1
|
Pepin ME, Konrad PJM, Nazir S, Bazgir F, Maack C, Nickel A, Gorman J, Hohl M, Schreiter F, Dewenter M, de Britto Chaves Filho A, Schulze A, Karlstaedt A, Frey N, Seidman C, Seidman J, Backs J. Mitochondrial NNT Promotes Diastolic Dysfunction in Cardiometabolic HFpEF. Circ Res 2025. [PMID: 40340422 DOI: 10.1161/circresaha.125.326154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Clinical management of heart failure with preserved ejection fraction (HFpEF) is hindered by a lack of disease-modifying therapies capable of altering its distinct pathophysiology. Despite the widespread implementation of a 2-hit model of cardiometabolic HFpEF to inform precision therapy, which utilizes ad libitum high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester, we observe that C57BL6/J mice exhibit less cardiac diastolic dysfunction in response to high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester. METHODS Genetic strain-specific single-nucleus transcriptomic analysis identified disease-relevant genes that enrich oxidative metabolic pathways within cardiomyocytes. Because C57BL/6J mice are known to harbor a loss-of-function mutation affecting the inner mitochondrial membrane protein Nnt (nicotinamide nucleotide transhydrogenase), we used an isogenic model of Nnt loss-of-function to determine whether intact NNT is necessary for the pathological cardiac manifestations of high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester. Twelve-week-old mice cross-bred to isolate wild-type (Nnt+/+) or loss-of-function (Nnt-/-) Nnt in the C57BL/6N background were challenged with high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester for 9 weeks (n=6-10). RESULTS Nnt+/+ mice exhibited impaired ventricular diastolic relaxation and pathological remodeling, as assessed via E/e' (42.8 versus 21.5, P=1.2×10-10), E/A (2.3 versus 1.4, P=4.1×10-2), diastolic stiffness (0.09 versus 0.04 mm Hg/μL, P=5.1×10-3), and myocardial fibrosis (P=2.3×10-2). Liquid chromatography and mass spectroscopy exposed a 40.0% reduction in NAD+ (P=8.4×10-3) and a 38.8% reduction in glutathione:GSSG (P=2.6×10-2) among Nnt+/+ mice after high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester feeding. Using single-nucleus ligand-receptor analysis, we implicate Fgf1 (fibroblast growth factor 1) as a putative NNT-dependent mediator of cardiomyocyte-to-fibroblast signaling of myocardial fibrosis. CONCLUSIONS Together, these findings underscore the pivotal role of mitochondrial dysfunction in HFpEF pathogenesis, implicating both NNT and Fgf1 as novel therapeutic targets.
Collapse
Affiliation(s)
- Mark E Pepin
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
- Broad Institute of Harvard and MIT, Boston, MA (M.E.P.)
- Division of Cardiovascular Medicine, Stanford University Hospital, CA (M.E.P.)
| | - Philipp J M Konrad
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine III, Heidelberg University Hospital, Germany. (P.J.M.K., N.F.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Sumra Nazir
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Farhad Bazgir
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Germany (C.M., A.N.)
- Medical Clinic I, University Clinic Würzburg, Germany (C.M.)
| | - Alexander Nickel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Germany (C.M., A.N.)
| | - Joshua Gorman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Mathias Hohl
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H.)
| | - Friederike Schreiter
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Matthias Dewenter
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Germany and Heidelberg University, Germany (M.D., J.B.)
| | | | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg (A.d.B.C.F., A.S.)
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (A.K.)
| | - Norbert Frey
- Department of Internal Medicine III, Heidelberg University Hospital, Germany. (P.J.M.K., N.F.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Christine Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Johannes Backs
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Heidelberg University, Germany. (J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Germany and Heidelberg University, Germany (M.D., J.B.)
| |
Collapse
|
2
|
Sharma G, Chaurasia SS, Carlson MA, Mishra PK. Recent advances associated with cardiometabolic remodeling in diabetes-induced heart failure. Am J Physiol Heart Circ Physiol 2024; 327:H1327-H1342. [PMID: 39453429 PMCID: PMC11684949 DOI: 10.1152/ajpheart.00539.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Diabetes mellitus (DM) is characterized by chronic hyperglycemia, and despite intensive glycemic control, the risk of heart failure in patients with diabetes remains high. Diabetes-induced heart failure (DHF) presents a unique metabolic challenge, driven by significant alterations in cardiac substrate metabolism, including increased reliance on fatty acid oxidation, reduced glucose utilization, and impaired mitochondrial function. These metabolic alterations lead to oxidative stress, lipotoxicity, and energy deficits, contributing to the progression of heart failure. Emerging research has identified novel mechanisms involved in the metabolic remodeling of diabetic hearts, such as autophagy dysregulation, epigenetic modifications, polyamine regulation, and branched-chain amino acid (BCAA) metabolism. These processes exacerbate mitochondrial dysfunction and metabolic inflexibility, further impairing cardiac function. Therapeutic interventions targeting these pathways-such as enhancing glucose oxidation, modulating fatty acid metabolism, and optimizing ketone body utilization-show promise in restoring metabolic homeostasis and improving cardiac outcomes. This review explores the key molecular mechanisms driving metabolic remodeling in diabetic hearts, highlights advanced methodologies, and presents the latest therapeutic strategies for mitigating the progression of DHF. Understanding these emerging pathways offers new opportunities to develop targeted therapies that address the root metabolic causes of heart failure in diabetes.
Collapse
Affiliation(s)
- Gaurav Sharma
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, United States
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, Texas, United States
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Shyam S Chaurasia
- Ocular Immunology and Angiogenesis Lab, Department Ophthalmology & Visual Sciences, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Mark A Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
3
|
Koleini N, Meddeb M, Zhao L, Keykhaei M, Kwon S, Farshidfar F, Hahn VS, Pearce EL, Sharma K, Kass DA. Landscape of glycolytic metabolites and their regulating proteins in myocardium from human heart failure with preserved ejection fraction. Eur J Heart Fail 2024; 26:1941-1951. [PMID: 39119952 PMCID: PMC11563863 DOI: 10.1002/ejhf.3389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/13/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
AIMS Heart failure (HF) with preserved ejection fraction (HFpEF) reflects half of all clinical HF yet has few therapies. Obesity and diabetes are now common comorbidities which have focused attention towards underlying myocardial metabolic defects. The profile of a major metabolic pathway, glycolytic intermediates and their regulating enzymes and ancillary pathways, remains unknown. METHODS AND RESULTS Endomyocardial biopsies from HFpEF (n = 37) and non-failing controls (n = 21) were assayed by non-targeted or targeted metabolomics and immunoblot to determine glycolytic and ancillary pathway metabolites and protein expression of their regulating enzymes. Glucose and GLUT1 expression were higher in HFpEF, but prominent glycolytic metabolites: glucose-6-phosphate, fructose-1,6-biphosphate (F1,6bP), and 3-phosphoglycerate were reduced by -78%, -91%, and -73%, respectively, versus controls. Expression of their corresponding synthesizing enzymes hexokinase, phospho-fructokinase, and phosphoglycerate kinase were also significantly lower (all p < 0.0005). Pentose phosphate and hexosamine biosynthetic pathway metabolites were reduced while glycogen content increased. Despite proximal reduction in key glycolytic intermediates, pyruvate increased but mitochondrial pyruvate transporter (MPC1) expression was reduced. Pyruvate dehydrogenase converting pyruvate to acetyl-CoA was more activated but some Krebs cycle intermediates were reduced. This HFpEF glycolytic profile persisted after adjusting for body mass index (BMI), diabetes, age, and sex, or in subgroup analysis with controls and HFpEF matched for BMI and diabetes/insulin history. In HFpEF, BMI but not glycated haemoglobin negatively correlated with F1,6bP (p = 7e-5, r = -0.61) and phosphoenolpyruvate (p = 0.006, r = -0.46). CONCLUSIONS Human HFpEF myocardium exhibits reduced glycolytic and ancillary pathway intermediates and expression of their synthesizing proteins. This combines features reported in HF with reduced ejection fraction and obesity/diabetes that likely exacerbate metabolic inflexibility.
Collapse
Affiliation(s)
- Navid Koleini
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mariam Meddeb
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liang Zhao
- Department of Chemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Complete Omics Inc, Baltimore, MD, USA
| | - Mohammad Keykhaei
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seoyoung Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Farnaz Farshidfar
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erika L Pearce
- Department of Chemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Takata T, Inoue S, Masauji T, Miyazawa K, Motoo Y. Generation and Accumulation of Various Advanced Glycation End-Products in Cardiomyocytes May Induce Cardiovascular Disease. Int J Mol Sci 2024; 25:7319. [PMID: 39000424 PMCID: PMC11242264 DOI: 10.3390/ijms25137319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Cardiomyocyte dysfunction and cardiovascular diseases (CVDs) can be classified as ischemic or non-ischemic. We consider the induction of cardiac tissue dysfunction by intracellular advanced glycation end-products (AGEs) in cardiomyocytes as a novel type of non-ischemic CVD. Various types of AGEs can be generated from saccharides (glucose and fructose) and their intermediate/non-enzymatic reaction byproducts. Recently, certain types of AGEs (Nε-carboxymethyl-lycine [CML], 2-ammnonio-6-[4-(hydroxymetyl)-3-oxidopyridinium-1-yl]-hexanoate-lysine [4-hydroxymethyl-OP-lysine, hydroxymethyl-OP-lysine], and Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine [MG-H1]) were identified and quantified in the ryanodine receptor 2 (RyR2) and F-actin-tropomyosin filament in the cardiomyocytes of mice or patients with diabetes and/or heart failure. Under these conditions, the excessive leakage of Ca2+ from glycated RyR2 and reduced contractile force from glycated F-actin-tropomyosin filaments induce cardiomyocyte dysfunction. CVDs are included in lifestyle-related diseases (LSRDs), which ancient people recognized and prevented using traditional medicines (e.g., Kampo medicines). Various natural compounds, such as quercetin, curcumin, and epigallocatechin-3-gallate, in these drugs can inhibit the generation of intracellular AGEs through mechanisms such as the carbonyl trap effect and glyoxalase 1 activation, potentially preventing CVDs caused by intracellular AGEs, such as CML, hydroxymethyl-OP, and MG-H1. These investigations showed that bioactive herbal extracts obtained from traditional medicine treatments may contain compounds that prevent CVDs.
Collapse
Affiliation(s)
- Takanobu Takata
- Division of Molecular and Genetic Biology, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293, Japan;
| | - Shinya Inoue
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan; (S.I.); (K.M.)
| | - Togen Masauji
- Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293, Japan;
| | - Katsuhito Miyazawa
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan; (S.I.); (K.M.)
| | - Yoshiharu Motoo
- Department of Internal Medicine, Fukui Saiseikai Hospital, Wadanaka, Fukui 918-8503, Japan
| |
Collapse
|
5
|
Lou S, Zhu W, Yu T, Zhang Q, Wang M, Jin L, Xiong Y, Xu J, Wang Q, Chen G, Liang G, Hu X, Luo W. Compound SJ-12 attenuates streptozocin-induced diabetic cardiomyopathy by stabilizing SERCA2a. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167140. [PMID: 38548092 DOI: 10.1016/j.bbadis.2024.167140] [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: 10/04/2023] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/01/2024]
Abstract
Heart failure (HF) is one of the major causes of death among diabetic patients. Although studies have shown that curcumin analog C66 can remarkably relieve diabetes-associated cardiovascular and kidney complications, the role of SJ-12, SJ-12, a novel curcumin analog, in diabetic cardiomyopathy and its molecular targets are unknown. 7-week-old male C57BL/6 mice were intraperitoneally injected with single streptozotocin (STZ) (160 mg/kg) to develop diabetic cardiomyopathy (DCM). The diabetic mice were then treated with SJ-12 via gavage for two months. Body weight, fast blood glucose, cardiac utrasonography, myocardial injury markers, pathological morphology of the heart, hypertrophic and fibrotic markers were assessed. The potential target of SJ-12 was evaluated via RNA-sequencing analysis. The O-GlcNAcylation levels of SP1 were detected via immunoprecipitation. SJ-12 effectively suppressed myocardial hypertrophy and fibrosis, thereby preventing heart dysfunction in mice with STZ-induced heart failure. RNA-sequencing analysis revealed that SJ-12 exerted its therapeutic effects through the modulation of the calcium signaling pathway. Furthermore, SJ-12 reduced the O-GlcNAcylation levels of SP1 by inhibiting O-linked N-acetylglucosamine transferase (OGT). Also, SJ-12 stabilized Sarcoplasmic/Endoplasmic Reticulum Calcium ATPase 2a (SERCA2a), a crucial regulator of calcium homeostasis, thus reducing hypertrophy and fibrosis in mouse hearts and cultured cardiomyocytes. However, the anti-fibrotic effects of SJ-12 were not detected in SERCA2a or OGT-silenced cardiomyocytes, indicating that SJ-12 can prevent DCM by targeting OGT-dependent O-GlcNAcylation of SP1.These findings indicate that SJ-12 can exert cardioprotective effects in STZ-induced mice by reducing the O-GlcNAcylation levels of SP1, thus stabilizing SERCA2a and reducing myocardial fibrosis and hypertrophy. Therefore, SJ-12 can be used for the treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shuaijie Lou
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Weiwei Zhu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Tianxiang Yu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Qianhui Zhang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Minxiu Wang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Leiming Jin
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Yongqiang Xiong
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiachen Xu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Qinyan Wang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Gaozhi Chen
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Guang Liang
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| | - Xiang Hu
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Wu Luo
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
6
|
Chen S, Zou Y, Song C, Cao K, Cai K, Wu Y, Zhang Z, Geng D, Sun W, Ouyang N, Zhang N, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches. Basic Res Cardiol 2023; 118:48. [PMID: 37938421 PMCID: PMC10632287 DOI: 10.1007/s00395-023-01018-w] [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: 05/24/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
Cardiovascular disease (CVD) is a major threat to human health, accounting for 46% of non-communicable disease deaths. Glycolysis is a conserved and rigorous biological process that breaks down glucose into pyruvate, and its primary function is to provide the body with the energy and intermediate products needed for life activities. The non-glycolytic actions of enzymes associated with the glycolytic pathway have long been found to be associated with the development of CVD, typically exemplified by metabolic remodeling in heart failure, which is a condition in which the heart exhibits a rapid adaptive response to hypoxic and hypoxic conditions, occurring early in the course of heart failure. It is mainly characterized by a decrease in oxidative phosphorylation and a rise in the glycolytic pathway, and the rise in glycolysis is considered a hallmark of metabolic remodeling. In addition to this, the glycolytic metabolic pathway is the main source of energy for cardiomyocytes during ischemia-reperfusion. Not only that, the auxiliary pathways of glycolysis, such as the polyol pathway, hexosamine pathway, and pentose phosphate pathway, are also closely related to CVD. Therefore, targeting glycolysis is very attractive for therapeutic intervention in CVD. However, the relationship between glycolytic pathway and CVD is very complex, and some preclinical studies have confirmed that targeting glycolysis does have a certain degree of efficacy, but its specific role in the development of CVD has yet to be explored. This article aims to summarize the current knowledge regarding the glycolytic pathway and its key enzymes (including hexokinase (HK), phosphoglucose isomerase (PGI), phosphofructokinase-1 (PFK1), aldolase (Aldolase), phosphoglycerate metatase (PGAM), enolase (ENO) pyruvate kinase (PKM) lactate dehydrogenase (LDH)) for their role in cardiovascular diseases (e.g., heart failure, myocardial infarction, atherosclerosis) and possible emerging therapeutic targets.
Collapse
Affiliation(s)
- Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Nanxiang Ouyang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
7
|
Norlin S, Axelsson J, Ericsson M, Edlund H. O304 ameliorates hyperglycemia in mice by dually promoting muscle glucose effectiveness and preserving β-cell function. Commun Biol 2023; 6:877. [PMID: 37626210 PMCID: PMC10457357 DOI: 10.1038/s42003-023-05255-6] [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: 10/04/2022] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Although insulin mediated glucose uptake in skeletal muscle is a major mechanism ensuring glucose disposal in humans, glucose effectiveness, i.e., the ability of glucose itself to stimulate its own uptake independent of insulin, accounts for roughly half of the glucose disposed during an oral glucose tolerance test. Both insulin dependent and insulin independent skeletal muscle glucose uptake are however reduced in individuals with diabetes. We here show that AMPK activator O304 stimulates insulin independent glucose uptake and utilization in skeletal muscle and heart in vivo, while preventing glycogen accumulation. Combined glucose uptake and utilization requires an increased metabolic demand and we show that O304 acts as a mitochondrial uncoupler, i.e., generates a metabolic demand. O304 averts gene expression changes associated with metabolic inflexibility in skeletal muscle and heart of diabetic mice and reverts diabetic cardiomyopathy. In Type 2 diabetes, insulin resistance elicits compensatory insulin hypersecretion, provoking β-cell stress and eventually compensatory failure. In db/db mice O304 preserves β-cell function by preventing decline in insulin secretion, β-cell mass, and pancreatic insulin content. Thus, as a dual AMPK activator and mitochondrial uncoupler O304 mitigates two central defects of T2D; impaired glucose uptake/utilization and β-cell failure, which today lack effective treatment.
Collapse
Affiliation(s)
- Stefan Norlin
- Umeå Centre for Molecular Medicine, Umeå University, SE-901 87, Umeå, Sweden
| | - Jan Axelsson
- Department of Radiation Sciences, Radiation Physics, Umeå University, SE-901 87, Umeå, Sweden
| | - Madelene Ericsson
- Umeå Centre for Molecular Medicine, Umeå University, SE-901 87, Umeå, Sweden
| | - Helena Edlund
- Umeå Centre for Molecular Medicine, Umeå University, SE-901 87, Umeå, Sweden.
| |
Collapse
|
8
|
AlTamimi JZ, AlFaris NA, Alshammari GM, Alagal RI, Aljabryn DH, Yahya MA. The Protective Effect of 11-Keto-β-Boswellic Acid against Diabetic Cardiomyopathy in Rats Entails Activation of AMPK. Nutrients 2023; 15:nu15071660. [PMID: 37049501 PMCID: PMC10097356 DOI: 10.3390/nu15071660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/25/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
This study examined the protective effect of 11-keto-β-boswellic acid (AKBA) against streptozotocin (STZ)-induced diabetic cardiomyopathy (DC) in rats and examined the possible mechanisms of action. Male rats were divided into 5 groups (n = 8/each): (1) control, AKBA (10 mg/kg, orally), STZ (65 mg/kg, i.p.), STZ + AKBA (10 mg/kg, orally), and STZ + AKBA + compound C (CC/an AMPK inhibitor, 0.2 mg/kg, i.p.). AKBA improved the structure and the systolic and diastolic functions of the left ventricles (LVs) of STZ rats. It also attenuated the increase in plasma glucose, plasma insulin, and serum and hepatic levels of triglycerides (TGs), cholesterol (CHOL), and free fatty acids (FFAs) in these diabetic rats. AKBA stimulated the ventricular activities of phosphofructokinase (PFK), pyruvate dehydrogenase (PDH), and acetyl CoA carboxylase (ACC); increased levels of malonyl CoA; and reduced levels of carnitine palmitoyltransferase I (CPT1), indicating improvement in glucose and FA oxidation. It also reduced levels of malondialdehyde (MDA); increased mitochondria efficiency and ATP production; stimulated mRNA, total, and nuclear levels of Nrf2; increased levels of glutathione (GSH), heme oxygenase (HO-1), superoxide dismutase (SOD), and catalase (CAT); but reduced the expression and nuclear translocation of NF-κB and levels of tumor-necrosis factor-α (TNF-α) and interleukin-6 (IL-6). These effects were concomitant with increased activities of AMPK in the LVs of the control and STZ-diabetic rats. Treatment with CC abolished all these protective effects of AKBA. In conclusion, AKBA protects against DC in rats, mainly by activating the AMPK-dependent control of insulin release, cardiac metabolism, and antioxidant and anti-inflammatory effects.
Collapse
|
9
|
Han S, Hou Y, Liu H, Zhao Q. The efficacy and safety of traditional Chinese medicine treating diabetic cardiomyopathy: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e31269. [PMID: 36451502 PMCID: PMC9704940 DOI: 10.1097/md.0000000000031269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Diabetic cardiomyopathy, secondary to diabetes, is the main cause of death in patients with diabetes. In China, traditional Chinese medicine has achieved good performance in treating diabetic cardiomyopathy. However, to date, no systematic review or meta-analysis has been published on the treatment of diabetic cardiomyopathy by traditional Chinese medicine. METHODS This study strictly followed the preferred guidelines for systematic review. Two researchers searched seven databases: EMbase, PubMed, Web of Science, Cochrane Library, China National Knowledge Infrastructure, Chinese Scientific Journal Database, and WANFANG Database. The retrieval time limit ranged from the establishment of the database to August 2022. All clinical randomized controlled trials that met the inclusion and exclusion criteria were included in this study. Statistical analysis was performed using RevMan 5.3. RESULTS This study analyzed the clinical efficacy and safety of traditional Chinese medicine in the treatment of diabetic cardiomyopathy. CONCLUSION The results of this study provide evidence-based medical evidence for the clinical use of traditional Chinese medicine in the treatment of diabetic heart disease in the future.
Collapse
Affiliation(s)
- Shuo Han
- Zibo Branch of the 960th Hospital of the Chinese People’s Liberation Army, Zibo, China
- * Correspondence: Shuo Han, Zibo Branch of the 960th Hospital of the Chinese People’s Liberation Army, Zibo 255300, China (e-mail: )
| | - Yuan Hou
- Shandong University of Chinese Medicine, Jinan, China
| | - Huaman Liu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Quanlin Zhao
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
10
|
Awolaja OO, Lawal AO, Folorunso IM, Elekofehinti OO, Umar HI. Silibinin ameliorates the cardiovascular oxidative and inflammatory effects of type-2-diabetic rats exposed to air particulate matter. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2123536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Olamide O. Awolaja
- Molecular Biology and Bioinformatics Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, Akure, Nigeria
| | - Akeem O. Lawal
- Molecular Biology and Bioinformatics Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, Akure, Nigeria
| | - Ibukun M. Folorunso
- Molecular Biology and Bioinformatics Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, Akure, Nigeria
| | - Olusola O. Elekofehinti
- Molecular Biology and Bioinformatics Unit, Department of Biochemistry, School of Sciences, Federal University of Technology, Akure, Nigeria
| | - Haruna I. Umar
- Computer-Aided Therapeutic Discovery and Design Group, FUTA, Akure, Nigeria
| |
Collapse
|
11
|
Bosma KJ, Ghosh M, Andrei SR, Zhong L, Dunn JC, Ricciardi VF, Burkett JB, Hatzopoulos AK, Damron DS, Gannon M. Pharmacological modulation of prostaglandin E 2 (PGE 2 ) EP receptors improves cardiomyocyte function under hyperglycemic conditions. Physiol Rep 2022; 10:e15212. [PMID: 35403369 PMCID: PMC8995713 DOI: 10.14814/phy2.15212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023] Open
Abstract
Type 2 diabetes (T2D) affects >30 million Americans and nearly 70% of individuals with T2D will die from cardiovascular disease (CVD). Circulating levels of the inflammatory signaling lipid, prostaglandin E2 (PGE2 ), are elevated in the setting of obesity and T2D and are associated with decreased cardiac function. The EP3 and EP4 PGE2 receptors have opposing actions in several tissues, including the heart: overexpression of EP3 in cardiomyocytes impairs function, while EP4 overexpression improves function. Here we performed complementary studies in vitro with isolated cardiomyocytes and in vivo using db/db mice, a model of T2D, to analyze the effects of EP3 inhibition or EP4 activation on cardiac function. Using echocardiography, we found that 2 weeks of systemic treatment of db/db mice with 20 mg/kg of EP3 antagonist, beginning at 6 weeks of age, improves ejection fraction and fractional shortening (with no effect on heart rate). We further show that either EP3 blockade or EP4 activation enhances contractility and calcium cycling in isolated mouse cardiomyocytes cultured in both normal and high glucose. Thus, peak [Ca2+ ]I transient amplitude was increased, while time to peak [Ca2+ ]I and [Ca2+ ]I decay were decreased. These data suggest that modulation of EP3 and EP4 activity has beneficial effects on cardiomyocyte contractility and overall heart function.
Collapse
Affiliation(s)
- Karin J. Bosma
- Department of Veterans Affairs Tennessee Valley AuthorityNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Monica Ghosh
- Department of Biological SciencesSchool of Biomedical SciencesKent State UniversityKentOhioUSA
| | - Spencer R. Andrei
- Department of Veterans Affairs Tennessee Valley AuthorityNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Lin Zhong
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jennifer C. Dunn
- Department of Veterans Affairs Tennessee Valley AuthorityNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Juliann B. Burkett
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennesseeUSA
| | - Antonis K. Hatzopoulos
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Derek S. Damron
- Department of Biological SciencesSchool of Biomedical SciencesKent State UniversityKentOhioUSA
| | - Maureen Gannon
- Department of Veterans Affairs Tennessee Valley AuthorityNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennesseeUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTennesseeUSA
| |
Collapse
|
12
|
Bloomgarden Z. The world congress on insulin resistance, diabetes, and cardiovascular disease (WCIRDC). J Diabetes 2022; 14:163-166. [PMID: 35191189 PMCID: PMC9060065 DOI: 10.1111/1753-0407.13260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
13
|
Stanton AM, Vaduganathan M, Chang LS, Turchin A, Januzzi JL, Aroda VR. Asymptomatic Diabetic Cardiomyopathy: an Underrecognized Entity in Type 2 Diabetes. Curr Diab Rep 2021; 21:41. [PMID: 34580767 DOI: 10.1007/s11892-021-01407-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Type 2 diabetes (T2D) is associated with an increased risk of heart failure (HF), with diabetic cardiomyopathy (DbCM) referring to abnormal heart structure in the absence of other driving cardiac factors such as hypertension, coronary artery disease (CAD), and valvular heart disease. Stage B DbCM is commonly asymptomatic and represents a form of stage B HF; DbCM thus represents a transitional phenotype prior to onset of symptomatic HF. The pathogenesis of DbCM is not fully elucidated but involves hyperglycemia, insulin resistance, increased free fatty acids (FFA), lipotoxicity, oxidative stress, advanced glycation end product (AGE) formation, activation of the renin-angiotensin-aldosterone system (RAAS) with an increase in angiotensin II, and dyshomeostasis of calcium, which all contribute to left ventricular hypertrophy (LVH) and cardiac systolic and diastolic dysfunction. Although DbCM is an established pathogenic process, it is underrecognized clinically due to its commonly asymptomatic nature. Raising awareness to identify high-risk individuals with stage B HF due to DbCM, who may subsequently progress to overt HF (stage C/D HF), as well as identifying new pharmacological agents and approaches to prevent functional decline, may help reduce this global health problem. The aim of this review is to focus on stage B DbCM; provide data on diagnostic approaches, current therapies, and potential therapies under investigation; and highlight the need to raise awareness and interdisciplinary dialogue among clinicians and researchers. RECENT FINDINGS There are no currently approved therapeutic strategies to treat or prevent progression of stage B DbCM, but multiple attempts are being made to target different pathogenic mechanisms involved in the development of DbCM. Recent cardiovascular (CV) outcome trials (CVOTs) have identified newer therapeutic agents with CV benefit, such as sodium-glucose cotransporter-2 (SGLT-2) inhibitors that reduce hospitalization for HF and glucagon-like peptide-1 (GLP-1) receptor agonists that reduce major adverse CV events (MACE), though without consistent effect on HF outcomes. Recent clinical practice guidelines recommend screening patients at high risk for HF. Further definition and interdisciplinary discussion of high-yield populations to screen, appropriate subsequent evaluation and intervention are needed to advance this area. DbCM is a complex entity that results from multiple pathogenic mechanisms triggered by impairment of glucose and lipid metabolism over many years. DbCM is commonly asymptomatic and represents a form of stage B HF. It is an underrecognized process that may progress to functional decline and overt HF. Although newer medications approved for the treatment of T2D may play an important role in reducing the risk of HF complications, less focus has been placed on earlier recognition and treatment of DbCM while asymptomatic. Additional efforts should be made to further study and target this stage in order to decrease the overall burden of HF.
Collapse
Affiliation(s)
- Ana Maria Stanton
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, 221 Longwood Ave, Office 381, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Muthiah Vaduganathan
- Harvard Medical School, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lee-Shing Chang
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, 221 Longwood Ave, Office 381, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alexander Turchin
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, 221 Longwood Ave, Office 381, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - James L Januzzi
- Harvard Medical School, Boston, MA, USA
- Cardiology Division, Massachusetts General Hospital, Baim Institute for Clinical Research, Boston, MA, USA
| | - Vanita R Aroda
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, 221 Longwood Ave, Office 381, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Tayanloo-Beik A, Roudsari PP, Rezaei-Tavirani M, Biglar M, Tabatabaei-Malazy O, Arjmand B, Larijani B. Diabetes and Heart Failure: Multi-Omics Approaches. Front Physiol 2021; 12:705424. [PMID: 34421642 PMCID: PMC8378451 DOI: 10.3389/fphys.2021.705424] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetes and heart failure, as important global issues, cause substantial expenses to countries and medical systems because of the morbidity and mortality rates. Most people with diabetes suffer from type 2 diabetes, which has an amplifying effect on the prevalence and severity of many health problems such as stroke, neuropathy, retinopathy, kidney injuries, and cardiovascular disease. Type 2 diabetes is one of the cornerstones of heart failure, another health epidemic, with 44% prevalence. Therefore, finding and targeting specific molecular and cellular pathways involved in the pathophysiology of each disease, either in diagnosis or treatment, will be beneficial. For diabetic cardiomyopathy, there are several mechanisms through which clinical heart failure is developed; oxidative stress with mediation of reactive oxygen species (ROS), reduced myocardial perfusion due to endothelial dysfunction, autonomic dysfunction, and metabolic changes, such as impaired glucose levels caused by insulin resistance, are the four main mechanisms. In the field of oxidative stress, advanced glycation end products (AGEs), protein kinase C (PKC), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) are the key mediators that new omics-driven methods can target. Besides, diabetes can affect myocardial function by impairing calcium (Ca) homeostasis, the mechanism in which reduced protein phosphatase 1 (PP1), sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a), and phosphorylated SERCA2a expressions are the main effectors. This article reviewed the recent omics-driven discoveries in the diagnosis and treatment of type 2 diabetes and heart failure with focus on the common molecular mechanisms.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyvand Parhizkar Roudsari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ozra Tabatabaei-Malazy
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Kaleem A, Javed S, Rehman N, Abdullah R, Iqtedar M, Aftab MN, Hoessli DC, Haq IU. Phosphorylated and O-GlcNAc Modified IRS-1 (Ser1101) and -2 (Ser1149) Contribute to Human Diabetes Type II. Protein Pept Lett 2021; 28:333-339. [PMID: 32798372 DOI: 10.2174/0929866527666200813210407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence of the chronic metabolic disorder Type 2 diabetes mellitus (T2DM) is increasing steadily, and has even turned into an epidemic in some countries. T2DM results from defective responses to insulin and obesity is a major factor behind insulin resistance in T2DM. Insulin receptor substrate (IRS) proteins are adaptor proteins in the insulin receptor signalling pathway. The insulin signalling is controlled through tyrosine phosphorylation of IRS-1 and IRS-2, and dysregulation of IRS proteins signalling may lead to glucose intolerance and eventually insulin resistance. OBJECTIVE In this work, we suggest that both glycosylation (O-GlcNAc modification) and phosphorylation of IRS-1 and -2 are involved in the pathogenesis of T2DM. METHODS Phosphorylation and O-GlcNAc modifications (Ser1101 in IRS-1 and Ser1149 in IRS-2) proteins were determined experimentally by sandwich ELISA with specific antibodies and with bioinformatics tools. RESULTS When IRS-1 (on Ser1101) and IRS-2 (Ser1149) become glycosylated following an increase in UDP-GlcNAc pools, it may contribute to insulin resistance. Whereas when the same (IRS-1 on Ser1101 and IRS-2 on Ser1149) are phosphorylated, the insulin signalling is inhibited. DISCUSSION In this work OGlcNAc-modified proteins were specifically detected using O-Glc- NAc-specific antibodies, suggesting that elevated levels of O-GlcNAc-modified proteins are found, independently of their possible involvement in Advanced Glycation End products (AGEs). CONCLUSION This study suggests a mechanism, which is controlled by posttranslational modifications, and may contribute to the pathogenesis of type II diabetes.
Collapse
Affiliation(s)
- Afshan Kaleem
- Department of Biotechnology, Lahore College for Women University, Lahore, Punjab 54000, Pakistan
| | - Sabahat Javed
- Department of Biotechnology, Lahore College for Women University, Lahore, Punjab 54000, Pakistan
| | - Nayab Rehman
- Department of Biotechnology, Lahore College for Women University, Lahore, Punjab 54000, Pakistan
| | - Roheena Abdullah
- Department of Biotechnology, Lahore College for Women University, Lahore, Punjab 54000, Pakistan
| | - Mehwish Iqtedar
- Department of Biotechnology, Lahore College for Women University, Lahore, Punjab 54000, Pakistan
| | - Mohammad Nauman Aftab
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Daniel C Hoessli
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ikram-Ul Haq
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| |
Collapse
|
16
|
Takeuchi M, Sakasai-Sakai A, Takata T, Takino JI, Koriyama Y, Kikuchi C, Furukawa A, Nagamine K, Hori T, Matsunaga T. Intracellular Toxic AGEs (TAGE) Triggers Numerous Types of Cell Damage. Biomolecules 2021; 11:biom11030387. [PMID: 33808036 PMCID: PMC8001776 DOI: 10.3390/biom11030387] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The habitual intake of large amounts of sugar, which has been implicated in the onset/progression of lifestyle-related diseases (LSRD), induces the excessive production of glyceraldehyde (GA), an intermediate of sugar metabolism, in neuronal cells, hepatocytes, and cardiomyocytes. Reactions between GA and intracellular proteins produce toxic advanced glycation end-products (toxic AGEs, TAGE), the accumulation of which contributes to various diseases, such as Alzheimer’s disease, non-alcoholic steatohepatitis, and cardiovascular disease. The cellular leakage of TAGE affects the surrounding cells via the receptor for AGEs (RAGE), thereby promoting the onset/progression of LSRD. We demonstrated that the intracellular accumulation of TAGE triggered numerous cellular disorders, and also that TAGE leaked into the extracellular space, thereby increasing extracellular TAGE levels in circulating fluids. Intracellular signaling and the production of reactive oxygen species are affected by extracellular TAGE and RAGE interactions, which, in turn, facilitate the intracellular generation of TAGE, all of which may contribute to the pathological changes observed in LSRD. In this review, we discuss the relationships between intracellular TAGE levels and numerous types of cell damage. The novel concept of the “TAGE theory” is expected to open new perspectives for research into LSRD.
Collapse
Affiliation(s)
- Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan; (A.S.-S.); (T.T.)
- Correspondence: ; Tel.: +81-76-218-8456
| | - Akiko Sakasai-Sakai
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan; (A.S.-S.); (T.T.)
| | - Takanobu Takata
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan; (A.S.-S.); (T.T.)
| | - Jun-ichi Takino
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (J.-i.T.); (T.H.)
| | - Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8670, Japan; (Y.K.); (A.F.)
| | - Chigusa Kikuchi
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (C.K.); (T.M.)
| | - Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8670, Japan; (Y.K.); (A.F.)
| | - Kentaro Nagamine
- Department of Clinical Nutrition, Faculty of Health Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan;
| | - Takamitsu Hori
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (J.-i.T.); (T.H.)
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (C.K.); (T.M.)
| |
Collapse
|
17
|
Ying F, Liu H, Ching Tang EH, Lakhani I, Liu N, Xia Z, Liu S. Prostaglandin E receptor subtype 4 protects against diabetic cardiomyopathy by modulating cardiac fatty acid metabolism via FOXO1/CD36 signalling. Biochem Biophys Res Commun 2021; 548:196-203. [PMID: 33647796 DOI: 10.1016/j.bbrc.2021.01.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cardiac fatty acid metabolism is essential for maintaining normal cardiac function at baseline and in response to various disease stress, like diabetes. EP4 is widely expressed in cardiomyocytes and has been demonstrated to play a role in cardio function. However, its function in regulating cardiac fatty acid metabolism is remained unknown. METHODS Mice were fed with standard chow or high-fat for eight weeks. The effects of EP4 deficiency on cardiac function, cardiomyocytes hypertrophy and myocardial fibrosis were studied. The possible regulatory mechanisms were further investigated. RESULTS EP4-/- mice exhibited concentric hypertrophy and myocardial fibrosis with cardiac energy deprivation due to reduction of fatty acid uptake and inhibition of ATP generation mediated by FOXO1/CD36 signalling. Moreover, pharmacologically activated EP4 alleviated impaired fatty acid transport and insufficient ATP generation in cardiomyocytes. CONCLUSION EP4 tightly coordinates the rates of cardiac fatty acid uptake and ATP generation via FOXO1/CD36 signalling axis. Our study provides evidences for the link between EP4 and cardiac fatty acid transport and further pointed out that EP4 could be a potential target for modulating fatty acid metabolism and curbing cardiac tissue-specific impairment of function following diabetes.
Collapse
Affiliation(s)
- Fan Ying
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, PR China
| | - Hao Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, PR China
| | - Eva Hoi Ching Tang
- Department of Pharmacology and Pharmacy and State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ishan Lakhani
- Cardiovascular Analytics Group, Laboratory of Cardiovascular Physiology, Hong Kong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, PR China
| | - Zhengyuan Xia
- Department of Anesthesiology, State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, PR China.
| |
Collapse
|
18
|
Parijat P, Kondacs L, Alexandrovich A, Gautel M, Cobb AJA, Kampourakis T. High Throughput Screen Identifies Small Molecule Effectors That Modulate Thin Filament Activation in Cardiac Muscle. ACS Chem Biol 2021; 16:225-235. [PMID: 33315370 DOI: 10.1021/acschembio.0c00908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Current therapeutic interventions for both heart disease and heart failure are largely insufficient and associated with undesired side effects. Biomedical research has emphasized the role of sarcomeric protein function for the normal performance and energy efficiency of the heart, suggesting that directly targeting the contractile myofilaments themselves using small molecule effectors has therapeutic potential and will likely result in greater drug efficacy and selectivity. In this study, we developed a robust and highly reproducible fluorescence polarization-based high throughput screening (HTS) assay that directly targets the calcium-dependent interaction between cardiac troponin C (cTnC) and the switch region of cardiac troponin I (cTnISP), with the aim of identifying small molecule effectors of the cardiac thin filament activation pathway. We screened a commercially available small molecule library and identified several hit compounds with both inhibitory and activating effects. We used a range of biophysical and biochemical methods to characterize hit compounds and identified fingolimod, a sphingosin-1-phosphate receptor modulator, as a new troponin-based small molecule effector. Fingolimod decreased the ATPase activity and calcium sensitivity of demembranated cardiac muscle fibers in a dose-dependent manner, suggesting that the compound acts as a calcium desensitizer. We investigated fingolimod's mechanism of action using a combination of computational studies, biophysical methods, and synthetic chemistry, showing that fingolimod bound to cTnC repels cTnISP via mainly electrostatic repulsion of its positively charged tail. These results suggest that fingolimod is a potential new lead compound/scaffold for the development of troponin-directed heart failure therapeutics.
Collapse
Affiliation(s)
- Priyanka Parijat
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Laszlo Kondacs
- Department of Chemistry, King’s College London, 7 Trinity Street, London, SE1 1DB, United Kingdom
| | - Alexander Alexandrovich
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Alexander J. A. Cobb
- Department of Chemistry, King’s College London, 7 Trinity Street, London, SE1 1DB, United Kingdom
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| |
Collapse
|
19
|
Umbarawan Y, Kawakami R, Syamsunarno MRAA, Koitabashi N, Obinata H, Yamaguchi A, Hanaoka H, Hishiki T, Hayakawa N, Sunaga H, Matsui H, Kurabayashi M, Iso T. Reduced fatty acid uptake aggravates cardiac contractile dysfunction in streptozotocin-induced diabetic cardiomyopathy. Sci Rep 2020; 10:20809. [PMID: 33257783 PMCID: PMC7705707 DOI: 10.1038/s41598-020-77895-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022] Open
Abstract
Diabetes is an independent risk factor for the development of heart failure. Increased fatty acid (FA) uptake and deranged utilization leads to reduced cardiac efficiency and accumulation of cardiotoxic lipids, which is suggested to facilitate diabetic cardiomyopathy. We studied whether reduced FA uptake in the heart is protective against streptozotocin (STZ)-induced diabetic cardiomyopathy by using mice doubly deficient in fatty acid binding protein 4 (FABP4) and FABP5 (DKO mice). Cardiac contractile dysfunction was aggravated 8 weeks after STZ treatment in DKO mice. Although compensatory glucose uptake was not reduced in DKO-STZ hearts, total energy supply, estimated by the pool size in the TCA cycle, was significantly reduced. Tracer analysis with 13C6-glucose revealed that accelerated glycolysis in DKO hearts was strongly suppressed by STZ treatment. Levels of ceramides, cardiotoxic lipids, were similarly elevated by STZ treatment. These findings suggest that a reduction in total energy supply by reduced FA uptake and suppressed glycolysis could account for exacerbated contractile dysfunction in DKO-STZ hearts. Thus, enhanced FA uptake in diabetic hearts seems to be a compensatory response to reduced energy supply from glucose, and therefore, limited FA use could be detrimental to cardiac contractile dysfunction due to energy insufficiency.
Collapse
Affiliation(s)
- Yogi Umbarawan
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.,Department of Internal Medicine, Faculty of Medicine Universitas Indonesia, Jl. Salemba Raya no. 6, Jakarta, 10430, Indonesia
| | - Ryo Kawakami
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Mas Rizky A A Syamsunarno
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.,Department of Biochemistry and Molecular Biology, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM 21, Jatinangor, West Java, 45363, Indonesia
| | - Norimichi Koitabashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Aiko Yamaguchi
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hirofumi Hanaoka
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Clinical and Translational Research Center, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Noriyo Hayakawa
- Clinical and Translational Research Center, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroaki Sunaga
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.,Center for Liberal Arts and Sciences, Ashikaga University, 268-1 Omae-machi, Ashikaga, Tochigi, 326-8558, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tatsuya Iso
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
20
|
Shu H, Peng Y, Hang W, Nie J, Zhou N, Wang DW. The role of CD36 in cardiovascular disease. Cardiovasc Res 2020; 118:115-129. [PMID: 33210138 PMCID: PMC8752351 DOI: 10.1093/cvr/cvaa319] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
CD36, also known as the scavenger receptor B2, is a multifunctional receptor widely expressed in various organs. CD36 plays a crucial role in the uptake of long-chain fatty acids, the main metabolic substrate in myocardial tissue. The maturation and transportation of CD36 is regulated by post-translational modifications, including phosphorylation, ubiquitination, glycosylation, and palmitoylation. CD36 is decreased in pathological cardiac hypertrophy caused by ischaemia-reperfusion and pressure overload, and increased in diabetic cardiomyopathy and atherosclerosis. Deficiency of CD36 alleviates diabetic cardiomyopathy and atherosclerosis, while overexpression of CD36 eliminates ischaemia-reperfusion damage, together suggesting that CD36 is closely associated with the progression of cardiovascular diseases and may be a new therapeutic target. This review summarizes the regulation and post-translational modifications of CD36 and evaluates its role in cardiovascular diseases and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Weijian Hang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
21
|
Nirengi S, Peres Valgas da Silva C, Stanford KI. Disruption of energy utilization in diabetic cardiomyopathy; a mini review. Curr Opin Pharmacol 2020; 54:82-90. [PMID: 32980777 PMCID: PMC7770009 DOI: 10.1016/j.coph.2020.08.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes (T2D) substantially elevates the risk for heart failure, a major cause of death. In advanced T2D, energy metabolism in the heart is disrupted; glucose metabolism is decreased, fatty acid (FA) metabolism is enhanced to maintain ATP production, and cardiac function is impaired. This condition is termed diabetic cardiomyopathy (DCM). The exact cause of DCM is still unknown although altered metabolism is an important component. While type 2 diabetes is characterized by insulin resistance, the traditional antidiabetic agents that improve insulin stimulation or sensitivity only partially improve DCM-induced cardiac dysfunction. Recently, sodium-glucose transporter-2 (SGLT2) inhibitors have been identified as potential pharmacological agents to treat DCM. This review highlights the molecular mechanisms underlying cardiac energy metabolism in DCM, and the potential effects of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Shinsuke Nirengi
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Carmem Peres Valgas da Silva
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
22
|
Li Y, Duan JZ, He Q, Wang CQ. miR‑155 modulates high glucose‑induced cardiac fibrosis via the Nrf2/HO‑1 signaling pathway. Mol Med Rep 2020; 22:4003-4016. [PMID: 32901848 DOI: 10.3892/mmr.2020.11495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 01/29/2020] [Indexed: 11/05/2022] Open
Abstract
Cardiac fibrosis is a major pathological manifestation of diabetic cardiomyopathy, which is a leading cause of mortality in patients with diabetes. MicroRNA (miR)‑155 is upregulated in cardiomyocytes in cardiac fibrosis, and the aim of the present study was to investigate if the inhibition of miR‑155 was able to ameliorate cardiac fibrosis by targeting the nuclear factor erythroid‑2‑related factor 2 (Nrf2)/heme oxygenase‑1 (HO‑1) signaling pathway. H9C2 rat cardiomyocytes were cultured with high glucose (HG; 30 mM) to establish an in vitro cardiac fibrosis model that mimicked diabetic conditions; a miR‑155 inhibitor and a miR‑155 mimic were transfected into H9C2 cells. Following HG treatment, H9C2 cells exhibited increased expression levels of miR‑155 and the fibrosis markers collagen I and α‑smooth muscle actin (α‑SMA). In addition, the expression levels of endonuclear Nrf2 and HO‑1 were decreased, but the expression level of cytoplasmic Nrf2 was increased. Moreover, oxidative stress, mitochondrial damage and cell apoptosis were significantly increased, as indicated by elevated reactive oxygen species, malonaldehyde and monomeric JC‑1 expression levels. In addition, superoxide dismutase expression was attenuated and there was an increased expression level of released cytochrome‑c following HG treatment. Furthermore, it was demonstrated that expression levels of Bcl‑2 and uncleaved Poly (ADP‑ribose) polymerase were downregulated, whereas Bax, cleaved caspase‑3 and caspase‑9 were upregulated after HG treatment. However, the miR‑155 inhibitor significantly restored Nrf2 and HO‑1 expression levels, and reduced oxidative stress levels, the extent of mitochondrial damage and the number of cells undergoing apoptosis. Additionally, the miR‑155 inhibitor significantly reversed the expression levels of collagen I and α‑SMA, thus ameliorating fibrosis. Furthermore, the knockdown of Nrf2 reversed the above effects induced by the miR‑155 inhibitor. In conclusion, the miR‑155 inhibitor may ameliorate diabetic cardiac fibrosis by reducing the accumulation of oxidative stress‑related molecules, and preventing mitochondrial damage and cardiomyocyte apoptosis by enhancing the Nrf2/HO‑1 signaling pathway. This mechanism may facilitate the development of novel targets to prevent cardiac fibrosis in patients with diabetes.
Collapse
Affiliation(s)
- Yu Li
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jing-Zhu Duan
- Department of Respiratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qian He
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Chong-Quan Wang
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
23
|
Brahma MK, Ha C, Pepin ME, Mia S, Sun Z, Chatham JC, Habegger KM, Abel ED, Paterson AJ, Young ME, Wende AR. Increased Glucose Availability Attenuates Myocardial Ketone Body Utilization. J Am Heart Assoc 2020; 9:e013039. [PMID: 32750298 PMCID: PMC7792234 DOI: 10.1161/jaha.119.013039] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/05/2020] [Indexed: 02/06/2023]
Abstract
Background Perturbations in myocardial substrate utilization have been proposed to contribute to the pathogenesis of cardiac dysfunction in diabetic subjects. The failing heart in nondiabetics tends to decrease reliance on fatty acid and glucose oxidation, and increases reliance on ketone body oxidation. In contrast, little is known regarding the mechanisms mediating this shift among all 3 substrates in diabetes mellitus. Therefore, we tested the hypothesis that changes in myocardial glucose utilization directly influence ketone body catabolism. Methods and Results We examined ventricular-cardiac tissue from the following murine models: (1) streptozotocin-induced type 1 diabetes mellitus; (2) high-fat-diet-induced glucose intolerance; and transgenic inducible cardiac-restricted expression of (3) glucose transporter 4 (transgenic inducible cardiac restricted expression of glucose transporter 4); or (4) dominant negative O-GlcNAcase. Elevated blood glucose (type 1 diabetes mellitus and high-fat diet mice) was associated with reduced cardiac expression of β-hydroxybutyrate-dehydrogenase and succinyl-CoA:3-oxoacid CoA transferase. Increased myocardial β-hydroxybutyrate levels were also observed in type 1 diabetes mellitus mice, suggesting a mismatch between ketone body availability and utilization. Increased cellular glucose delivery in transgenic inducible cardiac restricted expression of glucose transporter 4 mice attenuated cardiac expression of both Bdh1 and Oxct1 and reduced rates of myocardial BDH1 activity and β-hydroxybutyrate oxidation. Moreover, elevated cardiac protein O-GlcNAcylation (a glucose-derived posttranslational modification) by dominant negative O-GlcNAcase suppressed β-hydroxybutyrate dehydrogenase expression. Consistent with the mouse models, transcriptomic analysis confirmed suppression of BDH1 and OXCT1 in patients with type 2 diabetes mellitus and heart failure compared with nondiabetic patients. Conclusions Our results provide evidence that increased glucose leads to suppression of cardiac ketolytic capacity through multiple mechanisms and identifies a potential crosstalk between glucose and ketone body metabolism in the diabetic myocardium.
Collapse
Affiliation(s)
- Manoja K. Brahma
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - Chae‐Myeong Ha
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - Mark E. Pepin
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
- Biomedical EngineeringUniversity of Alabama at BirminghamALUSA
| | - Sobuj Mia
- Medicine, Division of Cardiovascular DiseasesUniversity of Alabama at BirminghamALUSA
| | - Zhihuan Sun
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - John C. Chatham
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
| | - Kirk M. Habegger
- Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamALUSA
| | - Evan Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIAUSA
| | - Andrew J. Paterson
- Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamALUSA
| | - Martin E. Young
- Medicine, Division of Cardiovascular DiseasesUniversity of Alabama at BirminghamALUSA
| | - Adam R. Wende
- Departments of PathologyDivision of Molecular and Cellular PathologyUniversity of Alabama at BirminghamALUSA
- Biomedical EngineeringUniversity of Alabama at BirminghamALUSA
| |
Collapse
|
24
|
Waldman M, Arad M, Abraham NG, Hochhauser E. The Peroxisome Proliferator-Activated Receptor-Gamma Coactivator-1α-Heme Oxygenase 1 Axis, a Powerful Antioxidative Pathway with Potential to Attenuate Diabetic Cardiomyopathy. Antioxid Redox Signal 2020; 32:1273-1290. [PMID: 32027164 PMCID: PMC7232636 DOI: 10.1089/ars.2019.7989] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
Significance: From studies of diabetic animal models, the downregulation of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α)-heme oxygenase 1 (HO-1) axis appears to be a crucial event in the development of obesity and diabetic cardiomyopathy (DCM). In this review, we discuss the role of metabolic and biochemical stressors in the rodent and human pathophysiology of DCM. A crucial contributor for many cardiac pathologies is excessive production of reactive oxygen species (ROS) pathologies, which lead to extensive cellular damage by impairing mitochondrial function and directly oxidizing DNA, proteins, and lipid membranes. We discuss the role of ROS production and inflammatory pathways with multiple contributing and confounding factors leading to DCM. Recent Advances: The relevant biochemical pathways that are critical to a therapeutic approach to treat DCM, specifically caloric restriction and its relation to the PGC-1α-HO-1 axis in the attenuation of DCM, are elucidated. Critical Issues: The increased prevalence of diabetes mellitus type 2, a major contributor to unique cardiomyopathy characterized by cardiomyocyte hypertrophy with no effective clinical treatment. This review highlights the role of mitochondrial dysfunction in the development of DCM and potential oxidative targets to attenuate oxidative stress and attenuate DCM. Future Directions: Targeting the PGC-1α-HO-1 axis is a promising approach to ameliorate DCM through improvement in mitochondrial function and antioxidant defenses. A pharmacological inducer to activate PGC-1α and HO-1 described in this review may be a promising therapeutic approach in the clinical setting.
Collapse
Affiliation(s)
- Maayan Waldman
- Cardiac Research Laboratory, Felsenstein Medical Research Institute at Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
- Cardiac Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Ramat Gan, Israel
| | - Michael Arad
- Cardiac Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Ramat Gan, Israel
| | - Nader G. Abraham
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute at Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
25
|
Glatz JFC, Nabben M, Young ME, Schulze PC, Taegtmeyer H, Luiken JJFP. Re-balancing cellular energy substrate metabolism to mend the failing heart. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165579. [PMID: 31678200 PMCID: PMC7586321 DOI: 10.1016/j.bbadis.2019.165579] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
Fatty acids and glucose are the main substrates for myocardial energy provision. Under physiologic conditions, there is a distinct and finely tuned balance between the utilization of these substrates. Using the non-ischemic heart as an example, we discuss that upon stress this substrate balance is upset resulting in an over-reliance on either fatty acids or glucose, and that chronic fuel shifts towards a single type of substrate appear to be linked with cardiac dysfunction. These observations suggest that interventions aimed at re-balancing a tilted substrate preference towards an appropriate mix of substrates may result in restoration of cardiac contractile performance. Examples of manipulating cellular substrate uptake as a means to re-balance fuel supply, being associated with mended cardiac function underscore this concept. We also address the molecular mechanisms underlying the apparent need for a fatty acid-glucose fuel balance. We propose that re-balancing cellular fuel supply, in particular with respect to fatty acids and glucose, may be an effective strategy to treat the failing heart.
Collapse
Affiliation(s)
- Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands.
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
26
|
Prakoso D, De Blasio MJ, Tate M, Kiriazis H, Donner DG, Qian H, Nash D, Deo M, Weeks KL, Parry LJ, Gregorevic P, McMullen JR, Ritchie RH. Gene therapy targeting cardiac phosphoinositide 3-kinase (p110α) attenuates cardiac remodeling in type 2 diabetes. Am J Physiol Heart Circ Physiol 2020; 318:H840-H852. [PMID: 32142359 DOI: 10.1152/ajpheart.00632.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Diabetic cardiomyopathy is a distinct form of heart disease that represents a major cause of death and disability in diabetic patients, particularly, the more prevalent type 2 diabetes patient population. In the current study, we investigated whether administration of recombinant adeno-associated viral vectors carrying a constitutively active phosphoinositide 3-kinase (PI3K)(p110α) construct (rAAV6-caPI3K) at a clinically relevant time point attenuates diabetic cardiomyopathy in a preclinical type 2 diabetes (T2D) model. T2D was induced by a combination of a high-fat diet (42% energy intake from lipid) and low-dose streptozotocin (three consecutive intraperitoneal injections of 55 mg/kg body wt), and confirmed by increased body weight, mild hyperglycemia, and impaired glucose tolerance (all P < 0.05 vs. nondiabetic mice). After 18 wk of untreated diabetes, impaired left ventricular (LV) systolic dysfunction was evident, as confirmed by reduced fractional shortening and velocity of circumferential fiber shortening (Vcfc, all P < 0.01 vs. baseline measurement). A single tail vein injection of rAAV6-caPI3K gene therapy (2×1011vector genomes) was then administered. Mice were followed for an additional 8 wk before end point. A single injection of cardiac targeted rAAV6-caPI3K attenuated diabetes-induced cardiac remodeling by limiting cardiac fibrosis (reduced interstitial and perivascular collagen deposition, P < 0.01 vs. T2D mice) and cardiomyocyte hypertrophy (reduced cardiomyocyte size and Nppa gene expression, P < 0.001 and P < 0.05 vs. T2D mice, respectively). The diabetes-induced LV systolic dysfunction was reversed with rAAV6-caPI3K, as demonstrated by improved fractional shortening and velocity of circumferential fiber shortening (all P < 0.05 vs pre-AAV measurement). This cardioprotection occurred in combination with reduced LV reactive oxygen species (P < 0.05 vs. T2D mice) and an associated decrease in markers of endoplasmic reticulum stress (reduced Grp94 and Chop, all P < 0.05 vs. T2D mice). Together, our findings demonstrate that a cardiac-selective increase in PI3K(p110α), via rAAV6-caPI3K, attenuates T2D-induced diabetic cardiomyopathy, providing proof of concept for potential translation to the clinic.NEW & NOTEWORTHY Diabetes remains a major cause of death and disability worldwide (and its resultant heart failure burden), despite current care. The lack of existing management of heart failure in the context of the poorer prognosis of concomitant diabetes represents an unmet clinical need. In the present study, we now demonstrate that delayed intervention with PI3K gene therapy (rAAV6-caPI3K), administered as a single dose in mice with preexisting type 2 diabetes, attenuates several characteristics of diabetic cardiomyopathy, including diabetes-induced impairments in cardiac remodeling, oxidative stress, and function. Our discovery here contributes to the previous body of work, suggesting the cardioprotective effects of PI3K(p110α) could be a novel therapeutic approach to reduce the progression to heart failure and death in diabetes-affected patients.
Collapse
Affiliation(s)
- Darnel Prakoso
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,School of Biosciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Miles J De Blasio
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,School of Biosciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Mitchel Tate
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Hongwei Qian
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - David Nash
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Minh Deo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| | - Laura J Parry
- School of Biosciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia.,Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Neurology, The University of Washington, Seattle, Washington
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Physiology, Monash University, Clayton, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| | - Rebecca Helen Ritchie
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Affiliation(s)
- Jin Hwa Kim
- Department of Endocrinology and Metabolism, Chosun University Hospital, Chosun University College of Medicine, Gwangju, Korea.
| |
Collapse
|
28
|
Abstract
It has been estimated that by 2030, the number of patients with diabetes aged > 64 years will be > 82 million in underdeveloped countries, and > 48 million in developed countries. Chronic hyperglycemia delays wound healing by reducing the expression of growth factors in the wound fluid and re-epithelialization. Impaired wound healing in patients with diabetes has also been associated with inhibition of the production of stromal cell-derived factor-1alpha by several tissues including bone marrow, brain, heart, spleen, and gingivae. Chronic hyperglycemia interferes with the osseointegration of implants by deferring the expression of fibronectin and integrins. Results from experimental studies have shown a significantly higher bone-to-implant contact around implants placed in healthy animals compared with animals with streptozotocin-induced diabetes. Moreover, persistent hyperglycemia plays a role in abnormal differentiation of osteoclasts, thereby making bone tissue more susceptible to resorption. Furthermore, persistent hyperglycemia has also been associated with increased peri-implant soft tissue inflammation (increased peri-implant bleeding on probing and probing depth) and crestal bone loss. Clinical studies have shown that under optimal glycemic control dental implants can show success and survival rates of up to 100% in patients diagnosed with diabetes. Although patients with diabetes can undergo dental implant therapy and can exhibit implant survival similar to those in systemically healthy individuals, the contribution of glycemic control and regular oral hygiene maintenance cannot be disregarded.
Collapse
Affiliation(s)
- Fawad Javed
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, New York.,Department of Oral Surgery and Implant Dentistry, Dental School, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Georgios E Romanos
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, New York.,Department of Oral Surgery and Implant Dentistry, Dental School, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
29
|
Wang Z, Li J, Wang Y, Liu Q. Palbociclib improves cardiac dysfunction in diabetic cardiomyopathy by regulating Rb phosphorylation. Am J Transl Res 2019; 11:3481-3489. [PMID: 31312360 PMCID: PMC6614619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/16/2019] [Indexed: 06/10/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a condition associated with significant structural changes including cardiac tissue necrosis, localized fibrosis, and hypertrophy of cardiomyocytes. This study sought to assess whether and how CDK4/6 inhibitor, Palbociclib, can attenuate DCM using a streptozotocin (STZ)-induced DCM model system. In this study, we found CDK4 and CDK6 expression are significantly increased the cardiac tissue of these mice. Palbociclib treatment after initial STZ administration attenuated oxidative stress and inflammation, thereby reducing cardiomyocyte death and preserving cardiac function in these animals. In addition, Rb phosphorylation induction was found in STZ-treated mice, which was inhibited by Palbociclib treatment. In summary, Palbociclib protects mice from damage associated with DCM pathway activation, making Palbociclib is a relevant therapeutic target in the context of DCM.
Collapse
Affiliation(s)
- Zhenggui Wang
- Department of Cardiovascular Center, The First Hospital of Jilin University Changchun 130021, Jilin, China
| | - Jing Li
- Department of Cardiovascular Center, The First Hospital of Jilin University Changchun 130021, Jilin, China
| | - Yonggang Wang
- Department of Cardiovascular Center, The First Hospital of Jilin University Changchun 130021, Jilin, China
| | - Quan Liu
- Department of Cardiovascular Center, The First Hospital of Jilin University Changchun 130021, Jilin, China
| |
Collapse
|
30
|
Pinckard K, Baskin KK, Stanford KI. Effects of Exercise to Improve Cardiovascular Health. Front Cardiovasc Med 2019; 6:69. [PMID: 31214598 PMCID: PMC6557987 DOI: 10.3389/fcvm.2019.00069] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022] Open
Abstract
Obesity is a complex disease that affects whole body metabolism and is associated with an increased risk of cardiovascular disease (CVD) and Type 2 diabetes (T2D). Physical exercise results in numerous health benefits and is an important tool to combat obesity and its co-morbidities, including cardiovascular disease. Exercise prevents both the onset and development of cardiovascular disease and is an important therapeutic tool to improve outcomes for patients with cardiovascular disease. Some benefits of exercise include enhanced mitochondrial function, restoration and improvement of vasculature, and the release of myokines from skeletal muscle that preserve or augment cardiovascular function. In this review we will discuss the mechanisms through which exercise promotes cardiovascular health.
Collapse
Affiliation(s)
| | | | - Kristin I. Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
31
|
A new hyperpolarized 13C ketone body probe reveals an increase in acetoacetate utilization in the diabetic rat heart. Sci Rep 2019; 9:5532. [PMID: 30940842 PMCID: PMC6445118 DOI: 10.1038/s41598-019-39378-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/17/2019] [Indexed: 12/27/2022] Open
Abstract
Emerging studies have recently shown the potential importance of ketone bodies in cardio-metabolic health. However, techniques to determine myocardial ketone body utilization in vivo are lacking. In this work, we developed a novel method to assess myocardial ketone body utilization in vivo using hyperpolarized [3-13C]acetoacetate and investigated the alterations in myocardial ketone body metabolism in diabetic rats. Within a minute upon injection of [3-13C]acetoacetate, the production of [5-13C]glutamate and [1-13C] acetylcarnitine can be observed real time in vivo. In diabetic rats, the production of [5-13C]glutamate was elevated compared to controls, while [1-13C]acetylcarnitine was not different. This suggests an increase in ketone body utilization in the diabetic heart, with the produced acetyl-CoA channelled into the tricarboxylic acid cycle. This observation was corroborated by an increase activity of succinyl-CoA:3-ketoacid-CoA transferase (SCOT) activity, the rate-limiting enzyme of ketone body utilization, in the diabetic heart. The increased ketone body oxidation in the diabetic hearts correlated with cardiac hypertrophy and dysfunction, suggesting a potential coupling between ketone body metabolism and cardiac function. Hyperpolarized [3-13C]acetoacetate is a new probe with potential for non-invasive and real time monitoring of myocardial ketone body oxidation in vivo, which offers a powerful tool to follow disease progression or therapeutic interventions.
Collapse
|
32
|
Metabolomics biomarkers and the risk of overall mortality and ESRD in CKD: Results from the Progredir Cohort. PLoS One 2019; 14:e0213764. [PMID: 30883578 PMCID: PMC6422295 DOI: 10.1371/journal.pone.0213764] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/28/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Studies on metabolomics and CKD have primarily addressed CKD incidence defined as a decline on eGFR or appearance of albuminuria in the general population, with very few evaluating hard outcomes. In the present study, we investigated the association between metabolites and mortality and ESRD in a CKD cohort. SETTING AND METHODS Data on 454 participants of the Progredir Cohort Study, Sao Paulo, Brazil were used. Metabolomics was performed by GC-MS (Agilent MassHunter) and metabolites were identified using Agilent Fiehn GC/MS and NIST libraries. After excluding metabolites present in <50% of participants, 293 metabolites were analyzed. An FDR q value <0.05 criteria was applied in Cox models on the composite outcome (mortality or incident renal replacement therapy) adjusted for batch effect, resulting in 34 metabolites associated with the outcome. Multivariable-adjusted Cox models were then built for the composite outcome, death, and ESRD incident events. Competing risk analysis was also performed for ESRD. RESULTS Mean age was 68±12y, mean eGFR-CKDEPI was 38.4±14.6 ml/min/1.73m2 and 57% were diabetic. After adjustments (GC-MS batch, sex, age, DM and eGFR), 18 metabolites remained significantly associated with the composite outcome. Nine metabolites were independently associated with death: D-malic acid (HR 1.84, 95%CI 1.32-2.56, p = 0.0003), acetohydroxamic acid (HR 1.90, 95%CI 1.30-2.78, p = 0.0008), butanoic acid (HR 1.59, 95%CI 1.17-2.15, p = 0.003), and docosahexaenoic acid (HR 0.58, 95%CI 0.39-0.88, p = 0.009), among the top associations. Lactose (SHR 1.49, 95%CI 1.04-2.12, p = 0.03), 2-O-glycerol-α-D-galactopyranoside (SHR 1.76, 95%CI 1.06-2.92, p = 0.03), and tyrosine (SHR 0.52, 95%CI 0.31-0.88, p = 0.02) were associated to ESRD risk, while D-threitol, mannitol and myo-inositol presented strong borderline associations. CONCLUSION Our results identify specific metabolites related to hard outcomes in a CKD population. These findings point to the need of further exploration of these metabolites as biomarkers in CKD and the understanding of the underlying biological mechanisms related to the observed associations.
Collapse
|
33
|
Park S, Kang HJ, Jeon JH, Kim MJ, Lee IK. Recent advances in the pathogenesis of microvascular complications in diabetes. Arch Pharm Res 2019; 42:252-262. [PMID: 30771210 DOI: 10.1007/s12272-019-01130-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/31/2019] [Indexed: 12/29/2022]
Abstract
Millions of people worldwide have diabetes, which is diagnosed by fasting blood glucose levels exceeding 126 mg/dL. Regardless of the type of diabetes, prolonged hyperglycemia is damaging to several organs including eyes, kidneys, nerve, and/or heart. The damages are associated with a high risk of morbidity and mortality. Diabetes has been implicated in ischemia in the microvasculature of the target tissues, which occurs due to the insufficient perfusion of tissues. The resulting occlusion and pain affect the quality of life. Multiple therapeutic approaches have been proposed for a long time to overcome these vascular complications. Apart from systemically controlling high glucose levels, other therapeutic strategies are not well understood. In this review, we summarize the recent literature for biochemical/cellular targets that are being utilized for the treatment of diabetic microvascular diseases. These targets, which are closely associated with mitochondrial dysfunction, include the polyol and diacylglycerol-protein kinase C pathways, oxidative stress, non-enzymatic glycation and the formation of advanced glycation end products, and immune dysregulation/inflammation.
Collapse
Affiliation(s)
- Sungmi Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
| | - Hyeon-Ji Kang
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea
| | - Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea
| | - In-Kyu Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea.
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
34
|
Abstract
In the early 1980s, while using purified glycosyltransferases to probe glycan structures on surfaces of living cells in the murine immune system, we discovered a novel form of serine/threonine protein glycosylation (O-linked β-GlcNAc; O-GlcNAc) that occurs on thousands of proteins within the nucleus, cytoplasm, and mitochondria. Prior to this discovery, it was dogma that protein glycosylation was restricted to the luminal compartments of the secretory pathway and on extracellular domains of membrane and secretory proteins. Work in the last 3 decades from several laboratories has shown that O-GlcNAc cycling serves as a nutrient sensor to regulate signaling, transcription, mitochondrial activity, and cytoskeletal functions. O-GlcNAc also has extensive cross-talk with phosphorylation, not only at the same or proximal sites on polypeptides, but also by regulating each other's enzymes that catalyze cycling of the modifications. O-GlcNAc is generally not elongated or modified. It cycles on and off polypeptides in a time scale similar to phosphorylation, and both the enzyme that adds O-GlcNAc, the O-GlcNAc transferase (OGT), and the enzyme that removes O-GlcNAc, O-GlcNAcase (OGA), are highly conserved from C. elegans to humans. Both O-GlcNAc cycling enzymes are essential in mammals and plants. Due to O-GlcNAc's fundamental roles as a nutrient and stress sensor, it plays an important role in the etiologies of chronic diseases of aging, including diabetes, cancer, and neurodegenerative disease. This review will present an overview of our current understanding of O-GlcNAc's regulation, functions, and roles in chronic diseases of aging.
Collapse
Affiliation(s)
- Gerald W Hart
- From the Complex Carbohydrate Research Center and Biochemistry and Molecular Biology Department, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
35
|
Takata T, Sakasai-Sakai A, Ueda T, Takeuchi M. Intracellular toxic advanced glycation end-products in cardiomyocytes may cause cardiovascular disease. Sci Rep 2019; 9:2121. [PMID: 30765817 PMCID: PMC6375929 DOI: 10.1038/s41598-019-39202-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular disease (CVD) is a lifestyle-related disease (LSRD) and one of the largest public health issues. Risk factors for CVD correlate with an excessive intake of glucose and/or fructose, which has been shown to induce the production of advanced glycation end-products (AGEs). We previously identified AGEs derived from glyceraldehyde and named them toxic AGEs (TAGE) due to their cytotoxicities and relationship with LSRD. We also reported that extracellular TAGE in the vascular system may promote CVD and that serum TAGE levels are associated with risk factors for CVD. The mechanisms responsible for the onset and/or progression of CVD by extracellular TAGE or the above risk factors involve vascular disorders. In the present study, we revealed that rat primary cultured cardiomyocytes generated intracellular TAGE, which decreased beating rates and induced cell death. LC3-II/LC3-I, a factor of autophagy, also decreased. Although intracellular TAGE may be targets of degradation as cytotoxic proteins via autophagy, they may inhibit autophagy. Furthermore, the mechanisms by which intracellular TAGE decrease beating rates and induce cell death may involve the suppression of autophagy. The present results suggest that intracellular TAGE are generated in cardiomyocytes and directly damage them, resulting in CVD.
Collapse
Affiliation(s)
- Takanobu Takata
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293, Japan.
| | - Akiko Sakasai-Sakai
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293, Japan
| | - Tadashi Ueda
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293, Japan
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293, Japan
| |
Collapse
|
36
|
Zhang B, Xu D. Protective effects of astaxanthin on diabetic cardiomyopathy in rats. CYTA - JOURNAL OF FOOD 2018. [DOI: 10.1080/19476337.2018.1503617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Bingshan Zhang
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Di Xu
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Lugat A, Joubert M, Cariou B, Prieur X. [At the heart of diabetic cardiomyopathy: Bscl2 knockout mice to investigate glucotoxicity]. Med Sci (Paris) 2018; 34:563-570. [PMID: 30067203 DOI: 10.1051/medsci/20183406016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a well-recognized independent risk factor for heart failure (HF). T2DM is associated with altered cardiac energy metabolism, leading to ectopic lipid accumulation and glucose overload. However, the relative contribution of these two parameters remains unclear. In order to get new insight into the mechanism involved in diabetic cardiomyopathy, the cardiac phenotype of a unique T2DM mice model has been performed: the seipin knockout mice (SKO). Cardiac phenotyping revealed a diastolic dysfunction associated with hyperglycemia in these mice with a chronic activation of the hexosamine biosynthetic pathway (HBP), suggesting a glucose overload. An inhibitor of the renal sodium/glucose cotransporter 2 (SGLT2), dapagliflozin, successfully prevented the development of cardiomyopathy in SKO mice. This is particularly relevant, given that SGLT2i treatment reduces cardiovascular event in T2DM patients. Therefore, glucose lowering appears an important therapeutic target to prevent cardiac dysfunction associated with T2DM.
Collapse
Affiliation(s)
- Alexandre Lugat
- Institut du thorax, Inserm, CNRS, univ Nantes, CHU Nantes, 8, quai Moncousu, Nantes, F-44000, France
| | - Michael Joubert
- Service Endocrinologie, CHU de Caen, Caen, F-14003, France - EA4650 - Unicaen - GIP Cyceron - Caen, Caen, F-14074, France
| | - Bertrand Cariou
- Institut du thorax, Inserm, CNRS, univ Nantes, CHU Nantes, 8, quai Moncousu, Nantes, F-44000, France
| | - Xavier Prieur
- Institut du thorax, Inserm, CNRS, univ Nantes, Nantes, F-44000, France
| |
Collapse
|
38
|
Edhager AV, Povlsen JA, Løfgren B, Bøtker HE, Palmfeldt J. Proteomics of the Rat Myocardium during Development of Type 2 Diabetes Mellitus Reveals Progressive Alterations in Major Metabolic Pathways. J Proteome Res 2018; 17:2521-2532. [PMID: 29847139 DOI: 10.1021/acs.jproteome.8b00276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Congestive heart failure and poor clinical outcome after myocardial infarction are known complications in patients with type-2 diabetes mellitus (T2DM). Protein alterations may be involved in the mechanisms underlying these disarrays in the diabetic heart. Here we map proteins involved in intracellular metabolic pathways in the Zucker diabetic fatty rat heart as T2DM develops using MS based proteomics. The prediabetic state only induced minor pathway changes, whereas onset and late T2DM caused pronounced perturbations. Two actin-associated proteins, ARPC2 and TPM3, were up-regulated at the prediabetic state indicating increased actin dynamics. All differentially regulated proteins involved in fatty acid metabolism, both peroxisomal and mitochondrial, were up-regulated at late T2DM, whereas enzymes of branched chain amino acid degradation were all down-regulated. At both onset and late T2DM, two members of the serine protease inhibitor superfamily, SERPINA3K and SERPINA3L, were down-regulated. Furthermore, we found alterations in proteins involved in clearance of advanced glycation end-products and lipotoxicity, DCXR and CBR1, at both onset and late T2DM. These proteins deserve elucidation with regard to their role in T2DM pathogenesis and their respective role in the deterioration of the diabetic heart. Data are available via ProteomeXchange with identifiers PXD009538, PXD009554, and PXD009555.
Collapse
Affiliation(s)
- Anders Valdemar Edhager
- Research Unit for Molecular Medicine, Department of Clinical Medicine , Aarhus University and Aarhus University Hospital , 8200 , Aarhus N , Denmark
| | | | - Bo Løfgren
- Department of Cardiology , Aarhus University Hospital , 8200 , Aarhus N , Denmark.,Institute for Experimental Clinical Research , Aarhus University , 8000 , Aarhus C , Denmark
| | - Hans Erik Bøtker
- Department of Cardiology , Aarhus University Hospital , 8200 , Aarhus N , Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine , Aarhus University and Aarhus University Hospital , 8200 , Aarhus N , Denmark
| |
Collapse
|
39
|
Moser O, Eckstein ML, McCarthy O, Deere R, Bain SC, Haahr HL, Zijlstra E, Heise T, Bracken RM. Heart rate dynamics during cardio-pulmonary exercise testing are associated with glycemic control in individuals with type 1 diabetes. PLoS One 2018; 13:e0194750. [PMID: 29608593 PMCID: PMC5880363 DOI: 10.1371/journal.pone.0194750] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/07/2018] [Indexed: 12/04/2022] Open
Abstract
INTRODUCTION This study investigated the degree and direction (kHR) of the heart rate to performance curve (HRPC) during cardio-pulmonary exercise (CPX) testing and explored the relationship with diabetes markers, anthropometry and exercise physiological markers in type 1 diabetes (T1DM). MATERIAL AND METHODS Sixty-four people with T1DM (13 females; age: 34 ± 8 years; HbA1c: 7.8 ± 1% (62 ± 13 mmol.mol-1) performed a CPX test until maximum exhaustion. kHR was calculated by a second-degree polynomial representation between post-warm up and maximum power output. Adjusted stepwise linear regression analysis was performed to investigate kHR and its associations. Receiver operating characteristic (ROC) curve was performed based on kHR for groups kHR < 0.20 vs. > 0.20 in relation to HbA1c. RESULTS We found significant relationships between kHR and HbA1c (β = -0.70, P < 0.0001), age (β = -0.23, P = 0.03) and duration of diabetes (β = 0.20, P = 0.04). Stepwise linear regression resulted in an overall adjusted R2 of 0.57 (R = 0.79, P < 0.0001). Our data revealed also significant associations between kHR and percentage of heart rate at heart rate turn point from maximum heart rate (β = 0.43, P < 0.0001) and maximum power output relativized to bodyweight (β = 0.44, P = 0.001) (overall adjusted R2 of 0.44 (R = 0.53, P < 0.0001)). ROC curve analysis based on kHR resulted in a HbA1c threshold of 7.9% (62 mmol.mol-1). CONCLUSION Our data demonstrate atypical HRPC during CPX testing that were mainly related to glycemic control in people with T1DM.
Collapse
Affiliation(s)
- Othmar Moser
- Diabetes Research Group, Medical School, Swansea University, Swansea, United Kingdom
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Max L. Eckstein
- Diabetes Research Group, Medical School, Swansea University, Swansea, United Kingdom
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Olivia McCarthy
- Diabetes Research Group, Medical School, Swansea University, Swansea, United Kingdom
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Rachel Deere
- Diabetes Research Group, Medical School, Swansea University, Swansea, United Kingdom
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| | - Stephen C. Bain
- Diabetes Research Group, Medical School, Swansea University, Swansea, United Kingdom
| | | | | | | | - Richard M. Bracken
- Diabetes Research Group, Medical School, Swansea University, Swansea, United Kingdom
- Applied Sport, Technology, Exercise and Medicine Research Centre (A-STEM), College of Engineering, Swansea University, Swansea, United Kingdom
| |
Collapse
|
40
|
Yan L. Redox imbalance stress in diabetes mellitus: Role of the polyol pathway. Animal Model Exp Med 2018; 1:7-13. [PMID: 29863179 PMCID: PMC5975374 DOI: 10.1002/ame2.12001] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
In diabetes mellitus, the polyol pathway is highly active and consumes approximately 30% glucose in the body. This pathway contains 2 reactions catalyzed by aldose reductase (AR) and sorbitol dehydrogenase, respectively. AR reduces glucose to sorbitol at the expense of NADPH, while sorbitol dehydrogenase converts sorbitol to fructose at the expense of NAD+, leading to NADH production. Consumption of NADPH, accumulation of sorbitol, and generation of fructose and NADH have all been implicated in the pathogenesis of diabetes and its complications. In this review, the roles of this pathway in NADH/NAD+ redox imbalance stress and oxidative stress in diabetes are highlighted. A potential intervention using nicotinamide riboside to restore redox balance as an approach to fighting diabetes is also discussed.
Collapse
Affiliation(s)
- Liang‐jun Yan
- Department of Pharmaceutical SciencesUNT System College of PharmacyUniversity of North Texas Health Science CenterFort WorthTXUSA
| |
Collapse
|
41
|
Ducheix S, Magré J, Cariou B, Prieur X. Chronic O-GlcNAcylation and Diabetic Cardiomyopathy: The Bitterness of Glucose. Front Endocrinol (Lausanne) 2018; 9:642. [PMID: 30420836 PMCID: PMC6215811 DOI: 10.3389/fendo.2018.00642] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes (T2D) is a major risk factor for heart failure. Diabetic cardiomyopathy (DC) is characterized by diastolic dysfunction and left ventricular hypertrophy. Epidemiological data suggest that hyperglycaemia contributes to the development of DC. Several cellular pathways have been implicated in the deleterious effects of high glucose concentrations in the heart: oxidative stress, accumulation of advanced glycation end products (AGE), and chronic hexosamine biosynthetic pathway (HBP) activation. In the present review, we focus on the effect of chronic activation of the HBP on diabetic heart function. The HBP supplies N-acetylglucosamine moiety (O-GlcNAc) that is O-linked by O-GlcNAc transferase (OGT) to proteins on serine or threonine residues. This post-translational protein modification modulates the activity of the targeted proteins. In the heart, acute activation of the HBP in response to ischaemia-reperfusion injury appears to be protective. Conversely, chronic activation of the HBP in the diabetic heart affects Ca2+ handling, contractile properties, and mitochondrial function and promotes stress signaling, such as left ventricular hypertrophy and endoplasmic reticulum stress. Many studies have shown that O-GlcNAc impairs the function of key protein targets involved in these pathways, such as phospholamban, calmodulin kinase II, troponin I, and FOXO1. The data show that excessive O-GlcNAcylation is a major trigger of the glucotoxic events that affect heart function under chronic hyperglycaemia. Supporting this finding, pharmacological or genetic inhibition of the HBP in the diabetic heart improves heart function. In addition, the SGLT2 inhibitor dapagliflozin, a glucose lowering agent, has recently been shown to lower cardiac HBP in a lipodystophic T2D mice model and to concomitantly improve the diastolic dysfunction of these mice. Therefore, targeting cardiac-excessive O-GlcNAcylation or specific target proteins represents a potential therapeutic option to treat glucotoxicity in the diabetic heart.
Collapse
Affiliation(s)
- Simon Ducheix
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Jocelyne Magré
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Bertrand Cariou
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Xavier Prieur
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
- *Correspondence: Xavier Prieur
| |
Collapse
|
42
|
Al Hariri M, Elmedawar M, Zhu R, Jaffa MA, Zhao J, Mirzaei P, Ahmed A, Kobeissy F, Ziyadeh FN, Mechref Y, Jaffa AA. Proteome profiling in the aorta and kidney of type 1 diabetic rats. PLoS One 2017; 12:e0187752. [PMID: 29121074 PMCID: PMC5679573 DOI: 10.1371/journal.pone.0187752] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/25/2017] [Indexed: 01/04/2023] Open
Abstract
Diabetes is associated with a number of metabolic and cardiovascular risk factors that contribute to a high rate of microvascular and macrovascular complications. The risk factors and mechanisms that contribute to the development of micro- and macrovascular disease in diabetes are not fully explained. In this study, we employed mass spectrometric analysis using tandem LC-MS/MS to generate a proteomic profile of protein abundance and post-translational modifications (PTM) in the aorta and kidney of diabetic rats. In addition, systems biology analyses were employed to identify key protein markers that can provide insights into molecular pathways and processes that are differentially regulated in the aorta and kidney of type 1 diabetic rats. Our results indicated that 188 (111 downregulated and 77 upregulated) proteins were significantly identified in the aorta of diabetic rats compared to normal controls. A total of 223 (109 downregulated and 114 upregulated) proteins were significantly identified in the kidney of diabetic rats compared to normal controls. When the protein profiles from the kidney and aorta of diabetic and control rats were analyzed by principal component analysis, a distinct separation of the groups was observed. In addition, diabetes resulted in a significant increase in PTM (oxidation, phosphorylation, and acetylation) of proteins in the kidney and aorta and this effect was partially reversed by insulin treatment. Ingenuity pathway analysis performed on the list of differentially expressed proteins depicted mitochondrial dysfunction, oxidative phosphorylation and acute phase response signaling to be among the altered canonical pathways by diabetes in both tissues. The findings of the present study provide a global proteomics view of markers that highlight the mechanisms and putative processes that modulate renal and vascular injury in diabetes.
Collapse
Affiliation(s)
- Moustafa Al Hariri
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohamad Elmedawar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rui Zhu
- Department of Chemistry and Biochemistry, Texas Tech University, Memorial Circle & Boston, Lubbock, Texas, United States of America
| | - Miran A. Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, Beirut, Lebanon
| | - Jingfu Zhao
- Department of Chemistry and Biochemistry, Texas Tech University, Memorial Circle & Boston, Lubbock, Texas, United States of America
| | - Parvin Mirzaei
- Department of Chemistry and Biochemistry, Texas Tech University, Memorial Circle & Boston, Lubbock, Texas, United States of America
| | - Adnan Ahmed
- Center for Biotechnology & Genomics, Texas Tech University, Canton & Main, Experimental Sciences building, Lubbock, Texas, United States of America
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fuad N. Ziyadeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Memorial Circle & Boston, Lubbock, Texas, United States of America
| | - Ayad A. Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
43
|
Renu K, Abilash V, Tirupathi Pichiah P, Syeda TA, Arunachalam S. Adriamycin-induced cardiomyopathy can serve as a model for diabetic cardiomyopathy – a hypothesis. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
44
|
Resistance exercise improves cardiac function and mitochondrial efficiency in diabetic rat hearts. Pflugers Arch 2017; 470:263-275. [PMID: 29032504 DOI: 10.1007/s00424-017-2076-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/22/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022]
Abstract
Metabolic disturbance and mitochondrial dysfunction are a hallmark of diabetic cardiomyopathy (DC). Resistance exercise (RE) not only enhances the condition of healthy individuals but could also improve the status of those with disease. However, the beneficial effects of RE in the prevention of DC and mitochondrial dysfunction are uncertain. Therefore, this study investigated whether RE attenuates DC by improving mitochondrial function using an in vivo rat model of diabetes. Fourteen Otsuka Long-Evans Tokushima Fatty rats were assigned to sedentary control (SC, n = 7) and RE (n = 7) groups at 28 weeks of age. Long-Evans Tokushima Otsuka rats were used as the non-diabetic control. The RE rats were trained by 20 repetitions of climbing a ladder 5 days per week. RE rats exhibited higher glucose uptake and lower lipid profiles, indicating changes in energy metabolism. RE rats significantly increased the ejection fraction and fractional shortening compared with the SC rats. Isolated mitochondria in RE rats showed increase in mitochondrial numbers, which were accompanied by higher expression of mitochondrial biogenesis proteins such as proliferator-activated receptor-γ coactivator-1α and TFAM. Moreover, RE rats reduced proton leakage and reactive oxygen species production, with higher membrane potential. These results were accompanied by higher superoxide dismutase 2 and lower uncoupling protein 2 (UCP2) and UCP3 levels in RE rats. These data suggest that RE is effective at ameliorating DC by improving mitochondrial function, which may contribute to the maintenance of diabetic cardiac contractility.
Collapse
|