölndal, Sweden
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28, Skövde, Sweden
| | - Peter Sartipy
- Systems Biology Research Center, School of Bioscience, University of Skövde, SE-541 28, Skövde, Sweden
- Global Medicines Development, CVRM, AstraZeneca Gothenburg, SE-431 50, Mölndal, Sweden
| |
Collapse
|
29
|
Understanding the Role of Obesity Trajectories in the Development of Left Ventricular Dysfunction: Another Step on the Path to Heart Failure Screening? J Am Soc Echocardiogr 2018; 31:1270-1271. [PMID: 30522603 DOI: 10.1016/j.echo.2018.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Indexed: 11/23/2022]
|
30
|
Borghetti G, von Lewinski D, Eaton DM, Sourij H, Houser SR, Wallner M. Diabetic Cardiomyopathy: Current and Future Therapies. Beyond Glycemic Control. Front Physiol 2018; 9:1514. [PMID: 30425649 PMCID: PMC6218509 DOI: 10.3389/fphys.2018.01514] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus and the associated complications represent a global burden on human health and economics. Cardiovascular diseases are the leading cause of death in diabetic patients, who have a 2–5 times higher risk of developing heart failure than age-matched non-diabetic patients, independent of other comorbidities. Diabetic cardiomyopathy is defined as the presence of abnormal cardiac structure and performance in the absence of other cardiac risk factors, such coronary artery disease, hypertension, and significant valvular disease. Hyperglycemia, hyperinsulinemia, and insulin resistance mediate the pathological remodeling of the heart, characterized by left ventricle concentric hypertrophy and perivascular and interstitial fibrosis leading to diastolic dysfunction. A change in the metabolic status, impaired calcium homeostasis and energy production, increased inflammation and oxidative stress, as well as an accumulation of advanced glycation end products are among the mechanisms implicated in the pathogenesis of diabetic cardiomyopathy. Despite a growing interest in the pathophysiology of diabetic cardiomyopathy, there are no specific guidelines for diagnosing patients or structuring a treatment strategy in clinical practice. Anti-hyperglycemic drugs are crucial in the management of diabetes by effectively reducing microvascular complications, preventing renal failure, retinopathy, and nerve damage. Interestingly, several drugs currently in use can improve cardiac health beyond their ability to control glycemia. GLP-1 receptor agonists and sodium-glucose co-transporter 2 inhibitors have been shown to have a beneficial effect on the cardiovascular system through a direct effect on myocardium, beyond their ability to lower blood glucose levels. In recent years, great improvements have been made toward the possibility of modulating the expression of specific cardiac genes or non-coding RNAs in vivo for therapeutic purpose, opening up the possibility to regulate the expression of key players in the development/progression of diabetic cardiomyopathy. This review summarizes the pathogenesis of diabetic cardiomyopathy, with particular focus on structural and molecular abnormalities occurring during its progression, as well as both current and potential future therapies.
Collapse
Affiliation(s)
- Giulia Borghetti
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Dirk von Lewinski
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Deborah M Eaton
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Markus Wallner
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
31
|
Abstract
Aging and diabetes mellitus are 2 well-known risk factors for cardiovascular disease (CVD). During the past 50 years, there has been an dramatic increase in life expectancy with a simultaneous increase in the prevalence of diabetes mellitus in the older population. This large number of older individuals with diabetes mellitus is problematic given that CVD risk associated with aging and diabetes mellitus. In this review, we summarize epidemiological data relating to diabetes mellitus and CVD, with an emphasis on the aging population. We then present data on hyperglycemia as a risk factor for CVD and review the current knowledge of age-related changes in glucose metabolism. Next, we review the role of obesity in the pathogenesis of age-related glucose dysregulation, followed by a summary of the results from major randomized controlled trials that focus on cardiovascular risk reduction through glycemic control, with a special emphasis on older adults. We then conclude with our proposed model of aging that body composition changes and insulin resistance link possible dysregulation of physiological pathways leading to obesity and diabetes mellitus-both forms of accelerated aging-and risks for CVD.
Collapse
Affiliation(s)
- Chee W Chia
- From the Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Josephine M Egan
- From the Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Luigi Ferrucci
- From the Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| |
Collapse
|
32
|
Chen D, Li X, Zhang L, Zhu M, Gao L. A high-fat diet impairs mitochondrial biogenesis, mitochondrial dynamics, and the respiratory chain complex in rat myocardial tissues. J Cell Biochem 2018; 119:9602. [PMID: 30171706 PMCID: PMC6220867 DOI: 10.1002/jcb.27068] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 04/23/2018] [Indexed: 12/15/2022]
Abstract
A high‐fat diet (HFD) has been associated with heart failure and arrhythmias; however, the molecular mechanisms underlying these associations are poorly understood. The mitochondria play an essential role in optimal heart performance, most of the energy for which is obtained from the oxidation of fatty acids. As such, chronic exposure to excess fatty acids may cause mitochondrial dysfunction and heart failure. To investigate the effects of a HFD on the mitochondrial function in the myocardium, 40 male rats were randomly divided into two groups and fed with either a normal diet or a HFD for 28 weeks. The myocardial lipid content, cardiac parameters and function, and mitochondrial morphology and function were evaluated. The expression of a number of genes involved in mitochondrial dynamics was measured using quantitative polymerase chain reaction and Western blot analyses. Proteomic analysis was also performed to identify the proteins affected by HFD treatment. Significant fat deposition in the myocardia, cardiac hypertrophy, and cardiac dysfunction were all observed in HFD‐treated rats. Electron microscopy showed abnormal mitochondrial density and morphology. In addition, abnormal expression of genes involved in mitochondrial dynamics, decreased mitochondrial DNA copy numbers, reduced complex I‐III and citrate synthase activities, and decreased mitochondrial respiration were observed in HFD‐treated rats. High performance liquid chromatography showed downregulated adenosine triphosphate (ATP) and adenosine diphosphate levels and an increased adenosine monophosphate (AMP)/ATP ratio. Proteomic analysis confirmed the alteration of mitochondrial function and impaired expression of proteins involved in mitochondrial dynamics in HFD‐treated rats. Mitochondrial dysfunction and impaired mitochondrial dynamics play an important role in heart dysfunction induced by a HFD, thus presenting a potential therapeutic target for the treatment of heart disease.
Collapse
Affiliation(s)
- Dan Chen
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China.,Department of Electrocardiographic, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Xia Li
- Department of Ultrasound, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - LiTing Zhang
- Department of Electrocardiographic, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Mei Zhu
- Department of Ultrasound, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China.,Department of Electrocardiographic, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
33
|
Rozenbaum Z, Topilsky Y, Khoury S, Pereg D, Laufer-Perl M. Association of body mass index and diastolic function in metabolically healthy obese with preserved ejection fraction. Int J Cardiol 2018; 277:147-152. [PMID: 30097335 DOI: 10.1016/j.ijcard.2018.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 07/23/2018] [Accepted: 08/03/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Small scale cohorts demonstrated an association between body mass index (BMI) and diastolic function in a metabolically healthy population. We aimed to characterize the relation between BMI and diastolic function in a relatively large cohort of metabolically healthy obese with preserved ejection fraction. METHODS AND RESULTS Echocardiograms of metabolically healthy patients between 2011 and 2016, who had no significant valvulopathies or atrial fibrillation, and had preserved ejection fraction, were retrospectively identified and analyzed. Metabolically healthy was defined as lack of known diabetes mellitus, hypertension, and hyperlipidemia. Patients were categorized into 4 groups according to BMI - normal BMI 18.5-25, overweight 25.01-30, obese 30.01-35, morbidly obese >35 kg/m2. The cohort consisted of 7057 individuals, 54.9% males, with a mean age 54 years. Patients in higher BMI groups more commonly demonstrated abnormalities in most echocardiographic parameters associated with diastolic dysfunction, including left atrial volume index>34 ml/m2, E/e'>14, e' lateral<10 cm/s, e' septal<7 cm/s, tricuspid regurgitation velocity>2.8 m/s and systolic pulmonary artery pressure≥36 mmHg (p<0.01 for all comparisons). Morbidly obese carried the highest risk compared to those with normal BMI. There were no significant differences between the groups in rates of readmission due to heart failure. CONCLUSION High BMI is associated with increased risk of diastolic dysfunction even in metabolically healthy patients. Additional trials are needed in order to evaluate whether these echocardiographic findings translate into clinical implications.
Collapse
Affiliation(s)
- Zach Rozenbaum
- Department of Cardiology, Tel Aviv Medical Center, Israel.
| | - Yan Topilsky
- Department of Cardiology, Tel Aviv Medical Center, Israel
| | - Shafik Khoury
- Department of Cardiology, Tel Aviv Medical Center, Israel
| | - David Pereg
- Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| | | |
Collapse
|
34
|
Abstract
INTRODUCTION Obesity is recognized as a risk factor for cardiovascular disease, expending independent adverse effects on the cardiovascular system. This relationship is complex due to several associations with cardiovascular disease risk factors/markers such as hypertension, dyslipidemia, insulin resistance/dysglycemia, or type 2 diabetes mellitus. Obesity induces a variety of cardiovascular system structural adaptations, from subclinical myocardial dysfunction to severe left ventricular systolic heart failure. Abnormalities in cardiac metabolism and subsequent cardiac energy, have been proposed as major contributors to obesity-related cardiovascular disease. Ectopic fat depots play an important role in several of the hypotheses postulated to explain the association between obesity, cardiac metabolism and cardiac dysfunction. AREAS COVERED In this review, we addressed with contemporary studies how obesity-associated metabolic conditions and ectopic cardiac fat accumulation, translate into cardiac energy metabolism disturbances that may lead to adverse effects on the cardiovascular system. EXPERT COMMENTARY Obesity and ectopic fat accumulation has long been related to metabolic diseases and adverse cardiovascular outcomes. Recent imaging advances have just started to address the complex interplays between obesity, ectopic fat depots, cardiac metabolism and the risk of obesity-related cardiovascular disease. A better comprehension of these obesity-associated metabolic disturbances will lead to earlier detection of patients at increased risk of cardiovascular disease and to the development of novel therapeutic metabolic targets to treat a wide variety of cardiovascular diseases.
Collapse
Affiliation(s)
- Marie-Eve Piché
- a Quebec Heart and Lung Institute , Laval University , Quebec , Canada
- b Faculty of Medicine , Laval University , Quebec , Canada
| | - Paul Poirier
- a Quebec Heart and Lung Institute , Laval University , Quebec , Canada
- c Faculty of Pharmacy , Laval University , Quebec , Canada
| |
Collapse
|
35
|
Piché ME, Poirier P, Lemieux I, Després JP. Overview of Epidemiology and Contribution of Obesity and Body Fat Distribution to Cardiovascular Disease: An Update. Prog Cardiovasc Dis 2018; 61:103-113. [PMID: 29964067 DOI: 10.1016/j.pcad.2018.06.004] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 06/21/2018] [Indexed: 01/14/2023]
Abstract
Obesity is recognized as a heterogeneous condition in which individuals with similar body mass index may have distinct metabolic and cardiovascular risk profiles. Susceptibility to obesity-related cardiometabolic complications is not solely mediated by overall body fat mass, but is largely dependent upon individual differences in regional body fat distribution and ability of subcutaneous adipose tissue to expand. The present review will discuss to what extent the individual variation in body fat distribution is one of the clinical key variables explaining the metabolic heterogeneity of obesity and its related cardiovascular risk. We will present the evidence for the complex nature of the relationship between obesity and cardiovascular disease, outline our current understanding of the mechanisms involved, and identify future direction of research pertinent to this interaction.
Collapse
Affiliation(s)
- Marie-Eve Piché
- Quebec Heart and Lung Institute, Québec, Canada; Faculty of Medicine, Université Laval, Québec, Canada.
| | - Paul Poirier
- Quebec Heart and Lung Institute, Québec, Canada; Faculty of Pharmacy, Université Laval, Québec, Canada
| | | | - Jean-Pierre Després
- Quebec Heart and Lung Institute, Québec, Canada; Faculty of Medicine, Université Laval, Québec, Canada
| |
Collapse
|
36
|
Levelt E, Gulsin G, Neubauer S, McCann GP. MECHANISMS IN ENDOCRINOLOGY: Diabetic cardiomyopathy: pathophysiology and potential metabolic interventions state of the art review. Eur J Endocrinol 2018; 178:R127-R139. [PMID: 29440374 PMCID: PMC5863473 DOI: 10.1530/eje-17-0724] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 02/12/2018] [Indexed: 12/22/2022]
Abstract
Heart failure is a major cause of morbidity and mortality in type 2 diabetes. Type 2 diabetes contributes to the development of heart failure through a variety of mechanisms, including disease-specific myocardial structural, functional and metabolic changes. This review will focus on the contemporary contributions of state of the art non-invasive technologies to our understanding of diabetic cardiomyopathy, including data on cardiac disease phenotype, cardiac energy metabolism and energetic deficiency, ectopic and visceral adiposity, diabetic liver disease, metabolic modulation strategies and cardiovascular outcomes with new classes of glucose-lowering therapies.
Collapse
Affiliation(s)
- Eylem Levelt
- British Heart Foundation Cardiovascular Research CentreUniversity of Leicester, Glenfield Hospital, Leicester, UK
- (E Levelt is now at Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science DepartmentLeeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK)
- Correspondenceshould be addressed to E Levelt;
| | - Gaurav Gulsin
- British Heart Foundation Cardiovascular Research CentreUniversity of Leicester, Glenfield Hospital, Leicester, UK
| | - Stefan Neubauer
- University of Oxford Centre for Clinical Magnetic Resonance ResearchUniversity of Oxford, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Gerry P McCann
- British Heart Foundation Cardiovascular Research CentreUniversity of Leicester, Glenfield Hospital, Leicester, UK
| |
Collapse
|
37
|
Gong TA, Hall SA. Targeting Other Modifiable Risk Factors for the Prevention of Heart Failure: Diabetes, Smoking, Obesity, and Inactivity. CURRENT CARDIOVASCULAR RISK REPORTS 2018. [DOI: 10.1007/s12170-018-0574-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
38
|
The relative contribution of metabolic and structural abnormalities to diastolic dysfunction in obesity. Int J Obes (Lond) 2017; 42:441-447. [PMID: 28974742 PMCID: PMC5880580 DOI: 10.1038/ijo.2017.239] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/25/2017] [Accepted: 09/03/2017] [Indexed: 01/05/2023]
Abstract
Background: Obesity causes diastolic dysfunction, and is one of the leading causes of heart failure with preserved ejection fraction. Myocardial relaxation is determined by both active metabolic processes such as impaired energetic status and steatosis, as well as intrinsic myocardial remodelling. However, the relative contribution of each to diastolic dysfunction in obesity is currently unknown. Methods: Eighty adult subjects (48 male) with no cardiovascular risk factors across a wide range of body mass indices (18.4–53.0 kg m−2) underwent magnetic resonance imaging for abdominal visceral fat, left ventricular geometry (LV mass:volume ratio) and diastolic function (peak diastolic strain rate), and magnetic resonance spectroscopy for PCr/ATP and myocardial triglyceride content. Results: Increasing visceral obesity was related to diastolic dysfunction (peak diastolic strain rate, r=−0.46, P=0.001). Myocardial triglyceride content (β=−0.2, P=0.008), PCr/ATP (β=−0.22, P=0.04) and LV mass:volume ratio (β=−0.61, P=0.04) all independently predicted peak diastolic strain rate (model R2 0.36, P<0.001). Moderated multiple regression confirmed the full mediating roles of PCr/ATP, myocardial triglyceride content and LV mass:volume ratio in the relationship between visceral fat and peak diastolic strain rate. Of the negative effect of visceral fat on diastolic function, 40% was explained by increased myocardial triglycerides, 39% by reduced PCr/ATP and 21% by LV concentric remodelling. Conclusions: Myocardial energetics and steatosis are more important in determining LV diastolic function than concentric hypertrophy, accounting for more of the negative effect of obesity on diastolic function than LV geometric remodelling. Targeting these metabolic processes is an attractive strategy to treat diastolic dysfunction in obesity.
Collapse
|
39
|
Rodriguez Flores M, Aguilar Salinas C, Piché ME, Auclair A, Poirier P. Effect of bariatric surgery on heart failure. Expert Rev Cardiovasc Ther 2017; 15:567-579. [PMID: 28714796 DOI: 10.1080/14779072.2017.1352471] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Obesity increases the risk of heart failure (HF), which continues to be a significant proportion of all cardiovascular diseases and affects increasingly younger populations. The cross-talk between adipose and the heart involves insulin resistance, adipokine signaling and inflammation, with the capacity of adipose tissue to mediate hemodynamic signals, promoting progressive cardiomyopathy. Areas covered: From a therapeutic perspective, there is not yet a single obesity-related pathway that when addressed, can ameliorate cardiomyopathy in obese patients and this is a matter of ongoing research. There is poor evidence of the beneficial long-term effect of small nonsurgical intentional weight loss on HF outcomes, in contrast to the field of HF accompanying severe obesity where observational studies have shown that bariatric surgery is associated with improved cardiac structure/function in severely obese patients with HF and preserved ejection fraction (HFpEF) as well as with improved cardiac structure/function in those with HF and reduced ejection fraction (HFrEF). Few studies report positive outcomes in subjects with obesity and HF, both severe, who underwent bariatric surgery as a rescue treatment, including bridge to heart transplantation. Expert commentary: The fast growing prevalence of obesity will continue to require the development of appropriate interventions directed at controlling or slowing pathways of cardiac damage in these patients, but at present, bariatric surgery should be considered an option to try to decrease morbidity associated with HF in severely obese adults.
Collapse
Affiliation(s)
- Marcela Rodriguez Flores
- a Endocrinology Department , Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , Mexico
| | - Carlos Aguilar Salinas
- a Endocrinology Department , Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , Mexico
| | - Marie-Eve Piché
- b Cardiology Department , Institut Universitaire de Cardiologie et de Pneumologie de Québec , Québec , Canada.,c Faculty of Medicine , Laval University , Québec , Canada
| | - Audrey Auclair
- b Cardiology Department , Institut Universitaire de Cardiologie et de Pneumologie de Québec , Québec , Canada
| | - Paul Poirier
- b Cardiology Department , Institut Universitaire de Cardiologie et de Pneumologie de Québec , Québec , Canada.,d Faculty of Pharmacy , Laval University , Québec , Canada
| |
Collapse
|
40
|
Zafirovic S, Obradovic M, Sudar-Milovanovic E, Jovanovic A, Stanimirovic J, Stewart AJ, Pitt SJ, Isenovic ER. 17β-Estradiol protects against the effects of a high fat diet on cardiac glucose, lipid and nitric oxide metabolism in rats. Mol Cell Endocrinol 2017; 446:12-20. [PMID: 28163099 DOI: 10.1016/j.mce.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/17/2017] [Accepted: 02/01/2017] [Indexed: 12/26/2022]
Abstract
The aim of this study was to investigate the in vivo effects of 17β-estradiol (E2) on myocardial metabolism and inducible nitric oxide synthase (iNOS) expression/activity in obese rats. Male Wistar rats were fed with a normal or a high fat (HF) diet (42% fat) for 10 weeks. Half of the HF fed rats were treated with a single dose of E2 while the other half were placebo-treated. 24 h after treatment animals were sacrificed. E2 reduced cardiac free fatty acid (FFA) (p < 0.05), L-arginine (p < 0.01), iNOS mRNA (p < 0.01), and protein (p < 0.05) levels and translocation of the FFA transporter (CD36) (p < 0.01) to the plasma membrane (PM) in HF fed rats. In contrast, Akt phosphorylation at Thr308 (p < 0.05) and translocation of the glucose transporter GLUT4 (p < 0.05) to the PM increased after E2 treatment in HF rats. Our results indicate that E2 acts via the PI3K/Akt signalling pathway to partially protect myocardial metabolism by attenuating the detrimental effects of increased iNOS expression/activity in HF fed rats.
Collapse
Affiliation(s)
- Sonja Zafirovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Milan Obradovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Emina Sudar-Milovanovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Aleksandra Jovanovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Julijana Stanimirovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| | - Alan J Stewart
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, United Kingdom.
| | - Samantha J Pitt
- School of Medicine, University of St Andrews, North Haugh, St Andrews, KY16 9TF, United Kingdom.
| | - Esma R Isenovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia.
| |
Collapse
|
41
|
Tune JD, Goodwill AG, Sassoon DJ, Mather KJ. Cardiovascular consequences of metabolic syndrome. Transl Res 2017; 183:57-70. [PMID: 28130064 PMCID: PMC5393930 DOI: 10.1016/j.trsl.2017.01.001] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 01/18/2023]
Abstract
The metabolic syndrome (MetS) is defined as the concurrence of obesity-associated cardiovascular risk factors including abdominal obesity, impaired glucose tolerance, hypertriglyceridemia, decreased HDL cholesterol, and/or hypertension. Earlier conceptualizations of the MetS focused on insulin resistance as a core feature, and it is clearly coincident with the above list of features. Each component of the MetS is an independent risk factor for cardiovascular disease and the combination of these risk factors elevates rates and severity of cardiovascular disease, related to a spectrum of cardiovascular conditions including microvascular dysfunction, coronary atherosclerosis and calcification, cardiac dysfunction, myocardial infarction, and heart failure. While advances in understanding the etiology and consequences of this complex disorder have been made, the underlying pathophysiological mechanisms remain incompletely understood, and it is unclear how these concurrent risk factors conspire to produce the variety of obesity-associated adverse cardiovascular diseases. In this review, we highlight current knowledge regarding the pathophysiological consequences of obesity and the MetS on cardiovascular function and disease, including considerations of potential physiological and molecular mechanisms that may contribute to these adverse outcomes.
Collapse
Affiliation(s)
- Johnathan D Tune
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Ind.
| | - Adam G Goodwill
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Ind
| | - Daniel J Sassoon
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Ind
| | - Kieren J Mather
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Ind; Department of Medicine, Indiana University School of Medicine, Indianapolis, Ind
| |
Collapse
|
42
|
Kolwicz SC. Lipid partitioning during cardiac stress. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:1472-80. [PMID: 27040509 DOI: 10.1016/j.bbalip.2016.03.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 01/11/2023]
Abstract
It is well documented that fatty acids serve as the primary fuel substrate for the contracting myocardium. However, extensive research has identified significant changes in the myocardial oxidation of fatty acids during acute or chronic cardiac stress. As a result, the redistribution or partitioning of fatty acids due to metabolic derangements could have biological implications. Fatty acids can be stored as triacylglycerols, serve as critical components for biosynthesis of phospholipid membranes, and form the potent signaling molecules, diacylglycerol and ceramides. Therefore, the contribution of lipid metabolism to health and disease is more intricate than a balance of uptake and oxidation. In this review, the available data regarding alterations that occur in endogenous cardiac lipid pathways during the pathological stressors of ischemia-reperfusion and pathological hypertrophy/heart failure are highlighted. In addition, changes in endogenous lipids observed in exercise training models are presented for comparison. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Stephen C Kolwicz
- Mitochondria and Metabolism Center, University of Washington, School of Medicine, 850 Republican St., Seattle, WA 98109, United States.
| |
Collapse
|
43
|
Cell Death and Heart Failure in Obesity: Role of Uncoupling Proteins. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9340654. [PMID: 27642497 PMCID: PMC5011521 DOI: 10.1155/2016/9340654] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022]
Abstract
Metabolic diseases such as obesity, metabolic syndrome, and type II diabetes are often characterized by increased reactive oxygen species (ROS) generation in mitochondrial respiratory complexes, associated with fat accumulation in cardiomyocytes, skeletal muscle, and hepatocytes. Several rodents studies showed that lipid accumulation in cardiac myocytes produces lipotoxicity that causes apoptosis and leads to heart failure, a dynamic pathological process. Meanwhile, several tissues including cardiac tissue develop an adaptive mechanism against oxidative stress and lipotoxicity by overexpressing uncoupling proteins (UCPs), specific mitochondrial membrane proteins. In heart from rodent and human with obesity, UCP2 and UCP3 may protect cardiomyocytes from death and from a state progressing to heart failure by downregulating programmed cell death. UCP activation may affect cytochrome c and proapoptotic protein release from mitochondria by reducing ROS generation and apoptotic cell death. Therefore the aim of this review is to discuss recent findings regarding the role that UCPs play in cardiomyocyte survival by protecting against ROS generation and maintaining bioenergetic metabolism homeostasis to promote heart protection.
Collapse
|
44
|
Mather KJ, Hutchins GD, Perry K, Territo W, Chisholm R, Acton A, Glick-Wilson B, Considine RV, Moberly S, DeGrado TR. Assessment of myocardial metabolic flexibility and work efficiency in human type 2 diabetes using 16-[18F]fluoro-4-thiapalmitate, a novel PET fatty acid tracer. Am J Physiol Endocrinol Metab 2016; 310:E452-60. [PMID: 26732686 PMCID: PMC4796267 DOI: 10.1152/ajpendo.00437.2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/19/2015] [Indexed: 01/13/2023]
Abstract
Altered myocardial fuel selection likely underlies cardiac disease risk in diabetes, affecting oxygen demand and myocardial metabolic flexibility. We investigated myocardial fuel selection and metabolic flexibility in human type 2 diabetes mellitus (T2DM), using positron emission tomography to measure rates of myocardial fatty acid oxidation {16-[(18)F]fluoro-4-thia-palmitate (FTP)} and myocardial perfusion and total oxidation ([(11)C]acetate). Participants underwent paired studies under fasting conditions, comparing 3-h insulin + glucose euglycemic clamp conditions (120 mU·m(-2)·min(-1)) to 3-h saline infusion. Lean controls (n = 10) were compared with glycemically controlled volunteers with T2DM (n = 8). Insulin augmented heart rate, blood pressure, and stroke index in both groups (all P < 0.01) and significantly increased myocardial oxygen consumption (P = 0.04) and perfusion (P = 0.01) in both groups. Insulin suppressed available nonesterified fatty acids (P < 0.0001), but fatty acid concentrations were higher in T2DM under both conditions (P < 0.001). Insulin-induced suppression of fatty acid oxidation was seen in both groups (P < 0.0001). However, fatty acid oxidation rates were higher under both conditions in T2DM (P = 0.003). Myocardial work efficiency was lower in T2DM (P = 0.006) and decreased in both groups with the insulin-induced increase in work and shift in fuel utilization (P = 0.01). Augmented fatty acid oxidation is present under baseline and insulin-treated conditions in T2DM, with impaired insulin-induced shifts away from fatty acid oxidation. This is accompanied by reduced work efficiency, possibly due to greater oxygen consumption with fatty acid metabolism. These observations suggest that improved fatty acid suppression, or reductions in myocardial fatty acid uptake and retention, could be therapeutic targets to improve myocardial ischemia tolerance in T2DM.
Collapse
Affiliation(s)
- K J Mather
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - G D Hutchins
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - K Perry
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - W Territo
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - R Chisholm
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - A Acton
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - B Glick-Wilson
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - R V Considine
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - S Moberly
- Indiana University School of Medicine, Indianapolis, Indiana; and
| | - T R DeGrado
- Indiana University School of Medicine, Indianapolis, Indiana; and Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
45
|
Liao PA, Lin G, Tsai SY, Wang CH, Juan YH, Lin YC, Wu MT, Yang LY, Liu MH, Chang TC, Lin YC, Huang YC, Huang PC, Wang JJ, Ng SH, Ng KK. Myocardial triglyceride content at 3 T cardiovascular magnetic resonance and left ventricular systolic function: a cross-sectional study in patients hospitalized with acute heart failure. J Cardiovasc Magn Reson 2016; 18:9. [PMID: 26850626 PMCID: PMC4744377 DOI: 10.1186/s12968-016-0228-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/25/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increased myocardial triglyceride (TG) content has been recognized as a risk factor for cardiovascular disease. However, its relation with cardiac function in patients on recovery from acute heart failure (HF) remains unclear. In this cross-sectional study, we sought to investigate the association between myocardial TG content measured on magnetic resonance spectroscopy ((1)H-MRS) and left ventricular (LV) function assessed on cardiovascular magnetic resonance (CMR) in patients who were hospitalized with HF. METHODS A total of 50 patients who were discharged after hospitalization for acute HF and 21 age- and sex-matched controls were included in the study. Myocardial TG content and LV parameters (function and mass) were measured on a 3.0 T MR scanner. Fatty acid (FA) and unsaturated fatty acid (UFA) content was normalized against water (W) using the LC-Model algorithm. The patient population was dichotomized according to the left ventricular ejection fraction (LVEF, <50% or ≥ 50%). RESULTS H-MRS data were available for 48 patients and 21 controls. Of the 48 patients, 25 had a LVEF <50% (mean, 31.2%), whereas the remaining 23 had a normal LVEF (mean, 60.2%). Myocardial UFA/W ratio was found to differ significantly in patients with low LVEF, normal LVEF, and controls (0.79% vs. 0.21% vs. 0.14%, respectively, p = 0.02). The myocardial UFA/TG ratio was associated with LV mass (r = 0.39, p < 0.001) and modestly related to LV end-diastolic volume (LVEDV; r = 0.24, p = 0.039). We also identified negative correlations of the myocardial FA/TG ratio with both LV mass (r = -0.39, p < 0.001) and LVEDV (r = -0.24, p = 0.039). CONCLUSIONS As compared with controls, patients who were discharged after hospitalization for acute HF had increased myocardial UFA content; furthermore, UFA was inversely related with LVEF, LV mass and, to a lesser extent, LVEDV. Our study may stimulate further research on the measure of myocardial UFA content by (1)H-MRS for outcome prediction. TRIAL REGISTRATION ClinicalTrial.gov: NCT02378402 . Registered 27/02/2015.
Collapse
Affiliation(s)
- Pen-An Liao
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou and Chang Gung University, 5 Fuhsing Street, Gueishan, Taoyuan, 333, Taiwan.
| | - Gigin Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou and Chang Gung University, 5 Fuhsing Street, Gueishan, Taoyuan, 333, Taiwan.
- Institute for Radiological Research, Chang Gung University, Taoyuan, Taiwan.
| | - Shang-Yueh Tsai
- Graduate Institute of Applied Physics, National Chengchi University, Taipei, Taiwan.
| | - Chao-Hung Wang
- Department of Cardiology and Heart Failure Center, Chang Gung Memorial Hospital, Keelung, Taiwan.
| | - Yu-Hsiang Juan
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou and Chang Gung University, 5 Fuhsing Street, Gueishan, Taoyuan, 333, Taiwan.
| | - Yu-Ching Lin
- Department of Radiology, Chang Gung Memorial Hospital, Keelung and Chang Gung University, Keelung, Taiwan.
| | - Ming-Ting Wu
- Department of Radiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.
| | - Lan-Yan Yang
- Clinical Trial Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Min-Hui Liu
- Department of Cardiology and Heart Failure Center, Chang Gung Memorial Hospital, Keelung, Taiwan.
| | - Tsun-Ching Chang
- Department of Radiology, Chang Gung Memorial Hospital, Keelung and Chang Gung University, Keelung, Taiwan.
| | - Yu-Chun Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou and Chang Gung University, 5 Fuhsing Street, Gueishan, Taoyuan, 333, Taiwan.
| | - Yu-Chieh Huang
- Department of Radiology, Chang Gung Memorial Hospital, Keelung and Chang Gung University, Keelung, Taiwan.
| | - Pei-Ching Huang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou and Chang Gung University, 5 Fuhsing Street, Gueishan, Taoyuan, 333, Taiwan.
| | - Jiun-Jie Wang
- Institute for Radiological Research, Chang Gung University, Taoyuan, Taiwan.
| | - Shu-Hang Ng
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou and Chang Gung University, 5 Fuhsing Street, Gueishan, Taoyuan, 333, Taiwan.
| | - Koon-Kwan Ng
- Department of Radiology, Chang Gung Memorial Hospital, Keelung and Chang Gung University, Keelung, Taiwan.
| |
Collapse
|
46
|
Sahraoui A, Dewachter C, de Medina G, Naeije R, Aouichat Bouguerra S, Dewachter L. Myocardial Structural and Biological Anomalies Induced by High Fat Diet in Psammomys obesus Gerbils. PLoS One 2016; 11:e0148117. [PMID: 26840416 PMCID: PMC4740502 DOI: 10.1371/journal.pone.0148117] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 01/13/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Psammomys obesus gerbils are particularly prone to develop diabetes and obesity after brief period of abundant food intake. A hypercaloric high fat diet has been shown to affect cardiac function. Here, we sought to determine whether a short period of high fat feeding might alter myocardial structure and expression of calcium handling proteins in this particular strain of gerbils. METHODS Twenty Psammomys obesus gerbils were randomly assigned to receive a normal plant diet (controls) or a high fat diet. At baseline and 16-week later, body weight, plasma biochemical parameters (including lipid and carbohydrate levels) were evaluated. Myocardial samples were collected for pathobiological evaluation. RESULTS Sixteen-week high fat dieting resulted in body weight gain and hyperlipidemia, while levels of carbohydrates remained unchanged. At myocardial level, high fat diet induced structural disorganization, including cardiomyocyte hypertrophy, lipid accumulation, interstitial and perivascular fibrosis and increased number of infiltrating neutrophils. Myocardial expressions of pro-apoptotic Bax-to-Bcl-2 ratio, pro-inflammatory cytokines [interleukin (IL)-1β and tumor necrosis factor (TNF)-α], intercellular (ICAM1) and vascular adhesion molecules (VCAM1) increased, while gene encoding cardiac muscle protein, the alpha myosin heavy polypeptide (MYH6), was downregulated. Myocardial expressions of sarco(endo)plasmic calcium-ATPase (SERCA2) and voltage-dependent calcium channel (Cacna1c) decreased, while protein kinase A (PKA) and calcium-calmodulin-dependent protein kinase (CaMK2D) expressions increased. Myocardial expressions of ryanodine receptor, phospholamban and sodium/calcium exchanger (Slc8a1) did not change. CONCLUSIONS We conclude that a relative short period of high fat diet in Psammomys obesus results in severe alterations of cardiac structure, activation of inflammatory and apoptotic processes, and altered expression of calcium-cycling determinants.
Collapse
Affiliation(s)
- Abdelhamid Sahraoui
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Team of Cellular and Molecular Physiopathology, Faculty of Biological Sciences, Houari Boumediene University of Sciences and Technology, El Alia, Algiers, Algeria
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Geoffrey de Medina
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Souhila Aouichat Bouguerra
- Team of Cellular and Molecular Physiopathology, Faculty of Biological Sciences, Houari Boumediene University of Sciences and Technology, El Alia, Algiers, Algeria
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
47
|
Rayner JJ, Neubauer S, Rider OJ. The paradox of obesity cardiomyopathy and the potential for weight loss as a therapy. Obes Rev 2015; 16:679-90. [PMID: 26096833 DOI: 10.1111/obr.12292] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/08/2015] [Accepted: 04/23/2015] [Indexed: 12/18/2022]
Abstract
Obesity is an independent risk factor for developing heart failure and the combination of the two disease states will prove to be a significant health burden over the coming years. Obesity is likely to contribute to the development of heart failure through a variety of mechanisms, including structural and functional changes, lipotoxicity and steatosis and altered substrate selection. However, once heart failure has developed, it seems that obesity confers a beneficial influence on prognosis in what has been termed the 'obesity paradox'. This may be a statistical phenomenon, but it should be considered that there is truly a protective state in the physiology of obesity. There is little evidence regarding the impact of weight loss in obese heart failure and whether or not this is beneficial. There have been small studies regarding the cardiovascular effects of both dietary weight loss and bariatric surgery, but few in heart failure. This is an important and increasingly relevant clinical question which must be addressed.
Collapse
Affiliation(s)
- J J Rayner
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - S Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - O J Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Kolwicz SC, Liu L, Goldberg IJ, Tian R. Enhancing Cardiac Triacylglycerol Metabolism Improves Recovery From Ischemic Stress. Diabetes 2015; 64:2817-27. [PMID: 25858561 PMCID: PMC4512225 DOI: 10.2337/db14-1943] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 04/02/2015] [Indexed: 01/11/2023]
Abstract
Elevated cardiac triacylglycerol (TAG) content is traditionally equated with cardiolipotoxicity and suggested to be a culprit in cardiac dysfunction. However, previous work demonstrated that myosin heavy-chain-mediated cardiac-specific overexpression of diacylglycerol transferase 1 (MHC-DGAT1), the primary enzyme for TAG synthesis, preserved cardiac function in two lipotoxic mouse models despite maintaining high TAG content. Therefore, we examined whether increased cardiomyocyte TAG levels due to DGAT1 overexpression led to changes in cardiac TAG turnover rates under normoxia and ischemia-reperfusion conditions. MHC-DGAT1 mice had elevated TAG content and synthesis rates, which did not alter cardiac function, substrate oxidation, or myocardial energetics. MHC-DGAT1 hearts had ischemia-induced lipolysis; however, when a physiologic mixture of long-chain fatty acids was provided, enhanced TAG turnover rates were associated with improved functional recovery from low-flow ischemia. Conversely, exogenous supply of palmitate during reperfusion suppressed elevated TAG turnover rates and impaired recovery from ischemia in MHC-DGAT1 hearts. Collectively, this study shows that elevated TAG content, accompanied by enhanced turnover, does not adversely affect cardiac function and, in fact, provides cardioprotection from ischemic stress. In addition, the results highlight the importance of exogenous supply of fatty acids when assessing cardiac lipid metabolism and its relationship with cardiac function.
Collapse
Affiliation(s)
- Stephen C Kolwicz
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA
| | - Li Liu
- Institute of Systems Biomedicine, Peking University, Beijing, China
| | - Ira J Goldberg
- Endocrinology, Diabetes, and Metabolism, New York University Langone Medical Center, New York, NY
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA
| |
Collapse
|
49
|
Le Page LM, Rider OJ, Lewis AJ, Ball V, Clarke K, Johansson E, Carr CA, Heather LC, Tyler DJ. Increasing Pyruvate Dehydrogenase Flux as a Treatment for Diabetic Cardiomyopathy: A Combined 13C Hyperpolarized Magnetic Resonance and Echocardiography Study. Diabetes 2015; 64:2735-43. [PMID: 25795215 PMCID: PMC4516266 DOI: 10.2337/db14-1560] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/16/2015] [Indexed: 01/02/2023]
Abstract
Although diabetic cardiomyopathy is widely recognized, there are no specific treatments available. Altered myocardial substrate selection has emerged as a candidate mechanism behind the development of cardiac dysfunction in diabetes. As pyruvate dehydrogenase (PDH) activity appears central to the balance of substrate use, we aimed to investigate the relationship between PDH flux and myocardial function in a rodent model of type 2 diabetes and to explore whether or not increasing PDH flux, with dichloroacetate, would restore the balance of substrate use and improve cardiac function. All animals underwent in vivo hyperpolarized [1-(13)C]pyruvate magnetic resonance spectroscopy and echocardiography to assess cardiac PDH flux and function, respectively. Diabetic animals showed significantly higher blood glucose levels (10.8 ± 0.7 vs. 8.4 ± 0.5 mmol/L), lower PDH flux (0.005 ± 0.001 vs. 0.017 ± 0.002 s(-1)), and significantly impaired diastolic function (transmitral early diastolic peak velocity/early diastolic myocardial velocity ratio [E/E'] 12.2 ± 0.8 vs. 20 ± 2), which are in keeping with early diabetic cardiomyopathy. Twenty-eight days of treatment with dichloroacetate restored PDH flux to normal levels (0.018 ± 0.002 s(-1)), reversed diastolic dysfunction (E/E' 14 ± 1), and normalized blood glucose levels (7.5 ± 0.7 mmol/L). The treatment of diabetes with dichloroacetate therefore restored the balance of myocardial substrate selection, reversed diastolic dysfunction, and normalized blood glucose levels. This suggests that PDH modulation could be a novel therapy for the treatment and/or prevention of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Lydia M Le Page
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Oliver J Rider
- Division of Cardiovascular Medicine, University of Oxford, Oxford, U.K
| | - Andrew J Lewis
- Division of Cardiovascular Medicine, University of Oxford, Oxford, U.K
| | - Vicky Ball
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | | | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K.
| |
Collapse
|
50
|
Coppiello G, Collantes M, Sirerol-Piquer MS, Vandenwijngaert S, Schoors S, Swinnen M, Vandersmissen I, Herijgers P, Topal B, van Loon J, Goffin J, Prósper F, Carmeliet P, García-Verdugo JM, Janssens S, Peñuelas I, Aranguren XL, Luttun A. Meox2/Tcf15 heterodimers program the heart capillary endothelium for cardiac fatty acid uptake. Circulation 2015; 131:815-26. [PMID: 25561514 DOI: 10.1161/circulationaha.114.013721] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Microvascular endothelium in different organs is specialized to fulfill the particular needs of parenchymal cells. However, specific information about heart capillary endothelial cells (ECs) is lacking. METHODS AND RESULTS Using microarray profiling on freshly isolated ECs from heart, brain, and liver, we revealed a genetic signature for microvascular heart ECs and identified Meox2/Tcf15 heterodimers as novel transcriptional determinants. This signature was largely shared with skeletal muscle and adipose tissue endothelium and was enriched in genes encoding fatty acid (FA) transport-related proteins. Using gain- and loss-of-function approaches, we showed that Meox2/Tcf15 mediate FA uptake in heart ECs, in part, by driving endothelial CD36 and lipoprotein lipase expression and facilitate FA transport across heart ECs. Combined Meox2 and Tcf15 haplodeficiency impaired FA uptake in heart ECs and reduced FA transfer to cardiomyocytes. In the long term, this combined haplodeficiency resulted in impaired cardiac contractility. CONCLUSIONS Our findings highlight a regulatory role for ECs in FA transfer to the heart parenchyma and unveil 2 of its intrinsic regulators. Our insights could be used to develop new strategies based on endothelial Meox2/Tcf15 targeting to modulate FA transfer to the heart and remedy cardiac dysfunction resulting from altered energy substrate usage.
Collapse
Affiliation(s)
- Giulia Coppiello
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Maria Collantes
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - María Salomé Sirerol-Piquer
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Sara Vandenwijngaert
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Sandra Schoors
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Melissa Swinnen
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Ine Vandersmissen
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Paul Herijgers
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Baki Topal
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Johannes van Loon
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Jan Goffin
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Felipe Prósper
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Peter Carmeliet
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Jose Manuel García-Verdugo
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Stefan Janssens
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Iván Peñuelas
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Xabier L Aranguren
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Aernout Luttun
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium.
| |
Collapse
|