151
|
Liu F, Zhao L, Wu T, Yu W, Li J, Wang W, Huang C, Diao Z, Xu Y. Targeting autophagy with natural products as a potential therapeutic approach for diabetic microangiopathy. Front Pharmacol 2024; 15:1364616. [PMID: 38659578 PMCID: PMC11039818 DOI: 10.3389/fphar.2024.1364616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
As the quality of life improves, the incidence of diabetes mellitus and its microvascular complications (DMC) continues to increase, posing a threat to people's health and wellbeing. Given the limitations of existing treatment, there is an urgent need for novel approaches to prevent and treat DMC. Autophagy, a pivotal mechanism governing metabolic regulation in organisms, facilitates the removal of dysfunctional proteins and organelles, thereby sustaining cellular homeostasis and energy generation. Anomalous states in pancreatic β-cells, podocytes, Müller cells, cardiomyocytes, and Schwann cells in DMC are closely linked to autophagic dysregulation. Natural products have the property of being multi-targeted and can affect autophagy and hence DMC progression in terms of nutrient perception, oxidative stress, endoplasmic reticulum stress, inflammation, and apoptosis. This review consolidates recent advancements in understanding DMC pathogenesis via autophagy and proposes novel perspectives on treating DMC by either stimulating or inhibiting autophagy using natural products.
Collapse
Affiliation(s)
- Fengzhao Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijuan Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Yu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jixin Li
- Xi yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenru Wang
- Xi yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chengcheng Huang
- Department of Endocrinology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Zhihao Diao
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
152
|
Li M, Liu L, Zhang C, Deng L, Zhong Y, Liao B, Li X, Wan Y, Feng J. The latest emerging drugs for the treatment of diabetic cardiomyopathy. Expert Opin Pharmacother 2024; 25:641-654. [PMID: 38660817 DOI: 10.1080/14656566.2024.2347468] [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: 02/05/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Diabetic cardiomyopathy (DCM) is a serious complication of diabetes mellitus involving multiple pathophysiologic mechanisms. In addition to hypoglycemic agents commonly used in diabetes, metabolism-related drugs, natural plant extracts, melatonin, exosomes, and rennin-angiotensin-aldosterone system are cardioprotective in DCM. However, there is a lack of systematic summarization of drugs for DCM. AREAS COVERED In this review, the authors systematically summarize the most recent drugs used for the treatment of DCM and discusses them from the perspective of DCM pathophysiological mechanisms. EXPERT OPINION We discuss DCM drugs from the perspective of the pathophysiological mechanisms of DCM, mainly including inflammation and metabolism. As a disease with multiple pathophysiological mechanisms, the combination of drugs may be more advantageous, and we have discussed some of the current studies on the combination of drugs.
Collapse
Affiliation(s)
- Minghao Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Bin Liao
- Department of Cardiovascular Surgery, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiuying Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University; Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Ying Wan
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University; Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
153
|
Chen X, Qian J, Liang S, Qian J, Luo W, Shi Y, Zhu H, Hu X, Wu G, Li X, Liang G. Hyperglycemia activates FGFR1 via TLR4/c-Src pathway to induce inflammatory cardiomyopathy in diabetes. Acta Pharm Sin B 2024; 14:1693-1710. [PMID: 38572108 PMCID: PMC10985127 DOI: 10.1016/j.apsb.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 04/05/2024] Open
Abstract
Protein tyrosine kinases (RTKs) modulate a wide range of pathophysiological events in several non-malignant disorders, including diabetic complications. To find new targets driving the development of diabetic cardiomyopathy (DCM), we profiled an RTKs phosphorylation array in diabetic mouse hearts and identified increased phosphorylated fibroblast growth factor receptor 1 (p-FGFR1) levels in cardiomyocytes, indicating that FGFR1 may contribute to the pathogenesis of DCM. Using primary cardiomyocytes and H9C2 cell lines, we discovered that high-concentration glucose (HG) transactivates FGFR1 kinase domain through toll-like receptor 4 (TLR4) and c-Src, independent of FGF ligands. Knocking down the levels of either TLR4 or c-Src prevents HG-activated FGFR1 in cardiomyocytes. RNA-sequencing analysis indicates that the elevated FGFR1 activity induces pro-inflammatory responses via MAPKs-NFκB signaling pathway in HG-challenged cardiomyocytes, which further results in fibrosis and hypertrophy. We then generated cardiomyocyte-specific FGFR1 knockout mice and showed that a lack of FGFR1 in cardiomyocytes prevents diabetes-induced cardiac inflammation and preserves cardiac function in mice. Pharmacological inhibition of FGFR1 by a selective inhibitor, AZD4547, also prevents cardiac inflammation, fibrosis, and dysfunction in both type 1 and type 2 diabetic mice. These studies have identified FGFR1 as a new player in driving DCM and support further testing of FGFR1 inhibitors for possible cardioprotective benefits.
Collapse
Affiliation(s)
- Xiong Chen
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Wound Repair, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Shiqi Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yujuan Shi
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Hong Zhu
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiang Hu
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Wound Repair, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Guang Liang
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| |
Collapse
|
154
|
Radzioch E, Dąbek B, Balcerczyk-Lis M, Frąk W, Fularski P, Młynarska E, Rysz J, Franczyk B. Diabetic Cardiomyopathy-From Basics through Diagnosis to Treatment. Biomedicines 2024; 12:765. [PMID: 38672121 PMCID: PMC11048005 DOI: 10.3390/biomedicines12040765] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is the development of myocardial dysfunction in patients with diabetes despite the absence of comorbidities such as hypertension, atherosclerosis or valvular defect. The cardiovascular complications of poorly controlled diabetes are very well illustrated by the U.K. Prospective Diabetes Study (UKPDS), which showed a clear association between increasing levels of glycated hemoglobin and the development of heart failure (HF). The incidence of HF in patients with diabetes is projected to increase significantly, which is why its proper diagnosis and treatment is so important. Providing appropriate therapy focusing on antidiabetic and hypolipemic treatment with the consideration of pharmacotherapy for heart failure reduces the risk of CMD and reduces the incidence of cardiovascular complications. Health-promoting changes made by patients such as a low-carbohydrate diet, regular exercise and weight reduction also appear to be important in achieving appropriate outcomes. New hope for the development of therapies for DCM is offered by novel methods using stem cells and miRNA, which, however, require more thorough research to confirm their efficacy.
Collapse
Affiliation(s)
- Ewa Radzioch
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Marta Balcerczyk-Lis
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Frąk
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Piotr Fularski
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
155
|
Wang S, Li J, Zhao Y. Construction and analysis of a network of exercise-induced mitochondria-related non-coding RNA in the regulation of diabetic cardiomyopathy. PLoS One 2024; 19:e0297848. [PMID: 38547044 PMCID: PMC10977711 DOI: 10.1371/journal.pone.0297848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/09/2024] [Indexed: 04/02/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a major factor in the development of heart failure. Mitochondria play a crucial role in regulating insulin resistance, oxidative stress, and inflammation, which affect the progression of DCM. Regular exercise can induce altered non-coding RNA (ncRNA) expression, which subsequently affects gene expression and protein function. The mechanism of exercise-induced mitochondrial-related non-coding RNA network in the regulation of DCM remains unclear. This study seeks to construct an innovative exercise-induced mitochondrial-related ncRNA network. Bioinformatic analysis of RNA sequencing data from an exercise rat model identified 144 differentially expressed long non-coding RNA (lncRNA) with cutoff criteria of p< 0.05 and fold change ≥1.0. GSE6880 and GSE4745 were the differentially expressed mRNAs from the left ventricle of DCM rat that downloaded from the GEO database. Combined with the differentially expressed mRNA and MitoCarta 3.0 dataset, the mitochondrial located gene Pdk4 was identified as a target gene. The miRNA prediction analysis using miRanda and TargetScan confirmed that 5 miRNAs have potential to interact with the 144 lncRNA. The novel lncRNA-miRNA-Pdk4 network was constructed for the first time. According to the functional protein association network, the newly created exercise-induced ncRNA network may serve as a promising diagnostic marker and therapeutic target, providing a fresh perspective to understand the molecular mechanism of different exercise types for the prevention and treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shuo Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, China
| | - Jiacong Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, China
| | - Yungang Zhao
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
156
|
Juttla PK, Chege BM, Mwangi PW, Bukachi F. Dapagliflozin Pretreatment Prevents Cardiac Electrophysiological Changes in a Diet and Streptozotocin Induction of Type 2 Diabetes in Rats: A Potential New First-Line? J Exp Pharmacol 2024; 16:123-133. [PMID: 38525051 PMCID: PMC10961018 DOI: 10.2147/jep.s443169] [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: 10/15/2023] [Accepted: 02/22/2024] [Indexed: 03/26/2024] Open
Abstract
Purpose Dapagliflozin exerts cardioprotective effects in Type 2 Diabetes Mellitus (T2DM). However, whether these effects prevent electrocardiographic changes associated with T2DM altogether remain unknown. Our aim was to investigate the prophylactic effect of dapagliflozin pretreatment on the rat ECG using a high-fat, high-fructose (HFHf) diet and a low dose streptozotocin (STZ) model of T2DM. Methods Twenty-five (25) rats were randomized into five (5) groups: normal control receiving a normal diet while the other groups received an 8-week HFHf and 40mg/kg STZ on day 42, and either: saline for the diabetic control (1 mg/kg/d), low dose (1.0 mg/kg/d) and high dose dapagliflozin (1.6 mg/kg/d), or metformin (250 mg/kg/d). Oral glucose tolerance (OGT), electrocardiograms (ECGs), paracardial adipose mass, and left ventricular fibrosis were determined. Data were analyzed using GraphPad version 9.0.0.121, with the level of significance at p < 0.05. Results Compared to the diabetic control group, a high dose of dapagliflozin preserved the OGT (p = 0.0001), QRS-duration (p = 0.0263), QT-interval (p = 0.0399), and QTc intervals (p = 0.0463). Furthermore, the high dose dapagliflozin group had the lowest paracardial adipose mass (p = 0.0104) and fibrotic area (p = 0.0001). In contrast, while metformin showed favorable effects on OGT (p = 0.0025), paracardial adiposity (p = 0.0153) and ventricular fibrosis (p = 0.0291), it did not demonstrate significant antiarrhythmic effects. Conclusion Pretreatment with higher doses of Dapagliflozin exhibits prophylactic cardioprotective characteristics against diabetic cardiomyopathy that include antifibrotic and antiarrhythmic qualities. This suggests that higher doses of dapagliflozin could be a more effective initial therapeutic option in T2DM.
Collapse
Affiliation(s)
| | | | | | - Frederick Bukachi
- Department of Medical Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
157
|
Ma XM, Geng K, Wang P, Jiang Z, Law BYK, Xu Y. MCT4-dependent lactate transport: a novel mechanism for cardiac energy metabolism injury and inflammation in type 2 diabetes mellitus. Cardiovasc Diabetol 2024; 23:96. [PMID: 38486199 PMCID: PMC10941417 DOI: 10.1186/s12933-024-02178-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 02/24/2024] [Indexed: 03/17/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a major contributor to mortality in diabetic patients, characterized by a multifaceted pathogenesis and limited therapeutic options. While lactate, a byproduct of glycolysis, is known to be significantly elevated in type 2 diabetes, its specific role in DCM remains uncertain. This study reveals an abnormal upregulation of monocarboxylate transporter 4 (MCT4) on the plasma membrane of cardiomyocytes in type 2 diabetes, leading to excessive lactate efflux from these cells. The disruption in lactate transport homeostasis perturbs the intracellular lactate-pyruvate balance in cardiomyocytes, resulting in oxidative stress and inflammatory responses that exacerbate myocardial damage. Additionally, our findings suggest increased lactate efflux augments histone H4K12 lactylation in macrophages, facilitating inflammatory infiltration within the microenvironment. In vivo experiments have demonstrated that inhibiting MCT4 effectively alleviates myocardial oxidative stress and pathological damage, reduces inflammatory macrophage infiltration, and enhances cardiac function in type 2 diabetic mice. Furthermore, a clinical prediction model has been established, demonstrating a notable association between peripheral blood lactate levels and diastolic dysfunction in individuals with type 2 diabetes. This underscores the potential of lactate as a prognostic biomarker for DCM. Ultimately, our findings highlight the pivotal involvement of MCT4 in the dysregulation of cardiac energy metabolism and macrophage-mediated inflammation in type 2 diabetes. These insights offer novel perspectives on the pathogenesis of DCM and pave the way for the development of targeted therapeutic strategies against this debilitating condition.
Collapse
Affiliation(s)
- Xiu Mei Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, Sichuan, People's Republic of China
| | - Kang Geng
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, Sichuan, People's Republic of China
| | - Peng Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China
| | - Zongzhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, Sichuan, People's Republic of China
| | - Betty Yuen-Kwan Law
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China.
| | - Yong Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, People's Republic of China.
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
158
|
Zhu J, Bao Z, Hu Z, Wu S, Tian C, Zhou Y, Ding Z, Tan X. Myricetin alleviates diabetic cardiomyopathy by regulating gut microbiota and their metabolites. Nutr Diabetes 2024; 14:10. [PMID: 38472186 PMCID: PMC10933338 DOI: 10.1038/s41387-024-00268-4] [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: 09/16/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The gut microbiota is involved in the pathogenesis of diabetic cardiomyopathy (DCM). Myricetin protects cardiac function in DCM. However, the low bioavailability of myricetin fails to explain its pharmacological mechanisms thoroughly. Research has shown that myricetin has a positive effect on the gut microbiota. We hypothesize that myricetin improves the development of DCM via regulating gut microbiota. METHODS DCM mice were induced with streptozotocin and fed a high-fat diet, and then treated with myricetin by gavage and high-fat diet for 16 weeks. Indexes related to gut microbiota composition, cardiac structure, cardiac function, intestinal barrier function, and inflammation were detected. Moreover, the gut contents were transplanted to DCM mice, and the effect of fecal microbiota transplantation (FMT) on DCM mice was assessed. RESULTS Myricetin could improve cardiac function in DCM mice by decreasing cardiomyocyte hypertrophy and interstitial fibrosis. The composition of gut microbiota, especially for short-chain fatty acid-producing bacteria involving Roseburia, Faecalibaculum, and Bifidobacterium, was more abundant by myricetin treatment in DCM mice. Myricetin increased occludin expression and the number of goblet cells in DCM mice. Compared with DCM mice unfed with gut content, the cardiac function, number of goblet cells, and expression of occludin in DCM mice fed by gut contents were elevated, while cardiomyocyte hypertrophy and TLR4/MyD88 pathway-related proteins were decreased. CONCLUSIONS Myricetin can prevent DCM development by increasing the abundance of beneficial gut microbiota and restoring the gut barrier function.
Collapse
Affiliation(s)
- Jinxiu Zhu
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
- Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), 518172, Shenzhen, Guangdong, China
| | - Zhijun Bao
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Zuoqi Hu
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Shenglin Wu
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Cuihong Tian
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Yueran Zhou
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Zipeng Ding
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Xuerui Tan
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China.
- Clinical Research Center, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China.
| |
Collapse
|
159
|
Xie SY, Liu SQ, Zhang T, Shi WK, Xing Y, Fang WX, Zhang M, Chen MY, Xu SC, Fan MQ, Li LL, Zhang H, Zhao N, Zeng ZX, Chen S, Zeng XF, Deng W, Tang QZ. USP28 Serves as a Key Suppressor of Mitochondrial Morphofunctional Defects and Cardiac Dysfunction in the Diabetic Heart. Circulation 2024; 149:684-706. [PMID: 37994595 DOI: 10.1161/circulationaha.123.065603] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND The majority of people with diabetes are susceptible to cardiac dysfunction and heart failure, and conventional drug therapy cannot correct diabetic cardiomyopathy progression. Herein, we assessed the potential role and therapeutic value of USP28 (ubiquitin-specific protease 28) on the metabolic vulnerability of diabetic cardiomyopathy. METHODS The type 2 diabetes mouse model was established using db/db leptin receptor-deficient mice and high-fat diet/streptozotocin-induced mice. Cardiac-specific knockout of USP28 in the db/db background mice was generated by crossbreeding db/m and Myh6-Cre+/USP28fl/fl mice. Recombinant adeno-associated virus serotype 9 carrying USP28 under cardiac troponin T promoter was injected into db/db mice. High glucose plus palmitic acid-incubated neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes were used to imitate diabetic cardiomyopathy in vitro. The molecular mechanism was explored through RNA sequencing, immunoprecipitation and mass spectrometry analysis, protein pull-down, chromatin immunoprecipitation sequencing, and chromatin immunoprecipitation assay. RESULTS Microarray profiling of the UPS (ubiquitin-proteasome system) on the basis of db/db mouse hearts and diabetic patients' hearts demonstrated that the diabetic ventricle presented a significant reduction in USP28 expression. Diabetic Myh6-Cre+/USP28fl/fl mice exhibited more severe progressive cardiac dysfunction, lipid accumulation, and mitochondrial disarrangement, compared with their controls. On the other hand, USP28 overexpression improved systolic and diastolic dysfunction and ameliorated cardiac hypertrophy and fibrosis in the diabetic heart. Adeno-associated virus serotype 9-USP28 diabetic mice also exhibited less lipid storage, reduced reactive oxygen species formation, and mitochondrial impairment in heart tissues than adeno-associated virus serotype 9-null diabetic mice. As a result, USP28 overexpression attenuated cardiac remodeling and dysfunction, lipid accumulation, and mitochondrial impairment in high-fat diet/streptozotocin-induced type 2 diabetes mice. These results were also confirmed in neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes. RNA sequencing, immunoprecipitation and mass spectrometry analysis, chromatin immunoprecipitation assays, chromatin immunoprecipitation sequencing, and protein pull-down assay mechanistically revealed that USP28 directly interacted with PPARα (peroxisome proliferator-activated receptor α), deubiquitinating and stabilizing PPARα (Lys152) to promote Mfn2 (mitofusin 2) transcription, thereby impeding mitochondrial morphofunctional defects. However, such cardioprotective benefits of USP28 were largely abrogated in db/db mice with PPARα deletion and conditional loss-of-function of Mfn2. CONCLUSIONS Our findings provide a USP28-modulated mitochondria homeostasis mechanism that involves the PPARα-Mfn2 axis in diabetic hearts, suggesting that USP28 activation or adeno-associated virus therapy targeting USP28 represents a potential therapeutic strategy for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Sai-Yang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Shi-Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Wen-Ke Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Yun Xing
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Wen-Xi Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Min Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Meng-Ya Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Si-Chi Xu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China (S.-c.X.)
| | - Meng-Qi Fan
- College of Life Sciences, Wuhan University, P.R. China (M.-q.F.)
| | - Lan-Lan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Zhao-Xiang Zeng
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai Jiaotong University, P.R. China (Z.-x.Z)
- Department of Cardiac Surgery, Changhai Hospital, Navy Medical University, Shanghai, P.R. China (Z.-x.Z)
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, P.R. China (S.C., X.-f.Z.)
| | - Xiao-Feng Zeng
- Cardiovascular Research Institute of Wuhan University, P.R. China (S.C., X.-f.Z.)
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| |
Collapse
|
160
|
Guo X, Chen K, Ji L, Wang S, Ye X, Xu L, Feng L. Ultrasound-targeted microbubble technology facilitates SAHH gene delivery to treat diabetic cardiomyopathy by activating AMPK pathway. iScience 2024; 27:108852. [PMID: 38303706 PMCID: PMC10831940 DOI: 10.1016/j.isci.2024.108852] [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: 09/12/2023] [Revised: 11/13/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a cardiovascular complication with no known cure. In this study, we evaluated the combination of ultrasound-targeted microbubble destruction (UTMD) and cationic microbubbles (CMBs) for cardiac S-adenosyl homocysteine hydrolase (SAHH) gene transfection as potential DCM therapy. Models of high glucose/fat (HG/HF)-induced H9C2 cells and streptozotocin-induced DCM rats were established. Ultrasound-mediated SAHH delivery using CMBs was a safe and noninvasive approach for spatially localized drug administration both in vitro and in vivo. Notably, SAHH overexpression increased cell viability and antioxidative stress and inhibited apoptosis of HG/HF-induced H9C2 cells. Likewise, UTMD-mediated SAHH delivery attenuated apoptosis, oxidative stress, cardiac fibrosis, and myocardial dysfunction in DCM rats. Activation of the AMPK/FOXO3/SIRT3 signaling pathway may be a key mechanism mediating the role of SAHH in regulating myocardial injury. Thus, UTMD-mediated SAHH transfection may be an important advancement in cardiac gene therapy for restoring ventricular function after DCM.
Collapse
Affiliation(s)
- Xiaohui Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Harbin Medical University, Harbin 150081, P.R. China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150086, P.R. China
| | - Kegong Chen
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, P.R. China
| | - Lin Ji
- Department of Orthopedics, The First Hospital of Harbin, Harbin 150010, P.R. China
| | - Shanjie Wang
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150086, P.R. China
| | - Xiangmei Ye
- Department of Clinical Laboratory, First Affiliated Hospital of Harbin Medical University, Harbin 150081, P.R. China
| | - Liang Xu
- Department of Cardiology, The Second Hospital of Harbin, Harbin 150056, P.R. China
| | - Leiguang Feng
- Department of Clinical Laboratory, First Affiliated Hospital of Harbin Medical University, Harbin 150081, P.R. China
| |
Collapse
|
161
|
Târtea G, Popa-Wagner A, Sfredel V, Mitran SI, Dan AO, Țucă AM, Preda AN, Raicea V, Țieranu E, Cozma D, Vătășescu R. Chitosan Versus Dapagliflozin in a Diabetic Cardiomyopathy Mouse Model. Int J Mol Sci 2024; 25:2118. [PMID: 38396795 PMCID: PMC10888683 DOI: 10.3390/ijms25042118] [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: 01/09/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Diabetes mellitus is a metabolic disorder with global economic implications that can lead to complications such as diabetic cardiomyopathy. The aim of this study was to compare the effects of chitosan versus dapagliflozin in mouse diabetic cardiomyopathy. We used 32 C57Bl/6 male mice aged between 8 and 10 weeks, which were randomly divided into Control-without diabetes mellitus (DM), type 1 DM (T1DM), T1DM + Chitosan, and T1DM + Dapapgliflozin groups. We induced diabetes with streptozotocin and treated the animals for 12 weeks. The analysis showed a reduction in intramyocardial fibrosis in the T1DM + Dapapgliflozin compared to T1DM animals. In T1DM + CHIT, a reduction in intramyocardial fibrosis was observed although, accordingly, there was also no significant decrease in blood glucose. The level of oxidative stress was reduced in the groups of treated animals compared to T1DM. All these observed changes in the structure and function of hearts were highlighted in the echocardiographic examination. In the treated groups, there was delayed appearance of left ventricular (LV) hypertrophy, a slight decrease in the ejection fraction of the LV, and an improved diastolic profile. The results demonstrate that chitosan has promising effects on diabetic cardiomyopathy that are comparable to the beneficial effects of dapagliflozin.
Collapse
Affiliation(s)
- Georgică Târtea
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (G.T.); (V.S.); (S.I.M.); (A.O.D.); (A.-M.Ț.)
| | - Aurel Popa-Wagner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Veronica Sfredel
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (G.T.); (V.S.); (S.I.M.); (A.O.D.); (A.-M.Ț.)
| | - Smaranda Ioana Mitran
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (G.T.); (V.S.); (S.I.M.); (A.O.D.); (A.-M.Ț.)
| | - Alexandra Oltea Dan
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (G.T.); (V.S.); (S.I.M.); (A.O.D.); (A.-M.Ț.)
| | - Anca-Maria Țucă
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (G.T.); (V.S.); (S.I.M.); (A.O.D.); (A.-M.Ț.)
| | - Alexandra Nicoleta Preda
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (G.T.); (V.S.); (S.I.M.); (A.O.D.); (A.-M.Ț.)
| | - Victor Raicea
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (V.R.); (E.Ț.)
| | - Eugen Țieranu
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (V.R.); (E.Ț.)
| | - Dragoș Cozma
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Radu Vătășescu
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
162
|
Januszewski AS, Blake R, Zhang M, Ma B, Anand S, Pinkert CA, Kelly DJ, Jenkins AJ, Trounce IA. Increased Diabetes Complications in a Mouse Model of Oxidative Stress Due to 'Mismatched' Mitochondrial DNA. Antioxidants (Basel) 2024; 13:187. [PMID: 38397785 PMCID: PMC10886269 DOI: 10.3390/antiox13020187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Associations between chronic diabetes complications and mitochondrial dysfunction represent a subject of major importance, given the diabetes pandemic and high personal and socioeconomic costs of diabetes and its complications. Modelling diabetes complications in inbred laboratory animals is challenging due to incomplete recapitulation of human features, but offer mechanistic insights and preclinical testing. As mitochondrial-based oxidative stress is implicated in human diabetic complications, herein we evaluate diabetes in a unique mouse model that harbors a mitochondrial DNA from a divergent mouse species (the 'xenomitochondrial mouse'), which has mild mitochondrial dysfunction and increased oxidative stress. We use the streptozotocin-induced diabetes model with insulin supplementation, with 20-weeks diabetes. We compare C57BL/6 mice and the 'xenomitochondrial' mouse, with measures of heart and kidney function, histology, and skin oxidative stress markers. Compared to C57BL/6 mice, the xenomitochondrial mouse has increased diabetic heart and kidney damage, with cardiac dysfunction, and increased cardiac and renal fibrosis. Our results show that mitochondrial oxidative stress consequent to divergent mtDNA can worsen diabetes complications. This has implications for novel therapeutics to counter diabetes complications, and for genetic studies of risk, as mtDNA genotypes may contribute to clinical outcomes.
Collapse
Affiliation(s)
- Andrzej S. Januszewski
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; (A.S.J.); (M.Z.); (B.M.); (D.J.K.); (A.J.J.)
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rachel Blake
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia; (R.B.); (S.A.)
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Michael Zhang
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; (A.S.J.); (M.Z.); (B.M.); (D.J.K.); (A.J.J.)
| | - Ben Ma
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; (A.S.J.); (M.Z.); (B.M.); (D.J.K.); (A.J.J.)
| | - Sushma Anand
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia; (R.B.); (S.A.)
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA;
| | - Darren J. Kelly
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; (A.S.J.); (M.Z.); (B.M.); (D.J.K.); (A.J.J.)
| | - Alicia J. Jenkins
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; (A.S.J.); (M.Z.); (B.M.); (D.J.K.); (A.J.J.)
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Ian A. Trounce
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; (A.S.J.); (M.Z.); (B.M.); (D.J.K.); (A.J.J.)
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC 3002, Australia; (R.B.); (S.A.)
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
163
|
Ma CY, Luo YM, Zhang TY, Hao YD, Xie XQ, Liu XW, Ren XL, He XL, Han YM, Deng KJ, Yan D, Yang H, Tang H, Lin H. Predicting coronary heart disease in Chinese diabetics using machine learning. Comput Biol Med 2024; 169:107952. [PMID: 38194779 DOI: 10.1016/j.compbiomed.2024.107952] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/15/2023] [Accepted: 01/01/2024] [Indexed: 01/11/2024]
Abstract
Diabetes, a common chronic disease worldwide, can induce vascular complications, such as coronary heart disease (CHD), which is also one of the main causes of human death. It is of great significance to study the factors of diabetic patients complicated with CHD for understanding the occurrence of diabetes/CHD comorbidity. In this study, by analyzing the risk of CHD in more than 300,000 diabetes patients in southwest China, an artificial intelligence (AI) model was proposed to predict the risk of diabetes/CHD comorbidity. Firstly, we statistically analyzed the distribution of four types of features (basic demographic information, laboratory indicators, medical examination, and questionnaire) in comorbidities, and evaluated the predictive performance of three traditional machine learning methods (eXtreme Gradient Boosting, Random Forest, and Logistic regression). In addition, we have identified nine important features, including age, WHtR, BMI, stroke, smoking, chronic lung disease, drinking and MSP. Finally, the model produced an area under the receiver operating characteristic curve (AUC) of 0.701 on the test samples. These findings can provide personalized guidance for early CHD warning for diabetic populations.
Collapse
Affiliation(s)
- Cai-Yi Ma
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Ya-Mei Luo
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, 646000, China
| | - Tian-Yu Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yu-Duo Hao
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xue-Qin Xie
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xiao-Wei Liu
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xiao-Lei Ren
- Sichuan Chuanjiang Science and Technology Research Institute Co., Ltd, Luzhou, 646000, China
| | - Xiao-Lin He
- Sichuan Chuanjiang Science and Technology Research Institute Co., Ltd, Luzhou, 646000, China
| | - Yu-Mei Han
- Beijing Physical Examination Center, Beijing, China
| | - Ke-Jun Deng
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Hui Yang
- School of Computer Science, Chengdu University of Information Technology, Chengdu, 610225, China.
| | - Hua Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, 646000, China.
| | - Hao Lin
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
164
|
Cai L, Tan Y, Holland B, Wintergerst K. Diabetic Cardiomyopathy and Cell Death: Focus on Metal-Mediated Cell Death. Cardiovasc Toxicol 2024; 24:71-84. [PMID: 38321349 PMCID: PMC11517829 DOI: 10.1007/s12012-024-09836-7] [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: 06/29/2023] [Accepted: 01/27/2024] [Indexed: 02/08/2024]
Abstract
Cardiac myocyte death is an essential initiator of the pathogenesis and progression of various etiological cardiomyopathies, including diabetic cardiomyopathy (DCM), a disease that has been reported since 1972. Cardiac cell death has been detected in the hearts of patients with diabetes and in animal models, and the role of cell death in the pathogenesis of DCM has been extensively investigated. The first review by the authors, specifically focusing on "Cell death and diabetic cardiomyopathy," was published in the journal, Cardiovascular Toxicology in 2003. Over the past two decades, studies investigating the role of cardiac cell death in the pathogenesis of DCM have gained significant attention, resulting in the discovery of several new kinds of cell death involving different mechanisms, including apoptosis, necroptosis, pyroptosis, autophagy, ferroptosis, and cuproptosis. After the 20th anniversary of the review published in 2003, we now provide an update with a focus on the potential role of metal-mediated cell death, ferroptosis, and cuproptosis in the development of DCM in compliance with this special issue. The intent of our review is to further stimulate work in the field to advance the body of knowledge and continue to drive efforts to develop more advanced therapeutic approaches to prevent cell death, particularly metal-dependent cell death, and, ultimately, to reduce or prevent the development of DCM.
Collapse
Affiliation(s)
- Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, 570 S. Preston Street, Baxter I, Rm: 304F, Louisville, KY, USA.
- Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, USA.
- Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
- Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Yi Tan
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, 570 S. Preston Street, Baxter I, Rm: 304F, Louisville, KY, USA
- Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, USA
- Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Brian Holland
- Division of Cardiology, Department of Pediatrics, Norton Children's Hospital, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kupper Wintergerst
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, 570 S. Preston Street, Baxter I, Rm: 304F, Louisville, KY, USA
- Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, USA
- Division of Endocrinology, Department of Pediatrics, Norton Children's Hospital, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
165
|
Khan AR, Alnoud MAH, Ali H, Ali I, Ahmad S, Ul Hassan SS, Shaikh AL, Hussain T, Khan MU, Khan SU, Khan MS, Khan SU. Beyond the beat: A pioneering investigation into exercise modalities for alleviating diabetic cardiomyopathy and enhancing cardiac health. Curr Probl Cardiol 2024; 49:102222. [PMID: 38000567 DOI: 10.1016/j.cpcardiol.2023.102222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
Patients with preexisting cardiovascular disease or those at high risk for developing the condition are often offered exercise as a form of therapy. Patients with cancer who are at an increased risk for cardiovascular issues are increasingly encouraged to participate in exercise-based, interdisciplinary programs due to the positive correlation between these interventions and clinical outcomes following myocardial infarction. Diabetic cardiomyopathy (DC) is a cardiac disorder that arises due to disruptions in the homeostasis of individuals with diabetes. One of the primary reasons for mortality in individuals with diabetes is the presence of cardiac structural damage and functional abnormalities, which are the primary pathological features of DC. The aetiology of dilated cardiomyopathy is multifaceted and encompasses a range of processes, including metabolic abnormalities, impaired mitochondrial function, dysregulation of calcium ion homeostasis, excessive cardiomyocyte death, and fibrosis. In recent years, many empirical investigations have demonstrated that exercise training substantially impacts the prevention and management of diabetes. Exercise has been found to positively impact the recovery of diabetes and improve several metabolic problem characteristics associated with DC. One potential benefit of exercise is its ability to increase systolic activity, which can enhance cardiometabolic and facilitate the repair of structural damage to the heart caused by DC, leading to a direct improvement in cardiac health. In contrast, exercise has the potential to indirectly mitigate the pathological progression of DC through its ability to decrease circulating levels of sugar and fat while concurrently enhancing insulin sensitivity. A more comprehensive understanding of the molecular mechanism via exercise facilitates the restoration of DC disease must be understood. Our goal in this review was to provide helpful information and clues for developing new therapeutic techniques for motion alleviation DC by examining the molecular mechanisms involved.
Collapse
Affiliation(s)
- Ahsan Riaz Khan
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Mohammed A H Alnoud
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Hamid Ali
- Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad 44000, Pakistan
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Hawally 32093, Kuwait
| | - Saleem Ahmad
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans 70112 LA, USA
| | - Syed Shams Ul Hassan
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310002, China
| | | | - Talib Hussain
- Women Dental College Abbottabad, KPK, 22020, Pakistan
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Muhammad Shehzad Khan
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Shatin city, (HKSAR), Hong Kong
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing 400715, China; Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, 22080, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
166
|
Januzzi JL, Del Prato S, Rosenstock J, Butler J, Ezekowitz J, Ibrahim NE, Lam CSP, Marwick T, Wilson Tang WH, Liu Y, Mohebi R, Urbinati A, Zannad F, Perfetti R. Characterizing diabetic cardiomyopathy: baseline results from the ARISE-HF trial. Cardiovasc Diabetol 2024; 23:49. [PMID: 38302936 PMCID: PMC10835978 DOI: 10.1186/s12933-024-02135-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DbCM) is a form of Stage B heart failure (HF) at high risk for progression to overt disease. Using baseline characteristics of study participants from the Aldose Reductase Inhibition for Stabilization of Exercise Capacity in Heart Failure (ARISE-HF) Trial we sought to characterize clinical characteristics of individuals with findings consistent with DbCM. METHODS Among study participants meeting inclusion criteria, clinical characteristics, laboratory testing, imaging, Kansas City Cardiomyopathy Questionnaire (KCCQ), Physical Activity Scale of the Elderly (PASE) and cardiopulmonary exercise testing (CPET) results were tabulated. Cluster phenogroups were identified. RESULTS Among 691 study participants (mean age 67.4 years; 50% were female), mean duration of type 2 diabetes mellitus (T2DM) was 14.5 years. The median (Q1, Q3) N-terminal pro-B type natriuretic peptide and high sensitivity cardiac troponin T were 71 (35, 135) ng/L and 9 [6, 12] ng/L. The most common echocardiographic abnormalities were reduced global longitudinal strain in 25.3% and impaired diastolic relaxation in 17.7%. Despite rather well-preserved KCCQ scores the average PASE score was markedly impaired at 155 accompanied by an average maximal oxygen consumption of 15.7 mL/Kg/minute on CPET. In K-means clustering, 4 phenogroups were identified including a higher-risk group with more advanced age, greater elevation of cardiac biomarkers, and more prevalent evidence for diastolic dysfunction and left ventricular hypertrophy. CONCLUSIONS Baseline data from the ARISE-HF Trial provide clinical characterization of individuals with T2DM and features of stage B HF, and may help clarify the diagnosis of DbCM. TRIAL REGISTRATION ARISE-HF, NCT04083339.
Collapse
Affiliation(s)
- James L Januzzi
- Heart Failure Trials, Baim Institute for Clinical Research, Boston, MA, USA.
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 02114, Boston, MA, USA.
| | - Stefano Del Prato
- Interdisciplinary Research Center 'Health Science', Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Julio Rosenstock
- Velocity Clinical Research at Medical City and University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Javed Butler
- Baylor Scott and White Research Institute, , Dallas, TX, USA
- University of Mississippi, Jackson, MS, USA
| | - Justin Ezekowitz
- Canadian VIGOUR Centre, University of Alberta, Edmonton, AB, Canada
| | - Nasrien E Ibrahim
- Cardiology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolyn S P Lam
- National Heart Centre Singapore and Duke-National University of Singapore, Singapore, Singapore
| | - Thomas Marwick
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Menzies Institute for Medical Research, Hobart, Australia
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuxi Liu
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 02114, Boston, MA, USA
| | - Reza Mohebi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, 02114, Boston, MA, USA
| | | | - Faiez Zannad
- Université de Lorraine, CIC Inserm and CHRU Nancy, Lorraine, France
| | | |
Collapse
|
167
|
Liu P, Zhang Z, Chen H, Chen Q. Pyroptosis: Mechanisms and links with diabetic cardiomyopathy. Ageing Res Rev 2024; 94:102182. [PMID: 38182080 DOI: 10.1016/j.arr.2023.102182] [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: 08/10/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease characterized by hyperglycaemia that seriously affects human health. Diabetic cardiomyopathy (DCM) is a major cardiovascular complication and one of the main causes of death in patients with DM. Although DCM attracts great attention, and new therapeutic methods are continuously developed, there is a lack of effective treatment strategies. Therefore, exploring and targeting new signalling pathways related to the evolution of DCM becomes a hotspot and difficulty in the prevention and treatment of DCM. Pyroptosis is a newly discovered regulated cell death that is heavily dependent on the formation of plasma membrane pores by members of the gasdermin protein family and is reported to be involved in the occurrence, development, and pathogenesis of DCM. In this review, we focus on the molecular mechanisms of pyroptosis, its involvement in the relevant signalling pathways of DCM, and potential pyroptosis-targeting therapeutic strategies for the treatment of DCM. Our review provides new insights into the use of pyroptosis as a useful tool for the prevention and treatment of DCM and clarifies future research directions.
Collapse
Affiliation(s)
- Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China; Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China
| | - Huizhen Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China.
| |
Collapse
|
168
|
Lin LC, Liu ZY, Yang JJ, Zhao JY, Tao H. Lipid metabolism reprogramming in cardiac fibrosis. Trends Endocrinol Metab 2024; 35:164-175. [PMID: 37949734 DOI: 10.1016/j.tem.2023.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
Cardiac fibrosis is a critical pathophysiological process that occurs with diverse types of cardiac injury. Lipids are the most important bioenergy substrates for maintaining optimal heart performance and act as second messengers to transduce signals within cardiac cells. However, lipid metabolism reprogramming is a double-edged sword in the regulation of cardiomyocyte homeostasis and heart function. Moreover, lipids can exert diverse effects on cardiac fibrosis through different signaling pathways. In this review, we provide a brief overview of aberrant cardiac lipid metabolism and recent progress in pharmacological research targeting lipid metabolism alterations in cardiac fibrosis.
Collapse
Affiliation(s)
- Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
169
|
Zou Y, Li S, Chen W, Xu J. Urine-derived stem cell therapy for diabetes mellitus and its complications: progress and challenges. Endocrine 2024; 83:270-284. [PMID: 37801228 DOI: 10.1007/s12020-023-03552-y] [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: 05/05/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Diabetes mellitus (DM) is a chronic and relentlessly progressive metabolic disease characterized by a relative or absolute deficiency of insulin in the body, leading to increased production of advanced glycosylation end products that further enhance oxidative and nitrosative stresses, often leading to multiple macrovascular (cardiovascular disease) and microvascular (e.g., diabetic nephropathy, diabetic retinopathy, and neuropathy) complications, representing the ninth leading cause of death worldwide. Existing medical treatments do not provide a complete cure for DM; thus, stem cell transplantation therapy has become the focus of research on DM and its complications. Urine-derived stem cells (USCs), which are isolated from fresh urine and have biological properties similar to those of mesenchymal stem cells (MSCs), were demonstrated to exert antiapoptotic, antifibrotic, anti-inflammatory, and proangiogenic effects through direct differentiation or paracrine mechanisms and potentially treat patients with DM. USCs also have the advantages of simple noninvasive sample collection procedures, minimal ethical issues, low cost, and easy cell isolation methods and thus have received more attention in regenerative therapies in recent years. This review outlines the biological properties of USCs and the research progress and current limitations of their role in DM and related complications. In summary, USCs have shown good versatility in treating hyperglycemia-impaired target organs in preclinical models, and many challenges remain in translating USC therapies to the clinic.
Collapse
Affiliation(s)
- Yun Zou
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shanshan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
170
|
D’Haese S, Claes L, de Laat I, Van Campenhout S, Deluyker D, Heeren E, Haesen S, Lambrichts I, Wouters K, Schalkwijk CG, Hansen D, Eijnde BO, Bito V. Moderate-Intensity and High-Intensity Interval Exercise Training Offer Equal Cardioprotection, with Different Mechanisms, during the Development of Type 2 Diabetes in Rats. Nutrients 2024; 16:431. [PMID: 38337716 PMCID: PMC10856993 DOI: 10.3390/nu16030431] [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: 01/08/2024] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Endurance exercise training is a promising cardioprotective strategy in type 2 diabetes mellitus (T2DM), but the impact of its intensity is not clear. We aimed to investigate whether and how isocaloric moderate-intensity exercise training (MIT) and high-intensity interval exercise training (HIIT) could prevent the adverse cardiac remodeling and dysfunction that develop T2DM in rats. Male rats received a Western diet (WD) to induce T2DM and underwent a sedentary lifestyle (n = 7), MIT (n = 7) or HIIT (n = 8). Insulin resistance was defined as the HOMA-IR value. Cardiac function was assessed with left ventricular (LV) echocardiography and invasive hemodynamics. A qPCR and histology of LV tissue unraveled underlying mechanisms. We found that MIT and HIIT halted T2DM development compared to in sedentary WD rats (p < 0.05). Both interventions prevented increases in LV end-systolic pressure, wall thickness and interstitial collagen content (p < 0.05). In LV tissue, HIIT tended to upregulate the gene expression of an ROS-generating enzyme (NOX4), while both modalities increased proinflammatory macrophage markers and cytokines (CD86, TNF-α, IL-1β; p < 0.05). HIIT promoted antioxidant and dicarbonyl defense systems (SOD2, glyoxalase 1; p < 0.05) whereas MIT elevated anti-inflammatory macrophage marker expression (CD206, CD163; p < 0.01). We conclude that both MIT and HIIT limit WD-induced T2DM with diastolic dysfunction and pathological LV hypertrophy, possibly using different adaptive mechanisms.
Collapse
Affiliation(s)
- Sarah D’Haese
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Lisa Claes
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Iris de Laat
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Sven Van Campenhout
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Dorien Deluyker
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Ellen Heeren
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Sibren Haesen
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Ivo Lambrichts
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| | - Kristiaan Wouters
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Casper G. Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Dominique Hansen
- UHasselt, Faculty of Rehabilitation Sciences, REVAL Rehabilitation Research Centre, Agoralaan, 3590 Diepenbeek, Belgium;
- Department of Cardiology, Heart Centre Hasselt, Jessa Hospital, Stadsomvaart 11, 3500 Hasselt, Belgium
| | - BO Eijnde
- SMRc-Sports Medicine Research Center, BIOMED-Biomedical Research Institute, Faculty of Medicine & Life Sciences, Hasselt University, 3500 Diepenbeek, Belgium;
- Division of Sport Science, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Virginie Bito
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (I.L.)
| |
Collapse
|
171
|
Zhong L, Li J, Yu J, Cao X, Du J, Liang L, Yang M, Yue Y, Zhao M, Zhou T, Lin J, Wang X, Shen X, Zhong Y, Wang Y, Shu Z. Anemarrhena asphodeloides Bunge total saponins ameliorate diabetic cardiomyopathy by modifying the PI3K/AKT/HIF-1α pathway to restore glycolytic metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117250. [PMID: 37832811 DOI: 10.1016/j.jep.2023.117250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on the theory of traditional Chinese medicine (TCM), diabetic cardiomyopathy (DCM) belongs to the category of "Xiaoke disease" according to the symptoms, and "stasis-heat" is the main pathogenesis of DCM. The Chinese medicine Anemarrhena asphodeloides Bunge (AAB), as a representative of heat-clearing and engendering fluid, is often used clinically in the treatment of DCM. Anemarrhena asphodeloides Bunge total saponins (RATS) are the main bioactive components of AAB, the modern pharmacologic effects of RATS are anti-inflammatory, hypoglycemic, and cardioprotective. However, the potential protective mechanisms of RATS against DCM remain largely undiscovered. AIM OF THE STUDY The primary goal of this study was to explore the effect of RATS on DCM and its mechanism of action. MATERIALS AND METHODS Streptozotocin and a high-fat diet were used to induce DCM in rats. UHPLC/Q-TOF-MS was used to determine the chemical components of RATS. The degenerative alterations and apoptotic cells in the heart were assessed by HE staining and TUNEL. Network pharmacology was used to anticipate the probable targets and important pathways of RATS. The alterations in metabolites and main metabolic pathways in heart tissue were discovered using 1 H-NMR metabolomics. Ultimately, immunohistochemistry was used to find critical pathway protein expression. RESULTS First of all, UHPLC/Q-TOF-MS analysis showed that RATS contained 11 active ingredients. In animal experiments, we found that RATS lowered blood glucose and lipid levels in DCM rats, and alleviated cardiac pathological damage, and decreased cardiomyocyte apoptosis. Furthermore, the study found that RATS effectively reduced inflammatory factor release and the level of oxidative stress. Mechanistically, RATS downregulated the expression levels of PI3K, AKT, HIF-1α, LDHA, and GLUT4 proteins. Additionally, glycolysis was discovered to be a crucial pathway for RATS in the therapy of DCM. CONCLUSIONS Our findings suggest that the protective effect of RATS on DCM may be attributed to the inhibition of the PI3K/AKT/HIF-1α pathway and the correction of glycolytic metabolism.
Collapse
Affiliation(s)
- Luyang Zhong
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jianhua Li
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jiamin Yu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xia Cao
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jieyong Du
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lanyuan Liang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Mengru Yang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yimin Yue
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Mantong Zhao
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Tong Zhou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jiazi Lin
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiao Wang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xuejuan Shen
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yanmei Zhong
- New Drug Research and Development Center, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Yi Wang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Zunpeng Shu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
172
|
Lou X, Zhang Y, Guo J, Gao L, Ding Y, Zhuo X, Lei Q, Bian J, Lei R, Gong W, Zhang X, Jiao Q. What is the impact of ferroptosis on diabetic cardiomyopathy: a systematic review. Heart Fail Rev 2024; 29:1-11. [PMID: 37555989 DOI: 10.1007/s10741-023-10336-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Iron overload increases the production of harmful reactive oxygen species in the Fenton reaction, which causes oxidative stress in the body and lipid peroxidation in the cell membrane, and eventually leads to ferroptosis. Diabetes is associated with increased intracellular oxidative stress, inflammation, autophagy, microRNA alterations, and advanced glycation end products (AGEs), which cause cardiac remodeling and cardiac diastolic contractile dysfunction, leading to the development of diabetic cardiomyopathy (DCM). While these factors are also closely associated with ferroptosis, more and more studies have shown that iron-mediated ferroptosis is an important causative factor in DCM. In order to gain fresh insights into the functions of ferroptosis in DCM, this review methodically summarizes the traits and mechanisms connected with ferroptosis and DCM.
Collapse
Affiliation(s)
- Xiaokun Lou
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Yuanyuan Zhang
- Department of Cardiovascular Ultrasonic Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Junfeng Guo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Lina Gao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Yingying Ding
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Xinyu Zhuo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Qingqing Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Jing Bian
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Rumei Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Wenyan Gong
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China.
- Hangzhou Institute of Cardiovascular Disease, Hangzhou, 310000, China.
| | - Xingwei Zhang
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China.
- Hangzhou Institute of Cardiovascular Disease, Hangzhou, 310000, China.
| | - Qibin Jiao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China.
| |
Collapse
|
173
|
Wang M, Liang Y, Qin Y, Ma R, Yu H, Wang W, Zhang X. Active Ingredients and Mechanism of Gegen Qinlian Decoction in the Treatment of Diabetic Cardiomyopathy: A Network Pharmacology Study. Curr Pharm Des 2024; 30:2896-2910. [PMID: 39136516 DOI: 10.2174/0113816128312242240722080551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a common diabetes complication with limited medications. Gegen Qinlian decoction (GQD) has been used in the treatment of diabetes and its related complications in China for several decades. OBJECTIVE In this study, network pharmacology was employed to predict the active ingredients, key targets, and pathways involved in the treatment of DCM by GQD and to validate it by animal experiments. METHODS The active ingredients of GQD were retrieved from TCMSP and published literature. DCM-related gene targets were searched in Drugbank, Genecards, Disgenet, and OMIM disease databases. Protein-protein interaction networks were constructed using the STRING database and Cytoscape. GO analysis and KEGG pathway enrichment analysis were performed using the Metascape platform. Moreover, a diabetic mouse model was established to evaluate the therapeutic effects of GQD by measuring serum biochemical markers and inflammation levels. Finally, the expression of predicted key target genes was determined using real-time quantitative PCR. RESULTS A total of 129 active ingredients were screened from GQD. Moreover, 146 intersecting genes related to DCM were obtained, with key targets, including AKT1, TNF, IL6, and VEGFA. Lipid and atherosclerosis, AGE-RAGE, PI3K-AKT, and MAPK pathways were identified. Blood glucose control, decreased inflammatory factors, and serum CK-MB levels were restored after GQD intervention, and the same occurred with the expressions of PPAR-γ, AKT1, APOB, and GSK3B genes. CONCLUSION Quercetin, kaempferol, wogonin, 7-methoxy-2-methyl isoflavone, and formononetin may exert major therapeutic effects by regulating key factors, such as AKT1, APOB, and GSK3B, in the inflammatory reaction, glycolipid oxidation, and glycogen synthesis related signaling pathways.
Collapse
Affiliation(s)
- Min Wang
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| | - Yanbin Liang
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Yuce Qin
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Ruimian Ma
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| | - Huanting Yu
- College of Clinical Medicine, Guilin Medical University, Guilin 541004, China
| | - Weixue Wang
- Department of Traditional Chinese Medicine, Huai'an No. 3 People's Hospital, Huaian 223001, China
- Department of Traditional Chinese Medicine, Affiliated Hospital of Guilin Medical University, Guilin 541004, China
| | - Xiaoxi Zhang
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| |
Collapse
|
174
|
Ji L, Yang X, Jin Y, Li L, Yang B, Zhu W, Xu M, Wang Y, Wu G, Luo W, Lee K, Liang G. Blockage of DCLK1 in cardiomyocytes suppresses myocardial inflammation and alleviates diabetic cardiomyopathy in streptozotocin-induced diabetic mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166900. [PMID: 37778481 DOI: 10.1016/j.bbadis.2023.166900] [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: 07/04/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition triggered by diabetes mellitus and can lead to heart failure. Doublecortin-like kinase protein 1 (DCLK1) is a multifunctional protein kinase involved in the regulation of cell proliferation, differentiation, survival, and migration. Current studies on DCLK1 mainly focus on cancer development; however, its role in non-tumor diseases such as DCM is yet to be deciphered. Our analysis revealed that DCLK1 was upregulated in cardiomyocytes of streptozotocin (STZ)-induced type 1 diabetic mouse, suggesting a correlation between DCLK1 and DCM progression. It was further demonstrated that either cardiomyocyte-specific DCLK1 knockout or pharmacological DCLK1 inhibitor DCLK1-IN-1 significantly alleviated cardiac hypertrophy and fibrosis in STZ-induced diabetic mice. RNA-seq analysis of heart tissues revealed that DCLK1 regulated the nuclear factor kappa B (NF-κB)-mediated inflammatory response in DCM. In vitro, DCLK1 activated NF-κB and the inflammatory response by inducing the IKKβ phosphorylation in high-concentration glucose (HG)-challenged cardiomyocytes. DCLK1-IN-1 also prevented HG-induced IKKβ/NF-κB activation and inflammatory injuries in cardiomyocytes. In conclusion, this study highlights the novel role of cardiomyocyte DCLK1 in regulating IKKβ/NF-κB, which aggravates inflammation to promote the pathogenesis of DCM. DCLK1 may serve as a new target for DCM treatment.
Collapse
Affiliation(s)
- Lijun Ji
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Xiaojing Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yiyi Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Li Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bin Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weiwei Zhu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mingjiang Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gaojun Wu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wu Luo
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Kwangyoul Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Guang Liang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Zhejiang Provincial People's Hospital, the Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
175
|
Wang B, Zhao C, Wang Y, Tian X, Lin J, Zhu B, Zhou Y, Zhang X, Li N, Sun Y, Xu H, Zhao R. Exercise ameliorating myocardial injury in type 2 diabetic rats by inhibiting excessive mitochondrial fission involving increased irisin expression and AMP-activated protein kinase phosphorylation. J Diabetes 2024; 16:e13475. [PMID: 37721125 PMCID: PMC10809304 DOI: 10.1111/1753-0407.13475] [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: 06/20/2023] [Revised: 08/15/2023] [Accepted: 08/27/2023] [Indexed: 09/19/2023] Open
Abstract
PURPOSE Though exercise generates beneficial effects on diabetes-associated cardiac damage, the underlying mechanism is largely unclear. Therefore, we prescribed a program of 8-week treadmill training for type 2 diabetes mellitus (T2DM) rats and determined the role of irisin signaling, via interacting with AMP-activated protein kinase (AMPK), in mediating the effects of exercise on myocardial injuries and mitochondrial fission. METHODS Forty 8-week-old male Wistar rats were randomly divided into groups of control (Con), diabetes mellitus (DM), diabetes plus exercise (Ex), and diabetes plus exercise and Cyclo RGDyk (ExRg). Ex and ExRg rats received 8 weeks of treadmill running, and the rats in the ExRg group additionally were treated with a twice weekly injection of Cyclo RGDyk, an irisin receptor-αV/β5 antagonist. At the end of the experiment, murine blood samples and heart tissues were collected and analyzed with methods of ELISA, Western blot, real-time quantitative polymerase chain reaction, as well as immunofluorescence staining. RESULTS Exercise effectively mitigated T2DM-related hyperglycemia, hyperinsulinemia, lipid dysmetabolism, and inflammation, which could be diminished by Cyclo RGDyk treatment. Additionally, exercise alleviated T2DM-induced myocardial injury and excessive mitochondrial fission, whereas the beneficial effects were blocked by the administration of Cyclo RGDyk. T2DM significantly decreased serum irisin concentrations and fibronectin type III domain-containing protein 5 (FNDC5)/irisin gene and protein expression levels in the rat heart, whereas exercise could rescue T2DM-reduced FNDC5/irisin expression. Blocking irisin receptor signaling diminished the exercise-alleviated mitochondrial fission protein expression and elevated AMPK phosphorylation. CONCLUSION Exercise is effective in mitigating diabetes-related insulin resistance, metabolic dysfunction, and inflammation. Irisin signaling engages in exercise-associated beneficial effects on myocardial injury and excessive mitochondrial fission in diabetes rats involving elevated AMPK phosphorylation.
Collapse
Affiliation(s)
- Bin Wang
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Chen Zhao
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Yuanxin Wang
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Xin Tian
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Junjie Lin
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Baishu Zhu
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Yalan Zhou
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Xin Zhang
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Nan Li
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Yu Sun
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Haocheng Xu
- College of Physical EducationYangzhou UniversityYangzhouChina
| | - Renqing Zhao
- College of Physical EducationYangzhou UniversityYangzhouChina
| |
Collapse
|
176
|
Imaralu OE, Aluganti Narasimhulu C, Singal PK, Singla DK. Role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in diabetic complications. Can J Physiol Pharmacol 2024; 102:14-25. [PMID: 37748207 DOI: 10.1139/cjpp-2023-0223] [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] [Indexed: 09/27/2023]
Abstract
Cardiovascular disease (CVD) complications have remained a major cause of death among patients with diabetes. Hence, there is a need for effective therapeutics against diabetes-induced CVD complications. Since its discovery, proprotein convertase subtilisin/kexin type 9 (PCSK9) has been reported to be involved in the pathology of various CVDs, with studies showing a positive association between plasma levels of PCSK9, hyperglycemia, and dyslipidemia. PCSK9 regulates lipid homeostasis by interacting with low-density lipoprotein receptors (LDLRs) present in hepatocytes and subsequently induces LDLR degradation via receptor-mediated endocytosis, thereby reducing LDL uptake from circulation. In addition, PCSK9 also induces pro-inflammatory cytokine expression and apoptotic cell death in diabetic-CVD. Furthermore, therapies designed to inhibit PCSK9 effectively reduces diabetic dyslipidemia with clinical studies reporting reduced cardiovascular events in patients with diabetes and no significant adverse effect on glycemic controls. In this review, we discuss the role of PCSK9 in the pathogenesis of diabetes-induced CVD and the potential mechanisms by which PCSK9 inhibition reduces cardiovascular events in diabetic patients.
Collapse
Affiliation(s)
- Omonzejie E Imaralu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Chandrakala Aluganti Narasimhulu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Pawan K Singal
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| |
Collapse
|
177
|
Wu J, Yan Y. SIAH1 Promotes the Pyroptosis of Cardiomyocytes in Diabetic Cardiomyopathy via Regulating IκB-α/NF-κВ Signaling. Crit Rev Eukaryot Gene Expr 2024; 34:45-57. [PMID: 38842203 DOI: 10.1615/critreveukaryotgeneexpr.2024052773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Inflammation-mediated dysfunction of cardiomyocytes is the main cause of diabetic cardiomyopathy (DCM). The present study aimed to investigate the roles of siah E3 ubiquitin protein ligase 1 (SIAH1) in DCM. The online dataset GSE4172 was used to analyze the differentially expressed genes in myocardial inflammation of DCM patients. RT-qPCR was conducted to detect mRNA levels. Enzyme-Linked Immunosorbent Assay (ELISA) was performed to detect cytokine release. Western blot was used to detect protein expression. Lactate dehydrogenase (LDH) assay was used to determine cytotoxicity. In vitro ubiquitination assay was applied to determine the ubiquitination of nuclear factor kappa B inhibitor alpha (1κВ-α). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was used to detect the death of cardiomyocytes. Flow cytometry was applied for determining cardiomyocyte pyroptosis. The results showed that SIAH1 was overexpressed in human inflammatory cardiomyopathy. High expression of SIAH1 was associated with inflammatory response. SIAH1 was also overexpressed lipopolysaccharide (LPS)-induced inflammatory cardiomyopathy model in vitro. However, SIAH1 knockdown suppressed the inflammatory-related pyroptosis of cardiomyocytes. SIAH1 promoted the ubiquitination of 1κВ-α and activated nuclear factor kappa В (NF-κВ) signaling, which promoted the pyroptosis of cardiomyocytes. In conclusion, SIAH1 exacerbated the progression of human inflammatory cardiomyopathy via inducing the ubiquitination of 1κВ-α and activation of NF-κВ signaling. Therefore, SIAHI/IκB-α/NF-κB signaling may be a potential target for human inflammatory cardiomyopathy.
Collapse
Affiliation(s)
| | - Yaoming Yan
- Laboratory Department, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
178
|
Wu S, Zhou Y, Liang J, Ying P, Situ Q, Tan X, Zhu J. Upregulation of NF-κB by USP24 aggravates ferroptosis in diabetic cardiomyopathy. Free Radic Biol Med 2024; 210:352-366. [PMID: 38056575 DOI: 10.1016/j.freeradbiomed.2023.11.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Recent investigations have proposed a potential causal association between the occurrence of ferroptosis, nuclear factor kappa B (NF-κB) and ubiquitin-specific protease 24 (USP24). Nevertheless, the mechanism of USP24 and NF-κB regulation of ferroptosis in the context of diabetic cardiomyopathy (DCM) remain unclear. METHODS In this study, a high-fat diet and a streptozotocin-induced mouse DCM model were established, and high glucose and palmitic acid treatment of H9c2 cells and neonatal mouse primary cardiomyocytes (NMPCs) was used as an in vitro DCM models. Utilizing both the in vivo and in vitro DCM models, we assessed of USP24, NF-κB, and ferroptosis levels, and explored the relationship among them. RESULTS In in vivo and in vitro DCM models, increased expression of USP24, NF-κB, phosphorylated NF-κB (p-NF-κB) and fatty acid-CoA ligase 4 (FACL4) were detected, along with accumulated iron, as well as reduced ferritin heavy chain 1 (FTH1), solute carrier family 7 member 11 (SLC7A11) and antioxidant capacity. Knockdown of USP24 resulted in a reduction of NF-κB levels, while knockdown of NF-κB did not lead to a decrease in USP24 expression. Moreover, in H9c2 cells, knockdown of USP24 and NF-κB separately resulted in reduced levels of FACL4, increased levels of SLC7A11 and FTH1, as well as improved antioxidant capacity and cell viability. In shUSP24 knockdown H9c2 cells, administration of phorbol 12-myristate 13-acetate (PMA) activated NF-κB, subsequently reversing the previously observed effect caused by USP24 knockdown. CONCLUSIONS These findings show that USP24 upregulates NF-κB to promote ferroptosis in DCM.
Collapse
Affiliation(s)
- Shenglin Wu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yueran Zhou
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jiaquan Liang
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Pengxiang Ying
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Qiwei Situ
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xuerui Tan
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jinxiu Zhu
- Institute of Clinical Electrocardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China; Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), Shenzhen, Guangdong 518172, China.
| |
Collapse
|
179
|
Rai B, Srivastava J, Saxena P. The Functional Role of microRNAs and mRNAs in Diabetic Kidney Disease: A Review. Curr Diabetes Rev 2024; 20:e201023222412. [PMID: 37867275 DOI: 10.2174/0115733998270983231009094216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/03/2023] [Accepted: 09/08/2023] [Indexed: 10/24/2023]
Abstract
Diabetes is a group of diseases marked by poor control of blood glucose levels. Diabetes mellitus (DM) occurs when pancreatic cells fail to make insulin, which is required to keep blood glucose levels stable, disorders, and so on. High glucose levels in the blood induce diabetic effects, which can cause catastrophic damage to bodily organs such as the eyes and lower extremities. Diabetes is classified into many forms, one of which is controlled by hyperglycemia or Diabetic Kidney Disease (DKD), and another that is not controlled by hyperglycemia (nondiabetic kidney disease or NDKD) and is caused by other factors such as hypertension, hereditary. DKD is associated with diabetic nephropathy (DN), a leading cause of chronic kidney disease (CKD) and end-stage renal failure. The disease is characterized by glomerular basement membrane thickening, glomerular sclerosis, and mesangial expansion, resulting in a progressive decrease in glomerular filtration rate, glomerular hypertension, and renal failure or nephrotic syndrome. It is also represented by some microvascular complications such as nerve ischemia produced by intracellular metabolic changes, microvascular illness, and the direct impact of excessive blood glucose on neuronal activity. Therefore, DKD-induced nephrotic failure is worse than NDKD. MicroRNAs (miRNAs) are important in the development and progression of several diseases, including diabetic kidney disease (DKD). These dysregulated miRNAs can impact various cellular processes, including inflammation, fibrosis, oxidative stress, and apoptosis, all of which are implicated during DKD. MiRNAs can alter the course of DKD by targeting several essential mechanisms. Understanding the miRNAs implicated in DKD and their involvement in disease development might lead to identifying possible therapeutic targets for DKD prevention and therapy. Therefore, this review focuses specifically on DKD-associated DN, as well as how in-silico approaches may aid in improving the management of the disease.
Collapse
Affiliation(s)
- Bhuvnesh Rai
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Jyotika Srivastava
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Pragati Saxena
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
180
|
Liu B, Zhang J, Zhou Z, Feng B, He J, Yan W, Zhou X, Amponsah AE, Guo R, Du X, Liu X, Cui H, O'Brien T, Ma J. Preclinical Evidence for the Effectiveness of Mesenchymal Stromal Cells for Diabetic Cardiomyopathy: A Systematic Review and Meta-analysis. Curr Stem Cell Res Ther 2024; 19:220-233. [PMID: 37165495 DOI: 10.2174/1574888x18666230510111302] [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: 01/20/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a complication of diabetes mellitus that endangers human health. DCM results in cardiac dysfunction, which eventually progresses to heart failure. Mesenchymal stromal cells (MSCs), a type of multipotent stem cell, have shown promising therapeutic effects in various cardiovascular diseases and diabetic complications in preclinical studies due to their immunomodulatory and regenerative abilities. However, there is still a lack of evidence to summarize the effectiveness of MSCs in the treatment of DCM. Therefore, a meta-analysis and systematic review are warranted to evaluate the therapeutic potential of MSCs for DCM in preclinical studies. METHODS A comprehensive literature search in English or Chinese was conducted in PubMed, EMBASE, web of Science, Cochrane Library, and China National Knowledge Internet from inception to June 30, 2022. The summarized outcomes included echocardiography, morphology, and pathology. Data were independently extracted and analyzed by two authors. The software we adopted was Review Manager5.4.1. This systematic review was written in compliance with PRISMA 2020 and the review protocol was registered on PROSPERO, registration no. CRD42022350032. RESULTS We included 20 studies in our meta-analysis to examine the efficacy of MSCs in the treatment of DCM. The MSC-treated group showed a statistically significant effect on left ventricular ejection fraction (WMD=12.61, 95% CI 4.32 to 20.90, P=0.003) and short axis fractional shortening (WMD=6.84, 95% CI 4.09 to 9.59, P < 0.00001). The overall effects on the ratio of early to late diastolic mitral annular velocity, left ventricular end-diastolic pressure, maximum positive pressure development, maximum negative pressure development, left ventricular relaxation time constant, heart weight to body weight ratio, fibrosis area, and arteriole density were analyzed, suggesting that MSCs represent an effective therapy for the treatment of DCM. CONCLUSION Our results suggest a therapeutic role for MSCs in the treatment of DCM, and these results provide support for the use of MSCs in clinical trials of patients with DCM.
Collapse
Affiliation(s)
- Boxin Liu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Jinyu Zhang
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Zijing Zhou
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Baofeng Feng
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Jingjing He
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Wei Yan
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Xinghong Zhou
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Asiamah Ernest Amponsah
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
| | - Ruiyun Guo
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Xiaofeng Du
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Xin Liu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
| | - Huixian Cui
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Human Anatomy Department, Hebei Medical University, Shijiazhuang, 050017, Hebei Province
| | - Timothy O'Brien
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Jun Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Human Anatomy Department, Hebei Medical University, Shijiazhuang, 050017, Hebei Province
| |
Collapse
|
181
|
Wang J, Zhao J, Lin Q, Xu X, Jiang K, Li Y. ΔRDW Could Predict Major Adverse Cardiovascular Events in Patients with Heart Failure with Reduced Ejection Fraction After Sacubitril/Valsartan Treatment. Int J Gen Med 2023; 16:5989-6003. [PMID: 38144439 PMCID: PMC10748743 DOI: 10.2147/ijgm.s444585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/09/2023] [Indexed: 12/26/2023] Open
Abstract
Objective This study aimed to evaluate the association between red blood cell distribution width (RDW) changes and major adverse cardiovascular event (MACE) occurrences during sacubitril/valsartan treatment in patients with heart failure with reduced ejection fraction (HFrEF). Methods This study retrospectively analyzed the medical records of patients with HFrEF hospitalized from April 2018 to February 2021. The patients were divided into two groups according to the inclusion of sacubitril/valsartan in the personal drug treatment regimen, the traditional and the sacubitril/valsartan group. RDW values before and after sacubitril/valsartan treatment were recorded respectively as RDW1 and RDW2. ΔRDW was defined as the difference between RDW2 and RDW1. The patients in the sacubitril/valsartan group were divided into two subgroups according to ΔRDW >0 or ≤0. MACEs, such as readmission for HF, acute myocardial infarction, ischemic stroke, and malignant arrhythmia and death, were recorded during the 1-year follow-up period in each group. Results MACE development was lower in patients treated with sacubitril/valsartan than those treated with conventional therapy (log-rank, P<0.001). The incidence of cardiac events during the follow-up period was greater in the group with ΔRDW >0 than in the group with ΔRDW ≤0 (Breslow, P<0.001). Increased RDW was associated with a higher likelihood of developing MACE than decreased RDW (odds ratio [OR] =2.055, 95% confidence interval [CI]:1.301-3.246), and the risk of developing MACE increased by 22.1% for each unit increase in RDW (OR=1.221, 95% CI:1.074-1.389). Conclusion Sacubitril/valsartan treatment is effective in reducing the risk of MACEs in HFrEF. Additionally, RDW changes are predictors of MACEs after sacubitril/valsartan treatment.
Collapse
Affiliation(s)
- Jingsheng Wang
- Department of Cardiology, the Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, People’s Republic of China
| | - Jian Zhao
- Department of Cardiology, the Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, People’s Republic of China
| | - Quanqiang Lin
- Department of Cardiology, the Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, People’s Republic of China
| | - Xiuxiu Xu
- Department of Cardiology, the Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, People’s Republic of China
| | - Ke Jiang
- Department of Cardiology, the Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, People’s Republic of China
| | - Yuanmin Li
- Department of Cardiology, the Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, People’s Republic of China
| |
Collapse
|
182
|
Zhao M, Gao J, Chen S, Yao S, Wang M, Wang C, Zhang S, Feng Z, Tian L, Li Y, Liu Y, Wu S, Xue H. Association Between New-Onset Type 2 Diabetes and Cardiac Conduction Diseases: A Prospective Cohort Study. J Am Heart Assoc 2023; 12:e032237. [PMID: 38063148 PMCID: PMC10863756 DOI: 10.1161/jaha.123.032237] [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: 08/15/2023] [Accepted: 10/31/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Cardiac conduction diseases can lead to life-threatening outcomes. However, the evidence on risk factors for conduction disease that is needed to underpin prevention strategies is limited. The present study aimed to determine the association between type 2 diabetes and cardiac conduction diseases. METHODS AND RESULTS This study included 101 080 participants free of prevalent diabetes and cardiac conduction diseases at baseline from the Kailuan Study. All participants were monitored biennially until December 31, 2020. During follow-up, 14 397 participants were diagnosed as having type 2 diabetes. For each case subject, 1 control subject was randomly selected, matched for age (±1 year) and sex. The final analysis comprised 10 744 case-control pairs. Cox regression models with age as the underlying time scale were used. During a median follow-up of 5.46 years, 571 incident events occurred, including 164 atrioventricular blocks, 414 bundle-branch blocks (BBBs), 274 right BBBs, and 210 left BBBs. After adjustment for potential confounders, participants with type 2 diabetes diagnosed had greater relative risks for most outcomes relative to controls, with hazard ratios of 1.42 (95% CI, 1.18-1.67) for conduction diseases, 1.40 (95% CI, 1.00-1.96) for atrioventricular blocks, 1.43 (95% CI, 1.16-1.75) for BBBs, and 1.69 (95% CI, 1.15-2.49) for left BBBs. In contrast, no association between diabetes and right BBB was observed. CONCLUSIONS In this study, participants with type 2 diabetes are at an increased risk of cardiac conduction disease but not associated with the development of right BBB.
Collapse
Affiliation(s)
- Maoxiang Zhao
- Department of Cardiology, The First Medical CenterChinese People’s Liberation Army Hospital, Medical School of Chinese People’s Liberation ArmyBeijingChina
| | - Jingli Gao
- Department of Intensive Care UnitKailuan General HospitalTangshanChina
| | - Shuohua Chen
- Department of CardiologyKailuan HospitalTangshanChina
| | - Siyu Yao
- Department of CardiologyThe Sixth Medical Center, Chinese People’s Liberation Army Hospital, Medical School of Chinese People’s Liberation ArmyBeijingChina
| | - Miao Wang
- School of MedicineNankai UniversityTianjinChina
| | - Chi Wang
- Department of CardiologyThe Sixth Medical Center, Chinese People’s Liberation Army Hospital, Medical School of Chinese People’s Liberation ArmyBeijingChina
| | - Sijin Zhang
- School of MedicineNankai UniversityTianjinChina
| | - Zekun Feng
- Department of CardiologyThe Sixth Medical Center, Chinese People’s Liberation Army Hospital, Medical School of Chinese People’s Liberation ArmyBeijingChina
| | - Lu Tian
- School of MedicineNankai UniversityTianjinChina
| | - Yanjie Li
- School of MedicineNankai UniversityTianjinChina
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public HealthChina Medical UniversityShenyangLiaoningChina
| | - Shouling Wu
- Department of CardiologyKailuan HospitalTangshanChina
| | - Hao Xue
- Department of CardiologyThe Sixth Medical Center, Chinese People’s Liberation Army Hospital, Medical School of Chinese People’s Liberation ArmyBeijingChina
| |
Collapse
|
183
|
Cai L, Horowitz M, Islam MS. Potential therapeutic targets for the prevention of diabetic nephropathy: Glycyrrhetinic acid. World J Diabetes 2023; 14:1717-1720. [PMID: 38222784 PMCID: PMC10784793 DOI: 10.4239/wjd.v14.i12.1717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 12/14/2023] Open
Abstract
Uncontrolled hyperglycemia or poorly managed disease increases the propensity for a number of diabetes-related complications targeting major organs including the heart, eyes, and kidney. Although the mechanisms by which diabetes induces cardiovascular diseases include oxidative stress and inflammation, when insulin resistance remains the key to the pathogenesis, as implicated in the two reviews in this issue. This editorial mainly comments on the potential preventive application of glycyrrhetinic acid (or 18β-GA) in relation to diabetic nephropathy. The thera-peutic or preventive effects of 18β-GA, as a hydrolytic product of glycyrrhizic acid that is a component of licorice, have been appreciated in other disorders, but have received much less attention in relation to diabetic complications. A study in this issue has identified 18β-GA as a therapeutic for preventing diabetic nephropathy and provides evidence to support efficacy in cultured human renal tubule cells in vitro. Although it represents a pilot study, the observations support a new therapeutic approach that warrants further ex-ploration.
Collapse
Affiliation(s)
- Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, United States
| | - Michael Horowitz
- Department of Medicine, University of Adelaide, Adelaide 5005, Australia
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban 4000, KwaZulu-Natal, South Africa
| |
Collapse
|
184
|
Song MW, Cui W, Lee CG, Cui R, Son YH, Kim YH, Kim Y, Kim HJ, Choi SE, Kang Y, Kim TH, Jeon JY, Lee KW. Protective effect of empagliflozin against palmitate-induced lipotoxicity through AMPK in H9c2 cells. Front Pharmacol 2023; 14:1228646. [PMID: 38116084 PMCID: PMC10728651 DOI: 10.3389/fphar.2023.1228646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have recently emerged as novel cardioprotective agents. However, their direct impact on cardiomyocyte injury is yet to be studied. In this work, we investigate the underlying molecular mechanisms of empagliflozin (EMPA), an SGLT2 inhibitor, in mitigating palmitate (PA)-induced cardiomyocyte injury in H9c2 cells. We found that EMPA significantly attenuated PA-induced impairments in insulin sensitivity, ER stress, inflammatory cytokine gene expression, and cellular apoptosis. Additionally, EMPA elevated AMP levels, activated the AMPK pathway, and increased carnitine palmitoyl transferase1 (CPT1) gene expression, which collectively enhanced fatty acid oxidation and reduced stress signals. This study reveals a novel mechanism of EMPA's protective effects against PA-induced cardiomyocyte injury, providing new therapeutic insights into EMPA as a cardioprotective agent.
Collapse
Affiliation(s)
- Min-Woo Song
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Wenhao Cui
- Department of Hematology, Yanbian University Hospital, Yanji, Jilin, China
| | - Chang-Gun Lee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju, Republic of Korea
| | - Rihua Cui
- Department of Hematology, Yanbian University Hospital, Yanji, Jilin, China
| | - Young Ho Son
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Young Ha Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yujin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sung-E. Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Tae Ho Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul Medical Center, Seoul, Republic of Korea
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Kwan-Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
185
|
Gao J, Xu M, Gong M, Jiang S, Yang Z, Jiang X, Chen M. Left ventricular longitudinal strain in patients with type 2 diabetes mellitus is independently associated with glycated hemoglobin level. Clin Cardiol 2023; 46:1578-1587. [PMID: 37621117 PMCID: PMC10716340 DOI: 10.1002/clc.24136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
OBJECTIVE Left ventricular and left atrial strain are sensitive and reliable markers for evaluating cardiac function in patients with type 2 diabetes mellitus (T2DM), with interactions between the two parameters. The present study aimed to analyze the correlation between global longitudinal strain (GLS) of the left ventricle and glycated hemoglobin (HbA1c) levels in patients with T2DM. METHODS A total of 292 patients clinically diagnosed with T2DM were selected and divided into three groups according to HbA1c level. The strains of the left atrium and left ventricle in the three groups of T2DM patients with different HbA1c levels were compared. Univariate and multivariate (including left atrial functional indicators) linear regression analyses were performed to assess the relationship between strain indicators and HbA1c levels. Generalized additive models were used to examine the relationship between strain indicators and HbA1c levels. RESULTS There were significant differences among the three groups in terms of age, microalbuminuria, total cholesterol, fasting blood glucose, postprandial blood glucose, and HbA1c level, and left atrial conduit longitudinal strain (LAScd) and GLS (p < .05). Univariate and multivariate linear regression analyses revealed that, as HbA1c levels increased, the absolute value of GLS gradually decreased (p < .001). Curve fitting revealed a positive correlation between HbA1c level and GLS, which was not affected by left atrial function. CONCLUSION Left ventricular GLS was independently correlated with HbA1c level in patients with T2DM and was not affected by left atrial function.
Collapse
Affiliation(s)
- Jinmei Gao
- Department of Echocardiography and CardiologyThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| | - Min Xu
- Department of Echocardiography and CardiologyThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| | - Mingxia Gong
- Department of Echocardiography and CardiologyThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| | - Shu Jiang
- Department of Echocardiography and CardiologyThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| | - Zhenni Yang
- Department of Echocardiography and CardiologyThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| | - Xiaohong Jiang
- Department of EndocrinologyThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| | - Ming Chen
- Department of Medical ImagingThe Third Affiliated Hospital of Soochow UniversityChang ZhouChina
| |
Collapse
|
186
|
Xu N, Liu S, Zhang Y, Chen Y, Zuo Y, Tan X, Liao B, Li P, Feng J. Oxidative stress signaling in the pathogenesis of diabetic cardiomyopathy and the potential therapeutic role of antioxidant naringenin. Redox Rep 2023; 28:2246720. [PMID: 37747066 PMCID: PMC10538464 DOI: 10.1080/13510002.2023.2246720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Diabetes mellitus (DM) is one of the most prevalent metabolic disorders that poses a global threat to human health. It can lead to complications in multiple organs and tissues, owing to its wide-ranging impact on the human body. Diabetic cardiomyopathy (DCM) is a specific cardiac manifestation of DM, which is characterized by heart failure in the absence of coronary heart disease, hypertension and valvular heart disease. Given that oxidative stress is a key factor in the pathogenesis of DCM, intervening to mitigate oxidative stress may serve as a therapeutic strategy for managing DCM. Naringenin is a natural product with anti-oxidative stress properties that can suppress oxidative damage by regulating various oxidative stress signaling pathways. In this review, we address the relationship between oxidative stress and its primary signaling pathways implicated in DCM, and explores the therapeutic potential of naringenin in DCM.
Collapse
Affiliation(s)
- Nan Xu
- Department of Cardiology, The First People's Hospital of Neijiang, Neijiang, People’s Republic of China
| | - Siqi Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yongqiang Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yujing Chen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yumei Zuo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Xiaoqiu Tan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
| | - Pengyun Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, People’s Republic of China
| |
Collapse
|
187
|
Zhang K, Tian XM, Li W, Hao LY. Ferroptosis in cardiac hypertrophy and heart failure. Biomed Pharmacother 2023; 168:115765. [PMID: 37879210 DOI: 10.1016/j.biopha.2023.115765] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Heart failure has become a public health problem that we cannot avoid choosing to face in today's context. In the case of heart failure, pathological cardiac hypertrophy plays a major role because of its condition of absolute increase in ventricular mass under various stresses. Ferroptosis, it could be defined as regulatory mechanisms that regulate cell death in the absence of apoptosis in iron-dependent cells. This paper introduces various new research findings on the use of different regulatory mechanisms of cellular ferroptosis for the treatment of heart failure and cardiac hypertrophy, providing new therapeutic targets and research directions for clinical treatment. The role and mechanism of ferroptosis in the field of heart failure has been increasingly demonstrated, and the relationship between cardiac hypertrophy, which is one of the causes of heart failure, is also an area of research that we should focus on. In addition, the latest applications and progress of inducers and inhibitors of ferroptosis are reported in this paper, updating the breakthroughs in their fields.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xin-Miao Tian
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Wei Li
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Li-Ying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
188
|
Jasińska-Stroschein M. The current state of preclinical modeling of human diabetic cardiomyopathy using rodents. Biomed Pharmacother 2023; 168:115843. [PMID: 37939616 DOI: 10.1016/j.biopha.2023.115843] [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: 09/10/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023] Open
Abstract
The prevalence of diabetic cardiomyopathy (DCM), a specific cardiovascular complication of diabetes mellitus, has recently increased. Its pathogenesis is not fully understood, and no consensus regarding therapeutic options has been reached. Experimental studies on rodents are expected to yield further data at the preclinical stage. The present paper describes and quantitatively compares the experimental protocols intended to mimic human DCM. Experimental articles (conducted between 1990 and 2022) were identified from online electronic databases according to the PRISMA Protocol. The Cochrane Q-test was used to estimate study heterogeneity; the quality of each individual study was assessed using SYRCLE's risk of bias tool for animal studies. Sensitivity analysis was performed according to the leave-one-out method. Publication bias across studies was assessed using Egger's weighted regression and Duval and Tweedie 'trim and fill' method. A wide spectrum of protocols - from 651 papers, was examined (type 1 or 2 diabetes mellitus, as well as obesity models). They were found to vary in their presentation of DCM according to a variety of hemodynamic, echocardiographic, histopathologic and metabolic parameters. Particular attention was paid to comorbid conditions, and cardiac performance featured as systolic, diastolic dysfunction, or refractory heart failure. The majority of models displayed diastolic dysfunction, as well as myocardial fibrosis and left ventricle hypertrophy, which mimics early stage DCM. Unlike in humans, animal DCM rarely progressed to the symptomatic heart failure with reduced ejection fraction (HFrEF). The ability of individual procedures to reflect refractory heart failure or biventricular dysfunction - in the end-stage DCM has remained unclear.
Collapse
|
189
|
Costantino S, Mengozzi A, Velagapudi S, Mohammed SA, Gorica E, Akhmedov A, Mongelli A, Pugliese NR, Masi S, Virdis A, Hülsmeier A, Matter CM, Hornemann T, Melina G, Ruschitzka F, Luscher TF, Paneni F. Treatment with recombinant Sirt1 rewires the cardiac lipidome and rescues diabetes-related metabolic cardiomyopathy. Cardiovasc Diabetol 2023; 22:312. [PMID: 37957697 PMCID: PMC10644415 DOI: 10.1186/s12933-023-02057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Metabolic cardiomyopathy (MCM), characterized by intramyocardial lipid accumulation, drives the progression to heart failure with preserved ejection fraction (HFpEF). Although evidence suggests that the mammalian silent information regulator 1 (Sirt1) orchestrates myocardial lipid metabolism, it is unknown whether its exogenous administration could avoid MCM onset. We investigated whether chronic treatment with recombinant Sirt1 (rSirt1) could halt MCM progression. METHODS db/db mice, an established model of MCM, were supplemented with intraperitoneal rSirt1 or vehicle for 4 weeks and compared with their db/ + heterozygous littermates. At the end of treatment, cardiac function was assessed by cardiac ultrasound and left ventricular samples were collected and processed for molecular analysis. Transcriptional changes were evaluated using a custom PCR array. Lipidomic analysis was performed by mass spectrometry. H9c2 cardiomyocytes exposed to hyperglycaemia and treated with rSirt1 were used as in vitro model of MCM to investigate the ability of rSirt1 to directly target cardiomyocytes and modulate malondialdehyde levels and caspase 3 activity. Myocardial samples from diabetic and nondiabetic patients were analysed to explore Sirt1 expression levels and signaling pathways. RESULTS rSirt1 treatment restored cardiac Sirt1 levels and preserved cardiac performance by improving left ventricular ejection fraction, fractional shortening and diastolic function (E/A ratio). In left ventricular samples from rSirt1-treated db/db mice, rSirt1 modulated the cardiac lipidome: medium and long-chain triacylglycerols, long-chain triacylglycerols, and triacylglycerols containing only saturated fatty acids were reduced, while those containing docosahexaenoic acid were increased. Mechanistically, several genes involved in lipid trafficking, metabolism and inflammation, such as Cd36, Acox3, Pparg, Ncoa3, and Ppara were downregulated by rSirt1 both in vitro and in vivo. In humans, reduced cardiac expression levels of Sirt1 were associated with higher intramyocardial triacylglycerols and PPARG-related genes. CONCLUSIONS In the db/db mouse model of MCM, chronic exogenous rSirt1 supplementation rescued cardiac function. This was associated with a modulation of the myocardial lipidome and a downregulation of genes involved in lipid metabolism, trafficking, inflammation, and PPARG signaling. These findings were confirmed in the human diabetic myocardium. Treatments that increase Sirt1 levels may represent a promising strategy to prevent myocardial lipid abnormalities and MCM development.
Collapse
Affiliation(s)
- Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Department of Cardiology, Zurich University Hospital, Zurich, Switzerland
| | - Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Pisa, Italy
| | | | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andreas Hülsmeier
- Institute for Clinical Chemistry, University Hospital and University of Zürich, Zurich, Switzerland
| | - Christian Matthias Matter
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Department of Cardiology, Zurich University Hospital, Zurich, Switzerland
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital and University of Zürich, Zurich, Switzerland
| | - Giovanni Melina
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Department of Cardiology, Zurich University Hospital, Zurich, Switzerland
| | - Thomas Felix Luscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland.
- Department of Cardiology, Zurich University Hospital, Zurich, Switzerland.
| |
Collapse
|
190
|
Wang LF, Li Q, Wen K, Zhao QH, Zhang YT, Zhao JL, Ding Q, Guan XH, Xiao YF, Deng KY, Xin HB. CD38 Deficiency Alleviates Diabetic Cardiomyopathy by Coordinately Inhibiting Pyroptosis and Apoptosis. Int J Mol Sci 2023; 24:16008. [PMID: 37958991 PMCID: PMC10650707 DOI: 10.3390/ijms242116008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetic cardiomyopathy is one of the diabetes mellitus-induced cardiovascular complications that can result in heart failure in severe cases, which is characterized by cardiomyocyte apoptosis, local inflammation, oxidative stress, and myocardial fibrosis. CD38, a main hydrolase of NAD+ in mammals, plays an important role in various cardiovascular diseases, according to our previous studies. However, the role of CD38 in diabetes-induced cardiomyopathy is still unknown. Here, we report that global deletion of the CD38 gene significantly prevented diabetic cardiomyopathy induced by high-fat diet plus streptozotocin (STZ) injection in CD38 knockout (CD38-KO) mice. We observed that CD38 expression was up-regulated, whereas the expression of Sirt3 was down-regulated in the hearts of diabetic mice. CD38 deficiency significantly promoted glucose metabolism and improved cardiac functions, exemplified by increased left ventricular ejection fraction and fractional shortening. In addition, we observed that CD38 deficiency markedly decreased diabetes or high glucose and palmitic acid (HG + PA)-induced pyroptosis and apoptosis in CD38 knockout hearts or cardiomyocytes, respectively. Furthermore, we found that the expression levels of Sirt3, mainly located in mitochondria, and its target gene FOXO3a were increased in CD38-deficient hearts and cardiomyocytes with CD38 knockdown under diabetic induction conditions. In conclusion, we demonstrated that CD38 deficiency protected mice from diabetes-induced diabetic cardiomyopathy by reducing pyroptosis and apoptosis via activating NAD+/Sirt3/FOXO3a signaling pathways.
Collapse
Affiliation(s)
- Ling-Fang Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.L.); (K.W.); (Q.-H.Z.); (Y.-T.Z.); (J.-L.Z.); (Q.D.); (X.-H.G.); (Y.-F.X.); (K.-Y.D.)
| | | | | | | | | | | | | | | | | | | | - Hong-Bo Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.L.); (K.W.); (Q.-H.Z.); (Y.-T.Z.); (J.-L.Z.); (Q.D.); (X.-H.G.); (Y.-F.X.); (K.-Y.D.)
| |
Collapse
|
191
|
Jiang C, Jin X, Li C, Wen L, Wang Y, Li X, Zhang Z, Tan R. Roles of IL-33 in the Pathogenesis of Cardiac Disorders. Exp Biol Med (Maywood) 2023; 248:2167-2174. [PMID: 37828753 PMCID: PMC10800126 DOI: 10.1177/15353702231198075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 cytokine family and is believed to play important roles in different diseases by binding to its specific receptor suppression of tumorigenicity 2 (ST2). In the heart, IL-33 is expressed in different cells including cardiomyocytes, fibroblasts, endothelium, and epithelium. Although many studies have been devoted to investigating the effects of IL-33 on heart diseases, its roles in myocardial injuries remain obscure, and thus further studies are mandatory to unravel the underlying molecular mechanisms. We highlighted the current knowledge of the molecular and cellular characteristics of IL-33 and then summarized its major roles in different myocardial injuries, mainly focusing on infection, heart transplantation, coronary atherosclerosis, myocardial infarction, and diabetic cardiomyopathy. This narrative review will summarize current understanding and insights regarding the implications of IL-33 in cardiac diseases and its diagnostic and therapeutic potential for cardiac disease management.
Collapse
Affiliation(s)
- Chunjie Jiang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Xuemei Jin
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji 133002, China
| | - Chunlei Li
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Luona Wen
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Yuqi Wang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Xiaojian Li
- Department of Burns, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220 China
| | - Zhi Zhang
- Department of Burns, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220 China
| | - Rongshao Tan
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| |
Collapse
|
192
|
Zhao X, Gao Z, Wen W, Zheng S. Impacts of delta 9-tetrahydrocannabinol against myocardial ischemia/reperfusion injury in diabetic rats: Role of PTEN/PI3K/Akt signaling pathway. CHINESE J PHYSIOL 2023; 66:446-455. [PMID: 38149557 DOI: 10.4103/cjop.cjop-d-23-00103] [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] [Indexed: 12/28/2023] Open
Abstract
Despite the current optimal therapy, patients with myocardial ischemia/reperfusion (IR) injury still experience a high mortality rate, especially when diabetes mellitus is present as a comorbidity. Investigating potential treatments aimed at improving the outcomes of myocardial IR injury in diabetic patients is necessary. Our objective was to ascertain the cardioprotective effect of delta 9-tetrahydrocannabinol (THC) against myocardial IR injury in diabetic rats and examine the role of phosphatase and tensin homolog (PTEN)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway in mediating this effect. Diabetes was induced in male Wistar rats (8-10 weeks old, 200-250 g; n = 60) by a single injection of streptozotocin. The duration of the diabetic period was 10 weeks. During the last 4 weeks of diabetic period, rats were treated with THC (1.5 mg/kg/day; intraperitoneally), either alone or in combination with LY294002, and then underwent IR intervention. After 24 h of reperfusion, infarct size, cardiac function, lactate dehydrogenase (LDH) and cardiac-specific isoform of troponin-I (cTn-I) levels, myocardial apoptosis, oxidative stress markers, and expression of PTEN, PI3K, and Akt proteins were evaluated. THC pretreatment resulted in significant improvements in infarct size and cardiac function and decreases in LDH and cTn-I levels (P < 0.05). It also reduced myocardial apoptosis and oxidative stress, accompanied by the downregulation of PTEN expression and activation of the PI3K/Akt signaling pathway (P < 0.05). LY294002 pretreatment abolished the cardioprotective action of THC. This study revealed the cardioprotective effects of THC against IR-induced myocardial injury in diabetic rats and also suggested that the mechanism may be associated with enhanced activity of the PI3K/Akt signaling pathway through the reduction of PTEN phosphorylation.
Collapse
Affiliation(s)
- Xiaohua Zhao
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, China
| | - Zhao Gao
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, China
| | - Wenbin Wen
- Department of Nephrology, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Shikang Zheng
- Department of Cardiology, Ankang Central Hospital, Ankang, China
| |
Collapse
|
193
|
Hu S, Luo J, Guo P, Du T, Liu X, He M, Li J, Ma T, Liu B, Huang M, Fang Q, Wang Y. Lentinan alleviates diabetic cardiomyopathy by suppressing CAV1/SDHA-regulated mitochondrial dysfunction. Biomed Pharmacother 2023; 167:115645. [PMID: 37804808 DOI: 10.1016/j.biopha.2023.115645] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/30/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), characterized by mitochondrial dysfunction and impaired energetics as contributing factors, significantly contributes to high mortality in patients with diabetes. Targeting key proteins involved in mitochondrial dysfunction might offer new therapeutic possibilities for DCM. Lentinan (LNT), a β-(1,3)-glucan polysaccharide obtained from lentinus edodes, has demonstrated biological activity in modulating metabolic syndrome. In this study, the authors investigate LNT's pharmacological effects on and mechanisms against DCM. The results demonstrate that administering LNT to db/db mice reduces cardiomyocyte apoptosis and mitochondrial dysfunction, thereby preventing DCM. Notably, these effects are fully negated by Caveolin-1 (CAV1) overexpression both in vivo and in vitro. Further studies and bioinformatics analysis uncovered that CAV1 bound with Succinate dehydrogenase subunit A (SDHA), triggering the following ubiquitination and degradation of SDHA, which leads to mitochondrial dysfunction and mitochondria-derived apoptosis under PA condition. Silencing CAV1 leads to reduced apoptosis and improved mitochondrial function, which is blocked by SDHA knockdown. In conclusion, CAV1 directly interacts with SDHA to promote ubiquitination and proteasomal degradation, resulting in mitochondrial dysfunction and mitochondria-derived apoptosis, which was depressed by LNT administration. Therefore, LNT may be a potential pharmacological agent in preventing DCM, and targeting the CAV1/SDHA pathway may be a promising therapeutic approach for DCM.
Collapse
Affiliation(s)
- Shuiqing Hu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Guo
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Tingyi Du
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiaohui Liu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Miaomiao He
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jie Li
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Tingqiong Ma
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Bo Liu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Man Huang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Qin Fang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Yan Wang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
194
|
Han X, Zhou W, Zhang J, Tu Y, Wei J, Zheng R, Zhu J, Xu D, Ying H, Wu G, Shi Q, Liang G. Linderalactone mitigates diabetic cardiomyopathy in mice via suppressing the MAPK/ATF6 pathway. Int Immunopharmacol 2023; 124:110984. [PMID: 37757635 DOI: 10.1016/j.intimp.2023.110984] [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: 07/12/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a challenging diabetic complication that manifests as chronic inflammation. Yet, the mechanism underlying diabetes-associated myocardial injury is not fully understood. We investigated the pharmacological effects and mechanisms of linderalactone, a natural compound that can prevent diabetes-induced cardiomyopathy in mice. Diabetes was induced by a single dose of streptozotocin (120 mg/kg, i.p.). Diabetic mice were administrated with linderalactone (2.5 or 5 mg/kg) by gavage for five weeks. Harvested heart tissues were then subjected to RNA-sequencing analysis to explore the potential mechanism of linderalactone. Linderalactone prevented heart dysfunction by inhibiting myocardial hypertrophy, fibrosis, and inflammation, without altering blood glucose. RNA-sequencing indicated that linderalactone exerted its cardioprotective effects mainly by affecting the mitogen-activated protein kinase (MAPK)/ activating transcription factor 6 (ATF6) pathway. Linderalactone also suppressed endoplasmic reticulum (ER) stress mediated by the diabetes-activated MAPKs/ATF6 pathway, thereby reducing myocardial hypertrophy and inflammation in heart tissues and in cultured cardiomyocytes. Inhibition of MAPKs or a deficiency of ATF6 in cardiomyocytes mimicked the linderalactone-associated decreases in high glucose-induced hypertrophy and inflammation. Linderalactone showed beneficial effects in alleviating diabetic cardiomyopathy, in part by modulating the MAPK/ATF6 signaling pathway to mitigate myocardial hypertrophy and inflammation. Linderalactone may have clinical utility in the treatment for diabetes-associated cardiomyopathy.
Collapse
Affiliation(s)
- Xue Han
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenwei Zhou
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Jiajia Zhang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Yu Tu
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Jiajia Wei
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Ruyi Zheng
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Jian Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Diyun Xu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huazhong Ying
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Gaojun Wu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Qiaojuan Shi
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China.
| | - Guang Liang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
195
|
Li D, Li Y, Ding H, Wang Y, Xie Y, Zhang X. Cellular Senescence in Cardiovascular Diseases: From Pathogenesis to Therapeutic Challenges. J Cardiovasc Dev Dis 2023; 10:439. [PMID: 37887886 PMCID: PMC10607269 DOI: 10.3390/jcdd10100439] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Cellular senescence (CS), classically considered a stable cell cycle withdrawal, is hallmarked by a progressive decrease in cell growth, differentiation, and biological activities. Senescent cells (SNCs) display a complicated senescence-associated secretory phenotype (SASP), encompassing a variety of pro-inflammatory factors that exert influence on the biology of both the cell and surrounding tissue. Among global mortality causes, cardiovascular diseases (CVDs) stand out, significantly impacting the living quality and functional abilities of patients. Recent data suggest the accumulation of SNCs in aged or diseased cardiovascular systems, suggesting their potential role in impairing cardiovascular function. CS operates as a double-edged sword: while it can stimulate the restoration of organs under physiological conditions, it can also participate in organ and tissue dysfunction and pave the way for multiple chronic diseases under pathological states. This review explores the mechanisms that underlie CS and delves into the distinctive features that characterize SNCs. Furthermore, we describe the involvement of SNCs in the progression of CVDs. Finally, the study provides a summary of emerging interventions that either promote or suppress senescence and discusses their therapeutic potential in CVDs.
Collapse
Affiliation(s)
- Dan Li
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Hong Ding
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yuqin Wang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yafei Xie
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Xiaowei Zhang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| |
Collapse
|
196
|
Duan Y, Ye L, Shu Q, Huang Y, Zhang H, Zhang Q, Ding G, Deng Y, Li C, Yin L, Wang Y. Abnormal left ventricular systolic reserve function detected by treadmill exercise stress echocardiography in asymptomatic type 2 diabetes. Front Cardiovasc Med 2023; 10:1253440. [PMID: 37928757 PMCID: PMC10622805 DOI: 10.3389/fcvm.2023.1253440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Aims Subclinical left ventricular (LV) dysfunction may occur in T2DM patients at the early asymptomatic stage, and LV reserve function is a sensitive index to detect subtle LV dysfunction. The purpose of our study is (1) to assess the LV reserve function using treadmill exercise stress echocardiography (ESE) in asymptomatic type 2 diabetes mellitus (T2DM) patients; (2) to explore the link of serum biological parameters and LV reserve function. Methods This study included 84 patients with asymptomatic T2DM from September 2021 to July 2022 and 41 sex- and age-matched healthy controls during the corresponding period. All subjects completed treadmill ESE, LV systolic function-related parameters such as global longitudinal strain (GLS) and systolic strain rate (SRs), as well as diastolic function-related parameters such as E wave (E), early diastolic velocity (e'), E/e' ratio, early diastolic SR (SRe), and late diastolic SR (SRa) were compared at rest and immediately after exercise. The difference between LV functional parameters after treadmill exercise and its corresponding resting value was used to compute LV reserve function. In addition, the associations of LV reserve function and serum biological parameters were analyzed. Results Patients with T2DM did not significantly vary from the controls in terms of alterations in LV diastolic reserve measures, the changes of LVGLS and SRs (ΔGLS: 2.19 ± 2.72% vs. 4.13 ± 2.79%, P < 0.001 and ΔSRs:0.78 ± 0.33 s-1 vs. 1.02 ± 0.28 s-1, P < 0.001) in the T2DM group were both lower than those in the control group. Glycated hemoglobin (HbA1c), N-terminal pro-brain natriuretic peptide (NTproBNP), waist circumference, and high-sensitive C-reactive protein (hsCRP) were identified as independent predictors of LV systolic reserve by stepwise multiple linear regression analysis. Conclusion LV systolic reserve function, as measured by pre- and post-exercise differences in GLS and SRs were significantly impaired in patients with asymptomatic T2DM, whereas diastolic reserve remained normal during exercise and was comparable to that of the control group. This was different from previous findings. High levels of HbA1c, NTproBNP, hsCRP, and increasing waist circumference were independent predictors of LV systolic reserve.
Collapse
Affiliation(s)
- Yuyou Duan
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Luwei Ye
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qinglan Shu
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Huang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hongmei Zhang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qingfeng Zhang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Geqi Ding
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Deng
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunmei Li
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lixue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Wang
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
197
|
Cai L, Tan Y, Watson S, Wintergerst K. Diabetic cardiomyopathy - Zinc preventive and therapeutic potentials by its anti-oxidative stress and sensitizing insulin signaling pathways. Toxicol Appl Pharmacol 2023; 477:116694. [PMID: 37739320 PMCID: PMC10616760 DOI: 10.1016/j.taap.2023.116694] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Oxidative stress and insulin resistance are two key mechanisms for the development of diabetic cardiomyopathy (DCM, cardiac remodeling and dysfunction). In this review, we discussed how zinc and metallothionein (MT) protect the heart from type 1 or type 2 diabetes (T1D or T2D) through its anti-oxidative function and insulin-mediated PI3K/Akt signaling activation. Both T1D and T2D-induced DCM, shown by cardiac structural remodeling and dysfunction, in wild-type mice, but not in cardiomyocyte-specific overexpressing MT mice. In contrast, mice with global MT gene deletion were more susceptible to the development of DCM. When we used zinc to treat mice with either T1D or T2D, cardiac remodeling and dysfunction were significantly prevented along with increased cardiac MT expression. To support the role of zinc homeostasis in insulin signaling pathways, treatment of diabetic mice with zinc showed the preservation of phosphorylation levels of insulin-mediated glucose metabolism-related Akt2 and GSK-3β and even rescued cardiac pathogenesis induced by global deletion of Akt2 gene in a MT-dependent manner. These results suggest the protection by zinc from DCM is through both the induction of MT and sensitization of insulin signaling. Combined our own and other works, this review comprehensively summarized the roles of zinc homeostasis in the development and progression of DCM and its therapeutic implications. At the end, we provided pre-clinical and clinical evidence for the preventive and therapeutic potential of zinc supplementation through its anti-oxidative stress and sensitizing insulin signaling actions. Understanding the intricate connections between zinc and DCM provides insights for the future interventional approaches.
Collapse
Affiliation(s)
- Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America; Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Sara Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| | - Kupper Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| |
Collapse
|
198
|
ALTamimi JZ, AlFaris NA, Alshammari GM, Alagal RI, Aljabryn DH, Yahya MA. Esculeoside A Decreases Diabetic Cardiomyopathy in Streptozotocin-Treated Rats by Attenuating Oxidative Stress, Inflammation, Fibrosis, and Apoptosis: Impressive Role of Nrf2. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1830. [PMID: 37893548 PMCID: PMC10608477 DOI: 10.3390/medicina59101830] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: This experiment evaluated the preventative influence of the tomato-derived Esculeoside A (ESA) on diabetic cardiomyopathy in type 1 diabetes mellitus (T1DM) in rats induced by streptozotocin (STZ). It also examined whether the activation of Nrf2 signaling affords this protection. Materials and Methods: Adult male Wistar control nondiabetic rats and rats with T1DM (STZ-T1DM) were given either carboxymethylcellulose as a vehicle or ESA (100 mg/kg) (eight rats/group) orally daily for 12 weeks. A group of STZ-T1DM rats was also treated with 100 mg/kg ESA and co-treated i.p. with 2 mg/kg (twice/week), brusatol, and Nrf2 inhibitors for 12 weeks. Results and Conclusions: Treatment with ESA prevented the gain in heart weight and cardiomyocyte hypertrophy and improved the left ventricular (LV) systolic and diastolic function (LV) in the STZ-T1DM rat group. Likewise, it reduced their serum levels of triglycerides, cholesterol, and low-density lipoproteins (LDL-c), as well as their LV mRNA, cytoplasmic total, and nuclear total levels of NF-κB. ESA also reduced the total levels of malondialdehyde, tumor necrosis factor-α, interleukine-6 (IL-6), Bax, cytochrome-c, and caspase-3 in the LV of the STZ-T1DM rats. In parallel, ESA enhanced the nuclear and cytoplasmic levels of Nrf2 and the levels of superoxide dismutase, glutathione, and heme oxygenase-1, but decreased the mRNA and cytoplasmic levels of keap-1 in the LVs of the STZ-T1DM rats. Interestingly, ESA did not affect the fasting insulin and glucose levels of the diabetic rats. All of these beneficially protective effects of ESA were not seen in the ESA-treated rats that received brusatol. In conclusion, ESA represses diabetic cardiomyopathy in STZ-diabetic hearts by activating the Nrf2/antioxidant/NF-κB axis.
Collapse
Affiliation(s)
- Jozaa Z. ALTamimi
- Department of Physical Sports Sciences, College of Education, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (J.Z.A.); (D.H.A.)
| | - Nora A. AlFaris
- Department of Physical Sports Sciences, College of Education, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (J.Z.A.); (D.H.A.)
| | - Ghedeir M. Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (G.M.A.); (M.A.Y.)
| | - Reham I. Alagal
- Department of Health Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Dalal H. Aljabryn
- Department of Physical Sports Sciences, College of Education, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (J.Z.A.); (D.H.A.)
| | - Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (G.M.A.); (M.A.Y.)
| |
Collapse
|
199
|
Saha S, Fang X, Green CD, Das A. mTORC1 and SGLT2 Inhibitors-A Therapeutic Perspective for Diabetic Cardiomyopathy. Int J Mol Sci 2023; 24:15078. [PMID: 37894760 PMCID: PMC10606418 DOI: 10.3390/ijms242015078] [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: 09/01/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic cardiomyopathy is a critical diabetes-mediated co-morbidity characterized by cardiac dysfunction and heart failure, without predisposing hypertensive or atherosclerotic conditions. Metabolic insulin resistance, promoting hyperglycemia and hyperlipidemia, is the primary cause of diabetes-related disorders, but ambiguous tissue-specific insulin sensitivity has shed light on the importance of identifying a unified target paradigm for both the glycemic and non-glycemic context of type 2 diabetes (T2D). Several studies have indicated hyperactivation of the mammalian target of rapamycin (mTOR), specifically complex 1 (mTORC1), as a critical mediator of T2D pathophysiology by promoting insulin resistance, hyperlipidemia, inflammation, vasoconstriction, and stress. Moreover, mTORC1 inhibitors like rapamycin and their analogs have shown significant benefits in diabetes and related cardiac dysfunction. Recently, FDA-approved anti-hyperglycemic sodium-glucose co-transporter 2 inhibitors (SGLT2is) have gained therapeutic popularity for T2D and diabetic cardiomyopathy, even acknowledging the absence of SGLT2 channels in the heart. Recent studies have proposed SGLT2-independent drug mechanisms to ascertain their cardioprotective benefits by regulating sodium homeostasis and mimicking energy deprivation. In this review, we systematically discuss the role of mTORC1 as a unified, eminent target to treat T2D-mediated cardiac dysfunction and scrutinize whether SGLT2is can target mTORC1 signaling to benefit patients with diabetic cardiomyopathy. Further studies are warranted to establish the underlying cardioprotective mechanisms of SGLT2is under diabetic conditions, with selective inhibition of cardiac mTORC1 but the concomitant activation of mTORC2 (mTOR complex 2) signaling.
Collapse
Affiliation(s)
- Sumit Saha
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Xianjun Fang
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Christopher D. Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.S.); (X.F.); (C.D.G.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
200
|
Titus AS, Sung EA, Zablocki D, Sadoshima J. Mitophagy for cardioprotection. Basic Res Cardiol 2023; 118:42. [PMID: 37798455 PMCID: PMC10556134 DOI: 10.1007/s00395-023-01009-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial function is maintained by several strictly coordinated mechanisms, collectively termed mitochondrial quality control mechanisms, including fusion and fission, degradation, and biogenesis. As the primary source of energy in cardiomyocytes, mitochondria are the central organelle for maintaining cardiac function. Since adult cardiomyocytes in humans rarely divide, the number of dysfunctional mitochondria cannot easily be diluted through cell division. Thus, efficient degradation of dysfunctional mitochondria is crucial to maintaining cellular function. Mitophagy, a mitochondria specific form of autophagy, is a major mechanism by which damaged or unnecessary mitochondria are targeted and eliminated. Mitophagy is active in cardiomyocytes at baseline and in response to stress, and plays an essential role in maintaining the quality of mitochondria in cardiomyocytes. Mitophagy is mediated through multiple mechanisms in the heart, and each of these mechanisms can partially compensate for the loss of another mechanism. However, insufficient levels of mitophagy eventually lead to mitochondrial dysfunction and the development of heart failure. In this review, we discuss the molecular mechanisms of mitophagy in the heart and the role of mitophagy in cardiac pathophysiology, with the focus on recent findings in the field.
Collapse
Affiliation(s)
- Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA.
| |
Collapse
|