1
|
Lee KCY, Williams AL, Wang L, Xie G, Jia W, Fujimoto A, Gerschenson M, Shohet RV. PKM2 regulates metabolic flux and oxidative stress in the murine heart. Physiol Rep 2024; 12:e70040. [PMID: 39256891 PMCID: PMC11387154 DOI: 10.14814/phy2.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
Cardiac metabolism ensures a continuous ATP supply, primarily using fatty acids in a healthy state and favoring glucose in pathological conditions. Pyruvate kinase muscle (PKM) controls the final step of glycolysis, with PKM1 being the main isoform in the heart. PKM2, elevated in various heart diseases, has been suggested to play a protective role in cardiac stress, but its function in basal cardiac metabolism remains unclear. We examined hearts from global PKM2 knockout (PKM2-/-) mice and found reduced intracellular glucose. Isotopic tracing of U-13C glucose revealed a shift to biosynthetic pathways in PKM2-/- cardiomyocytes. Total ATP content was two-thirds lower in PKM2-/- hearts, and functional analysis indicated reduced mitochondrial oxygen consumption. Total reactive oxygen species (ROS) and mitochondrial superoxide were also increased in PKM2-/- cardiomyocytes. Intriguingly, PKM2-/- hearts had preserved ejection fraction compared to controls. Mechanistically, increased calcium/calmodulin-dependent kinase II activity and phospholamban phosphorylation may contribute to higher sarcoendoplasmic reticulum calcium ATPase 2 pump activity in PKM2-/- hearts. Loss of PKM2 led to altered glucose metabolism, diminished mitochondrial function, and increased ROS in cardiomyocytes. These data suggest that cardiac PKM2 acts as an important rheostat to maintain ATP levels while limiting oxidative stress. Although loss of PKM2 did not impair baseline contractility, its absence may make hearts more sensitive to environmental stress or injury.
Collapse
Affiliation(s)
- Katie C. Y. Lee
- Department of Medicine, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
- Department of Cell and Molecular Biology, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Allison L. Williams
- Department of Medicine, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Lu Wang
- University of Hawaii Cancer CenterHonoluluHawaiiUSA
| | - Guoxiang Xie
- University of Hawaii Cancer CenterHonoluluHawaiiUSA
| | - Wei Jia
- University of Hawaii Cancer CenterHonoluluHawaiiUSA
| | - Anastasia Fujimoto
- Department of Cell and Molecular Biology, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Ralph V. Shohet
- Department of Medicine, John A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| |
Collapse
|
2
|
Quagliariello V, Canale ML, Bisceglia I, Iovine M, Paccone A, Maurea C, Scherillo M, Merola A, Giordano V, Palma G, Luciano A, Bruzzese F, Zito Marino F, Montella M, Franco R, Berretta M, Gabrielli D, Gallucci G, Maurea N. Sodium-glucose cotransporter 2 inhibitor dapagliflozin prevents ejection fraction reduction, reduces myocardial and renal NF-κB expression and systemic pro-inflammatory biomarkers in models of short-term doxorubicin cardiotoxicity. Front Cardiovasc Med 2024; 11:1289663. [PMID: 38818214 PMCID: PMC11138344 DOI: 10.3389/fcvm.2024.1289663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 04/09/2024] [Indexed: 06/01/2024] Open
Abstract
Background Anthracycline-mediated adverse cardiovascular events are among the leading causes of morbidity and mortality in patients with cancer. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) exert multiple cardiometabolic benefits in patients with/without type 2 diabetes, chronic kidney disease, and heart failure with reduced and preserved ejection fraction. We hypothesized that the SGLT2i dapagliflozin administered before and during doxorubicin (DOXO) therapy could prevent cardiac dysfunction and reduce pro-inflammatory pathways in preclinical models. Methods Cardiomyocytes were exposed to DOXO alone or combined with dapagliflozin (DAPA) at 10 and 100 nM for 24 h; cell viability, iATP, and Ca++ were quantified; lipid peroxidation products (malondialdehyde and 4-hydroxy 2-hexenal), NLRP3, MyD88, and cytokines were also analyzed through selective colorimetric and enzyme-linked immunosorbent assay (ELISA) methods. Female C57Bl/6 mice were treated for 10 days with a saline solution or DOXO (2.17 mg/kg), DAPA (10 mg/kg), or DOXO combined with DAPA. Systemic levels of ferroptosis-related biomarkers, galectin-3, high-sensitivity C-reactive protein (hs-CRP), and pro-inflammatory chemokines (IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12, IL17-α, IL-18, IFN-γ, TNF-α, G-CSF, and GM-CSF) were quantified. After treatments, immunohistochemical staining of myocardial and renal p65/NF-kB was performed. Results DAPA exerts cytoprotective, antioxidant, and anti-inflammatory properties in human cardiomyocytes exposed to DOXO by reducing iATP and iCa++ levels, lipid peroxidation, NLRP-3, and MyD88 expression. Pro-inflammatory intracellular cytokines were also reduced. In preclinical models, DAPA prevented the reduction of radial and longitudinal strain and ejection fraction after 10 days of treatment with DOXO. A reduced myocardial expression of NLRP-3 and MyD-88 was seen in the DOXO-DAPA group compared to DOXO mice. Systemic levels of IL-1β, IL-6, TNF-α, G-CSF, and GM-CSF were significantly reduced after treatment with DAPA. Serum levels of galectine-3 and hs-CRP were strongly enhanced in the DOXO group; on the other hand, their expression was reduced in the DAPA-DOXO group. Troponin-T, B-type natriuretic peptide (BNP), and N-Terminal Pro-BNP (NT-pro-BNP) were strongly reduced in the DOXO-DAPA group, revealing cardioprotective properties of SGLT2i. Mice treated with DOXO and DAPA exhibited reduced myocardial and renal NF-kB expression. Conclusion The overall picture of the study encourages the use of DAPA in the primary prevention of cardiomyopathies induced by anthracyclines in patients with cancer.
Collapse
Affiliation(s)
- V. Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - M. L. Canale
- Cardiology Division, Azienda USL Toscana Nord-Ovest, Versilia Hospital, Lido di Camaiore, Italy
| | - I. Bisceglia
- Integrated Cardiology Services, Department of Cardio-Thoracic-Vascular, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - M. Iovine
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - A. Paccone
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - C. Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, Naples, Italy
| | - M. Scherillo
- Cardiology Department, San Pio Hospital, Benevento, Italy
| | - A. Merola
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - V. Giordano
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - G. Palma
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - A. Luciano
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - F. Bruzzese
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - F. Zito Marino
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - M. Montella
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - R. Franco
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - M. Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - D. Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlani-ni, Roma—Fondazione per il Tuo Cuore—Heart Care Foundation, Firenze, Italy
| | - G. Gallucci
- Cardio-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - N. Maurea
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| |
Collapse
|
3
|
Ke ZP, Tao WQ, Zhao G, Cheng K. Role of PPAR-related genes in chronic heart failure: evidence from large populations. BMC Cardiovasc Disord 2023; 23:552. [PMID: 37950149 PMCID: PMC10638691 DOI: 10.1186/s12872-023-03554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The role of PPAR signaling and its associated genes in the pathogenesis and progression of chronic heart failure (CHF) remains elusive. METHODS We accessed the gene expression profile and relevant baseline information of CHF samples from the Gene Expression Omnibus (GEO) database, specifically from the GSE57338 project. RESULTS From GSE57338 project, we derived the expression value of 126 PPAR-related genes. A protein-protein interaction network was then established to illustrate potential protein interactions. ClueGO analysis results revealed that these genes predominantly participate in functions such as export across plasma membrane, regulation of lipid metabolic process, fatty acid metabolism, circulatory system vascular processes, alcohol metabolism, triglyceride metabolism and regulation of lipid localization and response to nutrient. Using the cytohubba plug-in in Cytoscape, we pinpointed ACADM, PPARG and CPT2 as potential central molecules in HF pathogenesis and progression. Subsequent Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis delved into the potential biological role of these three genes in CHF. Immune infiltration analysis suggested that the infiltration level of neutrophils and M2 macrophages might be notably influenced by these genes, thereby playing a role in the CHF mechanism. CONCLUSIONS Our research provides a comprehensive insight into the significance of PPAR associated genes in CHF development. Notably, the genes ACADM, PPARG and CPT2 emerged as potential targets for clinical interventions.
Collapse
Affiliation(s)
- Zun-Ping Ke
- Department of Geriatrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wen-Qi Tao
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Gang Zhao
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Kuan Cheng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Naasner L, Froese N, Hofmann W, Galuppo P, Werlein C, Shymotiuk I, Szaroszyk M, Erschow S, Amanakis G, Bähre H, Kühnel MP, Jonigk DD, Geffers R, Seifert R, Ricke-Hoch M, Wende AR, Blaner WS, Abel ED, Bauersachs J, Riehle C. Vitamin A preserves cardiac energetic gene expression in a murine model of diet-induced obesity. Am J Physiol Heart Circ Physiol 2022; 323:H1352-H1364. [PMID: 36399384 DOI: 10.1152/ajpheart.00514.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Perturbed vitamin-A metabolism is associated with type 2 diabetes and mitochondrial dysfunction that are pathophysiologically linked to the development of diabetic cardiomyopathy (DCM). However, the mechanism, by which vitamin A might regulate mitochondrial energetics in DCM has previously not been explored. To test the hypothesis that vitamin-A deficiency accelerates the onset of cardiomyopathy in diet-induced obesity (DIO), we subjected mice with lecithin retinol acyltransferase (Lrat) germline deletion, which exhibit impaired vitamin-A stores, to vitamin A-deficient high-fat diet (HFD) feeding. Wild-type mice fed with a vitamin A-sufficient HFD served as controls. Cardiac structure, contractile function, and mitochondrial respiratory capacity were preserved despite vitamin-A deficiency following 20 wk of HFD feeding. Gene profiling by RNA sequencing revealed that vitamin A is required for the expression of genes involved in cardiac fatty acid oxidation, glycolysis, tricarboxylic acid cycle, and mitochondrial oxidative phosphorylation in DIO as expression of these genes was relatively preserved under vitamin A-sufficient HFD conditions. Together, these data identify a transcriptional program, by which vitamin A preserves cardiac energetic gene expression in DIO that might attenuate subsequent onset of mitochondrial and contractile dysfunction.NEW & NOTEWORTHY The relationship between vitamin-A status and the pathogenesis of diabetic cardiomyopathy has not been studied in detail. We assessed cardiac mitochondrial respiratory capacity, contractile function, and gene expression by RNA sequencing in a murine model of combined vitamin-A deficiency and diet-induced obesity. Our study identifies a role for vitamin A in preserving cardiac energetic gene expression that might attenuate subsequent development of mitochondrial and contractile dysfunction in diet-induced obesity.
Collapse
Affiliation(s)
- Lea Naasner
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Natali Froese
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Winfried Hofmann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Paolo Galuppo
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | - Ivanna Shymotiuk
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Malgorzata Szaroszyk
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Sergej Erschow
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Georgios Amanakis
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Heike Bähre
- Research Core Unit Metabolomics, Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Mark P Kühnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH), German Lung Research Centre (DZL), Hannover, Germany
| | - Danny D Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH), German Lung Research Centre (DZL), Hannover, Germany
| | - Robert Geffers
- Research Group Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Roland Seifert
- Research Core Unit Metabolomics, Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - William S Blaner
- Department of Medicine, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine and UCLA Health, Los Angeles, California
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Ke J, Pan J, Lin H, Gu J. Diabetic cardiomyopathy: a brief summary on lipid toxicity. ESC Heart Fail 2022; 10:776-790. [PMID: 36369594 PMCID: PMC10053269 DOI: 10.1002/ehf2.14224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/30/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes mellitus (DM) is a serious epidemic around the globe, and cardiovascular diseases account for the majority of deaths in patients with DM. Diabetic cardiomyopathy (DCM) is defined as a cardiac dysfunction derived from DM without the presence of coronary artery diseases and hypertension. Patients with either type 1 or type 2 DM are at high risk of developing DCM and even heart failure. Metabolic disorders of obesity and insulin resistance in type 2 diabetic environments result in dyslipidaemia and subsequent lipid-induced toxicity (lipotoxicity) in organs including the heart. Although various mechanisms have been proposed underlying DCM, it remains incompletely understood how lipotoxicity alters cardiac function and how DM induces clinical heart syndrome. With recent progress, we here summarize the latest discoveries on lipid-induced cardiac toxicity in diabetic hearts and discuss the underlying therapies and controversies in clinical DCM.
Collapse
Affiliation(s)
- Jiahan Ke
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jianan Pan
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Hao Lin
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jun Gu
- Department of Cardiology Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
6
|
Switching to a Standard Chow Diet at Weaning Improves the Effects of Maternal and Postnatal High-Fat and High-Sucrose Diet on Cardiometabolic Health in Adult Male Mouse Offspring. Metabolites 2022; 12:metabo12060563. [PMID: 35736495 PMCID: PMC9230726 DOI: 10.3390/metabo12060563] [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: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiac mitochondrial dysfunction contributes to obesity-associated heart disease. Maternal and postnatal diet plays an important role in cardiac function, yet the impacts of a mismatch between prenatal and postweaning diet on cardiometabolic function are not well understood. We tested the hypothesis that switching to a standard chow diet after weaning would attenuate systemic metabolic disorders and cardiac and mitochondrial dysfunction associated with maternal and postnatal high-fat/high-sucrose (HFHS) diet in mice. Six-month-old male CD1 offspring from dams fed a HFHS diet and weaned to the same HFHS diet (HH) or switched to a standard chow diet (HC) were compared to offspring from dams fed a low-fat/low-sucrose diet and maintained on the same diet (LL). HC did not decrease body weight (BW) but normalized glucose tolerance, plasma cholesterol, LDL, and insulin levels compared to the HH. Systolic function indicated by the percent fractional shortening was not altered by diet. In freshly isolated cardiac mitochondria, maximal oxidative phosphorylation-linked respiratory capacity and coupling efficiency were significantly higher in the HC in the presence of fatty acid substrate compared to LL and HH, with modification of genes associated with metabolism and mitochondrial function. Switching to a standard chow diet at weaning can attenuate the deleterious effects of long-term HFHS in adult male mouse offspring.
Collapse
|
7
|
Tadinada SM, Weatherford ET, Collins GV, Bhardwaj G, Cochran J, Kutschke W, Zimmerman K, Bosko A, O'Neill BT, Weiss RM, Abel ED. Functional resilience of C57BL/6J mouse heart to dietary fat overload. Am J Physiol Heart Circ Physiol 2021; 321:H850-H864. [PMID: 34477461 PMCID: PMC8616610 DOI: 10.1152/ajpheart.00419.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 01/22/2023]
Abstract
Molecular mechanisms underlying cardiac dysfunction and subsequent heart failure in diabetic cardiomyopathy are incompletely understood. Initially we intended to test the role of G protein-coupled receptor kinase 2 (GRK2), a potential mediator of cardiac dysfunction in diabetic cardiomyopathy, but found that control animals on HFD did not develop cardiomyopathy. Cardiac function was preserved in both wild-type and GRK2 knockout animals fed high-fat diet as indicated by preserved left ventricular ejection fraction (LVEF) although heart mass was increased. The absence of cardiac dysfunction led us to rigorously evaluate the utility of diet-induced obesity to model diabetic cardiomyopathy in mice. Using pure C57BL/6J animals and various diets formulated with different sources of fat-lard (32% saturated fat, 68% unsaturated fat) or hydrogenated coconut oil (95% saturated fat), we consistently observed left ventricular hypertrophy, preserved LVEF, and preserved contractility measured by invasive hemodynamics in animals fed high-fat diet. Gene expression patterns that characterize pathological hypertrophy were not induced, but a modest induction of various collagen isoforms and matrix metalloproteinases was observed in heart with high-fat diet feeding. PPARα-target genes that enhance lipid utilization such as Pdk4, CD36, AcadL, and Cpt1b were induced, but mitochondrial energetics was not impaired. These results suggest that although long-term fat feeding in mice induces cardiac hypertrophy and increases cardiac fatty acid metabolism, it may not be sufficient to activate pathological hypertrophic mechanisms that impair cardiac function or induce cardiac fibrosis. Thus, additional factors that are currently not understood may contribute to the cardiac abnormalities previously reported by many groups.NEW & NOTEWORTHY Dietary fat overload (DFO) is widely used to model diabetic cardiomyopathy but the utility of this model is controversial. We comprehensively characterized cardiac contractile and mitochondrial function in C57BL6/J mice fed with lard-based or saturated fat-enriched diets initiated at two ages. Despite cardiac hypertrophy, contractile and mitochondrial function is preserved, and molecular adaptations likely limit lipotoxicity. The resilience of these hearts to DFO underscores the need to develop robust alternative models of diabetic cardiomyopathy.
Collapse
MESH Headings
- Age Factors
- Animals
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/physiopathology
- Diet, High-Fat
- Disease Models, Animal
- Energy Metabolism
- Female
- Fibrosis
- G-Protein-Coupled Receptor Kinase 2/genetics
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Myocardium/enzymology
- Myocardium/pathology
- Obesity/complications
- Stroke Volume
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
- Mice
Collapse
Affiliation(s)
- Satya Murthy Tadinada
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Eric T Weatherford
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Greg V Collins
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jesse Cochran
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - William Kutschke
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Kathy Zimmerman
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Alyssa Bosko
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Brian T O'Neill
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| | - Robert M Weiss
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Division of Cardiology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - E Dale Abel
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
8
|
Mitochondrial Uncoupling Proteins (UCPs) as Key Modulators of ROS Homeostasis: A Crosstalk between Diabesity and Male Infertility? Antioxidants (Basel) 2021; 10:antiox10111746. [PMID: 34829617 PMCID: PMC8614977 DOI: 10.3390/antiox10111746] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022] Open
Abstract
Uncoupling proteins (UCPs) are transmembrane proteins members of the mitochondrial anion transporter family present in the mitochondrial inner membrane. Currently, six homologs have been identified (UCP1-6) in mammals, with ubiquitous tissue distribution and multiple physiological functions. UCPs are regulators of key events for cellular bioenergetic metabolism, such as membrane potential, metabolic efficiency, and energy dissipation also functioning as pivotal modulators of ROS production and general cellular redox state. UCPs can act as proton channels, leading to proton re-entry the mitochondrial matrix from the intermembrane space and thus collapsing the proton gradient and decreasing the membrane potential. Each homolog exhibits its specific functions, from thermogenesis to regulation of ROS production. The expression and function of UCPs are intimately linked to diabesity, with their dysregulation/dysfunction not only associated to diabesity onset, but also by exacerbating oxidative stress-related damage. Male infertility is one of the most overlooked diabesity-related comorbidities, where high oxidative stress takes a major role. In this review, we discuss in detail the expression and function of the different UCP homologs. In addition, the role of UCPs as key regulators of ROS production and redox homeostasis, as well as their influence on the pathophysiology of diabesity and potential role on diabesity-induced male infertility is debated.
Collapse
|
9
|
Chen X, Ashraf S, Ashraf N, Harmancey R. UCP3 (Uncoupling Protein 3) Insufficiency Exacerbates Left Ventricular Diastolic Dysfunction During Angiotensin II-Induced Hypertension. J Am Heart Assoc 2021; 10:e022556. [PMID: 34533037 PMCID: PMC8649532 DOI: 10.1161/jaha.121.022556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Left ventricular diastolic dysfunction, an early stage in the pathogenesis of heart failure with preserved ejection fraction, is exacerbated by joint exposure to hypertension and obesity; however, the molecular mechanisms involved remain uncertain. The mitochondrial UCP3 (uncoupling protein 3) is downregulated in the heart with obesity. Here, we used a rat model of UCP3 haploinsufficiency (ucp3+/‐) to test the hypothesis that decreased UCP3 promotes left ventricular diastolic dysfunction during hypertension. Methods and Results Ucp3+/‐ rats and ucp3+/+ littermates fed a high‐salt diet (HS; 2% NaCl) and treated with angiotensin II (190 ng/kg per min for 28 days) experienced a similar rise in blood pressure (158±4 versus 155±7 mm Hg). However, UCP3 insufficiency worsened diastolic dysfunction according to echocardiographic assessment of left ventricular filling pressures (E/e’; 18.8±1.0 versus 14.9±0.6; P<0.05) and the isovolumic relaxation time (24.7±0.6 versus 21.3±0.5 ms; P<0.05), as well as invasive monitoring of the diastolic time constant (Tau; 15.5±0.8 versus 12.7±0.2 ms; P<0.05). Exercise tolerance on a treadmill also decreased for HS/angiotensin II‐treated ucp3+/‐ rats. Histological and molecular analyses further revealed that UCP3 insufficiency accelerated left ventricular concentric remodeling, detrimental interstitial matrix remodeling, and fetal gene reprogramming during hypertension. Moreover, UCP3 insufficiency increased oxidative stress and led to greater impairment of protein kinase G signaling. Conclusions Our findings identified UCP3 insufficiency as a cause for increased incidence of left ventricular diastolic dysfunction during hypertension. The results add further support to the use of antioxidants targeting mitochondrial reactive oxygen species as an adjuvant therapy for preventing heart failure with preserved ejection fraction in individuals with obesity.
Collapse
Affiliation(s)
- Xu Chen
- Department of Physiology and Biophysics University of Mississippi Medical Center Jackson MS.,Mississippi Center for Obesity Research University of Mississippi Medical Center Jackson MS
| | - Sadia Ashraf
- Department of Physiology and Biophysics University of Mississippi Medical Center Jackson MS.,Mississippi Center for Obesity Research University of Mississippi Medical Center Jackson MS
| | | | - Romain Harmancey
- Department of Physiology and Biophysics University of Mississippi Medical Center Jackson MS.,Mississippi Center for Obesity Research University of Mississippi Medical Center Jackson MS
| |
Collapse
|
10
|
Elrashidy RA, Ibrahim SE. Cinacalcet as a surrogate therapy for diabetic cardiomyopathy in rats through AMPK-mediated promotion of mitochondrial and autophagic function. Toxicol Appl Pharmacol 2021; 421:115533. [PMID: 33848515 DOI: 10.1016/j.taap.2021.115533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022]
Abstract
Decreased activity of AMP-activated protein kinase (AMPK) is implicated in the pathogenesis of diabetic cardiomyopathy (DCM). Recent evidence suggests a crosstalk between cinacalcet and AMPK activation. This study investigated the effects of cinacalcet on cardiac remodeling and dysfunction in type 2 diabetic rats (T2DM). High fat diet for 4 weeks combined with single intraperitoneal injection of streptozotocin (30 mg/kg) was used to induce type 2 diabetes in rats. Diabetic rats were either orally treated with vehicle, 5 or 10 mg/kg cinacalcet for 4 weeks. Control rats were fed standard chow diet and intraperitoneally injected with citrate buffer. T2DM rats showed lower body weight (BW), hyperglycemia and dyslipidemia, along with increased heart weight (HW) and HW/BW ratio. Masson's trichrome stained cardiac sections revealed massive fibrosis in T2DM rats. There were increased TGF-β1 and hydroxyproline levels, coupled with up-regulation of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) in hearts of T2DM rats. These alterations were associated with redox imbalance and impaired cardiac functions. Decreased phosphorylation of AMPK at threonine172 residue was found in T2DM hearts. Cinacalcet for 4 weeks significantly activated AMPK and alleviated cardiac remodeling and dysfunction in a dose-dependent manner, without affecting blood glucose, serum calcium and phosphorus levels. Cinacalcet increased the mitochondrial DNA content, and expressions of PGC-1α, UCP-3, beclin-1 and LC3-II/LC3-I ratio. Cinacalcet decreased the pro-apoptotic Bax, while increased the anti-apoptotic Bcl-2 in cardiac tissue of T2DM rats. These findings might highlight cinacalcet as an alternative therapy to combat the development and progression of DCM.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Animals
- Apoptosis/drug effects
- Autophagy/drug effects
- Cinacalcet/pharmacology
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/prevention & control
- Fibrosis
- Hemodynamics/drug effects
- Male
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Rats, Wistar
- Signal Transduction
- Streptozocin
- Ventricular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Rania A Elrashidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Samah E Ibrahim
- Physiology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
11
|
Bowman PRT, Smith GL, Gould GW. Run for your life: can exercise be used to effectively target GLUT4 in diabetic cardiac disease? PeerJ 2021; 9:e11485. [PMID: 34113491 PMCID: PMC8162245 DOI: 10.7717/peerj.11485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/27/2021] [Indexed: 12/25/2022] Open
Abstract
The global incidence, associated mortality rates and economic burden of diabetes are now such that it is considered one of the most pressing worldwide public health challenges. Considerable research is now devoted to better understanding the mechanisms underlying the onset and progression of this disease, with an ultimate aim of improving the array of available preventive and therapeutic interventions. One area of particular unmet clinical need is the significantly elevated rate of cardiomyopathy in diabetic patients, which in part contributes to cardiovascular disease being the primary cause of premature death in this population. This review will first consider the role of metabolism and more specifically the insulin sensitive glucose transporter GLUT4 in diabetic cardiac disease, before addressing how we may use exercise to intervene in order to beneficially impact key functional clinical outcomes.
Collapse
Affiliation(s)
- Peter R T Bowman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
12
|
Abstract
Alterations in cardiac energy metabolism contribute to the severity of heart failure. However, the energy metabolic changes that occur in heart failure are complex and are dependent not only on the severity and type of heart failure present but also on the co-existence of common comorbidities such as obesity and type 2 diabetes. The failing heart faces an energy deficit, primarily because of a decrease in mitochondrial oxidative capacity. This is partly compensated for by an increase in ATP production from glycolysis. The relative contribution of the different fuels for mitochondrial ATP production also changes, including a decrease in glucose and amino acid oxidation, and an increase in ketone oxidation. The oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in heart failure associated with diabetes and obesity, myocardial fatty acid oxidation increases, while in heart failure associated with hypertension or ischemia, myocardial fatty acid oxidation decreases. Combined, these energy metabolic changes result in the failing heart becoming less efficient (ie, a decrease in cardiac work/O2 consumed). The alterations in both glycolysis and mitochondrial oxidative metabolism in the failing heart are due to both transcriptional changes in key enzymes involved in these metabolic pathways, as well as alterations in NAD redox state (NAD+ and nicotinamide adenine dinucleotide levels) and metabolite signaling that contribute to posttranslational epigenetic changes in the control of expression of genes encoding energy metabolic enzymes. Alterations in the fate of glucose, beyond flux through glycolysis or glucose oxidation, also contribute to the pathology of heart failure. Of importance, pharmacological targeting of the energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac efficiency, decreasing the energy deficit and improving cardiac function in the failing heart.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle (R.T.)
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.)
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City (E.D.A.).,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City (E.D.A.)
| |
Collapse
|
13
|
Ho KL, Karwi QG, Wagg C, Zhang L, Vo K, Altamimi T, Uddin GM, Ussher JR, Lopaschuk GD. Ketones can become the major fuel source for the heart but do not increase cardiac efficiency. Cardiovasc Res 2021; 117:1178-1187. [PMID: 32402081 PMCID: PMC7982999 DOI: 10.1093/cvr/cvaa143] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/16/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS Ketones have been proposed to be a 'thrifty' fuel for the heart and increasing cardiac ketone oxidation can be cardioprotective. However, it is unclear how much ketone oxidation can contribute to energy production in the heart, nor whether increasing ketone oxidation increases cardiac efficiency. Therefore, our goal was to determine to what extent high levels of the ketone body, β-hydroxybutyrate (βOHB), contributes to cardiac energy production, and whether this influences cardiac efficiency. METHODS AND RESULTS Isolated working mice hearts were aerobically perfused with palmitate (0.8 mM or 1.2 mM), glucose (5 mM) and increasing concentrations of βOHB (0, 0.6, 2.0 mM). Subsequently, oxidation of these substrates, cardiac function, and cardiac efficiency were assessed. Increasing βOHB concentrations increased myocardial ketone oxidation rates without affecting glucose or fatty acid oxidation rates where normal physiological levels of glucose (5 mM) and fatty acid (0.8 mM) are present. Notably, ketones became the major fuel source for the heart at 2.0 mM βOHB (at both low or high fatty acid concentrations), with the elevated ketone oxidation rates markedly increasing tricarboxylic acid (TCA) cycle activity, producing a large amount of reducing equivalents and finally, increasing myocardial oxygen consumption. However, the marked increase in ketone oxidation at high concentrations of βOHB was not accompanied by an increase in cardiac work, suggesting that a mismatch between excess reduced equivalents production from ketone oxidation and cardiac adenosine triphosphate production. Consequently, cardiac efficiency decreased when the heart was exposed to higher ketone levels. CONCLUSIONS We demonstrate that while ketones can become the major fuel source for the heart, they do not increase cardiac efficiency, which also underscores the importance of recognizing ketones as a major fuel source for the heart in times of starvation, consumption of a ketogenic diet or poorly controlled diabetes.
Collapse
Affiliation(s)
- Kim L Ho
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pharmacology, College of Medicine, University of Diyala, Diyala, Iraq
| | - Cory Wagg
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Liyan Zhang
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Katherina Vo
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Tariq Altamimi
- Diabetes and Obesity Center, University of Louisville, Louisville, KT, USA
| | - Golam M Uddin
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - John R Ussher
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
14
|
Belosludtsev KN, Belosludtseva NV, Dubinin MV. Diabetes Mellitus, Mitochondrial Dysfunction and Ca 2+-Dependent Permeability Transition Pore. Int J Mol Sci 2020; 21:ijms21186559. [PMID: 32911736 PMCID: PMC7555889 DOI: 10.3390/ijms21186559] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus is one of the most common metabolic diseases in the developed world, and is associated either with the impaired secretion of insulin or with the resistance of cells to the actions of this hormone (type I and type II diabetes, respectively). In both cases, a common pathological change is an increase in blood glucose—hyperglycemia, which eventually can lead to serious damage to the organs and tissues of the organism. Mitochondria are one of the main targets of diabetes at the intracellular level. This review is dedicated to the analysis of recent data regarding the role of mitochondrial dysfunction in the development of diabetes mellitus. Specific areas of focus include the involvement of mitochondrial calcium transport systems and a pathophysiological phenomenon called the permeability transition pore in the pathogenesis of diabetes mellitus. The important contribution of these systems and their potential relevance as therapeutic targets in the pathology are discussed.
Collapse
Affiliation(s)
- Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Mari El, Russia; (N.V.B.); (M.V.D.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Moscow Region, Russia
- Correspondence: ; Tel.: +7-929-913-8910
| | - Natalia V. Belosludtseva
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Mari El, Russia; (N.V.B.); (M.V.D.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Moscow Region, Russia
| | - Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Mari El, Russia; (N.V.B.); (M.V.D.)
| |
Collapse
|
15
|
Abstract
Heart failure is a growing health problem resulting in the decreased life expectancy of patients and severely increased the healthcare burden. Penetrating research on the pathogenesis and regulation mechanism of heart failure is important for treatment of heart failure. Epicardial adipose tissue (EAT) has been demonstrated as not only a dynamic organ with biological functions but also an inert lipid store with regulating systemic metabolism. EAT mediates physiological and pathophysiological processes of heart failure by regulating adipogenesis, cardiac remodeling, insulin resistance, cardiac output, and renin angiotensin aldosterone system (RAAS). Moreover, EAT secretes a wide range of adipokines, adrenomedullin, adiponectin, and miRNAs through paracrine, endocrine, and vasocrine pathways, which involve in various extracellular and intracellular mechanism of cardiac-related cells in the progress of cardiovascular disease especially in heart failure. Nevertheless, mechanisms and roles of EAT on heart failure are barely summarized. Understanding the regulating mechanisms of EAT on heart failure may give rise to novel therapeutic targets and will open up innovative strategies to myocardial injury as well as in heart failure.
Collapse
Affiliation(s)
- Ying Song
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China.,Xiamen Branch of Zhongshan Hospital, Fudan University, Xiamen, 361015, Fujian, China
| | - Fei Song
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China.
| |
Collapse
|
16
|
Makrecka‐Kuka M, Liepinsh E, Murray AJ, Lemieux H, Dambrova M, Tepp K, Puurand M, Käämbre T, Han WH, Goede P, O'Brien KA, Turan B, Tuncay E, Olgar Y, Rolo AP, Palmeira CM, Boardman NT, Wüst RCI, Larsen TS. Altered mitochondrial metabolism in the insulin-resistant heart. Acta Physiol (Oxf) 2020; 228:e13430. [PMID: 31840389 DOI: 10.1111/apha.13430] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022]
Abstract
Obesity-induced insulin resistance and type 2 diabetes mellitus can ultimately result in various complications, including diabetic cardiomyopathy. In this case, cardiac dysfunction is characterized by metabolic disturbances such as impaired glucose oxidation and an increased reliance on fatty acid (FA) oxidation. Mitochondrial dysfunction has often been associated with the altered metabolic function in the diabetic heart, and may result from FA-induced lipotoxicity and uncoupling of oxidative phosphorylation. In this review, we address the metabolic changes in the diabetic heart, focusing on the loss of metabolic flexibility and cardiac mitochondrial function. We consider the alterations observed in mitochondrial substrate utilization, bioenergetics and dynamics, and highlight new areas of research which may improve our understanding of the cause and effect of cardiac mitochondrial dysfunction in diabetes. Finally, we explore how lifestyle (nutrition and exercise) and pharmacological interventions can prevent and treat metabolic and mitochondrial dysfunction in diabetes.
Collapse
Affiliation(s)
| | | | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience University of Cambridge Cambridge UK
| | - Hélène Lemieux
- Department of Medicine Faculty Saint‐Jean, Women and Children's Health Research Institute University of Alberta Edmonton AB Canada
| | | | - Kersti Tepp
- National Institute of Chemical Physics and Biophysics Tallinn Estonia
| | - Marju Puurand
- National Institute of Chemical Physics and Biophysics Tallinn Estonia
| | - Tuuli Käämbre
- National Institute of Chemical Physics and Biophysics Tallinn Estonia
| | - Woo H. Han
- Faculty Saint‐Jean University of Alberta Edmonton AB Canada
| | - Paul Goede
- Laboratory of Endocrinology Amsterdam Gastroenterology & Metabolism Amsterdam University Medical Center University of Amsterdam Amsterdam The Netherlands
| | - Katie A. O'Brien
- Department of Physiology, Development and Neuroscience University of Cambridge Cambridge UK
| | - Belma Turan
- Laboratory of Endocrinology Amsterdam Gastroenterology & Metabolism Amsterdam University Medical Center University of Amsterdam Amsterdam The Netherlands
| | - Erkan Tuncay
- Department of Biophysics Faculty of Medicine Ankara University Ankara Turkey
| | - Yusuf Olgar
- Department of Biophysics Faculty of Medicine Ankara University Ankara Turkey
| | - Anabela P. Rolo
- Department of Life Sciences University of Coimbra and Center for Neurosciences and Cell Biology University of Coimbra Coimbra Portugal
| | - Carlos M. Palmeira
- Department of Life Sciences University of Coimbra and Center for Neurosciences and Cell Biology University of Coimbra Coimbra Portugal
| | - Neoma T. Boardman
- Cardiovascular Research Group Department of Medical Biology UiT the Arctic University of Norway Tromso Norway
| | - Rob C. I. Wüst
- Laboratory for Myology Department of Human Movement Sciences Faculty of Behavioural and Movement Sciences Amsterdam Movement Sciences Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Terje S. Larsen
- Cardiovascular Research Group Department of Medical Biology UiT the Arctic University of Norway Tromso Norway
| |
Collapse
|
17
|
Plaza A, Antonazzi M, Blanco-Urgoiti J, Del Olmo N, Ruiz-Gayo M. Potential Role of Leptin in Cardiac Steatosis Induced by Highly Saturated Fat Intake during Adolescence. Mol Nutr Food Res 2019; 63:e1900110. [PMID: 31298470 DOI: 10.1002/mnfr.201900110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/16/2019] [Indexed: 01/06/2023]
Abstract
SCOPE To identify the age-dependent effect of diets containing elevated amounts of either saturated or unsaturated fatty acids on cardiac steatosis in mice. METHODS AND RESULTS Five- and eight-week-old C57BL/6J mice cohorts are given free access to either a saturated or an unsaturated fatty-acid-enriched diet during 8 weeks. Body weight (BW) and food intake are monitored during this period. Cardiac lipid content, carnitine palmitoyltransferase-I (CPT-I) activity, and the amount of uncoupling proteins 2 and 3 (UCP2 and UCP3) are analyzed and correlated with blood leptin concentration. Leptin and PPARγ gene expression is quantified in white adipose tissue (WAT). Both diets have a similar effect on food intake, BW, and adiposity, independently of the age. Nevertheless, cardiac steatosis is specifically identified in adolescent mice consuming the saturated diet. These animals also display lower activity of cardiac CPT-I, a down-regulation of cardiac UCP2, together with lower concentration of plasma leptin. Accordingly, leptin gene expression is reduced in the visceral WAT. CONCLUSION Consumption of diets containing elevated amounts of saturated fat during adolescence and early adult life promotes cardiac steatosis in mice. An insufficient endocrine activity of WAT, in terms of leptin production, may account for such an effect.
Collapse
Affiliation(s)
- Adrián Plaza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28925, Madrid, Spain
| | - Marco Antonazzi
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28925, Madrid, Spain
| | | | - Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28925, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28925, Madrid, Spain
| |
Collapse
|
18
|
Pohl EE, Rupprecht A, Macher G, Hilse KE. Important Trends in UCP3 Investigation. Front Physiol 2019; 10:470. [PMID: 31133866 PMCID: PMC6524716 DOI: 10.3389/fphys.2019.00470] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 04/04/2019] [Indexed: 11/13/2022] Open
Abstract
Membrane uncoupling protein 3 (UCP3), a member of the mitochondrial uncoupling protein family, was discovered in 1997. UCP3's properties, such as its high homology to other mitochondrial carriers, especially to UCP2, its short lifetime and low specificity of UCP3 antibodies, have hindered progress in understanding its biological function and transport mechanism over decades. The abundance of UCP3 is highest in murine brown adipose tissue (BAT, 15.0 pmol/mg protein), compared to heart (2.7 pmol/mg protein) and the gastrocnemius muscle (1.7 pmol/mg protein), but it is still 400-fold lower than the abundance of UCP1, a biomarker for BAT. Investigation of UCP3 reconstituted in planar bilayer membranes revealed that it transports protons only when activated by fatty acids (FA). Although purine nucleotides (PN) inhibit UCP3-mediated transport, the molecular mechanism differs from that of UCP1. It remains a conundrum that two homologous proton-transporting proteins exist within the same tissue. Recently, we proposed that UCP3 abundance directly correlates with the degree of FA β-oxidation in cell metabolism. Further development in this field implies that UCP3 may have dual function in transporting substrates, which have yet to be identified, alongside protons. Evaluation of the literature with respect to UCP3 is a complex task because (i) UCP3 features are often extrapolated from its "twin" UCP2 without additional proof, and (ii) the specificity of antibodies against UCP3 used in studies is rarely evaluated. In this review, we primarily focus on recent findings obtained for UCP3 in biological and biomimetic systems.
Collapse
Affiliation(s)
- Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Anne Rupprecht
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| | - Gabriel Macher
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Karolina E. Hilse
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
19
|
Santos RAS, Oudit GY, Verano-Braga T, Canta G, Steckelings UM, Bader M. The renin-angiotensin system: going beyond the classical paradigms. Am J Physiol Heart Circ Physiol 2019; 316:H958-H970. [PMID: 30707614 PMCID: PMC7191626 DOI: 10.1152/ajpheart.00723.2018] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thirty years ago, a novel axis of the renin-angiotensin system (RAS) was unveiled by the discovery of angiotensin-(1−7) [ANG-(1−7)] generation in vivo. Later, angiotensin-converting enzyme 2 (ACE2) was shown to be the main mediator of this reaction, and Mas was found to be the receptor for the heptapeptide. The functional analysis of this novel axis of the RAS that followed its discovery revealed numerous protective actions in particular for cardiovascular diseases. In parallel, similar protective actions were also described for one of the two receptors of ANG II, the ANG II type 2 receptor (AT2R), in contrast to the other, the ANG II type 1 receptor (AT1R), which mediates deleterious actions of this peptide, e.g., in the setting of cardiovascular disease. Very recently, another branch of the RAS was discovered, based on angiotensin peptides in which the amino-terminal aspartate was replaced by alanine, the alatensins. Ala-ANG-(1−7) or alamandine was shown to interact with Mas-related G protein-coupled receptor D, and the first functional data indicated that this peptide also exerts protective effects in the cardiovascular system. This review summarizes the presentations given at the International Union of Physiological Sciences Congress in Rio de Janeiro, Brazil, in 2017, during the symposium entitled “The Renin-Angiotensin System: Going Beyond the Classical Paradigms,” in which the signaling and physiological actions of ANG-(1−7), ACE2, AT2R, and alatensins were reported (with a focus on noncentral nervous system-related tissues) and the therapeutic opportunities based on these findings were discussed.
Collapse
Affiliation(s)
- Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Federal University of Minas Gerais , Belo Horizonte, Minas Gerais , Brazil
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta , Edmonton , Canada
| | - Thiago Verano-Braga
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Federal University of Minas Gerais , Belo Horizonte, Minas Gerais , Brazil
| | - Giovanni Canta
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Federal University of Minas Gerais , Belo Horizonte, Minas Gerais , Brazil
| | - Ulrike Muscha Steckelings
- Department of Molecular Medicine, Cardiovascular & Renal Research, University of Southern Denmark, Odense, Denmark
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine , Berlin , Germany.,Deutsches Zentrum für Herz-Kreislaufforschung, Partner Site Berlin, Berlin , Germany.,Berlin Institute of Health , Berlin , Germany.,Charité-University Medicine, Berlin , Germany.,Institute for Biology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
20
|
Riehle C, Bauersachs J. Of mice and men: models and mechanisms of diabetic cardiomyopathy. Basic Res Cardiol 2018; 114:2. [PMID: 30443826 PMCID: PMC6244639 DOI: 10.1007/s00395-018-0711-0] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increases the risk of heart failure independent of co-existing hypertension and coronary artery disease. Although several molecular mechanisms for the development of diabetic cardiomyopathy have been identified, they are incompletely understood. The pathomechanisms are multifactorial and as a consequence, no causative treatment exists at this time to modulate or reverse the molecular changes contributing to accelerated cardiac dysfunction in diabetic patients. Numerous animal models have been generated, which serve as powerful tools to study the impact of type 1 and type 2 diabetes on the heart. Despite specific limitations of the models generated, they mimic various perturbations observed in the diabetic myocardium and continue to provide important mechanistic insight into the pathogenesis underlying diabetic cardiomyopathy. This article reviews recent studies in both diabetic patients and in these animal models, and discusses novel hypotheses to delineate the increased incidence of heart failure in diabetic patients.
Collapse
Affiliation(s)
- Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| |
Collapse
|
21
|
Uncoupling protein 3 deficiency impairs myocardial fatty acid oxidation and contractile recovery following ischemia/reperfusion. Basic Res Cardiol 2018; 113:47. [PMID: 30374710 PMCID: PMC6208686 DOI: 10.1007/s00395-018-0707-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/23/2018] [Indexed: 12/23/2022]
Abstract
Patients with insulin resistance and type 2 diabetes have poor cardiac outcomes following myocardial infarction (MI). The mitochondrial uncoupling protein 3 (UCP3) is down-regulated in the heart with insulin resistance. We hypothesized that decreased UCP3 levels contribute to poor cardiac recovery following ischemia/reperfusion (I/R). After confirming that myocardial UCP3 levels were systematically decreased by 20-49% in animal models of insulin resistance and type 2 diabetes, we genetically engineered Sprague-Dawley rats with partial loss of UCP3 (ucp3+/-). Wild-type littermates (ucp3+/+) were used as controls. Isolated working hearts from ucp3+/- rats were characterized by impaired recovery of cardiac power and decreased long-chain fatty acid (LCFA) oxidation following I/R. Mitochondria isolated from ucp3+/- hearts subjected to I/R in vivo displayed increased reactive oxygen species (ROS) generation and decreased respiratory complex I activity. Supplying ucp3+/- cardiac mitochondria with the medium-chain fatty acid (MCFA) octanoate slowed electron transport through the respiratory chain and reduced ROS generation. This was accompanied by improvement of cardiac LCFA oxidation and recovery of contractile function post ischemia. In conclusion, we demonstrated that normal cardiac UCP3 levels are essential to recovery of LCFA oxidation, mitochondrial respiratory capacity, and contractile function following I/R. These results reveal a potential mechanism for the poor prognosis of type 2 diabetic patients following MI and expose MCFA supplementation as a feasible metabolic intervention to improve recovery of these patients at reperfusion.
Collapse
|
22
|
Vakrou S, Fukunaga R, Foster DB, Sorensen L, Liu Y, Guan Y, Woldemichael K, Pineda-Reyes R, Liu T, Tardiff JC, Leinwand LA, Tocchetti CG, Abraham TP, O'Rourke B, Aon MA, Abraham MR. Allele-specific differences in transcriptome, miRNome, and mitochondrial function in two hypertrophic cardiomyopathy mouse models. JCI Insight 2018; 3:94493. [PMID: 29563334 DOI: 10.1172/jci.insight.94493] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 02/14/2018] [Indexed: 01/06/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) stems from mutations in sarcomeric proteins that elicit distinct biophysical sequelae, which in turn may yield radically different intracellular signaling and molecular pathologic profiles. These signaling events remain largely unaddressed by clinical trials that have selected patients based on clinical HCM diagnosis, irrespective of genotype. In this study, we determined how two mouse models of HCM differ, with respect to cellular/mitochondrial function and molecular biosignatures, at an early stage of disease. We show that hearts from young R92W-TnT and R403Q-αMyHC mutation-bearing mice differ in their transcriptome, miRNome, intracellular redox environment, mitochondrial antioxidant defense mechanisms, and susceptibility to mitochondrial permeability transition pore opening. Pathway analysis of mRNA-sequencing data and microRNA profiles indicate that R92W-TnT mutants exhibit a biosignature consistent with activation of profibrotic TGF-β signaling. Our results suggest that the oxidative environment and mitochondrial impairment in young R92W-TnT mice promote activation of TGF-β signaling that foreshadows a pernicious phenotype in young individuals. Of the two mutations, R92W-TnT is more likely to benefit from anti-TGF-β signaling effects conferred by angiotensin receptor blockers and may be responsive to mitochondrial antioxidant strategies in the early stage of disease. Molecular and functional profiling may therefore serve as aids to guide precision therapy for HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lars Sorensen
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yamin Liu
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Yufan Guan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kirubel Woldemichael
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Roberto Pineda-Reyes
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ting Liu
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jill C Tardiff
- Department of Internal Medicine and Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology and the BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Carlo G Tocchetti
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Theodore P Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Miguel A Aon
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - M Roselle Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| |
Collapse
|
23
|
Rines AK, Chang HC, Wu R, Sato T, Khechaduri A, Kouzu H, Shapiro J, Shang M, Burke MA, Abdelwahid E, Jiang X, Chen C, Rawlings TA, Lopaschuk GD, Schumacker PT, Abel ED, Ardehali H. Snf1-related kinase improves cardiac mitochondrial efficiency and decreases mitochondrial uncoupling. Nat Commun 2017; 8:14095. [PMID: 28117339 PMCID: PMC5286102 DOI: 10.1038/ncomms14095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022] Open
Abstract
Ischaemic heart disease limits oxygen and metabolic substrate availability to the heart, resulting in tissue death. Here, we demonstrate that the AMP-activated protein kinase (AMPK)-related protein Snf1-related kinase (SNRK) decreases cardiac metabolic substrate usage and mitochondrial uncoupling, and protects against ischaemia/reperfusion. Hearts from transgenic mice overexpressing SNRK have decreased glucose and palmitate metabolism and oxygen consumption, but maintained power and function. They also exhibit decreased uncoupling protein 3 (UCP3) and mitochondrial uncoupling. Conversely, Snrk knockout mouse hearts have increased glucose and palmitate oxidation and UCP3. SNRK knockdown in cardiac cells decreases mitochondrial efficiency, which is abolished with UCP3 knockdown. We show that Tribbles homologue 3 (Trib3) binds to SNRK, and downregulates UCP3 through PPARα. Finally, SNRK is increased in cardiomyopathy patients, and SNRK reduces infarct size after ischaemia/reperfusion. SNRK also decreases cardiac cell death in a UCP3-dependent manner. Our results suggest that SNRK improves cardiac mitochondrial efficiency and ischaemic protection. The Snf1-related kinase (SNRK) is widely expressed and yet its function is poorly understood. Here the authors show that SNRK regulates mitochondrial coupling via the Trib3-PPARα-UCP3 pathway and that cardiac overexpression of SNRK decreases metabolic substrate usage and oxygen consumption but maintains cardiac function and energy in mice.
Collapse
Affiliation(s)
- Amy K Rines
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Hsiang-Chun Chang
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Rongxue Wu
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Tatsuya Sato
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Arineh Khechaduri
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Hidemichi Kouzu
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Jason Shapiro
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Meng Shang
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Michael A Burke
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Eltyeb Abdelwahid
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Xinghang Jiang
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Chunlei Chen
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Tenley A Rawlings
- Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine, University of Utah, School of Medicine, Salt Lake City, Utah 84132, USA
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada T6G 2B7
| | - Paul T Schumacker
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - E Dale Abel
- Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine, University of Utah, School of Medicine, Salt Lake City, Utah 84132, USA
| | - Hossein Ardehali
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
24
|
Lee TW, Bai KJ, Lee TI, Chao TF, Kao YH, Chen YJ. PPARs modulate cardiac metabolism and mitochondrial function in diabetes. J Biomed Sci 2017; 24:5. [PMID: 28069019 PMCID: PMC5223385 DOI: 10.1186/s12929-016-0309-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023] Open
Abstract
Diabetic cardiomyopathy is a major complication of diabetes mellitus (DM). Currently, effective treatments for diabetic cardiomyopathy are limited. The pathophysiology of diabetic cardiomyopathy is complex, whereas mitochondrial dysfunction plays a vital role in the genesis of diabetic cardiomyopathy. Metabolic regulation targeting mitochondrial dysfunction is expected to be a reasonable strategy for treating diabetic cardiomyopathy. Peroxisome proliferator-activated receptors (PPARs) are master executors in regulating glucose and lipid homeostasis and also modulate mitochondrial function. However, synthetic PPAR agonists used for treating hyperlipidemia and DM have shown controversial effects on cardiovascular regulation. This article reviews our updated understanding of the beneficial and detrimental effects of PPARs on mitochondria in diabetic hearts.
Collapse
Affiliation(s)
- Ting-Wei Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Jen Bai
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tze-Fan Chao
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan. .,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
Lee J, Harris AN, Holley CL, Mahadevan J, Pyles KD, Lavagnino Z, Scherrer DE, Fujiwara H, Sidhu R, Zhang J, Huang SCC, Piston DW, Remedi MS, Urano F, Ory DS, Schaffer JE. Rpl13a small nucleolar RNAs regulate systemic glucose metabolism. J Clin Invest 2016; 126:4616-4625. [PMID: 27820699 DOI: 10.1172/jci88069] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/29/2016] [Indexed: 12/22/2022] Open
Abstract
Small nucleolar RNAs (snoRNAs) are non-coding RNAs that form ribonucleoproteins to guide covalent modifications of ribosomal and small nuclear RNAs in the nucleus. Recent studies have also uncovered additional non-canonical roles for snoRNAs. However, the physiological contributions of these small RNAs are largely unknown. Here, we selectively deleted four snoRNAs encoded within the introns of the ribosomal protein L13a (Rpl13a) locus in a mouse model. Loss of Rpl13a snoRNAs altered mitochondrial metabolism and lowered reactive oxygen species tone, leading to increased glucose-stimulated insulin secretion from pancreatic islets and enhanced systemic glucose tolerance. Islets from mice lacking Rpl13a snoRNAs demonstrated blunted oxidative stress responses. Furthermore, these mice were protected against diabetogenic stimuli that cause oxidative stress damage to islets. Our study illuminates a previously unrecognized role for snoRNAs in metabolic regulation.
Collapse
|
26
|
Patel VB, Zhong JC, Grant MB, Oudit GY. Role of the ACE2/Angiotensin 1-7 Axis of the Renin-Angiotensin System in Heart Failure. Circ Res 2016; 118:1313-26. [PMID: 27081112 DOI: 10.1161/circresaha.116.307708] [Citation(s) in RCA: 592] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) remains the most common cause of death and disability, and a major economic burden, in industrialized nations. Physiological, pharmacological, and clinical studies have demonstrated that activation of the renin-angiotensin system is a key mediator of HF progression. Angiotensin-converting enzyme 2 (ACE2), a homolog of ACE, is a monocarboxypeptidase that converts angiotensin II into angiotensin 1-7 (Ang 1-7) which, by virtue of its actions on the Mas receptor, opposes the molecular and cellular effects of angiotensin II. ACE2 is widely expressed in cardiomyocytes, cardiofibroblasts, and coronary endothelial cells. Recent preclinical translational studies confirmed a critical counter-regulatory role of ACE2/Ang 1-7 axis on the activated renin-angiotensin system that results in HF with preserved ejection fraction. Although loss of ACE2 enhances susceptibility to HF, increasing ACE2 level prevents and reverses the HF phenotype. ACE2 and Ang 1-7 have emerged as a key protective pathway against HF with reduced and preserved ejection fraction. Recombinant human ACE2 has been tested in phase I and II clinical trials without adverse effects while lowering and increasing plasma angiotensin II and Ang 1-7 levels, respectively. This review discusses the transcriptional and post-transcriptional regulation of ACE2 and the role of the ACE2/Ang 1-7 axis in cardiac physiology and in the pathophysiology of HF. The pharmacological and therapeutic potential of enhancing ACE2/Ang 1-7 action as a novel therapy for HF is highlighted.
Collapse
Affiliation(s)
- Vaibhav B Patel
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Jiu-Chang Zhong
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Maria B Grant
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Gavin Y Oudit
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.).
| |
Collapse
|
27
|
Lark DS, Torres MJ, Lin CT, Ryan TE, Anderson EJ, Neufer PD. Direct real-time quantification of mitochondrial oxidative phosphorylation efficiency in permeabilized skeletal muscle myofibers. Am J Physiol Cell Physiol 2016; 311:C239-45. [PMID: 27335172 DOI: 10.1152/ajpcell.00124.2016] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/11/2016] [Indexed: 11/22/2022]
Abstract
Oxidative phosphorylation (OXPHOS) efficiency, defined as the ATP-to-O ratio, is a critical feature of mitochondrial function that has been implicated in health, aging, and disease. To date, however, the methods to measure ATP/O have primarily relied on indirect approaches or entail parallel rather than simultaneous determination of ATP synthesis and O2 consumption rates. The purpose of this project was to develop and validate an approach to determine the ATP/O ratio in permeabilized fiber bundles (PmFBs) from simultaneous measures of ATP synthesis (JATP) and O2 consumption (JO2 ) rates in real time using a custom-designed apparatus. JO2 was measured via a polarigraphic oxygen sensor and JATP via fluorescence using an enzyme-linked assay system (hexokinase II, glucose-6-phosphate dehydrogenase) linked to NADPH production. Within the dynamic linear range of the assay system, ADP-stimulated increases in steady-state JATP mirrored increases in steady-state JO2 (r(2) = 0.91, P < 0.0001, n = 57 data points). ATP/O ratio was less than one under low rates of respiration (15 μM ADP) but increased to more than two at moderate (200 μM ADP) and maximal (2,000 μM ADP) rates of respiration with an interassay coefficient of variation of 24.03, 16.72, and 11.99%, respectively. Absolute and relative (to mechanistic) ATP/O ratios were lower in PmFBs (2.09 ± 0.251, 84%) compared with isolated mitochondria (2.44 ± 0.124, 98%). ATP/O ratios in PmFBs were not affected by the activity of adenylate kinase or creatine kinase. These findings validate an enzyme-linked respiratory clamp system for measuring OXPHOS efficiency in PmFBs and provide evidence that OXPHOS efficiency increases as energy demand increases.
Collapse
Affiliation(s)
- Daniel S Lark
- Department of Kinesiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Maria J Torres
- Department of Kinesiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Chien-Te Lin
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Ethan J Anderson
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - P Darrell Neufer
- Department of Kinesiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
28
|
β-Sitosterol increases mitochondrial electron transport by fluidizing mitochondrial membranes and enhances mitochondrial responsiveness to increasing energy demand by the induction of uncoupling in C2C12 myotubes. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.02.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
29
|
Lewis AJ, Neubauer S, Tyler DJ, Rider OJ. Pyruvate dehydrogenase as a therapeutic target for obesity cardiomyopathy. Expert Opin Ther Targets 2016; 20:755-66. [PMID: 26617082 DOI: 10.1517/14728222.2016.1126248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Obesity cardiomyopathy is a major public health problem with few specific therapeutic options. Abnormal cardiac substrate metabolism with reduced pyruvate dehydrogenase (PDH) activity is associated with energetic and functional cardiac impairment and may be a therapeutic target. AREAS COVERED This review summarizes the changes to cardiac substrate and high energy phosphorus metabolism that occur in obesity and describes the links between abnormal metabolism and impairment of cardiac function. The available evidence for the currently available pharmacological options for selective metabolic therapy in obesity cardiomyopathy is reviewed. EXPERT OPINION Pharmacological restoration of PDH activity is in general associated with favourable effects upon cardiac substrate metabolism and function in both animal models and small scale human studies, supporting a potential role as a therapeutic target.
Collapse
Affiliation(s)
- Andrew Jm Lewis
- a Division of Cardiovascular Medicine, Radcliffe Department of Medicine , University of Oxford , Oxford , OX3 9DU , UK.,b Department of Physiology , Anatomy and Genetics, University of Oxford , Sherrington Road, Oxford , OX3 9DU , UK
| | - Stefan Neubauer
- a Division of Cardiovascular Medicine, Radcliffe Department of Medicine , University of Oxford , Oxford , OX3 9DU , UK
| | - Damian J Tyler
- a Division of Cardiovascular Medicine, Radcliffe Department of Medicine , University of Oxford , Oxford , OX3 9DU , UK.,b Department of Physiology , Anatomy and Genetics, University of Oxford , Sherrington Road, Oxford , OX3 9DU , UK
| | - Oliver J Rider
- a Division of Cardiovascular Medicine, Radcliffe Department of Medicine , University of Oxford , Oxford , OX3 9DU , UK.,b Department of Physiology , Anatomy and Genetics, University of Oxford , Sherrington Road, Oxford , OX3 9DU , UK
| |
Collapse
|
30
|
Patel VB, Mori J, McLean BA, Basu R, Das SK, Ramprasath T, Parajuli N, Penninger JM, Grant MB, Lopaschuk GD, Oudit GY. ACE2 Deficiency Worsens Epicardial Adipose Tissue Inflammation and Cardiac Dysfunction in Response to Diet-Induced Obesity. Diabetes 2016; 65. [PMID: 26224885 PMCID: PMC4686955 DOI: 10.2337/db15-0399] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is increasing in prevalence and is strongly associated with metabolic and cardiovascular disorders. The renin-angiotensin system (RAS) has emerged as a key pathogenic mechanism for these disorders; angiotensin (Ang)-converting enzyme 2 (ACE2) negatively regulates RAS by metabolizing Ang II into Ang 1-7. We studied the role of ACE2 in obesity-mediated cardiac dysfunction. ACE2 null (ACE2KO) and wild-type (WT) mice were fed a high-fat diet (HFD) or a control diet and studied at 6 months of age. Loss of ACE2 resulted in decreased weight gain but increased glucose intolerance, epicardial adipose tissue (EAT) inflammation, and polarization of macrophages into a proinflammatory phenotype in response to HFD. Similarly, human EAT in patients with obesity and heart failure displayed a proinflammatory macrophage phenotype. Exacerbated EAT inflammation in ACE2KO-HFD mice was associated with decreased myocardial adiponectin, decreased phosphorylation of AMPK, increased cardiac steatosis and lipotoxicity, and myocardial insulin resistance, which worsened heart function. Ang 1-7 (24 µg/kg/h) administered to ACE2KO-HFD mice resulted in ameliorated EAT inflammation and reduced cardiac steatosis and lipotoxicity, resulting in normalization of heart failure. In conclusion, ACE2 plays a novel role in heart disease associated with obesity wherein ACE2 negatively regulates obesity-induced EAT inflammation and cardiac insulin resistance.
Collapse
Affiliation(s)
- Vaibhav B Patel
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jun Mori
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada Departments of Pediatrics and Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Brent A McLean
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Ratnadeep Basu
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Subhash K Das
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Tharmarajan Ramprasath
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Nirmal Parajuli
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Maria B Grant
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN
| | - Gary D Lopaschuk
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada Departments of Pediatrics and Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada Department of Physiology, University of Alberta, Edmonton, Alberta, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
31
|
Harmancey R, Haight DL, Watts KA, Taegtmeyer H. Chronic Hyperinsulinemia Causes Selective Insulin Resistance and Down-regulates Uncoupling Protein 3 (UCP3) through the Activation of Sterol Regulatory Element-binding Protein (SREBP)-1 Transcription Factor in the Mouse Heart. J Biol Chem 2015; 290:30947-61. [PMID: 26555260 DOI: 10.1074/jbc.m115.673988] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Indexed: 01/22/2023] Open
Abstract
The risk for heart failure and death after myocardial infarction is abnormally high in diabetic subjects. We and others have shown previously that mitochondrial uncoupling protein 3 (UCP3) improves functional recovery of the rodent heart during reperfusion. Here, we demonstrate that pharmacological induction of hyperinsulinemia in mice down-regulates myocardial UCP3. Decreased UCP3 expression was linked to the development of selective insulin resistance in the heart, characterized by decreased basal activity of Akt but preserved activity of the p44/42 mitogen-activated protein kinase, and overactivation of the sterol regulatory element-binding protein (SREBP)-1-mediated lipogenic program. In cultured myocytes, insulin treatment and SREBP-1 overexpression decreased, whereas SREBP-1 interference increased, peroxisome proliferator-activated receptor-stimulated expression of UCP3. Promoter deletion and site-directed mutagenesis identified three functional sterol regulatory elements in the vicinity of a known complex intronic enhancer. Increased binding of SREBP-1 to this DNA region was confirmed in the heart of hyperinsulinemic mice. In conclusion, we describe a hitherto unknown regulatory mechanism by which insulin inhibits cardiac UCP3 expression through activation of the lipogenic factor SREBP-1. Sustained down-regulation of cardiac UCP3 by hyperinsulinemia may partly explain the poor prognosis of type 2 diabetic patients after myocardial infarction.
Collapse
Affiliation(s)
- Romain Harmancey
- From the Department of Internal Medicine, Division of Cardiology, University of Texas Medical School, University of Texas Health Science Center, Houston, Texas 77030 and the Department of Physiology and Biophysics, Mississippi Center for Obesity Research and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505
| | - Derek L Haight
- From the Department of Internal Medicine, Division of Cardiology, University of Texas Medical School, University of Texas Health Science Center, Houston, Texas 77030 and
| | - Kayla A Watts
- the Department of Physiology and Biophysics, Mississippi Center for Obesity Research and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505
| | - Heinrich Taegtmeyer
- From the Department of Internal Medicine, Division of Cardiology, University of Texas Medical School, University of Texas Health Science Center, Houston, Texas 77030 and
| |
Collapse
|
32
|
UCP-3 uncoupling protein confers hypoxia resistance to renal epithelial cells and is upregulated in renal cell carcinoma. Sci Rep 2015; 5:13450. [PMID: 26304588 PMCID: PMC4548255 DOI: 10.1038/srep13450] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 07/01/2015] [Indexed: 01/17/2023] Open
Abstract
Tumor cells can adapt to a hostile environment with reduced oxygen supply. The present study aimed to identify mechanisms that confer hypoxia resistance. Partially hypoxia/reoxygenation (H/R)-resistant proximal tubular (PT) cells were selected by exposing PT cultures to repetitive cycles of H/R. Thereafter, H/R-induced changes in mRNA and protein expression, inner mitochondrial membrane potential (ΔΨm), formation of superoxide, and cell death were compared between H/R-adapted and control PT cultures. As a result, H/R-adapted PT cells exhibited lower H/R-induced hyperpolarization of ΔΨm and produced less superoxide than the control cultures. Consequently, H/R triggered ΔΨm break-down and DNA degradation in a lower percentage of H/R-adapted than control PT cells. Moreover, H/R induced upregulation of mitochondrial uncoupling protein-3 (UCP-3) in H/R-adapted PT but not in control cultures. In addition, ionizing radiation killed a lower percentage of H/R-adapted as compared to control cells suggestive of an H/R-radiation cross-resistance developed by the selection procedure. Knockdown of UCP-3 decreased H/R- and radioresitance of the H/R-adapted cells. Finally, UCP-3 protein abundance of PT-derived clear cell renal cell carcinoma and normal renal tissue was compared in human specimens indicating upregulation of UCP-3 during tumor development. Combined, our data suggest functional significance of UCP-3 for H/R resistance.
Collapse
|
33
|
Hafstad AD, Boardman N, Aasum E. How exercise may amend metabolic disturbances in diabetic cardiomyopathy. Antioxid Redox Signal 2015; 22:1587-605. [PMID: 25738326 PMCID: PMC4449627 DOI: 10.1089/ars.2015.6304] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SIGNIFICANCE Over-nutrition and sedentary lifestyle has led to a worldwide increase in obesity, insulin resistance, and type 2 diabetes (T2D) associated with an increased risk of development of cardiovascular disorders. Diabetic cardiomyopathy, independent of hypertension or coronary disease, is induced by a range of systemic changes and may through multiple processes result in functional and structural cardiac derangements. The pathogenesis of this cardiomyopathy is complex and multifactorial, and it will eventually lead to reduced cardiac working capacity and increased susceptibility to ischemic injury. RECENT ADVANCES Metabolic disturbances such as altered lipid handling and substrate utilization, decreased mechanical efficiency, mitochondrial dysfunction, disturbances in nonoxidative glucose pathways, and increased oxidative stress are hallmarks of diabetic cardiomyopathy. Interestingly, several of these disturbances are found to precede the development of cardiac dysfunction. CRITICAL ISSUES Exercise training is effective in the prevention and treatment of obesity and T2D. In addition to its beneficial influence on diabetes/obesity-related systemic changes, it may also amend many of the metabolic disturbances characterizing the diabetic myocardium. These changes are due to both indirect effects, exercise-mediated systemic changes, and direct effects originating from the high contractile activity of the heart during physical training. FUTURE DIRECTIONS Revealing the molecular mechanisms behind the beneficial effects of exercise training is of considerable scientific value to generate evidence-based therapy and in the development of new treatment strategies.
Collapse
Affiliation(s)
- Anne D Hafstad
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Neoma Boardman
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ellen Aasum
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
34
|
Schilling JD. The mitochondria in diabetic heart failure: from pathogenesis to therapeutic promise. Antioxid Redox Signal 2015; 22:1515-26. [PMID: 25761843 PMCID: PMC4449623 DOI: 10.1089/ars.2015.6294] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Diabetes is an important risk factor for the development of heart failure (HF). Given the increasing prevalence of diabetes in the population, strategies are needed to reduce the burden of HF in these patients. RECENT ADVANCES Diabetes is associated with several pathologic findings in the heart including dysregulated metabolism, lipid accumulation, oxidative stress, and inflammation. Emerging evidence suggests that mitochondrial dysfunction may be a central mediator of these pathologic responses. The development of therapeutic approaches targeting mitochondrial biology holds promise for the management of HF in diabetic patients. CRITICAL ISSUES Despite significant data implicating mitochondrial pathology in diabetic cardiomyopathy, the optimal pharmacologic approach to improve mitochondrial function remains undefined. FUTURE DIRECTIONS Detailed mechanistic studies coupled with more robust clinical phenotyping will be necessary to develop novel approaches to improve cardiac function in diabetes. Moreover, understanding the interplay between diabetes and other cardiac stressors (hypertension, ischemia, and valvular disease) will be of the utmost importance for clinical translation of scientific discoveries made in this field.
Collapse
Affiliation(s)
- Joel D Schilling
- 1Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, Missouri.,2Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.,3Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
35
|
Westenbrink BD, Ling H, Divakaruni AS, Gray CBB, Zambon AC, Dalton ND, Peterson KL, Gu Y, Matkovich SJ, Murphy AN, Miyamoto S, Dorn GW, Heller Brown J. Mitochondrial reprogramming induced by CaMKIIδ mediates hypertrophy decompensation. Circ Res 2015; 116:e28-39. [PMID: 25605649 DOI: 10.1161/circresaha.116.304682] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RATIONALE Sustained activation of Gαq transgenic (Gq) signaling during pressure overload causes cardiac hypertrophy that ultimately progresses to dilated cardiomyopathy. The molecular events that drive hypertrophy decompensation are incompletely understood. Ca(2+)/calmodulin-dependent protein kinase II δ (CaMKIIδ) is activated downstream of Gq, and overexpression of Gq and CaMKIIδ recapitulates hypertrophy decompensation. OBJECTIVE To determine whether CaMKIIδ contributes to hypertrophy decompensation provoked by Gq. METHODS AND RESULTS Compared with Gq mice, compound Gq/CaMKIIδ knockout mice developed a similar degree of cardiac hypertrophy but exhibited significantly improved left ventricular function, less cardiac fibrosis and cardiomyocyte apoptosis, and fewer ventricular arrhythmias. Markers of oxidative stress were elevated in mitochondria from Gq versus wild-type mice and respiratory rates were lower; these changes in mitochondrial function were restored by CaMKIIδ deletion. Gq-mediated increases in mitochondrial oxidative stress, compromised membrane potential, and cell death were recapitulated in neonatal rat ventricular myocytes infected with constitutively active Gq and attenuated by CaMKII inhibition. Deep RNA sequencing revealed altered expression of 41 mitochondrial genes in Gq hearts, with normalization of ≈40% of these genes by CaMKIIδ deletion. Uncoupling protein 3 was markedly downregulated in Gq or by Gq expression in neonatal rat ventricular myocytes and reversed by CaMKIIδ deletion or inhibition, as was peroxisome proliferator-activated receptor α. The protective effects of CaMKIIδ inhibition on reactive oxygen species generation and cell death were abrogated by knock down of uncoupling protein 3. Conversely, restoration of uncoupling protein 3 expression attenuated reactive oxygen species generation and cell death induced by CaMKIIδ. Our in vivo studies further demonstrated that pressure overload induced decreases in peroxisome proliferator-activated receptor α and uncoupling protein 3, increases in mitochondrial protein oxidation, and hypertrophy decompensation, which were attenuated by CaMKIIδ deletion. CONCLUSIONS Mitochondrial gene reprogramming induced by CaMKIIδ emerges as an important mechanism contributing to mitotoxicity in decompensating hypertrophy.
Collapse
Affiliation(s)
- B Daan Westenbrink
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Haiyun Ling
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Ajit S Divakaruni
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Charles B B Gray
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Alexander C Zambon
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Nancy D Dalton
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Kirk L Peterson
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Yusu Gu
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Scot J Matkovich
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Anne N Murphy
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Shigeki Miyamoto
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Gerald W Dorn
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| | - Joan Heller Brown
- From the Department of Pharmacology (B.D.W., H.L., A.S.D., C.B.B.G., A.C.Z., A.N.M., J.H.B.), Department of Medicine (N.D.D., K.L.P., Y.G.), and Biomedical Sciences Graduate Program (C.B.B.G.), University of California San Diego; School of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO (S.J.M., G.W.D.); Department of Cardiology, University Medical Center Groningen, Unversity of Groningen, Groningen, The Netherlands (B.D.W.)
| |
Collapse
|
36
|
Sverdlov AL, Elezaby A, Behring JB, Bachschmid MM, Luptak I, Tu VH, Siwik DA, Miller EJ, Liesa M, Shirihai OS, Pimentel DR, Cohen RA, Colucci WS. High fat, high sucrose diet causes cardiac mitochondrial dysfunction due in part to oxidative post-translational modification of mitochondrial complex II. J Mol Cell Cardiol 2015; 78:165-73. [PMID: 25109264 PMCID: PMC4268348 DOI: 10.1016/j.yjmcc.2014.07.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Diet-induced obesity leads to metabolic heart disease (MHD) characterized by increased oxidative stress that may cause oxidative post-translational modifications (OPTM) of cardiac mitochondrial proteins. The functional consequences of OPTM of cardiac mitochondrial proteins in MHD are unknown. Our objective was to determine whether cardiac mitochondrial dysfunction in MHD due to diet-induced obesity is associated with cysteine OPTM. METHODS AND RESULTS Male C57BL/6J mice were fed either a high-fat, high-sucrose (HFHS) or control diet for 8months. Cardiac mitochondria from HFHS-fed mice (vs. control diet) had an increased rate of H2O2 production, a decreased GSH/GSSG ratio, a decreased rate of complex II substrate-driven ATP synthesis and decreased complex II activity. Complex II substrate-driven ATP synthesis and complex II activity were partially restored ex-vivo by reducing conditions. A biotin switch assay showed that HFHS feeding increased cysteine OPTM in complex II subunits A (SDHA) and B (SDHB). Using iodo-TMT multiplex tags we found that HFHS feeding is associated with reversible oxidation of cysteines 89 and 231 in SDHA, and 100, 103 and 115 in SDHB. CONCLUSIONS MHD due to consumption of a HFHS "Western" diet causes increased H2O2 production and oxidative stress in cardiac mitochondria associated with decreased ATP synthesis and decreased complex II activity. Impaired complex II activity and ATP production are associated with reversible cysteine OPTM of complex II. Possible sites of reversible cysteine OPTM in SDHA and SDHB were identified by iodo-TMT tag labeling. Mitochondrial ROS may contribute to the pathophysiology of MHD by impairing the function of complex II. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".
Collapse
Affiliation(s)
- Aaron L Sverdlov
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Aly Elezaby
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Jessica B Behring
- Vascular Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Markus M Bachschmid
- Vascular Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Ivan Luptak
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Vivian H Tu
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Deborah A Siwik
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Edward J Miller
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Marc Liesa
- Obesity and Nutrition Section, Mitochondria ARC, Boston University School of Medicine, Boston, MA, USA
| | - Orian S Shirihai
- Obesity and Nutrition Section, Mitochondria ARC, Boston University School of Medicine, Boston, MA, USA
| | - David R Pimentel
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Richard A Cohen
- Vascular Biology Unit, Boston University School of Medicine, Boston, MA, USA
| | - Wilson S Colucci
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
37
|
Banke NH, Lewandowski ED. Impaired cytosolic NADH shuttling and elevated UCP3 contribute to inefficient citric acid cycle flux support of postischemic cardiac work in diabetic hearts. J Mol Cell Cardiol 2014; 79:13-20. [PMID: 25450611 DOI: 10.1016/j.yjmcc.2014.10.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/18/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022]
Abstract
Diabetic hearts are subject to more extensive ischemia/reperfusion (ISC/REP) damage. This study examined the efficiency of citric acid cycle (CAC) flux and the transfer of cytosolic reducing equivalents into the mitochondria for oxidative support of cardiac work following ISC/REP in hearts of c57bl/6 (NORM) and type 2 diabetic, db/db mouse hearts. Flux through the CAC and malate-aspartate shuttle (MA) were monitored via dynamic (13)C NMR of isolated hearts perfused with (13)C palmitate+glucose. MA flux was lower in db/db than NORM. Oxoglutarate malate carrier (OMC) was elevated in the db/db heart, suggesting a compensatory response to low NADHc. Baseline CAC flux per unit work (rate-pressure-product, RPP) was similar between NORM and db/db, but ISC/REP reduced the efficiency of CAC flux/RPP by 20% in db/db. ISC/REP also increased UCP3 transcription, indicating potential for greater uncoupling. Therefore, ISC/REP induces inefficient carbon utilization through the CAC in hearts of diabetic mice due to the combined inefficiencies in NADHc transfer per OMC content and increased uncoupling via UCP3. Ischemia and reperfusion exacerbated pre-existing mitochondrial defects and metabolic limitations in the cytosol of diabetic hearts. These limitations and defects render diabetic hearts more susceptible to inefficient carbon fuel utilization for oxidative energy metabolism.
Collapse
Affiliation(s)
- Natasha H Banke
- Center for Cardiovascular Research and Department of Physiology and Biophysics, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - E Douglas Lewandowski
- Center for Cardiovascular Research and Department of Physiology and Biophysics, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
38
|
Nabben M, van Bree BWJ, Lenaers E, Hoeks J, Hesselink MKC, Schaart G, Gijbels MJJ, Glatz JFC, da Silva GJJ, de Windt LJ, Tian R, Mike E, Skapura DG, Wehrens XHT, Schrauwen P. Lack of UCP3 does not affect skeletal muscle mitochondrial function under lipid-challenged conditions, but leads to sudden cardiac death. Basic Res Cardiol 2014; 109:447. [PMID: 25344084 DOI: 10.1007/s00395-014-0447-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 10/07/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
UCP3's exact physiological function in lipid handling in skeletal and cardiac muscle remains unknown. Interestingly, etomoxir, a fat oxidation inhibitor and strong inducer of UCP3, is proposed for treating both diabetes and heart failure. We hypothesize that the upregulation of UCP3 upon etomoxir serves to protect mitochondria against lipotoxicity. To evaluate UCP3's role in skeletal muscle (skm) and heart under lipid-challenged conditions, the effect of UCP3 ablation was examined in a state of dysbalance between fat availability and oxidative capacity. Wild type (WT) and UCP3(-/-) mice were subjected to high-fat feeding for 14 days. From day 6 onwards, they were given either saline or etomoxir. Etomoxir treatment induced an increase in markers of lipotoxicity in skm compared to saline. This increase upon etomoxir was similar for both, WT and UCP3(-/-) mice, suggesting that UCP3 does not play a role in protection against lipotoxicity. Interestingly, we observed 25 % mortality in UCP3(-/-)s upon etomoxir administration vs. 11 % in WTs. This increased mortality in UCP3(-/-) compared to WT mice could not be explained by differences in cardiac lipotoxicity, apoptosis, fibrosis (histology, immunohistochemistry), oxidative capacity (respirometry) or function (echocardiography). Electrophysiology demonstrated, however, prolonged QRS and QTc intervals and greater susceptibility to ventricular tachycardia upon programmed electrical stimulation in etomoxir-treated UCP3(-/-)s versus WTs. Isoproterenol administration after pacing resulted in 75 % mortality in UCP3(-/-)s vs. 14 % in WTs. Our results argue against a protective role for UCP3 on skm metabolism under lipid overload, but suggest UCP3 to be crucial in prevention of arrhythmias upon lipid-challenged conditions.
Collapse
Affiliation(s)
- Miranda Nabben
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Bianca W J van Bree
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Ellen Lenaers
- Department of Human Movement Sciences, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Joris Hoeks
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Matthijs K C Hesselink
- Department of Human Movement Sciences, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Gert Schaart
- Department of Human Movement Sciences, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marion J J Gijbels
- Department of Molecular Genetics, CARIM School for Cardiovascular Research, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan F C Glatz
- Department of Molecular Genetics, CARIM School for Cardiovascular Research, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Gustavo J J da Silva
- Department of Cardiology, CARIM School for Cardiovascular Research, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leon J de Windt
- Department of Cardiology, CARIM School for Cardiovascular Research, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Elise Mike
- Department of Molecular Physiology and Biophysics and Medicine (Cardiology), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Darlene G Skapura
- Department of Molecular Physiology and Biophysics and Medicine (Cardiology), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Xander H T Wehrens
- Department of Molecular Physiology and Biophysics and Medicine (Cardiology), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Patrick Schrauwen
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, PO Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
39
|
Littlejohns B, Pasdois P, Duggan S, Bond AR, Heesom K, Jackson CL, Angelini GD, Halestrap AP, Suleiman MS. Hearts from mice fed a non-obesogenic high-fat diet exhibit changes in their oxidative state, calcium and mitochondria in parallel with increased susceptibility to reperfusion injury. PLoS One 2014; 9:e100579. [PMID: 24950187 PMCID: PMC4065057 DOI: 10.1371/journal.pone.0100579] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/26/2014] [Indexed: 11/20/2022] Open
Abstract
Rationale High-fat diet with obesity-associated co-morbidities triggers cardiac remodeling and renders the heart more vulnerable to ischemia/reperfusion injury. However, the effect of high-fat diet without obesity and associated co-morbidities is presently unknown. Objectives To characterize a non-obese mouse model of high-fat diet, assess the vulnerability of hearts to reperfusion injury and to investigate cardiac cellular remodeling in relation to the mechanism(s) underlying reperfusion injury. Methods and Results Feeding C57BL/6J male mice high-fat diet for 20 weeks did not induce obesity, diabetes, cardiac hypertrophy, cardiac dysfunction, atherosclerosis or cardiac apoptosis. However, isolated perfused hearts from mice fed high-fat diet were more vulnerable to reperfusion injury than those from mice fed normal diet. In isolated cardiomyocytes, high-fat diet was associated with higher diastolic intracellular Ca2+ concentration and greater damage to isolated cardiomyocytes following simulated ischemia/reperfusion. High-fat diet was also associated with changes in mitochondrial morphology and expression of some related proteins but not mitochondrial respiration or reactive oxygen species turnover rates. Proteomics, western blot and high-performance liquid chromatography techniques revealed that high-fat diet led to less cardiac oxidative stress, higher catalase expression and significant changes in expression of putative components of the mitochondrial permeability transition pore (mPTP). Inhibition of the mPTP conferred relatively more cardio-protection in the high-fat fed mice compared to normal diet. Conclusions This study shows for the first time that high-fat diet, independent of obesity-induced co-morbidities, triggers changes in cardiac oxidative state, calcium handling and mitochondria which are likely to be responsible for increased vulnerability to cardiac insults.
Collapse
Affiliation(s)
- Ben Littlejohns
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Philippe Pasdois
- Bioénergétique et Métabolisme, Institut de Rythmologie et Modélisation Cardiaque, Université de Bordeaux, Pessac, France
| | - Simon Duggan
- Oxford Heart Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Andrew R. Bond
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Kate Heesom
- Proteomics Facility, Faculty of Medical and Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Christopher L. Jackson
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Gianni D. Angelini
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Andrew P. Halestrap
- School of Biochemistry, Faculty of Medical and Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - M.-Saadeh Suleiman
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Tripathy S, Lytle KA, Stevens RD, Bain JR, Newgard CB, Greenberg AS, Huang LS, Jump DB. Fatty acid elongase-5 (Elovl5) regulates hepatic triglyceride catabolism in obese C57BL/6J mice. J Lipid Res 2014; 55:1448-64. [PMID: 24814977 DOI: 10.1194/jlr.m050062] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease is a major public health concern in the obese and type 2 diabetic populations. The high-fat lard diet induces obesity and fatty liver in C57BL/6J mice and suppresses expression of the PPAR-target gene, FA elongase 5 (Elovl5). Elovl5 plays a key role in MUFA and PUFA synthesis. Increasing hepatic Elovl5 activity in obese mice lowered hepatic TGs and endoplasmic reticulum stress markers (X-box binding protein 1 and cAMP-dependent transcription factor 6α) and increased TG catabolism and fatty acyl carnitines. Increased hepatic Elovl5 activity did not increase hepatic capacity for β-oxidation. Elovl5 effects on hepatic TG catabolism were linked to increased protein levels of adipocyte TG lipase (ATGL) and comparative gene identification 58 (CGI58). Elevated hepatic Elovl5 activity also induced the expression of some (pyruvate dehydrogenase kinase 4 and fibroblast growth factor 21), but not other cytochrome P450 4A10 (CYP4A10), PPAR-target genes. FA products of Elovl5 activity increased ATGL, but not CGI58, mRNA through PPARβ-dependent mechanisms in human HepG2 cells. Treatment of mouse AML12 hepatocytes with the PPARβ agonist (GW0742) decreased (14)C-18:2,n-6 in TGs but did not affect β-oxidation. These studies establish that Elovl5 activity regulates hepatic levels of FAs controlling PPARβ activity, ATGL expression, and TG catabolism, but not FA oxidation.
Collapse
Affiliation(s)
- Sasmita Tripathy
- School of Biological and Population Health Sciences and the Linus Pauling Institute, Oregon State University, Corvallis, OR 97331
| | - Kelli A Lytle
- School of Biological and Population Health Sciences and the Linus Pauling Institute, Oregon State University, Corvallis, OR 97331
| | - Robert D Stevens
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - Andrew S Greenberg
- Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center, Boston, MA 02111
| | - Li-Shin Huang
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY 10032
| | - Donald B Jump
- School of Biological and Population Health Sciences and the Linus Pauling Institute, Oregon State University, Corvallis, OR 97331
| |
Collapse
|
41
|
Hafstad AD, Lund J, Hadler-Olsen E, Höper AC, Larsen TS, Aasum E. High- and moderate-intensity training normalizes ventricular function and mechanoenergetics in mice with diet-induced obesity. Diabetes 2013; 62:2287-94. [PMID: 23493573 PMCID: PMC3712042 DOI: 10.2337/db12-1580] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although exercise reduces several cardiovascular risk factors associated with obesity/diabetes, the metabolic effects of exercise on the heart are not well-known. This study was designed to investigate whether high-intensity interval training (HIT) is superior to moderate-intensity training (MIT) in counteracting obesity-induced impairment of left ventricular (LV) mechanoenergetics and function. C57BL/6J mice with diet-induced obesity (DIO mice) displaying a cardiac phenotype with altered substrate utilization and impaired mechanoenergetics were subjected to a sedentary lifestyle or 8-10 weeks of isocaloric HIT or MIT. Although both modes of exercise equally improved aerobic capacity and reduced obesity, only HIT improved glucose tolerance. Hearts from sedentary DIO mice developed concentric LV remodeling with diastolic and systolic dysfunction, which was prevented by both HIT and MIT. Both modes of exercise also normalized LV mechanical efficiency and mechanoenergetics. These changes were associated with altered myocardial substrate utilization and improved mitochondrial capacity and efficiency, as well as reduced oxidative stress, fibrosis, and intracellular matrix metalloproteinase 2 content. As both modes of exercise equally ameliorated the development of diabetic cardiomyopathy by preventing LV remodeling and mechanoenergetic impairment, this study advocates the therapeutic potential of physical activity in obesity-related cardiac disorders.
Collapse
Affiliation(s)
- Anne D Hafstad
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| | | | | | | | | | | |
Collapse
|
42
|
Harmancey R, Vasquez HG, Guthrie PH, Taegtmeyer H. Decreased long-chain fatty acid oxidation impairs postischemic recovery of the insulin-resistant rat heart. FASEB J 2013; 27:3966-78. [PMID: 23825227 DOI: 10.1096/fj.13-234914] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Diabetic patients with acute myocardial infarction are more likely to die than nondiabetic patients. In the present study we examined the effect of insulin resistance on myocardial ischemia tolerance. Hearts of rats, rendered insulin resistant by high-sucrose feeding, were subjected to ischemia/reperfusion ex vivo. Cardiac power of control hearts from chow-fed rats recovered to 93%, while insulin-resistant hearts recovered only to 80% (P<0.001 vs. control). Unexpectedly, impaired contractile recovery did not result from an impairment of glucose oxidation (576±36 vs. 593±42 nmol/min/g dry weight; not significant), but from a failure to increase and to sustain oxidation of the long-chain fatty acid oleate on reperfusion (1878±56 vs. 2070±67 nmol/min/g dry weight; P<0.05). This phenomenon was due to a reduced ability to transport oleate into mitochondria and associated with a 38-58% decrease in the mitochondrial uncoupling protein 3 (UCP3) levels. Contractile function was rescued by replacing oleate with a medium-chain fatty acid or by restoring UCP3 levels with 24 h of food withdrawal. Lastly, the knockdown of UCP3 in rat L6 myocytes also decreased oleate oxidation by 13-18% following ischemia. Together the results expose UCP3 as a critical regulator of long-chain fatty acid oxidation in the stressed heart postischemia and identify octanoate as an intervention by which myocardial metabolism can be manipulated to improve function of the insulin-resistant heart.
Collapse
Affiliation(s)
- Romain Harmancey
- 1University of Texas Medical School at Houston, 6431 Fannin, MSB 1.246, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
43
|
Guzmán-Ruiz R, Gómez-Hurtado N, Gil-Ortega M, Somoza B, González MC, Aránguez I, Martín-Ramos M, González-Martín C, Delgado C, Fernández-Alfonso M, Ruiz-Gayo M. Remodeling of energy metabolism and absence of electrophysiological changes in the heart of obese hyperleptinemic mice. New insights into the pleiotropic role of leptin. Front Endocrinol (Lausanne) 2013; 4:175. [PMID: 24298268 PMCID: PMC3828673 DOI: 10.3389/fendo.2013.00175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/31/2013] [Indexed: 01/13/2023] Open
Abstract
Dietary treatment with high-fat diets (HFD) triggers diabetes and hyperleptinemia, concomitantly with a partial state of leptin resistance that affects hepatic and adipose tissue but not the heart. In this context, characterized by widespread steatosis, cardiac lipid content remains unchanged. As previously reported, HFD-evoked hyperleptinemia could be a pivotal element contributing to increase fatty-acid (FA) metabolism in the heart and to prevent cardiac steatosis. This metabolic adaptation might theoretically reduce energy efficiency in cardiomyocytes and lead to cardiac electrophysiological remodeling. Therefore the aim of the current study has been to investigate the impact of long-term HFD on cardiac metabolism and electrophysiological properties of the principal ionic currents responsible of the action potential duration in mouse cardiomyocytes. Male C57BL/6J mice were fed a control (10 kcal% from fat) or HFD (45 kcal% from fat) during 32 weeks. Quantification of enzymatic activities regulating mitochondrial uptake of pyruvate and FA showed an increase of both carnitine-palmitoyltransferase and citrate synthase activities together with a decrease of lactate dehydrogenase and pyruvate dehydrogenase activities. Increased expression of uncoupling protein-3, Mn-, and Cu/Zn-superoxide dismutases and catalase were also detected. Total glutathione/oxidized glutathione ratios were unaffected by HFD. These data suggest that HFD triggers adaptive mechanisms aimed at (i) facilitating FA catabolism, and (ii) preventing oxidative stress. All these changes did not affect the duration of action potentials in cardiomyocytes and only slightly modified electrocardiographic parameters.
Collapse
Affiliation(s)
- Rocío Guzmán-Ruiz
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
- Present address: Rocío Guzmán-Ruiz, Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Spain
| | - Nieves Gómez-Hurtado
- Departamento de Farmacología. Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - M. Carmen González
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma, Madrid, Spain
| | - Isabel Aránguez
- Departamento de Bioquímica, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Miriam Martín-Ramos
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Carmen González-Martín
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Carmen Delgado
- Departamento de Farmacología. Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Marisol Fernández-Alfonso
- Instituto Pluridisciplinar-Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
- *Correspondence: Mariano Ruiz-Gayo, Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Campus de Montepríncipe – Boadilla del Monte, 28668 Madrid, Spain e-mail:
| |
Collapse
|