1
|
Wang X, Tan X, Zhang T, Xu S, Zeng Y, Xu A, Li X, Zhang G, Jiang Y, Jiang H, Fan J, Bo X, Fan H, Zhou Y. Modeling diabetic cardiomyopathy using human cardiac organoids: Effects of high glucose and lipid conditions. Chem Biol Interact 2025; 411:111421. [PMID: 39984109 DOI: 10.1016/j.cbi.2025.111421] [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: 09/25/2024] [Revised: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a complex metabolic disorder resulting from chronic hyperglycemia and lipid toxicity, which leads to cardiac dysfunction, fibrosis, inflammation, and mitochondrial impairment. Traditional two-dimensional (2D) cell cultures and animal models have limitations in replicating human cardiac physiology and pathophysiology. In this study, we successfully developed a three-dimensional (3D) model of DCM using cardiac organoids generated from human induced pluripotent stem cells (hiPSCs). These organoids were treated with varying concentrations of glucose and sodium palmitate to mimic the high-glucose and high-lipid environment associated with diabetes. At lower concentrations, glucose and sodium palmitate enhanced cell viability, while higher concentrations induced significant cardiotoxic effects, including apoptosis, oxidative stress, and mitochondrial dysfunction. The cardiac organoids also exhibited increased expression of cardiac injury markers, fibrosis-related genes, and inflammatory cytokines under high-glucose and high-lipid conditions. Treatment with metformin, a widely used antidiabetic drug, mitigated these adverse effects, indicating the model's potential for drug testing and evaluation. Our findings demonstrate that this human-derived 3D cardiac organoid model provides a more physiologically relevant platform for studying DCM and can effectively complement traditional models. This model holds promise for advancing the understanding of diabetic heart disease and for assessing the efficacy of potential therapeutic interventions.
Collapse
Affiliation(s)
- Xiangyu Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Xin Tan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Ting Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China; Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Ahhui Medical University, Hefei, 230011, China
| | - Shuai Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Yiyao Zeng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Anchen Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Xian Li
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Hezi Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Jili Fan
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Xiaohong Bo
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Huimin Fan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou Dushu Lake Hospital, Suzhou, 215028, China.
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
2
|
Mo D, Zhang P, Wang M, Guan J, Dai H. Associations of the triglyceride-glucose index with short-term mortality in patients with cardiogenic shock: a cohort study. Lipids Health Dis 2025; 24:130. [PMID: 40186173 PMCID: PMC11969958 DOI: 10.1186/s12944-025-02548-z] [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: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Cardiogenic shock (CS) is a severe cardiac disorder with a high mortality rate. The triglyceride-glucose (TyG) index, a biomarker of insulin resistance, is associated with cardiovascular disease-related mortality. This study aimed to investigate the association between the TyG index and mortality in patients with CS. METHODS This retrospective cohort study analyzed 727 patients with CS from the Medical Information Mart for Intensive Care IV database. The TyG index was calculated as follows: ln[triglycerides (mg/dL) × fasting blood glucose (mg/dL)/2]. Outcomes included 28-day intensive care unit (ICU) mortality and 28-day in-hospital mortality. Kaplan-Meier survival curve models and Cox proportional hazards regression models were used to evaluate the prognostic significance of the TyG index. Receiver Operating Characteristic (ROC) curve analysis was used to determine the predictive efficacy of the TyG index for mortality. Subgroup analyses were conducted to determine the association between the TyG index and mortality across different groups. RESULTS Non-survivors had a significantly higher TyG index (ICU: 9.30 vs. 9.13, p = 0.008; in-hospital: 9.29 vs. 9.13, p = 0.004). Adjusted Cox models showed that each 1-unit increase in the TyG index increased ICU mortality risk by 24% (hazard ratio [HR] = 1.24, 95% confidence interval [CI]:1.04-1.48; p = 0.015) and in-hospital mortality by 44% (HR = 1.44, 95% CI:1.11-1.88; p = 0.007). The Quartile 4 TyG index ICU mortality was increased by 77% (HR = 1.77, 95% CI:1.09-2.89) compared to that for Quartile 1 and in-hospital mortality was increased by 61% (HR = 1.61, 95% CI:1.08-2.38). The area under the ROC curve (AUROC) showed a modest standalone predictive ability of 0.56, but when combined with clinical variables, the AUROC improved to 0.80 (ICU) and 0.78 (in-hospital). Subgroup analyses identified stronger associations in patients ≥ 60 years, females, non-septic, and those with acute myocardial infarction or heart failure. CONCLUSIONS The TyG index is significantly associated with short-term mortality in patients with CS and may serve as a useful biomarker for risk stratification. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Degang Mo
- School of Medicine, Qingdao University, Qingdao, 266000, China
- Department of Cardiology, Qingdao Municipal Hospital, No.5 Donghai Middle Road, Qingdao, 266071, China
| | - Peng Zhang
- School of Medicine, Qingdao University, Qingdao, 266000, China
- Department of Cardiology, Qingdao Municipal Hospital, No.5 Donghai Middle Road, Qingdao, 266071, China
| | - Mengmeng Wang
- School of Medicine, Qingdao University, Qingdao, 266000, China
| | - Jun Guan
- Department of Cardiology, Qingdao Municipal Hospital, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Hongyan Dai
- Department of Cardiology, Qingdao Municipal Hospital, No.5 Donghai Middle Road, Qingdao, 266071, China.
| |
Collapse
|
3
|
Guo Y, Zhang Z, Wen Z, Kang X, Wang D, Zhang L, Cheng M, Yuan G, Ren H. Mitochondrial SIRT2-mediated CPT2 deacetylation prevents diabetic cardiomyopathy by impeding cardiac fatty acid oxidation. Int J Biol Sci 2025; 21:725-744. [PMID: 39781464 PMCID: PMC11705638 DOI: 10.7150/ijbs.102834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/07/2024] [Indexed: 01/12/2025] Open
Abstract
Dysregulated energy metabolism, particularly lipid metabolism disorders, has been identified as a key factor in the development of diabetic cardiomyopathy (DCM). Sirtuin 2 (SIRT2) is a deacetylase involved in the regulation of metabolism and cellular energy homeostasis, yet its role in the progression of DCM remains unclear. We observed significantly reduced SIRT2 expression in DCM model mice. Cardiac-specific overexpression of SIRT2 protected mice from streptozotocin/high-fat diet (STZ/HFD)-induced insulin resistance (IR), cell apoptosis, and cardiac dysfunction, whereas its downregulation exacerbated these conditions. Moreover, we found that SIRT2 regulated cardiac lipid accumulation and fatty acid oxidation (FAO), and identified its localization in cardiac mitochondria. Mechanistically, we determined carnitine palmitoyltransferase 2 (CPT2) as a critical substrate of SIRT2, which is implicated in DCM. SIRT2-mediated deacetylation at K239 enhanced CPT2 ubiquitination, resulting in decreased protein stability and subsequent inhibition of FAO and reactive oxygen species (ROS) production. Taken together, these findings suggest that the SIRT2/CPT2 signaling pathway plays a crucial role in DCM progression.
Collapse
Affiliation(s)
- Yaoyao Guo
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Ziyin Zhang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Hubei, China
| | - Xiaonan Kang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Dan Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Lu Zhang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Mengke Cheng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Gang Yuan
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Huihui Ren
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| |
Collapse
|
4
|
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
|
5
|
Durumutla HB, Prabakaran AD, El Abdellaoui Soussi F, Akinborewa O, Latimer H, McFarland K, Piczer K, Werbrich C, Jain MK, Haldar SM, Quattrocelli M. Glucocorticoid chronopharmacology promotes glucose metabolism in heart through a cardiomyocyte-autonomous transactivation program. JCI Insight 2024; 9:e182599. [PMID: 39378111 PMCID: PMC11601906 DOI: 10.1172/jci.insight.182599] [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: 05/01/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024] Open
Abstract
Circadian time of intake gates the cardioprotective effects of glucocorticoid administration in both healthy and infarcted hearts. The cardiomyocyte-specific glucocorticoid receptor (GR) and its cofactor, Krüppel-like factor 15 (KLF15), play critical roles in maintaining normal heart function in the long term and serve as pleiotropic regulators of cardiac metabolism. Despite this understanding, the cardiomyocyte-autonomous metabolic targets influenced by the concerted epigenetic action of the GR/KLF15 axis remain undefined. Here, we demonstrated the critical roles of the cardiomyocyte-specific GR and KLF15 in orchestrating a circadian-dependent glucose oxidation program within the heart. Combining integrated transcriptomics and epigenomics with cardiomyocyte-specific inducible ablation of GR or KLF15, we identified their synergistic role in the activation of adiponectin receptor expression (Adipor1) and the mitochondrial pyruvate complex (Mpc1/2), thereby enhancing insulin-stimulated glucose uptake and pyruvate oxidation. Furthermore, in obese diabetic (db/db) mice exhibiting insulin resistance and impaired glucose oxidation, light-phase prednisone administration, as opposed to dark-phase prednisone dosing, restored cardiomyocyte glucose oxidation and improved diastolic function. These effects were blocked by combined in vivo knockdown of GR and KLF15 levels in db/db hearts. In summary, this study leveraged the circadian-dependent cardioprotective effects of glucocorticoids to identify cardiomyocyte-autonomous targets for the GR/KLF15 axis in glucose metabolism.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Mukesh K. Jain
- Department of Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Saptarsi M. Haldar
- Amgen Research, South San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
- Department of Medicine, Cardiology Division, UCSF, San Francisco, California, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| |
Collapse
|
6
|
Zhan J, Zhou Y, Chen Y, Jin K, Chen Z, Chen C, Li H, Wang DW. Subcellular mass spectrometric detection unveils hyperglycemic memory in the diabetic heart. J Diabetes 2024; 16:e70033. [PMID: 39539089 PMCID: PMC11561303 DOI: 10.1111/1753-0407.70033] [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: 03/10/2024] [Revised: 09/24/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Intensive glycemic control is insufficient to reduce the risk of heart failure in patients with diabetes mellitus. While the hyperglycemic memory in the diabetic cardiomyopathy has been well documented, its underlying mechanisms are not fully understood. The present study tried to investigate whether the dysregulated proteins/biological pathways, which persistently altered in diabetic hearts during normoglycemia, participate in the hyperglycemic memory. METHODS Hearts of streptozotocin-induced diabetic mice, with or without intensive glycemic control using slow-release insulin implants, were collected. Proteins from total heart samples and subcellular fractions were assessed by mass spectrometry, Western blotting, and KEGG pathway enrichment analysis. mRNA sequencing was used to determine whether the persistently altered proteins were regulated at the transcriptional or post-transcriptional level. RESULTS Western blot validation of several proteins with high pathophysiological importance, including MYH7, HMGCS2, PDK4, and BDH1, indicated that mass spectrometry was able to qualitatively, but not quantitatively, reflect the fold changes of certain proteins in diabetes. Pathway analysis revealed that the peroxisome, PPAR pathway, and fatty acid metabolism could be efficiently rescued by glycemic control. However, dysregulation of oxidative phosphorylation and reactive oxygen species persisted even after normalization of hyperglycemia. Notably, mRNA sequencing revealed that dysregulated proteins in the oxidative phosphorylation pathway were not accompanied by coordinated changes in mRNA levels, indicating post-transcriptional regulation. Moreover, literature review and bioinformatics analysis suggested that hyperglycemia-induced persistent alterations of miRNAs targeted genes from the persistently dysregulated oxidative phosphorylation pathway, whereas, oxidative phosphorylation dysfunction-induced ROS regulated miRNA expression, which thereby might sustained the dysregulation of miRNAs. CONCLUSIONS Glycemic control cannot rescue hyperglycemia-induced alterations of subcellular proteins in the diabetic heart, and persistently altered proteins are involved in multiple functional pathways, including oxidative phosphorylation. These findings might provide novel insights into hyperglycemic memory in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jiabing Zhan
- Division of Cardiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhanChina
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart DiseaseFujian Medical University Union HospitalFuzhouChina
| | - Yufei Zhou
- Division of Cardiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhanChina
| | - Yifan Chen
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart DiseaseFujian Medical University Union HospitalFuzhouChina
| | - Kunying Jin
- Division of Cardiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhanChina
| | - Zhaoyang Chen
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart DiseaseFujian Medical University Union HospitalFuzhouChina
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhanChina
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhanChina
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhanChina
| |
Collapse
|
7
|
Durumutla HB, Prabakaran AD, Soussi FEA, Akinborewa O, Latimer H, McFarland K, Piczer K, Werbrich C, Jain MK, Haldar SM, Quattrocelli M. Glucocorticoid chrono-pharmacology unveils novel targets for the cardiomyocyte-specific GR-KLF15 axis in cardiac glucose metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572210. [PMID: 38187555 PMCID: PMC10769285 DOI: 10.1101/2023.12.18.572210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Circadian time-of-intake gates the cardioprotective effects of glucocorticoid administration in both healthy and infarcted hearts. The cardiomyocyte-specific glucocorticoid receptor (GR) and its co-factor, Krüppel-like factor (Klf15), play critical roles in maintaining normal heart function in the long-term and serve as pleiotropic regulators of cardiac metabolism. Despite this understanding, the cardiomyocyte-autonomous metabolic targets influenced by the concerted epigenetic action of GR-Klf15 axis remain undefined. Here, we demonstrate the critical roles of the cardiomyocyte-specific GR and Klf15 in orchestrating a circadian-dependent glucose oxidation program within the heart. Combining integrated transcriptomics and epigenomics with cardiomyocyte-specific inducible ablation of GR or Klf15, we identified their synergistic role in the activation of adiponectin receptor expression (Adipor1) and the mitochondrial pyruvate complex (Mpc1/2), thereby enhancing insulin-stimulated glucose uptake and pyruvate oxidation. Furthermore, in obese diabetic (db/db) mice exhibiting insulin resistance and impaired glucose oxidation, light-phase prednisone administration, as opposed to dark-phase prednisone dosing, effectively restored cardiomyocyte glucose oxidation and improved diastolic function towards control-like levels in a sex-independent manner. Collectively, our findings uncover novel cardiomyocyte-autonomous metabolic targets of the GR-Klf15 axis. This study highlights the circadian-dependent cardioprotective effects of glucocorticoids on cardiomyocyte glucose metabolism, providing critical insights into chrono-pharmacological strategies for glucocorticoid therapy in cardiovascular disease.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mukesh K Jain
- Dept Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Saptarsi M Haldar
- Amgen Research, South San Francisco, CA, USA; Gladstone Institutes, San Francisco, CA, USA and Dept Medicine, Cardiology Division, UCSF, San Francisco, CA, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
8
|
Bora S, Adole PS, Vinod KV, Pillai AA, Ahmed S. GC-MS validation and analysis of targeted plasma metabolites related to carbonyl stress in type 2 diabetes mellitus patients with and without acute coronary syndrome. Biomed Chromatogr 2024; 38:e5952. [PMID: 38966927 DOI: 10.1002/bmc.5952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/18/2024] [Indexed: 07/06/2024]
Abstract
Methylglyoxal (MG) is responsible for advanced glycation end-product formation, the mechanisms leading to diabetes pathogenesis and complications like acute coronary syndrome (ACS). Sugar metabolites, amino acids and fatty acids are possible substrates for MG. The study aimed to measure plasma MG substrate levels using a validated gas chromatography-mass spectrometry (GC-MS) method and explore their association with ACS risk in type 2 diabetes mellitus (T2DM). The study included 150 T2DM patients with ACS as cases and 150 T2DM without ACS as controls for the analysis of glucose, fructose, ribulose, sorbitol, glycerol, pyruvate, lactate, glycine, serine, threonine, C16:0, C16:1, C18:0, C18:1, C18:2, C18:3, C20:0 and C22:6 by GC-MS. Validated GC-MS methods were accurate, precise and sensitive. Cases significantly differed in plasma MG and metabolite levels except for lactate, C16:0, C18:0, C18:2, and C18:3 levels compared with controls. On multivariable logistic regression, plasma C20:0, C18:1, glycine and glycerol levels had increased odds of ACS risk. On multivariate receiver operating characteristic analysis, a model containing plasma C20:0, C16:1, C18:1, C18:2, serine, glycerol, lactate and threonine levels had the highest area under the curve value (0.932) for ACS diagnosis. In conclusion, plasma C20:0, C16:1, C18:1, glycine, glycerol and sorbitol levels were associated with ACS risk in T2DM.
Collapse
Affiliation(s)
- Sushmita Bora
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Prashant Shankarrao Adole
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Kolar Vishwanath Vinod
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Ajith Ananthakrishna Pillai
- Department of Cardiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Shaheer Ahmed
- Department of Cardiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| |
Collapse
|
9
|
Mendez DA, Soñanez-Organis JG, Yang X, Vazquez-Anaya G, Nishiyama A, Ortiz RM. Exogenous thyroxine increases cardiac GLUT4 translocation in insulin resistant OLETF rats. Mol Cell Endocrinol 2024; 590:112254. [PMID: 38677465 DOI: 10.1016/j.mce.2024.112254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
During insulin resistance, the heart undergoes a metabolic shift in which fatty acids (FA) account for roughly about 99% of the ATP production. This metabolic shift is indicative of impaired glucose metabolism. A shift in FA metabolism with impaired glucose tolerance can increase reactive oxygen species (ROS), lipotoxicity, and mitochondrial dysfunction, ultimately leading to cardiomyopathy. Thyroid hormones (TH) may improve the glucose intolerance by increasing glucose reabsorption and metabolism in peripheral tissues, but little is known on its effects on cardiac tissue during insulin resistance. In the present study, insulin resistant Otsuka Long Evans Tokushima Fatty (OLETF) rats were used to assess the effects of exogenous thyroxine (T4) on glucose metabolism in cardiac tissue. Rats were assigned to four groups: (1) lean, Long Evans Tokushima Otsuka (LETO; n=6), (2) LETO + T4 (8 μg/100 g BM/d × 5 wks; n = 7), (3) untreated OLETF (n = 6), and (4) OLETF + T4 (8 μg/100 g BM/d × 5 wks; n = 7). T4 increased GLUT4 gene expression by 85% in OLETF and increased GLUT4 protein translocation to the membrane by 294%. Additionally, T4 increased p-AS160 by 285%, phosphofructokinase-1 (PFK-1) mRNA, the rate limiting step in glycolysis, by 98% and hexokinase II by 64% in OLETF. T4 decreased both CPT2 mRNA and protein expression in OLETF. The results suggest that exogenous T4 has the potential to increase glucose uptake and metabolism while simultaneously reducing fatty acid transport in the heart of insulin resistant rats. Thus, L-thyroxine may have therapeutic value to help correct the impaired substrate metabolism associated with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Dora A Mendez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA.
| | - José G Soñanez-Organis
- Division of Science and Engineering, Department of Chemical Biological and Agropecuary Sciences, University of Sonora, Navojoa, SON, Mexico
| | - Xue Yang
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA
| | - Guillermo Vazquez-Anaya
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Rudy M Ortiz
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, USA
| |
Collapse
|
10
|
Garg M, Gandhi K, Gera P, Jadhav SM, Mohanty B, Gurjar M, Sandupatla B, Gala R, Chaudhari P, Prasad M, Chinnaswamy G, Gota V. Implications of chronic moderate protein-deficiency malnutrition on doxorubicin pharmacokinetics and cardiotoxicity in early post-weaning stage. Life Sci 2024; 350:122765. [PMID: 38830506 DOI: 10.1016/j.lfs.2024.122765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Malnutrition is a common problem in developing countries, and the impact of severe malnutrition on optimal treatment outcomes of chemotherapy in pediatric cancer patients is well documented. However, despite being a more prevalent and distinct entity, moderate malnutrition is until now unexplored for its effects on treatment outcomes. AIMS In this study we aimed to investigate the molecular basis of altered pharmacokinetics and cardiotoxicity of doxorubicin observed in early-life chronic moderate protein deficiency malnutrition. MATERIALS AND METHODS We developed an animal model of early-life moderate protein-deficiency malnutrition and validated it using clinical samples. This model was used to study pharmacokinetic and toxicity changes and was further utilized to study the molecular changes in liver and heart to get mechanistic insights. KEY FINDINGS Here we show that moderate protein-deficiency malnutrition in weanling rats causes changes in drug disposition in the liver by modification of hepatic ABCC3 and MRP2 transporters through the TNFα signalling axis. Furthermore, malnourished rats in repeat-dose doxorubicin toxicity study showed higher toxicity and mortality. A higher accumulation of doxorubicin in the heart was observed which was associated with alterations in cardiac metabolic pathways and increased cardiotoxicity. SIGNIFICANCE Our findings indicate that moderate malnutrition causes increased susceptibility towards toxic side effects of chemotherapy. These results may necessitate further investigations and new guidelines on the dosing of chemotherapy in moderately malnourished pediatric cancer patients.
Collapse
Affiliation(s)
- Megha Garg
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India
| | - Khushboo Gandhi
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Poonam Gera
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Biorepository, Advanced Centre for Treatment Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Shraddha Mahesh Jadhav
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Bhabani Mohanty
- Department of Comparative Oncology and Small Animal Imaging Facility, ACTREC, Tata Memorial Center, Kharghar, Navi-Mumbai, Maharashtra 410210, India
| | - Murari Gurjar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Department of Clinical Pharmacology, Mahamana Pandit Madan Mohan Malviya Cancer Centre, Banaras Hindu University Campus, Varanasi, Uttar Pradesh 221005, India
| | | | - Rajul Gala
- Paediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Pradip Chaudhari
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Department of Comparative Oncology and Small Animal Imaging Facility, ACTREC, Tata Memorial Center, Kharghar, Navi-Mumbai, Maharashtra 410210, India
| | - Maya Prasad
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Paediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Girish Chinnaswamy
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India; Paediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Vikram Gota
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra 400094, India.
| |
Collapse
|
11
|
Tappia PS, Elimban V, Shah AK, Goyal RK, Dhalla NS. Improvement of Cardiac Function and Subcellular Defects Due to Chronic Diabetes upon Treatment with Sarpogrelate. J Cardiovasc Dev Dis 2024; 11:215. [PMID: 39057635 PMCID: PMC11276782 DOI: 10.3390/jcdd11070215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
In order to investigate the subcellular mechanisms underlying the beneficial effects of sarpogrelate-a 5-HT2A receptor antagonist-on diabetic cardiomyopathy, diabetes was induced in rats by injecting streptozotocin (65 mg/kg). Diabetic animals were treated with or without sarpogrelate (5 mg/kg daily) for 6 weeks; diabetic animals were also treated with insulin (10 units/kg daily) for comparison. Elevated plasma levels of glucose and lipids, depressed insulin levels, hemodynamic alterations and cardiac dysfunction in diabetic animals were partially or fully attenuated by sarpogrelate or insulin treatment. Diabetes-induced changes in myocardial high-energy phosphate stores, as well as depressed mitochondrial oxidative phosphorylation and Ca2+-uptake activities, were significantly prevented by these treatments. Reductions in sarcolemma Na+-K+ ATPase, Na+-Ca2+ exchange, Ca2+-channel density and Ca2+-uptake activities were also attenuated by treatments with sarpogrelate and insulin. In addition, decreases in diabetes-induced sarcoplasmic reticulum Ca2+-uptake, Ca2+-release and Ca2+-stimulated ATPase activities, myofibrillar Mg2+-ATPase and Ca2+-stimulated ATPase activities, and myosin Mg2+-ATPase and Ca2+-ATPase activities were fully or partially prevented by sarpogrelate and insulin treatments. Marked alterations in different biomarkers of oxidative stress, such as malondialdehyde, superoxide dismutase and glutathione peroxidase, in diabetic hearts were also attenuated by treating the animals with sarpogrelate or insulin. These observations suggest that therapy with sarpogrelate, like that with insulin, may improve cardiac function by preventing subcellular and metabolic defects as a consequence of a reduction in oxidative stress.
Collapse
Affiliation(s)
- Paramjit S. Tappia
- Institute of Cardiovascular Sciences, and Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
| | - Vijayan Elimban
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada;
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Anureet K. Shah
- Department of Nutrition and Food Sciences, California State University, Los Angeles, CA 90032, USA;
| | - Ramesh K. Goyal
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India;
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada;
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
12
|
Wu M, Tan J, Cao Z, Cai Y, Huang Z, Chen Z, He W, Liu X, Jiang Y, Gao Q, Deng B, Wang J, Yuan W, Zhang H, Chen Y. Sirt5 improves cardiomyocytes fatty acid metabolism and ameliorates cardiac lipotoxicity in diabetic cardiomyopathy via CPT2 de-succinylation. Redox Biol 2024; 73:103184. [PMID: 38718533 PMCID: PMC11091707 DOI: 10.1016/j.redox.2024.103184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 06/14/2024] Open
Abstract
RATIONALE The disruption of the balance between fatty acid (FA) uptake and oxidation (FAO) leads to cardiac lipotoxicity, serving as the driving force behind diabetic cardiomyopathy (DbCM). Sirtuin 5 (Sirt5), a lysine de-succinylase, could impact diverse metabolic pathways, including FA metabolism. Nevertheless, the precise roles of Sirt5 in cardiac lipotoxicity and DbCM remain unknown. OBJECTIVE This study aims to elucidate the role and underlying mechanism of Sirt5 in the context of cardiac lipotoxicity and DbCM. METHODS AND RESULTS The expression of myocardial Sirt5 was found to be modestly elevated in diabetic heart failure patients and mice. Cardiac dysfunction, hypertrophy and lipotoxicity were exacerbated by ablation of Sirt5 but improved by forced expression of Sirt5 in diabetic mice. Notably, Sirt5 deficiency impaired FAO without affecting the capacity of FA uptake in the diabetic heart, leading to accumulation of FA intermediate metabolites, which mainly included medium- and long-chain fatty acyl-carnitines. Mechanistically, succinylomics analyses identified carnitine palmitoyltransferase 2 (CPT2), a crucial enzyme involved in the reconversion of fatty acyl-carnitines to fatty acyl-CoA and facilitating FAO, as the functional succinylated substrate mediator of Sirt5. Succinylation of Lys424 in CPT2 was significantly increased by Sirt5 deficiency, leading to the inactivation of its enzymatic activity and the subsequent accumulation of fatty acyl-carnitines. CPT2 K424R mutation, which mitigated succinylation modification, counteracted the reduction of enzymatic activity in CPT2 mediated by Sirt5 deficiency, thereby attenuating Sirt5 knockout-induced FAO impairment and lipid deposition. CONCLUSIONS Sirt5 deficiency impairs FAO, leading to cardiac lipotoxicity in the diabetic heart through the succinylation of Lys424 in CPT2. This underscores the potential roles of Sirt5 and CPT2 as therapeutic targets for addressing DbCM.
Collapse
Affiliation(s)
- Maoxiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Jing Tan
- Laboratory Animal Center and Department of Biochemistry, Institute of Guangdong Engineering and Technology Research Center for Disease-Model Animals, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhengyu Cao
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Yangwei Cai
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhaoqi Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhiteng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Wanbing He
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuan Jiang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Qingyuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Bingqing Deng
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
| | - Woliang Yuan
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
| | - Haifeng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
| | - Yangxin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangzhou Key Laboratory of Molecular Mechanisms of Major Cardiovascular Disease, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
13
|
Zhang J, Zhou H, Lei F, Jiang K, Liao Y, Huang F, Chen M. Cholesterol 25-hydroxylase prevents type 2 diabetes mellitus induced cardiomyopathy by alleviating cardiac lipotoxicity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167158. [PMID: 38588780 DOI: 10.1016/j.bbadis.2024.167158] [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: 10/06/2023] [Revised: 02/28/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
OBJECTIVES Diabetic cardiomyopathy (DCM) is the leading cause of mortality in type 2 diabetes mellitus (T2DM) patients, with its underlying mechanisms still elusive. This study aims to investigate the role of cholesterol-25-monooxygenase (CH25H) in T2DM induced cardiomyopathy. METHODS High fat diet combined with streptozotocin (HFD/STZ) were used to establish a T2DM model. CH25H and its product 25-hydroxycholesterol (25HC) were detected in the hearts of T2DM model. Gain- or loss-of-function of CH25H were performed by receiving AAV9-cTNT-CH25H or CH25H knockout (CH25H-/-) mice with HFD/STZ treatment. Cardiac function was evaluated using echocardiography, and cardiac tissues were collected for immunoblot analysis, histological assessment and quantitative polymerase chain reaction (qPCR). Mitochondrial morphology and function were evaluated using transmission electron microscopy (TEM) and Seahorse XF Cell Mito Stress Test Kit. RNA-sequence analysis was performed to determine the molecular changes associated with CH25H deletion. RESULTS CH25H and 25HC were significantly decreased in the hearts of T2DM mice. CH25H-/- mice treated with HFD/STZ exhibited impaired mitochondrial function and structure, increased lipid accumulation, and aggregated cardiac dysfunction. Conversely, T2DM mice receiving AAV9-CH25H displayed cardioprotective effects. Mechanistically, RNA sequencing and qPCR analysis revealed that CH25H deficiency decreased peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and its target gene expression. Additionally, administration of ZLN005, a potent PGC-1α activator, partially protected against high glucose and palmitic acid induced mitochondria dysfunction and lipid accumulation in vitro. CONCLUSION Our study provides compelling evidence supporting the protective role of CH25H in T2DM-induced cardiomyopathy. Furthermore, the regulation of PGC-1α may be intricately involved in this cardioprotective process.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/prevention & control
- Diabetic Cardiomyopathies/etiology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Mice
- Mice, Knockout
- Male
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Steroid Hydroxylases/metabolism
- Steroid Hydroxylases/genetics
- Diet, High-Fat/adverse effects
- Mice, Inbred C57BL
- Hydroxycholesterols/metabolism
- Myocardium/metabolism
- Myocardium/pathology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
Collapse
Affiliation(s)
- Jialiang Zhang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Zhou
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Lei
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kexin Jiang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbiao Liao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fangyang Huang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Chen PH, Lee TW, Liu SH, Huynh TV, Chung CC, Yeh YH, Kao YH, Chen YJ. Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes. Exp Ther Med 2024; 27:126. [PMID: 38414784 PMCID: PMC10895620 DOI: 10.3892/etm.2024.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/11/2024] [Indexed: 02/29/2024] Open
Abstract
Acetyl-CoA carboxylase 2 plays a crucial role in regulating mitochondrial fatty acid oxidation in cardiomyocytes. Lithium, a monovalent cation known for its cardioprotective potential, has been investigated for its influence on mitochondrial bioenergetics. The present study explored whether lithium modulated acetyl-CoA carboxylase 2 and mitochondrial fatty acid metabolism in cardiomyocytes and the potential therapeutic applications of lithium in alleviating metabolic stress. Mitochondrial bioenergetic function, fatty acid oxidation, reactive oxygen species production, membrane potential and the expression of proteins involved in fatty acid metabolism in H9c2 cardiomyocytes treated with LiCl for 48 h was measured by using a Seahorse extracellular flux analyzer, fluorescence microscopy and western blotting. Small interfering RNA against glucose transporter type 4 was transfected into H9c2 cardiomyocytes for 48 h to induce metabolic stress mimicking insulin resistance. The results revealed that LiCl at a concentration of 0.3 mM (but not at a concentration of 0.1 or 1.0 mM) upregulated the expression of phosphorylated (p-)glycogen synthase kinase-3 beta and downregulated the expression of p-acetyl-CoA carboxylase 2 but did not affect the expression of adenosine monophosphate-activated protein kinase or calcineurin. Cotreatment with TWS119 (8 µM) and LiCl (0.3 mM) downregulated p-acetyl-CoA carboxylase 2 expression to a similar extent as did treatment with TWS119 (8 µM) alone. Moreover, LiCl (0.3 mM) inhibited mitochondrial fatty acid oxidation, improved coupling efficiency and the cellular respiratory control ratio, hindered reactive oxygen species production and proton leakage and restored mitochondrial membrane potential in glucose transporter type 4 knockdown-H9c2 cardiomyocytes. These findings suggested that low therapeutic levels of lithium can downregulate p-acetyl-CoA carboxylase 2, thus reducing mitochondrial fatty acid oxidation and oxidative stress in cardiomyocytes.
Collapse
Affiliation(s)
- Pao-Huan Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Psychiatry, Taipei Medical University Hospital, Taipei 11031, Taiwan, R.O.C
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Shuen-Hsin Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan, R.O.C
| | - Tin Van Huynh
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Interventional Cardiology, Thong Nhat Hospital, Ho Chi Minh City 700000, Vietnam
| | - Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Yung-Hsin Yeh
- Division of Cardiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan, R.O.C
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan, R.O.C
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| |
Collapse
|
15
|
Uddandrao VVS, Chandrasekaran P, Saravanan G, Brahmanaidu P, Sengottuvelu S, Ponmurugan P, Vadivukkarasi S, Kumar U. Phytoformulation with hydroxycitric acid and capsaicin protects against high-fat-diet-induced obesity cardiomyopathy by reducing cardiac lipid deposition and ameliorating inflammation and apoptosis in the heart. J Tradit Complement Med 2024; 14:162-172. [PMID: 38481548 PMCID: PMC10927456 DOI: 10.1016/j.jtcme.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/15/2023] [Accepted: 08/23/2023] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND AND AIM Phytoformulation therapy is a pioneering strategy for the treatment of metabolic disorders and related diseases. The aim of the present study was to investigate the protective effect of a phytoformulation consisting of hydroxycitric acid and capsaicin against obesity-related cardiomyopathy. EXPERIMENTAL PROCEDURE Sprague-Dawley rats were fed HFD for 21 weeks, and phytoformulation (100 mg/kg body weight) was administered orally for 45 days starting at week 16. RESULTS AND CONCLUSION We found that HFD supplementation resulted in significant hyperglycemia and caused an increase in cardiac lipid deposition, inflammation and apoptosis in the heart. Phytoformulation therapy not only significantly decreased blood levels of glucose, cholesterol, triglycerides, free fatty acids, and inflammatory cytokines in obese rats, but also protected cardiac tissue, as shown by histological analysis. Conversely, phytoformulation therapy decreased mRNA levels for sterol regulatory element-binding factor 1, fatty acid synthase, acetyl-CoA carboxylase, and fatty acid binding protein 1 genes involved in fatty acid synthesis and absorption in obese rats. It increased the levels of lysosomal acid lipase, hormone-sensitive lipase, and lipoprotein lipase genes involved in fatty acid degradation in the heart. In addition, the phytoformulation improved cardiac inflammation and apoptosis by downregulating the genes nuclear factor kappa-light-chain enhancer of activated B cells (NF-kB), tumour necrosis factor α, interleukin-6, toll-like receptor-4 (TLR-4), BCL2-associated X and caspase-3. In conclusion, our results show that the phytoformulation improved insulin sensitivity and attenuated myocardial lipid accumulation, inflammation, and apoptosis in the heart of HFD-induced obese rats by regulating fatty acid metabolism genes and downregulating NF-kB/TLR-4/caspase-3.
Collapse
Affiliation(s)
- V. V. Sathibabu Uddandrao
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - P. Chandrasekaran
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - G. Saravanan
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - Parim Brahmanaidu
- Animal Physiology and Biochemistry Laboratory, ICMR-National Animal Resource Facility for Biomedical Research (ICMR-NARFBR), Hyderabad, 500078, India
| | - S. Sengottuvelu
- Department of Pharmacology, Nandha College of Pharmacy, Erode, Tamilnadu, 638052, India
| | - P. Ponmurugan
- Department of Botany, Bharathiar University, Coimbatore, Tamilnadu, 641046, India
| | - S. Vadivukkarasi
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Ghaziabad, Uttar Pradesh, 201015, India
| |
Collapse
|
16
|
Quan Z, Yang Z, Tang X, Fu C, Zhou X, Huang L, Xia L, Zhang X. A double-tuned 1 H/ 31 P coil for rabbit heart metabolism detection at 3 T. NMR IN BIOMEDICINE 2024; 37:e5049. [PMID: 37767723 DOI: 10.1002/nbm.5049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/16/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
Magnetic resonance imaging (MRI)/magnetic resonance spectroscopy (MRS) employing proton nuclear resonance has emerged as a pivotal modality in clinical diagnostics and fundamental research. Nonetheless, the scope of MRI/MRS extends beyond protons, encompassing nonproton nuclei that offer enhanced metabolic insights. A notable example is phosphorus-31 (31 P) MRS, which provides valuable information on energy metabolites within the skeletal muscle and cardiac tissues of individuals affected by diabetes. This study introduces a novel double-tuned coil tailored for 1 H and 31 P frequencies, specifically designed for investigating cardiac metabolism in rabbits. The proposed coil design incorporates a butterfly-like coil for 31 P transmission, a four-channel array for 31 P reception, and an eight-channel array for 1 H reception, all strategically arranged on a body-conformal elliptic cylinder. To assess the performance of the double-tuned coil, a comprehensive evaluation encompassing simulations and experimental investigations was conducted. The simulation results demonstrated that the proposed 31 P transmit design achieved acceptable homogeneity and exhibited comparable transmit efficiency on par with a band-pass birdcage coil. In vivo experiments further substantiated the coil's efficacy, revealing that the rabbit with experimentally induced diabetes exhibited a lower phosphocreatine/adenosine triphosphate ratio compared with its normal counterpart. These findings emphasize the potential of the proposed coil design as a promising tool for investigating the therapeutic effects of novel diabetes drugs within the context of animal experimentation. Its capability to provide detailed metabolic information establishes it as an indispensable asset within this realm of research.
Collapse
Affiliation(s)
- Zhiyan Quan
- The Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University, Hangzhou, China
| | - Zhaoxia Yang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaocui Tang
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University, Hangzhou, China
| | - Caixia Fu
- Application Developments, Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen, China
| | - Xiaoyue Zhou
- Siemens Healthineers Digital Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Lu Huang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Xia
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaotong Zhang
- The Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University, Hangzhou, China
- Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- College of Electrical Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Weeks KL, Kiriazis H, Wadley GD, Masterman EI, Sergienko NM, Raaijmakers AJA, Trewin AJ, Harmawan CA, Yildiz GS, Liu Y, Drew BG, Gregorevic P, Delbridge LMD, McMullen JR, Bernardo BC. A gene therapy targeting medium-chain acyl-CoA dehydrogenase (MCAD) did not protect against diabetes-induced cardiac pathology. J Mol Med (Berl) 2024; 102:95-111. [PMID: 37987775 DOI: 10.1007/s00109-023-02397-2] [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: 03/20/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Diabetic cardiomyopathy describes heart disease in patients with diabetes who have no other cardiac conditions but have a higher risk of developing heart failure. Specific therapies to treat the diabetic heart are limited. A key mechanism involved in the progression of diabetic cardiomyopathy is dysregulation of cardiac energy metabolism. The aim of this study was to determine if increasing the expression of medium-chain acyl-coenzyme A dehydrogenase (MCAD; encoded by Acadm), a key regulator of fatty acid oxidation, could improve the function of the diabetic heart. Male mice were administered streptozotocin to induce diabetes, which led to diastolic dysfunction 8 weeks post-injection. Mice then received cardiac-selective adeno-associated viral vectors encoding MCAD (rAAV6:MCAD) or control AAV and were followed for 8 weeks. In the non-diabetic heart, rAAV6:MCAD increased MCAD expression (mRNA and protein) and increased Acadl and Acadvl, but an increase in MCAD enzyme activity was not detectable. rAAV6:MCAD delivery in the diabetic heart increased MCAD mRNA expression but did not significantly increase protein, activity, or improve diabetes-induced cardiac pathology or molecular metabolic and lipid markers. The uptake of AAV viral vectors was reduced in the diabetic versus non-diabetic heart, which may have implications for the translation of AAV therapies into the clinic. KEY MESSAGES: The effects of increasing MCAD in the diabetic heart are unknown. Delivery of rAAV6:MCAD increased MCAD mRNA and protein, but not enzyme activity, in the non-diabetic heart. Independent of MCAD enzyme activity, rAAV6:MCAD increased Acadl and Acadvl in the non-diabetic heart. Increasing MCAD cardiac gene expression alone was not sufficient to protect against diabetes-induced cardiac pathology. AAV transduction efficiency was reduced in the diabetic heart, which has clinical implications.
Collapse
Affiliation(s)
- Kate L Weeks
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
| | - Helen Kiriazis
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Emma I Masterman
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Nicola M Sergienko
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Antonia J A Raaijmakers
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Adam J Trewin
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Claudia A Harmawan
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Gunes S Yildiz
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Yingying Liu
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Brian G Drew
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Paul Gregorevic
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
- Centre for Muscle Research, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Julie R McMullen
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Bianca C Bernardo
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia.
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia.
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
18
|
Wu T, Qu Y, Xu S, Wang Y, Liu X, Ma D. SIRT6: A potential therapeutic target for diabetic cardiomyopathy. FASEB J 2023; 37:e23099. [PMID: 37462453 DOI: 10.1096/fj.202301012r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
The abnormal lipid metabolism in diabetic cardiomyopathy can cause myocardial mitochondrial dysfunction, lipotoxicity, abnormal death of myocardial cells, and myocardial remodeling. Mitochondrial homeostasis and normal lipid metabolism can effectively slow down the development of diabetic cardiomyopathy. Recent studies have shown that SIRT6 may play an important role in the pathological changes of diabetic cardiomyopathy such as myocardial cell death, myocardial hypertrophy, and myocardial fibrosis by regulating mitochondrial oxidative stress and glucose and lipid metabolism. Therefore, understanding the function of SIRT6 and its role in the pathogenesis of diabetic cardiomyopathy is of great significance for exploring and developing new targets and drugs for the treatment of diabetic cardiomyopathy. This article reviews the latest findings of SIRT6 in the pathogenesis of diabetic cardiomyopathy, focusing on the regulation of mitochondria and lipid metabolism by SIRT6 to explore potential clinical treatments.
Collapse
Affiliation(s)
- Tao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiwei Qu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shengjie Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Xue Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dufang Ma
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
19
|
Salau VF, Erukainure OL, Olofinsan KA, Msomi NZ, Ijomone OK, Islam MS. Ferulic acid mitigates diabetic cardiomyopathy via modulation of metabolic abnormalities in cardiac tissues of diabetic rats. Fundam Clin Pharmacol 2023; 37:44-59. [PMID: 35841183 PMCID: PMC10086938 DOI: 10.1111/fcp.12819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 01/25/2023]
Abstract
Cardiovascular abnormalities have been reported as a major contributor of diabetic mortality. The protective effect of ferulic acid on diabetic cardiomyopathy in fructose-streptozotocin induced type 2 diabetes (T2D) rat model was elucidated in this study. Type 2 diabetic rats were treated by oral administration of low (150 mg/kg b.w) and high (300 mg/kg b.w) doses of ferulic acid. Metformin was used as the antidiabetic drug. Rats were humanely euthanized after 5 weeks of treatment, and their blood and hearts were collected. Induction of T2D depleted the levels of reduced glutathione, glycogen, and HDL-cholesterol and the activities of superoxide dismutase, catalase, ENTPDase, and 5'nucleotidase. It simultaneously triggered increase in the levels of malondialdehyde, total cholesterol, triglyceride, LDL-cholesterol, creatinine kinase-MB as well as activities of acetylcholinesterase, angiotensin converting enzyme (ACE), ATPase, glucose-6-phopsphatase, fructose-1,6-bisphophatase, glycogen phosphorylase, and lipase. T2D induction further revealed an obvious degeneration of cardiac muscle morphology. However, treatment with ferulic acid markedly reversed the levels and activities of these biomarkers with concomitant improvement in myocardium structural morphology, which had favorable comparison with the standard drug, metformin. Additionally, T2D induction led to the depletion of 40%, 75%, and 33% of fatty acids, fatty esters, and steroids, respectively, with concomitant generation of eicosenoic acid, gamolenic acid, and vitamin E. Ferulic acid treatment restored eicosanoic acid, 2-hydroxyethyl ester, with concomitant generation of 6-octadecenoic acid, (Z)-, cis-11-eicosenoic acid, tridecanedioic acid, octadecanoic acid, 2-hydroxyethyl ester, ethyl 3-hydroxytridecanoate, dipalmitin, cholesterol isocaproate, cholest-5-ene, 3-(1-oxobuthoxy)-, cholesta-3,5-diene. These results suggest the cardioprotective potential of ferulic acid against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Veronica F Salau
- Department of Biochemistry, University of KwaZulu-Natal, Durban, South Africa.,Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | | | - Nontokozo Z Msomi
- Department of Biochemistry, University of KwaZulu-Natal, Durban, South Africa
| | - Olayemi K Ijomone
- Department of Anatomy, University of Medical Sciences, Ondo City, Nigeria
| | - Md Shahidul Islam
- Department of Biochemistry, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
20
|
Ushakov A, Ivanchenko V, Gagarina A. Heart Failure And Type 2 Diabetes Mellitus: Neurohumoral, Histological And Molecular Interconnections. Curr Cardiol Rev 2023; 19:e170622206132. [PMID: 35718961 PMCID: PMC10201898 DOI: 10.2174/1573403x18666220617121144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) is a global healthcare burden and a leading cause of morbidity and mortality worldwide. Type 2 diabetes mellitus (T2DM) appears to be one of the major risk factors that significantly worsen HF prognosis and increase the risk of fatal cardiovascular outcomes. Despite a great knowledge of pathophysiological mechanisms involved in HF development and progression, hospitalization rates in patients with HF and concomitant T2DM remain elevated. In this review, we discuss the complex interplay between systemic neurohumoral regulation and local cardiac mechanisms participating in myocardial remodeling and HF development in T2DM with special attention to cardiomyocyte energy metabolism, mitochondrial function and calcium metabolism, cardiomyocyte hypertrophy and death, extracellular matrix remodeling.
Collapse
Affiliation(s)
- A. Ushakov
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - V. Ivanchenko
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - A. Gagarina
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
21
|
Xi Y, Chen D, Dong Z, Zhang J, Lam H, He J, Du K, Chen C, Guo J, Xiao J. Multi-omics insights into potential mechanism of SGLT2 inhibitors cardiovascular benefit in diabetic cardiomyopathy. Front Cardiovasc Med 2022; 9:999254. [PMID: 36277768 PMCID: PMC9579694 DOI: 10.3389/fcvm.2022.999254] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background Metabolic and energy disorders are considered central to the etiology of diabetic cardiomyopathy (DCM). Sodium-glucose cotransporter-2 inhibitors (SGLT2i) can effectively reduce the risk of cardiovascular death and heart failure in patients with DCM. However, the underlying mechanism has not been elucidated. Methods We established a DCM rat model followed by treatment with empagliflozin (EMPA) for 12 weeks. Echocardiography, blood tests, histopathology, and transmission electron microscopy (TEM) were used to evaluate the phenotypic characteristics of the rats. The proteomics and metabolomics of the myocardium in the rat model were performed to identify the potential targets and signaling pathways associated with the cardiovascular benefit of SGLT2i. Results The diabetic rat showed pronounced DCM characterized by mitochondrial pleomorphic, impaired lipid metabolism, myocardial fibrosis, and associated diastolic and systolic functional impairments in the heart. To some extent, these changes were ameliorated after treatment with EMPA. A total of 43 proteins and 34 metabolites were identified as targets in the myocardium of diabetic rats treated with EMPA. The KEGG analysis showed that arachidonic acid is associated with the maximum number of related pathways and may be a potential target of EMPA treatment. Fatty acid (FA) metabolism was enhanced in diabetic hearts, and the perturbation of biosynthesis of unsaturated FAs and arachidonic acid metabolism was a potential enabler for the cardiovascular benefit of EMPA. Conclusion SGLT2i ameliorated lipid accumulation and mitochondrial damage in the myocardium of diabetic rats. The metabolomic and proteomic data revealed the potential targets and signaling pathways associated with the cardiovascular benefit of SGLT2i, which provides a valuable resource for the mechanism of SGLT2i.
Collapse
Affiliation(s)
- Yangbo Xi
- The First Clinical Medical College, Jinan University, Guangzhou, China,Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongping Chen
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China
| | - Zhihui Dong
- Central Laboratory, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China
| | - Jinhua Zhang
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Hingcheung Lam
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Jiading He
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Keyi Du
- The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Can Chen
- Department of Pathology, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China
| | - Jun Guo
- The First Clinical Medical College, Jinan University, Guangzhou, China,Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China,*Correspondence: Jun Guo,
| | - Jianmin Xiao
- The First Clinical Medical College, Jinan University, Guangzhou, China,Central Laboratory, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China,Department of Cardiology, Binhaiwan Central Hospital of Dongguan, The Dongguan Affiliated Hospital of Jinan University, Dongguan, China,Jianmin Xiao,
| |
Collapse
|
22
|
Purnama U, Castro-Guarda M, Sahoo OS, Carr CA. Modelling Diabetic Cardiomyopathy: Using Human Stem Cell-Derived Cardiomyocytes to Complement Animal Models. Metabolites 2022; 12:metabo12090832. [PMID: 36144236 PMCID: PMC9503602 DOI: 10.3390/metabo12090832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Diabetes is a global epidemic, with cardiovascular disease being the leading cause of death in diabetic patients. There is a pressing need for an in vitro model to aid understanding of the mechanisms driving diabetic heart disease, and to provide an accurate, reliable tool for drug testing. Human induced-pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have potential as a disease modelling tool. There are several factors that drive molecular changes inside cardiomyocytes contributing to diabetic cardiomyopathy, including hyperglycaemia, lipotoxicity and hyperinsulinemia. Here we discuss these factors and how they can be seen in animal models and utilised in cell culture to mimic the diabetic heart. The use of human iPSC-CMs will allow for a greater understanding of disease pathogenesis and open up new avenues for drug testing.
Collapse
Affiliation(s)
- Ujang Purnama
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Marcos Castro-Guarda
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713216, India
| | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: ; Tel.: +44-1865-282247
| |
Collapse
|
23
|
Mengstie MA, Abebe EC, Teklemariam AB, Mulu AT, Teshome AA, Zewde EA, Muche ZT, Azezew MT. Molecular and cellular mechanisms in diabetic heart failure: Potential therapeutic targets. Front Endocrinol (Lausanne) 2022; 13:947294. [PMID: 36120460 PMCID: PMC9478122 DOI: 10.3389/fendo.2022.947294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/12/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes Mellitus (DM) is a worldwide health issue that can lead to a variety of complications. DM is a serious metabolic disorder that causes long-term microvascular and macro-vascular complications, as well as the failure of various organ systems. Diabetes-related cardiovascular diseases (CVD) including heart failure cause significant morbidity and mortality worldwide. Concurrent hypertensive heart disease and/or coronary artery disease have been thought to be the causes of diabetic heart failure in DM patients. However, heart failure is extremely common in DM patients even in the absence of other risk factors such as coronary artery disease and hypertension. The occurrence of diabetes-induced heart failure has recently received a lot of attention. Understanding how diabetes increases the risk of heart failure and how it mediates major cellular and molecular alteration will aid in the development of therapeutics to prevent these changes. Hence, this review aimed to summarize the current knowledge and most recent findings in cellular and molecular mechanisms of diabetes-induced heart failure.
Collapse
Affiliation(s)
- Misganaw Asmamaw Mengstie
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Endeshaw Chekol Abebe
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Awgichew Behaile Teklemariam
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Anemut Tilahun Mulu
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Assefa Agegnehu Teshome
- Department of Anatomy, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Edgeit Abebe Zewde
- Department of Physiology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Zelalem Tilahun Muche
- Department of Physiology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Muluken Teshome Azezew
- Department of Physiology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
24
|
Yao PA, Wei KZ, Feng JH, Liu XN, Xu X, Cui HY, Zhang XC, Gao JP. Sodium houttuyfonate protects against cardiac injury by regulating cardiac energy metabolism in diabetic rats. Eur J Pharmacol 2022; 932:175236. [PMID: 36044971 DOI: 10.1016/j.ejphar.2022.175236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/03/2022]
Abstract
Diabetic cardiomyopathy is a diabetic complication with complicated pathophysiological changes and pathogenesis and difficult treatment. Sodium houttuyfonate is the adduct of sodium bisulfite and houttuynin, the main volatile component in Houttuynia cordata Thunb, possesses a variety of activities including multiple interventions on inhibiting ventricular remodeling. The study aims to explore effect of sodium houttuyfonate on diabetic myocardial injury and its underlying mechanisms. The diabetes model was established by intraperitoneal injection of streptozotocin at a dose of 85 mg/kg. By intragastric administration for 26 days, sodium houttuyfonate (50 and 100 mg/kg/d) reversed the abnormal serum levels of triglyceride, total cholesterol, low-density lipoprotein cholesterol and low-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio, improved the abnormal levels of serum aspartate aminotransferase and brain natriuretic peptide, reduced electrocardiogram P-R and QRS interval extension, accelerated the heart rate, decreased serum malondialdehyde content, up-regulated the myocardial energy metabolism including elevated the contents of ATP, ADP, total adenine nucleotides and phosphocreatine in myocardium, decreased AMP/ATP ratio, elevated myocardial Ca2+-Mg2+-ATPase activity, and down-regulated the mRNA expressions of AMP protein activation kinase α2 (AMPK-α2) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α). In a conclusion, these results suggest that sodium houttuyfonate can improve cardiac energy metabolism disorder caused by diabetes by increasing cardiac Ca2+-Mg2+-ATPase activity and regulating AMPK signaling pathway, and then attenuates cardiac injury caused by hyperglycemia. In addition, sodium houttuyfonate also has the effects of anti-oxidation and improving abnormal levels of blood lipid.
Collapse
Affiliation(s)
- Ping-An Yao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of Pharmacology, School of Pharmacy, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310053, China
| | - Ke-Zhao Wei
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia-Hua Feng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Ning Liu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Xu Xu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Hong-Yan Cui
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Chen Zhang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jian-Ping Gao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
25
|
Han R, Huang H, Xia W, Liu J, Luo H, Tang J, Xia Z. Perspectives for Forkhead box transcription factors in diabetic cardiomyopathy: Their therapeutic potential and possible effects of salvianolic acids. Front Cardiovasc Med 2022; 9:951597. [PMID: 36035917 PMCID: PMC9403618 DOI: 10.3389/fcvm.2022.951597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of morbidity and mortality in diabetic cardiovascular complications, which initially manifests as cardiac hypertrophy, myocardial fibrosis, dysfunctional remodeling, and diastolic dysfunction, followed by systolic dysfunction, and eventually end with acute heart failure. Molecular mechanisms underlying these pathological changes in diabetic hearts are complicated and multifactorial, including but not limited to insulin resistance, oxidative stress, lipotoxicity, cardiomyocytes apoptosis or autophagy, inflammatory response, and myocardial metabolic dysfunction. With the development of molecular biology technology, accumulating evidence illustrates that members of the class O of Forkhead box (FoxO) transcription factors are vital for maintaining cardiomyocyte metabolism and cell survival, and the functions of the FoxO family proteins can be modulated by a wide variety of post-translational modifications including phosphorylation, acetylation, ubiquitination, arginine methylation, and O-glycosylation. In this review, we highlight and summarize the most recent advances in two members of the FoxO family (predominately FoxO1 and FoxO3a) that are abundantly expressed in cardiac tissue and whose levels of gene and protein expressions change as DCM progresses, with the goal of providing valuable insights into the pathogenesis of diabetic cardiovascular complications and discussing their therapeutic potential and possible effects of salvianolic acids, a natural product.
Collapse
Affiliation(s)
- Ronghui Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hemeng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The Univerisity of Hong Kong, Hong Kong, China
- *Correspondence: Weiyi Xia,
| | - Jingjin Liu
- Department of Cardiology, Shenzhen People’s Hospital and The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Hui Luo
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang, China
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, China
- Zhengyuan Xia,
| |
Collapse
|
26
|
García-Vega D, González-Juanatey JR, Eiras S. Diabesity in Elderly Cardiovascular Disease Patients: Mechanisms and Regulators. Int J Mol Sci 2022; 23:7886. [PMID: 35887234 PMCID: PMC9318065 DOI: 10.3390/ijms23147886] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/04/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the world. In 2019, 550 million people were suffering from CVD and 18 million of them died as a result. Most of them had associated risk factors such as high fasting glucose, which caused 134 million deaths, and obesity, which accounted for 5.02 million deaths. Diabesity, a combination of type 2 diabetes and obesity, contributes to cardiac, metabolic, inflammation and neurohumoral changes that determine cardiac dysfunction (diabesity-related cardiomyopathy). Epicardial adipose tissue (EAT) is distributed around the myocardium, promoting myocardial inflammation and fibrosis, and is associated with an increased risk of heart failure, particularly with preserved systolic function, atrial fibrillation and coronary atherosclerosis. In fact, several hypoglycaemic drugs have demonstrated a volume reduction of EAT and effects on its metabolic and inflammation profile. However, it is necessary to improve knowledge of the diabesity pathophysiologic mechanisms involved in the development and progression of cardiovascular diseases for comprehensive patient management including drugs to optimize glucometabolic control. This review presents the mechanisms of diabesity associated with cardiovascular disease and their therapeutic implications.
Collapse
Affiliation(s)
- David García-Vega
- Cardiology and Intensive Cardiac Care Department, University Hospital, 15706 Santiago de Compostela, Spain;
- Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cardiology and Intensive Cardiac Care Department, University Hospital, 15706 Santiago de Compostela, Spain;
- Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain
- CIBERCV, 28029 Madrid, Spain
| | - Sonia Eiras
- CIBERCV, 28029 Madrid, Spain
- Translational Cardiology Group (Laboratory 6), Health Research Institute, 15706 Santiago de Compostela, Spain
| |
Collapse
|
27
|
Metabolic Determinants in Cardiomyocyte Function and Heart Regenerative Strategies. Metabolites 2022; 12:metabo12060500. [PMID: 35736435 PMCID: PMC9227827 DOI: 10.3390/metabo12060500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Heart disease is the leading cause of mortality in developed countries. The associated pathology is characterized by a loss of cardiomyocytes that leads, eventually, to heart failure. In this context, several cardiac regenerative strategies have been developed, but they still lack clinical effectiveness. The mammalian neonatal heart is capable of substantial regeneration following injury, but this capacity is lost at postnatal stages when cardiomyocytes become terminally differentiated and transit to the fetal metabolic switch. Cardiomyocytes are metabolically versatile cells capable of using an array of fuel sources, and the metabolism of cardiomyocytes suffers extended reprogramming after injury. Apart from energetic sources, metabolites are emerging regulators of epigenetic programs driving cell pluripotency and differentiation. Thus, understanding the metabolic determinants that regulate cardiomyocyte maturation and function is key for unlocking future metabolic interventions for cardiac regeneration. In this review, we will discuss the emerging role of metabolism and nutrient signaling in cardiomyocyte function and repair, as well as whether exploiting this axis could potentiate current cellular regenerative strategies for the mammalian heart.
Collapse
|
28
|
Mitochondria-Mediated Cardiovascular Benefits of Sodium-Glucose Co-Transporter 2 Inhibitors. Int J Mol Sci 2022; 23:ijms23105371. [PMID: 35628174 PMCID: PMC9140946 DOI: 10.3390/ijms23105371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
Several recent cardiovascular trials of SGLT 2 (sodium-glucose cotransporter 2) inhibitors revealed that they could reduce adverse cardiovascular events in patients with T2DM (type 2 diabetes mellitus). However, the exact molecular mechanism underlying the beneficial effects that SGLT2 inhibitors have on the cardiovascular system is still unknown. In this review, we focus on the molecular mechanisms of the mitochondria-mediated beneficial effects of SGLT2 inhibitors on the cardiovascular system. The application of SGLT2 inhibitors ameliorates mitochondrial dysfunction, dynamics, bioenergetics, and ion homeostasis and reduces the production of mitochondrial reactive oxygen species, which results in cardioprotective effects. Herein, we present a comprehensive overview of the impact of SGLT2 inhibitors on mitochondria and highlight the potential application of these medications to treat both T2DM and cardiovascular diseases.
Collapse
|
29
|
Wei Z, Jing Z, Pinfang K, Chao S, Shaohuan Q. Quercetin Inhibits Pyroptosis in Diabetic Cardiomyopathy through the Nrf2 Pathway. J Diabetes Res 2022; 2022:9723632. [PMID: 36624860 PMCID: PMC9825227 DOI: 10.1155/2022/9723632] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/02/2023] Open
Abstract
The present study investigated whether quercetin promotes the nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) to inhibit pyroptosis progression and ameliorate diabetic cardiomyopathy. We evaluated the protective effects of quercetin against diabetic cardiomyopathy by analyzing the expression of pyroptosis pathway proteins, myocardial cell apoptosis rate, degree of myocardial fibrosis, and serum inflammatory indices in the hearts of model rats with diabetes. We evaluated the expression of Nrf2 in the nucleus of cardiomyocytes and H9C2 cells to clarify the role of quercetin in promoting the nuclear translocation of Nrf2. In addition, we coincubated cardiomyocytes with the Nrf2 inhibitor ML385 to confirm that quercetin inhibits the diabetes-induced cardiomyocyte pyroptosis via the Nrf2 pathway. We found that quercetin promoted the nuclear translocation of Nrf2 in cardiac cells of diabetic rats, increased the expression of the antioxidant proteins HO-1, GCLC, and SOD, reduced the accumulation of ROS and the degree of cardiomyocyte apoptosis, and alleviated diabetes-induced cardiac fibrosis. The therapeutic effects of quercetin were further validated in H9C2 cardiomyocytes. Interestingly, ML385 prevented the beneficial effects of quercetin on diabetic cardiomyopathy, further indicating that the quercetin-mediated inhibition of pyroptosis requires the participation of the Nrf2 pathway. In conclusion, quercetin promoted the nuclear translocation of Nrf2, increased the expression of antioxidant factors in cells, and inhibited the progression of cell pyroptosis, thereby alleviating diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhang Wei
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Zhou Jing
- Department of Physiology of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Kang Pinfang
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
- Department of Physiology of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Shi Chao
- Department of Cardiac Surgery of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| | - Qian Shaohuan
- Department of Cardiovascular Medicine of The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui, China 233000
| |
Collapse
|