1
|
Zhu M, Liu W, Su S, Gong M, Liao G, Fu F, Chen G, Rao Z, Cheng J, Liu J, Lu Y, Chen Y. Reprogramming of lipids and amino acids metabolism is an early event in myocardium of type 1 diabetic rhesus monkeys. J Pharm Biomed Anal 2025; 258:116699. [PMID: 39914331 DOI: 10.1016/j.jpba.2025.116699] [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: 09/05/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/10/2025]
Abstract
Diabetic cardiomyopathy (DC) refers to the abnormal myocardial structure and performance induced by diabetes. Although numerous studies have been carried out, the pathophysiological mechanisms of cardiovascular disorders during diabetes have not been fully clarified. Here, we compared the cardiomyopathy of healthy rhesus monkeys and rhesus monkeys with a history of streptozocin induced type 1 diabetes (T1D) over 7 years. Through comparing the cardiac function using echocardiography, and detecting the serum biochemical indexes, and changes of left ventricle (LV), we found that decreased systolic function, higher blood glycosylated hemoglobin A1c (HbA1C) level, hyperglycemia, and hyperlipidemia were early events in diabetic rhesus monkeys. In addition, cardiac histological analysis showed mildly fibrosis and early myocardial hypertrophy, as evidenced by increased Sirius red stained area and cross-sectional area of left ventricle. Transcriptome results revealed that the nutrients metabolism and extracellular matrix related pathways were markedly changed in the left ventricle of diabetic monkeys. Targeted metabolomics and targeted lipid metabolomics further revealed that disturbed amino acid metabolism and lipid accumulation in the LV of diabetic monkeys manifested by accumulated branched chain amino acids (BCAAs) and triglycerides (TAGs), and reduced contents of sphingolipids, glycerophospholipids, cholesteryl esters and carnitines. In conclusion, we reported here for the first time that diabetes lasting for more than 7 years leads to some early pathological changes of myocardium in rhesus monkeys. The cardiac function is mildly compromised and the reprogramming of lipids and amino acids metabolism might play important roles in the progression of DC.
Collapse
Affiliation(s)
- Min Zhu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wen Liu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Shan Su
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Meng Gong
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, PR China; Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Guangneng Liao
- Animal experimental center of West China hospital, Sichuan University, Chengdu, PR China
| | - Fudong Fu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, PR China
| | - Gen Chen
- Development and Application of Human Major Disease Monkey Model Key Laboratory of Sichuan Province, Sichuan Hengshu Bio-Technology Co. Ltd., Sichuan 644600, PR China
| | - Zhiyong Rao
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jingqiu Cheng
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jingping Liu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yanrong Lu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Younan Chen
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, PR China.
| |
Collapse
|
2
|
Mahmood NMS, Mahmud AMR, Maulood IM. Vascular actions of Ang 1-7 and Ang 1-8 through EDRFs and EDHFs in non-diabetes and diabetes mellitus. Nitric Oxide 2025; 156:9-26. [PMID: 40032212 DOI: 10.1016/j.niox.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in regulating vascular homeostasis, while angiotensin 1-8 (Ang 1-8) traditionally dominates as a vasoconstrictor factor. However, the discovery of angiotensin 1-7 (Ang 1-7) and Ang 1-8 has revealed counter-regulatory mechanisms mediated through endothelial-derived relaxing factors (EDRFs) and endothelial-derived hyperpolarizing factors (EDHFs). This review delves into the vascular actions of Ang 1-7 and Ang 1-8 in both non-diabetes mellitus (non-DM) and diabetes mellitus (DM) conditions, highlighting their effects on vascular endothelial cell (VECs) function as well. In a non-DM vasculature context, Ang 1-8 demonstrate dual effect including vasoconstriction and vasodilation, respectively. Additionally, Ang 1-7 induces vasodilation upon nitric oxide (NO) production as a prominent EDRFs in distinct mechanisms. Further research elucidating the precise mechanisms underlying the vascular actions of Ang 1-7 and Ang 1-8 in DM will facilitate the development of tailored therapeutic interventions aimed at preserving vascular health and preventing cardiovascular complications.
Collapse
Affiliation(s)
- Nazar M Shareef Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
| | - Almas M R Mahmud
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ismail M Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
3
|
Li X, Qu S. Novel insights into the central protective role of ACE2 in diabetic cardiomyopathy: from underlying signaling pathways to therapeutic perspectives. Mol Cell Biochem 2025; 480:3535-3551. [PMID: 39928210 DOI: 10.1007/s11010-024-05196-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a cardiac complication specific to individuals with diabetes. It is defined as abnormalities of myocardial structure and function in diabetic patients who do not exhibit any obvious coronary artery disease, hypertensive heart disease, valvular heart disease, or inherited cardiomyopathy. A significant cardiovascular protective factor identified recently is angiotensin-converting enzyme 2 (ACE2), which is a rising star in the renin angiotensin system (RAS) and is responsible for the onset and progression of DCM. Nonetheless, there is not a comprehensive review outlining ACE2's effect on DCM. From the perspective of the pathogenesis of DCM, this review summarizes the myocardial protective role of ACE2 in the aspects of alleviating myocardial structure and dysfunction, correcting energy metabolism disorders, and restoring vascular function. Concurrently, we propose the connections between ACE2 and underlying signaling pathways, including ADAM17, Apelin/APJ, and Nrf2. Additionally, we highlight ACE2-related pharmaceutical treatment options and clinical application prospects for preventing and managing DCM. Further and underlying research is extensively required to completely comprehend the principal pathophysiological mechanism of DCM and the distinctive function of ACE2, switching experimental findings into clinical practice and identifying efficient therapeutic approaches.
Collapse
Affiliation(s)
- Xinyi Li
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China.
| |
Collapse
|
4
|
Zhang X, Sun J, Zhu X, Yang Z, Zhu Z, Zhou M, Li C, Yu H, Gan X. Low-magnitude high-frequency vibration ameliorates high glucose-induced endothelial injury by restoring mitochondrial function via AMPK/mTOR pathway. J Histotechnol 2025; 48:82-92. [PMID: 39564647 DOI: 10.1080/01478885.2024.2429855] [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: 06/14/2024] [Accepted: 11/10/2024] [Indexed: 11/21/2024]
Abstract
High glucose-induced dysfunction of endothelial cells is a critical and initiating factor in the genesis of diabetic vascular complications. Low-magnitude high-frequency vibration (LMHFV) is a non-invasive biophysical intervention. It has been reported that it exhibits protective effects on high glucose-induced osteoblast dysfunction, but little was known on diabetic vascular complications. In this work, we aim to clarify the role of LMHFV on high glucose-induced endothelial dysfunction and hypothesized that the protective effects functioned through adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway. We cultured primary murine aortic endothelial cells (MAECs) in normal or HG medium, respectively, before exposing to LMHFV. The tube formation, paracellular permeability assay, and aortic ring sprouting assay showed that the high glucose injured-function of MAECs was improved after LMHFV treatment. The intracellular ROS generation analysis, mitochondrial complex I activities measurement, ATP measurement and mitochondrial membrane potential (MMP), and mitochondrial ROS generation analysis of MAECs indicated that mitochondrial function was restored by LMHFV loading in a high glucose environment. Mechanically, western blot assays showed that AMPK phosphorylation was promoted and mTOR was inhibited in LMHFV-induced endothelial function restoration. After the administration of the AMPK inhibitor, Compound C, these protective effects resulting from LMHFV are reversed. These findings suggest that LMHFV plays a significant role in protecting endothelial cells' function and mitochondrial function in high glucose-induced injured MAECs via AMPK/mTOR signalling.
Collapse
Affiliation(s)
- Xidan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiyu Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiting Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhenghao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Min Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Yang T, Luo L, Luo X, Liu X. Metabolic crosstalk and therapeutic interplay between diabetes and hyperuricemia. Diabetes Res Clin Pract 2025; 224:112204. [PMID: 40294652 DOI: 10.1016/j.diabres.2025.112204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Hyperuricemia and diabetes mellitus (DM) are prevalent metabolic disorders with high comorbidity, imposing a substantial global public health burden. Their coexistence is not merely additive but synergistic, exacerbating metabolic dysregulation through mechanisms such as insulin resistance and β-cell apoptosis, ultimately establishing a vicious cycle. Both disorders induce acute and chronic damage to vital organs, particularly the cardiovascular, renal systems. Hyperuricemia aggravates diabetic complications, notably diabetic cardiomyopathy, nephropathy and retinopathy via oxidative stress, inflammation, and metabolic dysregulation.Current urate-lowering therapies (ULTs), such as xanthine oxidase inhibitors and urate transporter 1 (URAT1, also known as SLC22A12) antagonists, demonstrate potential benefits in ameliorating diabetic complications but face challenges including safety concerns and dose adjustments. Similarly, several glucose-lowering drugs also exhibit the benefits of improving hyperuricemia. This review summarizes the metabolic crosstalk and therapeutic interplay between hyperuricemia and DM, examines the pathogenic role of uric acid in diabetic complications, and discusses the benefits and challenges of existing ULTs and glucose-lowering drugs in disrupting this cycle of metabolic dysregulation and concurrent organ damage. We hope our findings deepen the comprehension of the intricate metabolic crosstalk between glucose and urate homeostasis, providing novel therapeutic insights for patients with comorbid DM and hyperuricemia.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China
| | - Lingyun Luo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030 Hubei, China
| | - Xuelian Luo
- Department of Oncology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Xiaolei Liu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030 Hubei, China.
| |
Collapse
|
6
|
Yao J, Zhou Y, Xie R, Wang C, Zhu Y, Li W, Zhang Y. Prophylactic administration of tirofiban prevents ischemic events in endovascular treatment of unruptured intracranial aneurysms. Neurosurg Rev 2025; 48:440. [PMID: 40410393 DOI: 10.1007/s10143-025-03601-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/03/2025] [Accepted: 05/17/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND Stents-assisted coiling (SAC) and flow diverters (FD) are widely used in the endovascular treatment of intracranial aneurysms. However, due to the thrombogenicity of metallic implants, their application may increase the risk of ischemic events. This study aims to evaluate the efficacy of prophylactic tirofiban with dual antiplatelet treatment (DAPT) in unruptured intracranial aneurysms (UIA) patients treated with SAC or FD. METHODS This single center retrospective study included patients with UIAs treated with SAC or FD. Data collected included demographic information, imaging findings, laboratory results, and perioperative complications. Multivariate logistic regression analysis was used to identify independent risk factors for ischemic events. Patients were stratified based on these risk factors, and the efficacy of tirofiban was evaluated across different risk groups. RESULTS A total of 420 patients were included in the study, of whom 22(5.2%) experienced ischemic events. Among them, eight patients (3.3%) were in the tirofiban group and 14 patients (8.0%) were in the DAPT (non-tirofiban) group. Multivariate logistic regression identified independent risk factors for ischemic events, including posterior circulation aneurysm (OR: 2.87, 95% CI: 1.06-7.78; P = 0.038) and diabetes (OR: 4.05, 95% CI: 1.50-10.96; P = 0.006). The prophylactic use of tirofiban combined with DAPT can effectively reduce postoperative ischemic events (OR: 0.35, 95% CI: 0.14-0.91; P = 0.032) without increasing the risk of hemorrhage. CONCLUSION This study demonstrates that prophylactic use of tirofiban can effectively reduce postoperative ischemic events in UIA patients receiving SAC or FD treatments without increasing the risk of hemorrhage.
Collapse
Affiliation(s)
- Jinbiao Yao
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yangyang Zhou
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruhang Xie
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chao Wang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yongnan Zhu
- Neurosurgery, Beijing Geriatric Hospital, Beijing, China
| | - Wenqiang Li
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Ying Zhang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
8
|
Nunan E, Huff DR, Gore JL, Wright CL, Harris T, Butler L, Padgett CA, Rochowski MT, Lovern PC, Boolani A, Valdez C, Butcher JT. Targeting Myostatin as an Adjunct Treatment for the Preservation of Cardiometabolic and Skeletal Muscle Function in Type 1 Diabetes. Int J Mol Sci 2025; 26:4830. [PMID: 40429969 PMCID: PMC12112738 DOI: 10.3390/ijms26104830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Type 1 Diabetes Mellitus (T1D) is a disease characterized by the destruction of pancreatic beta cells. The subsequent loss of insulin production results in hyperglycemia, muscle wasting, and vascular dysfunction. Due to an inability to appropriately maintain glucose homeostasis, patients afflicted with T1D suffer from increased morbidity and early mortality. Skeletal muscle is the body's largest metabolic reservoir, absorbing significant amounts of glucose from the bloodstream and physical exercise is known to improve and prevent the progression of pathological outcomes, but many T1D patients are unable to exercise at a level that conveys benefit. Thus, directly targeting muscle mass and function may prove beneficial for improving T1D patient outcomes, independent of exercise. A potent negative regulator of skeletal muscle has been identified as being upregulated in T1D patients, namely the myokine myostatin. Our hypothesis is that targeting myostatin (via genetic deletion) will prevent glucose dysfunction in a T1D model, preserve skeletal muscle function, and protect against vascular and renal dysfunction. Our methods utilized adult male mice with (WT) and without myostatin (Myo KO), in combination with the chemical induction of T1D (streptozotocin). Experimental outcomes included the assessment of glucose homeostasis (plasma glucose, HbA1c, IGTT), metabolism, muscle function (in vivo plantarflexion), and skeletal muscle vascular function (ex vivo pressure myography). Our results described systemic benefits from myostatin deletion in the T1D model, independent of insulin, including the following: inhibition of T1D-induced increases in plasma glucose, prevention of functional deficits in muscle performance, and preservation of fluid dynamics. Further, endothelial function was preserved with myostatin deletion. Taken together, these data inform upon the use of myostatin inhibition as a therapeutic target for effective treatment and management of the cardiometabolic and skeletal muscle dysfunction that occurs with T1D.
Collapse
Affiliation(s)
- Emily Nunan
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Denton R. Huff
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jillian L. Gore
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Carson L. Wright
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Tag Harris
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Landon Butler
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Caleb A. Padgett
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Matthew T. Rochowski
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Pamela C. Lovern
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Ali Boolani
- Human Performance and Nutrition Research Institute, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Cammi Valdez
- Department of Physical Sciences, Northeastern State University, Tahlequah, OK 74464, USA;
| | - Joshua T. Butcher
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
9
|
Quisi A, Nacar Quisi NS, Alıcı G, Donma İ, Yıldırım A, Genç Ö. Effect of dapagliflozin on the no-reflow phenomenon in patients with acute myocardial infarction and type II diabetes mellitus. Acta Cardiol 2025:1-9. [PMID: 40366712 DOI: 10.1080/00015385.2025.2500892] [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: 03/15/2025] [Revised: 04/07/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE This study aimed to assess the effect of dapagliflozin on the no-reflow phenomenon in patients with type II diabetes mellitus (T2DM) and acute myocardial infarction (AMI) who underwent percutaneous coronary intervention (PCI). METHODS This single-center, observational cohort study included a total of 829 consecutive T2DM patients who were diagnosed with AMI and underwent PCI within 24 h of the onset of symptoms. Only patients using dapagliflozin (10 mg per day) for more than one year were considered as patients using dapagliflozin. The no-reflow phenomenon was defined as inadequate myocardial perfusion within a segment of the coronary circulation without angiographic evidence of mechanical vessel obstruction, dissection, or residual stenosis after PCI. RESULTS Four hundred and thirty-four patients were diagnosed with ST-segment elevation myocardial infarction (STEMI), and 395 patients were diagnosed with non-ST-segment elevation myocardial infarction (NSTEMI). Forward conditional logistic regression analysis demonstrated that the estimated glomerular filtration rate (OR = 0.940, 95% CI: 0.900 to 0.982, p = 0.006), SYNTAX score I (OR = 1.338, 95% CI: 1.179 to 1.520, p < 0.001), and dapagliflozin use (OR = 0.030, 95% CI: 0.004 to 0.228, p = 0.001) were independent predictors of the no-reflow phenomenon in STEMI. However, dapagliflozin use (OR = 0.112, 95% CI: 0.013 to 0.933, p = 0.043) was the only independent predictor of the no-reflow phenomenon in NSTEMI. CONCLUSION Lower rates of the no-reflow phenomenon were observed in T2DM patients taking dapagliflozin, diagnosed with AMI, and underwent PCI. However, this finding requires further investigation.
Collapse
Affiliation(s)
- Alaa Quisi
- Department of Cardiology, Medline Adana Hospital, Adana, Turkey
| | | | - Gökhan Alıcı
- Department of Cardiology, Adana City Training and Research Hospital, Adana, Turkey
| | - İdil Donma
- Department of Cardiology, Adana City Training and Research Hospital, Adana, Turkey
| | - Abdullah Yıldırım
- Department of Cardiology, Adana City Training and Research Hospital, Adana, Turkey
| | - Ömer Genç
- Department of Cardiology, Basaksehir Cam & Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
10
|
Gao X, Chen T, Zhou F, Sun Y, Zhang J, Li X, Zhao W, Li Y, Shi Y, Niu K, Wang Y, Zhang Y, Zhang W. The association between different insulin resistance surrogates and all-cause mortality and cardiovascular mortality in patients with metabolic dysfunction-associated steatotic liver disease. Cardiovasc Diabetol 2025; 24:200. [PMID: 40346671 PMCID: PMC12065324 DOI: 10.1186/s12933-025-02758-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/26/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is closely associated with insulin resistance (IR). However, the prognostic value of different alternative IR surrogates in patients with MASLD remains unclear. This study aimed to evaluate the association between various IR indices and all-cause mortality and cardiovascular mortality in MASLD patients. METHODS A total of 8,753 adults aged ≥ 20 years with MASLD from the National Health and Nutrition Examination Survey (NHANES, 2003-2018) were included, and their mortality data were obtained from the National Death Index (NDI). Insulin resistance surrogates [including the triglyceride-glucose (TyG) index, TyG-body mass index (TyG-BMI), TyG-waist circumference index, TyG-waist-to-height ratio index, and Homeostatic Model Assessment for IR] were stratified into quartiles. Cox proportional hazards models, receiver operating characteristic (ROC) curve analysis, restricted cubic spline (RCS), mediation analyses, and subgroup analyses were used to explore the associations between these indices and all-cause mortality as well as cardiovascular mortality in MASLD patients. RESULTS During a median follow-up of 98 months, 1,234 deaths were observed, including 409 cardiovascular disease (CVD)-related deaths. In the fully adjusted model, higher quartiles of TyG-related indices were significantly associated with an increased risk of all-cause mortality in MASLD patients. Furthermore, the TyG-BMI index was associated with both all-cause mortality and CVD mortality [all-cause mortality: HR (95% CI) 2.84 (1.73-4.67), P < 0.001; CVD mortality: HR (95% CI) 5.32 (2.26-12.49), P < 0.001]. The RCS analyses indicated a U-shaped relationship between TyG-BMI and mortality, with a threshold value of 270.49. Subgroup analyses demonstrated that TyG-related indices had stronger associations with mortality in elderly MASLD patients. CONCLUSIONS Our findings highlight the prognostic value of IR indices, particularly TyG-BMI index, in predicting all-cause mortality and CVD mortality in MASLD patients.
Collapse
Affiliation(s)
- Xin Gao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Tianyi Chen
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Feilong Zhou
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yanmei Sun
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Jiaqi Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Xinhao Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Weijie Zhao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yunxin Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yanlong Shi
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Kaiyi Niu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yizhu Wang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China.
- Department of Hepatobiliary Surgery, The Second Hospital of Shangdong University, Jinan, Shandong Province, China.
| | - Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
11
|
Pinto FR, Cascos E, Pérez-López E, Baile-González M, Martín Rodríguez C, Pascual Cascón MJ, Luque M, Esquirol A, Calvo CM, Peña-Muñóz F, Fernando IH, Oiartzabal Ormtegi I, Sáez Marín AJ, Fernández-Luis S, Domínguez-García JJ, Fernández SV, López Lorenzo JL, Girón MFDS, Pinedo LG, García L, González-Rodriguez AP, Torrado T, Filaferro S, Basalobre P, López-García A, Ortí G, Jurado Chacón M, Queralt Salas M. Early cardiac events after haploidentical hematopoietic cell transplantation with post-transplant cyclophosphamide. subanalysis exploring cardiac toxicity conducted on behalf of GETH-TC. Front Immunol 2025; 16:1571678. [PMID: 40375982 PMCID: PMC12079671 DOI: 10.3389/fimmu.2025.1571678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/09/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Haploidentical allogeneic hematopoietic cell transplantation (haplo-HCT) using post-transplant cyclophosphamide (PTCY) has become a standard approach for patients lacking HLA-matched donors. While effective in reducing graft-versus-host disease (GVHD), concerns about PTCY-associated cardiovascular toxicity remain. This study investigates the incidence, predictors, and impact of early cardiac events (ECE) in haplo-HCT recipients. Methods We conducted a retrospective, multicenter analysis of 268 patients with acute myeloid leukemia (AML) treated with anthracycline-based induction regimens and undergoing their first haplo-HCT with PTCY (50 mg/kg/day on days +3 and +4) between 2011 and 2022. ECEs, defined as any new cardiac event within 100 days post-transplant, were analyzed using cumulative incidence functions considering death and relapse as competing risks. Risk factors and the impact on non-relapse mortality (NRM) and overall survival (OS) were assessed via univariate and multivariate regression models. Results The median patient age was 57 years (range: 18-79), and pre-transplant comorbidities included hypertension (22.4%), dyslipidemia (13.1%), diabetes mellitus (6.7%), and prior cardiac history (14.2%). ECEs occurred in 23 patients (8.6%) at a median of 19 days post-transplant (IQR: 5-66), with a day +100 cumulative incidence of 8.6% (95% CI: 6.1-12.3). The most frequent complications were pericardial effusion/pericarditis (43.5%), arrhythmias (30.4%), and heart failure (17.4%). Severe ECEs (CTCAE grade 3-4) were observed in 30.4% of cases, and four deaths (17.4%) were directly attributed to ECEs. Univariate analysis identified dyslipidemia (HR: 3.87, p=0.001), hypertension (HR: 2.76, p=0.015), and moderate-severe veno-occlusive disease (HR: 4.94, p=0.002) as significant predictors of ECE. ECEs were associated with lower OS (HR: 1.78, p=0.04) and higher NRM (HR: 2.87, p=0.005). Discussion While the incidence of ECEs following haplo-HCT with PTCY was relatively low, their occurrence significantly worsened transplant outcomes. These findings underscore the importance of cardiovascular risk assessment and structured cardiac monitoring to mitigate complications in haplo-HCT recipients.
Collapse
Affiliation(s)
- Filipe R. Pinto
- Clinical Hematology Department, Unidade Local de Saúde de Santo António – Hospital de Santo António, Oporto, Portugal
| | - Enric Cascos
- Cardiology Department, Institut Clínic Cardiovascular (ICCV), Hospital Clinic de Barcelona, Barcelona, Spain
| | - Estefanía Pérez-López
- Hematology Department, Complejo Asistencial Universitario de Salamanca/Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Mónica Baile-González
- Hematology Department, Complejo Asistencial Universitario de Salamanca/Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Carlos Martín Rodríguez
- Hematology Department, Complejo Asistencial Universitario de Salamanca/Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - Marta Luque
- Hematology Department, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Albert Esquirol
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Felipe Peña-Muñóz
- Hematology Department, Institut Català d’Oncologia - Hospital Duran i Reynals, Barcelona, Spain
| | | | | | | | - Sara Fernández-Luis
- Hematology Department, Hospital Universitario Marqués de Valdecilla (IDIVAL), Santander, Spain
| | | | | | | | | | - Leslie González Pinedo
- Hematology Department, Hospital Universitario de Gran Canaria Doctor Negrín, Gran Canaria, Spain
| | - Lucía García
- Hematology Department, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | | | - Tamara Torrado
- Hematology Department, Hospital Universitario de A Coruña, A Coruña, Spain
| | - Silvia Filaferro
- Grupo Español de Trasplante de Progenitores Hematopoyéticos y Terapia Celular, Madrid, Spain
| | - Pascual Basalobre
- Grupo Español de Trasplante de Progenitores Hematopoyéticos y Terapia Celular, Madrid, Spain
| | | | - Guillermo Ortí
- Grupo Español de Trasplante de Progenitores Hematopoyéticos y Terapia Celular, Madrid, Spain
- Hematology Department, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Manuel Jurado Chacón
- Grupo Español de Trasplante de Progenitores Hematopoyéticos y Terapia Celular, Madrid, Spain
- Hematology Department, Hospital Universitario Virgen de las Nieves de Granada, Granada, Spain
| | - María Queralt Salas
- Cardiology Department, Institut Clínic Cardiovascular (ICCV), Hospital Clinic de Barcelona, Barcelona, Spain
- Hematopoietic Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Shang W, Geng X, Sun X, Fan X, Li A, Zhang C, Kang Y, Liang Y, Zhang J. Non-coding RNAs modulate pyroptosis in diabetic cardiomyopathy: A comprehensive review. Int J Biol Macromol 2025; 309:142865. [PMID: 40188918 DOI: 10.1016/j.ijbiomac.2025.142865] [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: 01/01/2025] [Revised: 02/07/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of heart failure (HF) among individuals with diabetes, presenting a significant medical challenge due to its complex pathophysiology and the lack of targeted therapies. Pyroptosis, a pro-inflammatory form of programmed cell death (PCD), is the predominant mode of cell death in the primary resident cells involved in DCM. It has been reported to be critical in DCM's onset, progression, and pathogenesis. Non-coding RNAs (ncRNAs), diverse transcripts lacking protein-coding potential, are essential for cellular physiology and the progression of various diseases. Increasing evidence indicates that ncRNAs are pivotal in the pathogenesis of DCM by regulating pyroptosis. This observation suggests that targeting the regulation of pyroptosis by ncRNAs may offer a novel therapeutic approach for DCM. However, a comprehensive review of this topic is currently lacking. Our objective is to elucidate the regulatory role of ncRNAs in pyroptosis associated with DCM and to elucidate the relationships among these factors. Additionally, we explored how ncRNAs influence pyroptosis and contribute to the pathophysiology of DCM. By doing so, we aim to identify new research targets for the clinical diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Wenyu Shang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Xiaofei Geng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Xitong Sun
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Xinbiao Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Aolin Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Chi Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Yuxin Kang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Yongchun Liang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Junping Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China.
| |
Collapse
|
13
|
Zhong W, Chen R, Zhao J, Zhang Y, He J, Wang H, Zhu F, Fan C, Liu X. SETD7 drives diabetic endothelial dysfunction through FBXO45-mediated GPX4 ubiquitylation. Cardiovasc Diabetol 2025; 24:178. [PMID: 40275362 PMCID: PMC12023459 DOI: 10.1186/s12933-025-02740-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Vasculopathy is the most prevalent complication of diabetes. Endothelial damage, a primary contributor to hyperglycemic vascular complications, impacts macro- and micro-vasculatures, causing functional impairment of multiple organs. SETD7 was initially identified as a transcriptional activator based on its ability to methylate histone 3 lysine 4. However, its function in the context of diabetic endothelial dysfunction remains poorly understood. This study aims to elucidate the involvement and underlying mechanisms of SETD7 in diabetic endothelial dysfunction. METHODS SETD7 knockout mice were generated to investigate the effects of SETD7 on Streptozotocin (STZ)-induced hyperglycemia and vascular endothelial injury. Endothelial-specific SETD7 interruption adeno-associated virus (AAV) system was utilized to investigate the effects of SETD7 on diabetic vascular endothelial injury in BKS-DB(Lepr) KO/KO (db/db) mice. In vitro manipulation of SETD7 activation or knockdown was conducted to assess its regulation on the lipid peroxidation, oxidative stress, and cell function of primary rat aortic endothelial cells (RAECs) under high glucose conditions. RESULTS Our study revealed that knockout and endothelial deficiency of SETD7 partially restored damaged vascular function and attenuated the inflammatory response caused by high glucose in both STZ-induced and db/db mice. Moreover, SETD7 activation aggravated oxidative stress injury and resulted in profound dysfunction through Glutathione Peroxidase 4 (GPX4)-mediated lipid peroxidation in RAECs. Mechanistically, SETD7 deficiency reduced p53 mono-methylation and blocked FBXO45 transcription, thereby inhibiting the protein degradation of GPX4 and subsequent lipid peroxidation as well as oxidative stress. CONCLUSIONS In summary, our study demonstrates that SETD7-p53-FBXO45-GPX4 is involved in high glucose-induced oxidative stress injury and exacerbated endothelial dysfunction, which offering great significance for mitigating hyperglycemia-induced endothelial damage.
Collapse
MESH Headings
- Animals
- Mice, Knockout
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/pathology
- Oxidative Stress
- Ubiquitination
- Endothelial Cells/enzymology
- Endothelial Cells/pathology
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/deficiency
- Male
- Glutathione Peroxidase/metabolism
- Glutathione Peroxidase/genetics
- F-Box Proteins/metabolism
- F-Box Proteins/genetics
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/physiopathology
- Cells, Cultured
- Signal Transduction
- Mice, Inbred C57BL
- Rats
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/pathology
- Lipid Peroxidation
- Blood Glucose/metabolism
Collapse
Affiliation(s)
- Wen Zhong
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Ruoxue Chen
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jialin Zhao
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Yuyu Zhang
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jintao He
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Huibin Wang
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Feng Zhu
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Chunxiang Fan
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China.
| | - Xinhua Liu
- Phenome Research Center of TCM, Department of Traditional Chinese Medicine, Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, 201203, China.
| |
Collapse
|
14
|
Zhai Y, Shang H, Li Y, Zhang N, Zhang J, Wu S. A Systematic Review of risk factors for major adverse cardiovascular events in patients with coronary heart disease who underwent percutaneous coronary intervention. Front Physiol 2025; 16:1514585. [PMID: 40271216 PMCID: PMC12014560 DOI: 10.3389/fphys.2025.1514585] [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: 10/21/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Objective This study aims to systematically review the risk factors for major adverse cardiovascular events (MACE) in patients with coronary heart disease who have undergone percutaneous coronary intervention (PCI). Design Systematic review and meta-analysis. Data sources The Cochrane Library, PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), Wanfang Database, and VIP Database for Chinese Technical Periodicals (VIP) were screened until December 2024. Eligibility criteria for selecting studies Case-control studies or cohort studies on the risk factors for MACE in patients with coronary heart disease who underwent PCI. Data extraction and synthesis: The literature review, data extraction, and quality evaluation were conducted by two independent researchers, and the meta-analysis was performed using RevMan 5.4 software. Main outcomes The main outcome was that MACE occurred during the follow-up period. Results A total of 40 articles were included. The meta-analysis erevealed that dyslipidemia (OR = 1.50; 95% CI [1.19, 1.89], p = 0.0007), diabetes mellitus (OR = 1.70; 95% CI [1.43, 2.02], p < 0.00001), hypertension (OR = 1.62; 95% CI [1.35, 1.96], p < 0.0001), history of smoking (OR = 2.08; 95% CI [1.51, 2.85], p < 0.0001), poorer ventricular function (OR = 2.39; 95% CI [2.17-2.64], p < 0.0001), impaired left ventricular ejection fraction (LVEF) (OR = 1.86; 95% CI [1.71-2.03], p < 0.0001), door to balloon (D-to-B) time (OR = 0.61; 95% CI [0.42-0.88]; p = 0.009), thrombolysis in myocardial infarction (TIMI) (OR = 1.41; 95% CI [1.17, 1.70], p = 0.0004), renal dysfunction (OR = 1.82; 95% CI [1.37, 2.43], p < 0.0001), and multi-vessel coronary artery disease (OR = 0.41; 95% CI [0.37, 0.46], p < 0.0001) were significantly associated with MACE after PCI. Conclusion The independent risk factors of MACE after PCI are dyslipidemia, hypertension, diabetes mellitus, smoking history, Killip class > II, LVEF ≤40%, D-to-B time >90 min, TIMI flow grade ≤ II, renal insufficiency, and multivessel disease.
Collapse
Affiliation(s)
- You Zhai
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Nan Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jisi Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shangwen Wu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
15
|
Gantzel Nielsen C, Olsen MT, Lommer Kristensen P, Schønemann-Lund M, Johansson PI, Pedersen-Bjergaard U, Heiberg Bestle M. The Association Between Dysglycemia and Endotheliopathy in ICU Patients With and Without Diabetes: A Cohort Study. Crit Care Explor 2025; 7:e1229. [PMID: 40126923 PMCID: PMC11936623 DOI: 10.1097/cce.0000000000001229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
IMPORTANCE Dysglycemia in critically ill patients is associated with endotheliopathy. This relationship may be altered in patients with diabetes. OBJECTIVES Dysglycemia is common in critically ill patients and associated with increased mortality. Endotheliopathy is thought to play a role in this relationship; however, evidence is scarce. The aim of this study was to investigate the associations between dysglycemia and endotheliopathy to inform future glycemic management. DESIGN, SETTING, AND PARTICIPANTS This prospective observational study included 577 acutely admitted adult ICU patients at Copenhagen University Hospital-North Zealand, Denmark. MAIN OUTCOMES AND MEASURES Up to twenty-four hours of patient glycemia was paired with same-day levels of endothelial biomarkers measured after each 24-hour period for three consecutive days. Endotheliopathy was assessed by measurement of Syndecan-1, Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1), and soluble thrombomodulin (sTM). RESULTS Of the included patients, a total 57.5% were males, median age was 71 yr (interquartile range [IQR], 63-79), and 24.6% had diabetes prior to admission. Median admission time was 5 d (IQR, 3-10). Time above range (TAR) greater than 13.9 mmol/L, but not TAR 10.0-13.9 mmol/L, was associated with increase in sTM (0.01 ng/mL per %-point increase in TAR, p = 0.049) and PECAM-1 (0.01 ng/mL per %-point increase, p = 0.007). Glycemic variability was associated with increases in sTM (0.24 ng/mL per mmol/L increase in sd, p = 0.001 and 0.03 ng/mL per %-point increase in coefficient of variation, p < 0.001). Hypoglycemia 3.0-3.9 mmol/L was associated with increases in sTM (3.0 ng/mL, p < 0.001) and PECAM-1 (1.54 ng/mL, p < 0.001). CONCLUSIONS AND RELEVANCE In acutely admitted adult ICU patients, hypoglycemia was associated with endotheliopathy regardless of preadmission diabetes status. Hyperglycemia and high glycemic variability were associated with endotheliopathy in patients without diabetes. This suggests different responses to acute dysglycemia in patients with and without diabetes and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Christian Gantzel Nielsen
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital – North Zealand, Hilleroed, Denmark
| | - Mikkel Thor Olsen
- Department of Endocrinology and Nephrology, Copenhagen University Hospital – North Zealand, Hilleroed, Denmark
| | - Peter Lommer Kristensen
- Department of Endocrinology and Nephrology, Copenhagen University Hospital – North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin Schønemann-Lund
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital – North Zealand, Hilleroed, Denmark
| | - Pär Ingemar Johansson
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Pedersen-Bjergaard
- Department of Endocrinology and Nephrology, Copenhagen University Hospital – North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Morten Heiberg Bestle
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital – North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Abas ASM, Sherif MH, Ibrahim S. Effects of Naringin and Zinc Treatment on Biochemical, Molecular, and Histological Alterations in Stomach and Pancreatic Tissues of STZ-Induced Diabetic Rats. Adv Biol (Weinh) 2025; 9:e2400688. [PMID: 39957607 DOI: 10.1002/adbi.202400688] [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: 11/07/2024] [Revised: 01/23/2025] [Indexed: 02/18/2025]
Abstract
Diabetes mellitus is a chronic metabolic disorder that affects multiple organs, including the stomach. This research examines the effects of naringin and/or zinc on stomach and pancreatic tissues of streptozotocin-induced diabetic rats. Type 2 diabetes is induced by intraperitoneal injection of nicotinamide and streptozotocin. Three weeks after diabetes induction, rats receive eight weeks of treatment. Malondialdehyde and total antioxidant capacity are estimated colorimetrically. Asprosin and P-selectin levels are assessed via ELISA. Quantitative RT-PCR analysis of nuclear factor kappa B (NF-кB), peroxisome proliferator-activated receptor gamma (PPAR γ), and nuclear factor erythroid 2-related factor 2 (Nrf-2) genes is carried out. Tumor necrosis factor-alpha (TNF-α) is assessed immunohistochemically, and stomach and pancreatic tissues are examined histologically. Combined naringin and zinc treatment significantly reduces gastric Malondialdehyde, serum asprosin, and P-selectin levels in serum, stomach, and pancreas compared to diabetic rats. Additionally, gastric NF-кB expression is significantly lower, while PPAR γ and Nrf-2 expressions are significantly higher compared to diabetic rats. Immunohistochemical analysis and histopathological examination confirm these findings. In conclusion, combined naringin and zinc treatment significantly improves gastric alterations in diabetic rats by reducing oxidative stress and inflammation. Nonetheless, it shows no additional impacts on pancreatic tissue compared to naringin or zinc alone.
Collapse
Affiliation(s)
- Al-Shimaa M Abas
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed H Sherif
- Chemistry Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Sarah Ibrahim
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
17
|
Lin J, Zhang X, Ge W, Duan Y, Zhang X, Zhang Y, Dai X, Jiang M, Zhang X, Zhang J, Qiang H, Sun D. Rnd3 Ameliorates Diabetic Cardiac Microvascular Injury via Facilitating Trim40-Mediated Rock1 Ubiquitination. Diabetes 2025; 74:569-584. [PMID: 39792251 DOI: 10.2337/db24-0543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
ARTICLE HIGHLIGHTS Impaired cardiac microvascular function is a significant contributor to diabetic cardiomyopathy. Rnd3 expression is notably downregulated in cardiac microvascular endothelial cells under diabetic conditions. Rnd3 overexpression mitigates diabetic myocardial microvascular injury and improves cardiac function through the Rock1/myosin light chain signaling pathway. Rnd3 facilitates the recruitment and interaction with Trim40 to promote Rock1 ubiquitination, thereby preserving endothelial barrier integrity in the diabetic heart.
Collapse
Affiliation(s)
- Jie Lin
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Wen Ge
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
18
|
Deng W, Cao H, Sun T, Yuan P. Exploring the role of glycolysis in the pathogenesis of erectile dysfunction in diabetes. Transl Androl Urol 2025; 14:791-807. [PMID: 40226065 PMCID: PMC11986553 DOI: 10.21037/tau-2025-6] [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: 01/02/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025] Open
Abstract
Background Diabetes mellitus-related erectile dysfunction (DMED) is characterized by complicated pathogenesis and unsatisfactory therapeutic remedies. Glycolysis plays an essential role in diabetic complications and whether it is involved in the process of DMED has not been expounded. The aim of this study was to investigate the genetic profiling of glycolysis and explore potential mechanisms for DMED. Methods Glycolysis-related genes (GRGs) and gene expression matrix of DMED were obtained from the molecular signatures database and gene expression omnibus dataset. Differentially expressed analysis and support vector machine-recursive feature elimination (SVM-RFE) method were both used to obtain hub GRGs. Interactive network and functional enrichment analyses were performed to clarify the associated biological roles of these genes. The expression profile of hub GRGs was validated in cavernous endothelial cells, animals, and clinical patients. The subpopulation distribution of hub GRGs was further identified. In addition, a miRNA-GRGs network was constructed and expression patterns as well as molecular functions of relevant miRNAs were explored and validated. In addition, the relationship between hypoxia and DMED was also uncovered. Results Based on the combined analysis, 48 differentially expressed GRGs were obtained. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed that these genes were significantly enriched in carbon metabolism and oxidoreductase activities. Then hub GRGs including down-regulated as well as up-regulated genes in DMED were identified ultimately. Among them, ALDOC, ANGPTL4, and CITED2 were well-validated. In addition, 334 glycolysis-related miRNAs were verified and they were involved in endoplasmic reticulum membrane activity, smooth muscle cell differentiation and angiogenesis. After validation of miRNA signature in DMED patients, miR-222-5p, let-7e-5p, miR-184, and miR-122-3p were identified as the promising glycolysis-related miRNA biomarkers in DMED. Conclusions We clarified the expression signature of GRGs in DMED based on multi-omics analysis for the first time. It will be significantly important to reveal pathological mechanisms and promising treatments in DMED.
Collapse
Affiliation(s)
- Wenjia Deng
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Honggang Cao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Taotao Sun
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Penghui Yuan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Thal SC, Shityakov S, Salvador E, Förster CY. Heart Rate Variability, Microvascular Dysfunction, and Inflammation: Exploring the Potential of taVNS in Managing Heart Failure in Type 2 Diabetes Mellitus. Biomolecules 2025; 15:499. [PMID: 40305215 PMCID: PMC12024555 DOI: 10.3390/biom15040499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) predominantly experience mortality due to cardiovascular diseases (CVD), particularly in low- and middle-income nations. Among these, heart failure (HF) is the most severe cardiovascular complication in terms of prognosis and management. Despite advancements in individualized glycemic control and cardiovascular risk management, including the development of novel glucose- and lipid-lowering agents, the prevalence of HF in T2DM patients remains persistently high. This indicates that factors beyond hyperglycemia significantly contribute to the heightened risk of HF associated with T2DM. This review examines critical factors influencing CVD risk in T2DM, particularly the roles of reduced heart rate variability (HRV), a marker of autonomic dysfunction, and chronic inflammation, both of which play pivotal roles in HF pathogenesis. Recent evidence highlights the potential of vagus nerve activation to modulate these risk factors, underscoring its capacity to reduce T2DM-related cardiovascular complications. Specifically, we discuss the therapeutic promise of transcutaneous auricular vagus nerve stimulation (taVNS) as a non-invasive intervention to enhance vagal tone, decrease systemic inflammation, and improve cardiovascular outcomes in T2DM. By addressing the interplay among HRV, microvascular disease, and inflammation, this review provides a comprehensive perspective on the potential utility of taVNS in managing HF in T2DM.
Collapse
Affiliation(s)
- Serge C. Thal
- Department of Anesthesiology, Helios University Hospital, Witten/Herdecke University, 42283 Wuppertal, Germany;
| | - Sergey Shityakov
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, 197101 Saint-Petersburg, Russia;
| | - Ellaine Salvador
- Section Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Carola Y. Förster
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, Section Cerebrovascular Sciences and Neuromodulation, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
20
|
Rroji M, Spahia N, Figurek A, Spasovski G. Targeting Diabetic Atherosclerosis: The Role of GLP-1 Receptor Agonists, SGLT2 Inhibitors, and Nonsteroidal Mineralocorticoid Receptor Antagonists in Vascular Protection and Disease Modulation. Biomedicines 2025; 13:728. [PMID: 40149704 PMCID: PMC11940462 DOI: 10.3390/biomedicines13030728] [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: 02/09/2025] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Atherosclerosis is a closely related complication of diabetes mellitus (DM), driven by endothelial dysfunction, inflammation, and oxidative stress. The progression of atherosclerosis is accelerated by hyperglycemia, insulin resistance, and hyperlipidemia. Novel antidiabetic agents, SGLT2 inhibitors, and GLP-1 agonists improve glycemic control and offer cardiovascular protection, reducing the risk of major adverse cardiovascular events (MACEs) and heart failure hospitalization. These agents, along with nonsteroidal mineralocorticoid receptor antagonists (nsMRAs), promise to mitigate metabolic disorders and their impact on endothelial function, oxidative stress, and inflammation. This review explores the potential molecular mechanisms through which these drugs may prevent the development of atherosclerosis and cardiovascular disease (CVD), supported by a summary of preclinical and clinical evidence.
Collapse
Affiliation(s)
- Merita Rroji
- Department of Nephrology, University of Medicine Tirana, 1001 Tirana, Albania
- Department of Nephrology, University Hospital Center Mother Tereza, 1001 Tirana, Albania;
| | - Nereida Spahia
- Department of Nephrology, University Hospital Center Mother Tereza, 1001 Tirana, Albania;
| | - Andreja Figurek
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland;
| | - Goce Spasovski
- Department of Nephrology, University Sts. Cyril and Methodius, 1000 Skopje, North Macedonia;
| |
Collapse
|
21
|
Penna C, Pagliaro P. Endothelial Dysfunction: Redox Imbalance, NLRP3 Inflammasome, and Inflammatory Responses in Cardiovascular Diseases. Antioxidants (Basel) 2025; 14:256. [PMID: 40227195 PMCID: PMC11939635 DOI: 10.3390/antiox14030256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Endothelial dysfunction (ED) is characterized by an imbalance between vasodilatory and vasoconstrictive factors, leading to impaired vascular tone, thrombosis, and inflammation. These processes are critical in the development of cardiovascular diseases (CVDs) such as atherosclerosis, hypertension and ischemia/reperfusion injury (IRI). Reduced nitric oxide (NO) production and increased oxidative stress are key contributors to ED. Aging further exacerbates ED through mitochondrial dysfunction and increased oxidative/nitrosative stress, heightening CVD risk. Antioxidant systems like superoxide-dismutase (SOD), glutathione-peroxidase (GPx), and thioredoxin/thioredoxin-reductase (Trx/TXNRD) pathways protect against oxidative stress. However, their reduced activity promotes ED, atherosclerosis, and vulnerability to IRI. Metabolic syndrome, comprising insulin resistance, obesity, and hypertension, is often accompanied by ED. Specifically, hyperglycemia worsens endothelial damage by promoting oxidative stress and inflammation. Obesity leads to chronic inflammation and changes in perivascular adipose tissue, while hypertension is associated with an increase in oxidative stress. The NLRP3 inflammasome plays a significant role in ED, being triggered by factors such as reactive oxygen and nitrogen species, ischemia, and high glucose, which contribute to inflammation, endothelial injury, and exacerbation of IRI. Treatments, such as N-acetyl-L-cysteine, SGLT2 or NLRP3 inhibitors, show promise in improving endothelial function. Yet the complexity of ED suggests that multi-targeted therapies addressing oxidative stress, inflammation, and metabolic disturbances are essential for managing CVDs associated with metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
22
|
Shao F, Wieland J, Wang Y, Keles M, Meng Z, Lomada S, Qin M, Leiss V, Martin-Garrido A, Fuhrmann M, Qiu Y, Trogisch FA, Vettel C, Heineke J, Feng Y. Deficiency in nucleoside diphosphate kinase B leads to endothelial activation of the hexosamine biosynthesis pathway and cardiac dysfunction. Cardiovasc Diabetol 2025; 24:84. [PMID: 39985023 PMCID: PMC11846329 DOI: 10.1186/s12933-025-02633-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Nucleoside diphosphate kinase B (NDPKB) deficiency in endothelial cells (ECs) promotes the activation of the hexosamine biosynthesis pathway (HBP), leading to vascular damage in the retina. The aim of this study was to investigate the consequences of NDPKB deficiency in the mouse heart. METHODS NDPKB deficient mice were used in the study. Echocardiography was employed to assess cardiac function in vivo. Characterization of contractility in hiPSC-derived cardiomyocytes (hiPSC-CMs) was measured with the IonOptix contractility system. Immunoblotting and immunofluorescence were carried out to analyze the expression and localization of proteins in cultured cells and left ventricles (LVs). RESULTS NDPKB deficient mice displayed impaired glucose tolerance and increased heart weight compared to controls. Echocardiographic analysis revealed an increase in the diastolic diameter of the left ventricular posterior wall (LVPW), a decrease in the early diastolic mitral valve E and E' wave, and in the ratios of E/A and E'/A' in NDPKB deficient hearts, suggesting cardiac hypertrophy and diastolic dysfunction. In line with cardiac dysfunction, the phosphorylation of myocardial phospholamban (PLN) and the expression of sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2 (SERCA2) in the NDPKB deficient LVs were significantly reduced. Moreover, the accumulation of collagen, fibronectin as well as the upregulation of transforming growth factor β (TGF-β), were detected in NDPKB deficient LVs. In addition, activation of the HBP and its downstream O-GlcNAc cycle was observed in the LVs and cardiac ECs (CECs) isolated from the NDPKB-/- mice. Furthermore, a bipolar O-GlcNAc regulation was identified in CMs. O-GlcNAc was decreased in NDPKB-depleted CMs, while conditioned medium from NDPKB-depleted ECs significantly increased O-GlcNAc levels, along with contractile and relaxation dysfunction of the hiPSC-CMs, which was attenuated by inhibiting endothelial HBP activation. CONCLUSIONS Deficiency in NDPKB leads to endothelial activation of the HBP and cardiac dysfunction. Our findings may highlight the crucial role of proper endothelial HBP in maintaining cardiovascular homeostasis.
Collapse
MESH Headings
- Animals
- Hexosamines/biosynthesis
- Mice, Knockout
- Endothelial Cells/enzymology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NM23 Nucleoside Diphosphate Kinases/deficiency
- NM23 Nucleoside Diphosphate Kinases/genetics
- Ventricular Function, Left
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/diagnostic imaging
- Cells, Cultured
- Myocardial Contraction
- Disease Models, Animal
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Male
- Phosphorylation
- Mice, Inbred C57BL
- Signal Transduction
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Calcium-Binding Proteins/metabolism
- Mice
- Ventricular Remodeling
Collapse
Affiliation(s)
- Feng Shao
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Johanna Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Yixin Wang
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Merve Keles
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Zenghui Meng
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, 68167 Mannheim, Germany
| | - Santosh Lomada
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Miao Qin
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, University of Tübingen, 72074 Tübingen, Germany
| | - Abel Martin-Garrido
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Manuela Fuhrmann
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Yi Qiu
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Felix A Trogisch
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Christiane Vettel
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Joerg Heineke
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
23
|
Zhang YY, Li YJ, Hu XQ, Xue CD, Li S, Gao ZN, Qin KR. Unveiling the Negative Synergistic Effect of Wall Shear Stress and Insulin on Endothelial NO Dynamics by Mathematical Modeling. Bull Math Biol 2025; 87:46. [PMID: 39969626 DOI: 10.1007/s11538-025-01424-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Diabetic vascular complications (DVCs) are diabetes-induced vascular dysfunction and pathologies, leading to the major causes of morbidity and mortality in millions of diabetic patients worldwide. DVCs are provoked by endothelial dysfunction which is closely coordinated with two important hallmarks: one is the insufficient insulin secretion or insulin resistance, and another is the decrease in intracellular nitric oxide (NO) influenced by dynamic wall shear stress (WSS). Although the intracellular NO dynamics in endothelial cells (ECs) is crucial for endothelial function, the regulation of NO production by dynamic WSS and insulin is still poorly understood. In this study, we have proposed a mathematical model of intracellular NO production in ECs under the stimulation of dynamic WSS combined with insulin. The model integrates simultaneously the biochemical signaling pathways of insulin and the mechanotransduction pathways induced by dynamic WSS. The accuracy and reliability of the model to quantitatively describe NO production in ECs were compared and validated with reported experimental data. According to the validated model, inhibition of protein kinase B (AKT) phosphorylation and Ca2+ influx by dynamic oscillatory WSS disrupts the dual nature of endothelial nitric oxide synthase (eNOS) enzyme activation. This disruption leads to the decrease in NO production and the bimodal disappearance of NO waveforms. Moreover, the results reveal that dynamic WSS combined with insulin promote endothelial NO production through negative synergistic effects, which is resulted from the temporal differences in mechanical and biochemical signaling. In brief, the proposed model elucidates the mechanism of NO generation activated by dynamic WSS combined with insulin, providing a potential target and theoretical framework for future treatment of DVCs.
Collapse
Affiliation(s)
- Yu-Yuan Zhang
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, 116024, Liaoning, People's Republic of China
| | - Yong-Jiang Li
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China.
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, 116024, Liaoning, People's Republic of China.
| | - Xu-Qu Hu
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, 116024, Liaoning, People's Republic of China
| | - Chun-Dong Xue
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, 116024, Liaoning, People's Republic of China
| | - Shen Li
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China
| | - Zheng-Nan Gao
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China
| | - Kai-Rong Qin
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, 116033, Liaoning, People's Republic of China.
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, 116024, Liaoning, People's Republic of China.
| |
Collapse
|
24
|
Pan J, Chen MY, Jiang CY, Zhang ZY, Yan JL, Meng XF, Han YP, Lou YY, Yang JT, Qian LB. Luteolin alleviates diabetic cardiac injury related to inhibiting SHP2/STAT3 pathway. Eur J Pharmacol 2025; 989:177259. [PMID: 39788407 DOI: 10.1016/j.ejphar.2025.177259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/15/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
Diabetic cardiomyopathy, a heart disease resulting from diabetes mellitus, inflicts structural and functional damage to the heart. Recent studies have highlighted the potential role of luteolin, a flavonoid, in mitigating diabetic cardiovascular injuries. The Src homology 2-containing protein tyrosine phosphatase 2 (SHP2) is implicated in exacerbating diabetes- and obesity-related complications. Interestingly, luteolin has been shown to inhibit protein tyrosine phosphatases, but it's unclear how SHP2 relates to luteolin's protective effects against diabetic heart disease. Here, we hypothesized that the inhibition of SHP2 signaling could play a role in luteolin's protective action against diabetic heart injury. Diabetes was induced in male Sprague-Dawley rats through a high-fat diet followed by a single intraperitoneal dose of streptozotocin (30 mg/kg). Five weeks post-diabetes induction, these rats were intraperitoneally injected with luteolin at varying doses (5, 10, 20 mg/kg) every other day for an additional 5 weeks. Then cardiac function was assessed, and hearts were isolated for further analysis. We found that luteolin notably improved cardiac function, inhibited cardiac hypertrophy and fibrosis, reduced levels of inflammatory factors and reactive oxygen species, and activated superoxide dismutase. Importantly, luteolin treatment also reduced the expression of SHP2 and phosphorylated signal transducer and activator of transcription 3 (STAT3) in a dose-dependent manner. These findings suggest that luteolin protects the diabetic heart against inflammation, oxidative stress, hypertrophy, and fibrosis, which may relate to down-regulating cardiac SHP2/STAT3 signaling.
Collapse
Affiliation(s)
- Jie Pan
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Meng-Yuan Chen
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China; Department of Clinical Laboratory Medicine, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Chun-Yan Jiang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Zi-Yan Zhang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Jia-Lin Yan
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Xiang-Fei Meng
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yu-Peng Han
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yang-Yun Lou
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Jin-Ting Yang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Ling-Bo Qian
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, 310053, China.
| |
Collapse
|
25
|
Zhang Y, Li M, Liu H, Fan Y, Liu HH. The application of procyanidins in diabetes and its complications: a review of preclinical studies. Front Pharmacol 2025; 16:1532246. [PMID: 39995417 PMCID: PMC11847907 DOI: 10.3389/fphar.2025.1532246] [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: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetes mellitus (DM) and its various complications, including diabetic nephropathy, retinopathy, neuropathy, cardiovascular disease, and ulcers, pose significant challenges to global health. This review investigates the potential of procyanidins (PCs), a natural polyphenolic compound, in preventing and managing diabetes and its complications. PCs, recognized for their strong antioxidant, anti-inflammatory, and anti-hyperglycemic properties, play a crucial role in reducing oxidative stress and enhancing endothelial function, which are essential for managing diabetic complications. This review elucidates the molecular mechanisms by which PCs improve insulin sensitivity and endothelial health, thereby providing protection against the various complications of diabetes. The comprehensive analysis underscores the promising therapeutic role of PCs in diabetes care, indicating the need for further clinical studies to confirm and leverage their potential in comprehensive diabetes management strategies.
Collapse
Affiliation(s)
- Yongchuang Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengna Li
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Haoyuan Liu
- Rehabilitation Department, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Yongfu Fan
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Huan Huan Liu
- International institute for Traditional Chinese Medicine, Guanzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
26
|
Huang K, Mi B, Xiong Y, Fu Z, Zhou W, Liu W, Liu G, Dai G. Angiogenesis during diabetic wound repair: from mechanism to therapy opportunity. BURNS & TRAUMA 2025; 13:tkae052. [PMID: 39927093 PMCID: PMC11802347 DOI: 10.1093/burnst/tkae052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/23/2024] [Accepted: 08/09/2024] [Indexed: 02/11/2025]
Abstract
Diabetes mellitus, a pervasive chronic metabolic disorder, is often associated with complications such as impaired wound healing. Various factors, most notably vascular deficiency, govern the wound repair process in diabetic patients, significantly impeding diabetic wound healing; therefore, angiogenesis and its role in diabetic wound repair have emerged as important areas of research. This review aims to delve into the mechanisms of angiogenesis, the effects of diabetes on angiogenesis, and the association between angiogenesis and diabetic wound repair. This will ultimately offer valuable guidance regarding the ideal timing of diabetic wound treatment in a clinical setting.
Collapse
Affiliation(s)
- Kang Huang
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Bobin Mi
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Yuan Xiong
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Zicai Fu
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Wenyun Zhou
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Wanjun Liu
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Guohui Liu
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| | - Guandong Dai
- Department of Orthopedics, Southern Medical University Pingshan Hospital, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
- Department of Orthopedics, Pingshan District Peoples’Hospital of Shenzhen, No. 19 Renmin Street, Pingshan District, Shenzhen City, Guangdong Province, 518118, P.R. China
| |
Collapse
|
27
|
Harris DD, Stone C, Broadwin M, Kanuparthy M, Sabe SA, Nho JW, Hamze J, Abid MR, Sellke FW. Dipeptidyl peptidase-4 inhibitor linagliptin improves fibrosis, apoptosis, and cardiac function in a large animal model of chronic myocardial ischemia. J Pharmacol Exp Ther 2025; 392:100532. [PMID: 40023609 DOI: 10.1016/j.jpet.2024.100532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 12/16/2024] [Indexed: 03/04/2025] Open
Abstract
Interest is increasing in using novel diabetic medications, such as glucagon-like peptide 1 (GLP-1) receptor agonists, to manage coronary artery disease. Dipeptidyl peptidase-4 (DPP-4) inhibitors enhance GLP-1 activity through the same pathway as GLP-1 agonists; however, DPP-4 inhibitors have not been fully evaluated in the setting of ischemic heart disease. We chose to study the DPP-4 inhibitor linagliptin (LIN) in a porcine model of chronic coronary ischemia. Seventeen Yorkshire swine underwent left thoracotomy and ameroid constrictor placement over the left circumflex coronary artery at age 11 weeks. Two weeks thereafter, swine received either vehicle without drug (n = 9) or LIN 2.5 mg (n = 8). Following the elapse of 5 weeks of treatment, swine underwent terminal harvest. LIN significantly increased stroke volume, ejection fraction, cardiac output, and ischemic myocardial perfusion, while decreasing Tau (all P < .05). Trichrome staining showed a marked reduction in ischemic myocardial interstitial and perivascular fibrosis, accompanied by decreased levels of transforming growth factor-β (all P < .05). Apoptosis, measured by terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling staining, was significantly reduced, and accompanied by decreases in apoptosis-inducing factor, BCL2-associated agonist of cell death, caspase-9, and cleaved caspase-9 (all P < .05). Additionally, there were significant increases in phosphoinositide 3-kinase, phospho-protein kinase B, 5' adenosine monophosphate-activated protein kinase, phospho-5' adenosine monophosphate-activated protein kinase, and endothelial nitric oxide synthase, and significant reductions in collagen 18 and angiostatin (all P < .05). LIN significantly improved left ventricular function, cellular survival, and attenuated adverse remodeling, all likely secondary to augmented perfusion ischemic myocardial perfusion. Given that this increased perfusion occurred independently of changes in vascular density, treatment likely resulted in enhanced microvascular reactivity. These benefits warrant further investigation of LIN to fully understand its potential as a therapy for ischemic heart disease. SIGNIFICANCE STATEMENT: Linagliptin significantly improved cardiac cellular survival, left ventricular function, and attenuated adverse myocardial remodeling in a clinically relevant, large animal model of chronic ischemic cardiomyopathy. This warrants further investigation of linagliptin to fully understand its therapeutic potential.
Collapse
Affiliation(s)
- Dwight Douglas Harris
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Christopher Stone
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Meghamsh Kanuparthy
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Ju-Woo Nho
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Jad Hamze
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island.
| |
Collapse
|
28
|
Ebrahimi M, Ahmadieh H, Rezaei Kanavi M, Safi S, Alipour-Parsa S, Advani S, Sorenson CM, Sheibani N. Shared signaling pathways and comprehensive therapeutic approaches among diabetes complications. Front Med (Lausanne) 2025; 11:1497750. [PMID: 39845838 PMCID: PMC11750824 DOI: 10.3389/fmed.2024.1497750] [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: 09/17/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
The growing global prevalence of diabetes mellitus (DM), along with its associated complications, continues to rise. When clinically detected most DM complications are irreversible. It is therefore crucial to detect and address these complications early and systematically in order to improve patient care and outcomes. The current clinical practice often prioritizes DM complications by addressing one complication while overlooking others that could occur. It is proposed that the commonly targeted cell types including vascular cells, immune cells, glial cells, and fibroblasts that mediate DM complications, might share early responses to diabetes. In addition, the impact of one complication could be influenced by other complications. Recognizing and focusing on the shared early responses among DM complications, and the impacted cellular constituents, will allow to simultaneously address all DM-related complications and limit adverse treatment impacts. This review explores the current understanding of shared pathological signaling mechanisms among DM complications and recognizes new concepts that will benefit from further investigation in both basic and clinical settings. The ultimate goal is to develop more comprehensive treatment strategies, which effectively impact DM complications in multiple organs and improve patient care and outcomes.
Collapse
Affiliation(s)
- Moein Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sare Safi
- Ophthalmic Epidemiology Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Alipour-Parsa
- Cardiovascular Research Center, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soroor Advani
- Neurology Department, Shohada Tajrish Hospital, Shahid-Beheshti University of Medical Sciences, Tehran, Iran
| | - Christine M. Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
29
|
Zhao B, Tan WL, Yu BB, Fan J, Liu C, Liu J, Liu Z. Selenoprotein M protects cardiac endothelial cell integrity against high-glucose stress via enhancing Parkin-mediated mitophagy. Mol Cell Endocrinol 2025; 595:112392. [PMID: 39454935 DOI: 10.1016/j.mce.2024.112392] [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: 07/29/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Selenoprotein M (SELENOM) has emerged as a crucial factor in maintaining cellular redox homeostasis and mitigating oxidative damage. This study aims to investigate its protective role in cardiac endothelial cells under hyperglycemic stress, a condition commonly associated with diabetes mellitus and its cardiovascular complications. Diabetic mice model and human umbilical vein endothelial cells (HUVECs) were applied for in vivo and in vitro studies. Results reveal that hyperglycemia significantly downregulates SELENOM expression in both diabetic mouse hearts and primary cultured cardiac endothelial cells. Overexpression of SELENOM in HUVECs mitigated high-glucose-induced FITC-Dextran diffusion and the loss of transendothelial electrical resistance. Additionally, SELENOM overexpression decreased reactive oxygen species (ROS) levels, preserved tight junction protein expression, and maintained cellular structural integrity under hyperglycemic conditions. Furthermore, SELENOM overexpression attenuated high-glucose-induced mitochondrial apoptosis. High-glucose conditions decreased Parkin and increased p62 and Beclin1 expressions. SELENOM overexpression restored Parkin levels and promoted co-localization of LAMP1 and TOMM20. Knockdown of Parkin significantly attenuated these protective effects, suggesting the importance of Parkin in Selenoprotein M-mediated mitophagy. Collectively, these findings suggest that Selenoprotein M enhances Parkin-mediated mitophagy to protect endothelial cells from hyperglycemic stress, offering potential therapeutic insights for diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wen-Liang Tan
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bing-Bo Yu
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jun Fan
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chang Liu
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian Liu
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Zhen Liu
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Paudel P, McDonald F, Fronius M. Isolation of Endothelial Cells from Human Internal Mammary Artery. Methods Mol Biol 2025; 2894:21-34. [PMID: 39699807 DOI: 10.1007/978-1-0716-4342-6_3] [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: 12/20/2024]
Abstract
Endothelial cells are a major contributor to cardiovascular diseases. Studying their function and how they influence pathological processes remains an ongoing area of research. Although primary endothelial cells might be readily available from animals, translating results from these studies can be challenging due to species-dependent differences. A common source of human endothelial cells is human umbilical vein endothelial cells, but these cells might not represent the variety of endothelial cells from various vascular beds. Here we describe a protocol for the isolation and cultivation of endothelial cells from human internal mammary artery remains. These remains are commonly available from coronary artery bypass graft surgeries. The described tissue explant method combined with a magnetic sorting process provided a relatively high yield of endothelial cells that were subsequently passaged and used for studies involving mRNA and protein expression analyses, as well as fluorescence-based immunohistochemistry and patch-clamp electrophysiology. This protocol may help to extend the repertoire of studying human endothelial cells to understand their role and function in health and disease.
Collapse
Affiliation(s)
- Puja Paudel
- Department of Physiology, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Fiona McDonald
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Martin Fronius
- Department of Physiology, University of Otago, Dunedin, New Zealand.
- HeartOtago, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
31
|
Shen Q, Ma S, Li L, Xia Y. Tanshinone IIA attenuates fluoride-induced spinal cord injury by inhibiting ferroptosis and inflammation. Heliyon 2024; 10:e40549. [PMID: 39687171 PMCID: PMC11648119 DOI: 10.1016/j.heliyon.2024.e40549] [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: 06/29/2023] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Excessive fluoride exposure can lead to health problems, such as fluorosis and neurotoxicity. However, effective therapeutic strategies for neurofluorosis remain elusive due to a limited understanding of the underlying molecular mechanisms. This study aimed to investigate the effects of Tanshinone IIA on spinal cord injury induced by high-fluoride exposure. To identify dysregulated genes associated with ferroptosis, we conducted an intersection analysis between differentially expressed genes in fluoride-treated HOS cells (GSE70719) and ferroptosis-related genes from the FerrDb database. A rat model of fluoride-induced spinal cord injury was established, revealing evidence of aberrant molecular and structural changes. Furthermore, the study demonstrated that Tanshinone IIA restored the altered expression of nine ferroptosis-related genes, eight fluorosis-related inflammatory indicators, and the observed structural changes. Overall, these findings suggest that Tanshinone IIA therapeutic potential in the treatment of fluoride-induced spinal cord injury by inhibiting ferroptosis and inflammation.
Collapse
Affiliation(s)
- Qingfeng Shen
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Shibo Ma
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Lingbo Li
- Department of Orthopedics, Jingzhou Central Hospital, Jingzhou, 434020, Hubei Province, China
| | - Yingpeng Xia
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| |
Collapse
|
32
|
Seetharaman ATM, Owens CE, Gangaraju R. Cysteinyl Leukotriene Receptor Antagonism by Montelukast to Treat Visual Deficits. J Ocul Pharmacol Ther 2024; 40:617-628. [PMID: 39358316 DOI: 10.1089/jop.2024.0111] [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: 10/04/2024] Open
Abstract
Montelukast, a Food and Drug Administration-approved drug for asthma and allergic rhinitis modulates leukotriene (LT) receptors and serves as a critical anti-inflammatory agent. Recent research suggests that the LT signaling pathway targeted by montelukast has broader implications for diseases such as fibrosis, cardiovascular diseases, cancer, cerebrovascular disease, and immune defense. This expanded understanding highlights montelukast's potential for repurposing in conditions involving aberrant stress mechanisms, including ocular diseases marked by inflammation, oxidative stress, ER stress, and apoptosis, among several others. This review delves into montelukast's therapeutic mechanisms across various diseases, draws parallels to ocular conditions, and examines clinical trials and associated adverse effects to underscore the unmet need for cysteinyl LT receptor antagonism by montelukast as an effective therapy for visual deficits.
Collapse
Affiliation(s)
- Amritha T M Seetharaman
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Caroline E Owens
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, Anatomy & Neurobiology, Neuroscience Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
33
|
Zhang Y, Cao Y, Zhang X, Lin J, Jiang M, Zhang X, Dai X, Zhang X, Liu Y, Ge W, Qiang H, Li C, Sun D. Single-Cell RNA Sequencing Uncovers Pathological Processes and Crucial Targets for Vascular Endothelial Injury in Diabetic Hearts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405543. [PMID: 39475009 DOI: 10.1002/advs.202405543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/18/2024] [Indexed: 12/19/2024]
Abstract
Cardiovascular disease remains the leading cause of high mortality in individuals with diabetes mellitus. Endothelial injury is a major contributing factor for vascular dysfunction in diabetes. However, the precise mechanisms underlying endothelial cell injury and their heterogeneity in diabetes remains elusive. In this study, single-cell sequencing is performed in heart tissues from leptin receptor knock-out (db/db) diabetic mice at various pathological stages. Through cell cluster identification, differential gene analysis, intercellular communication analysis, pseudo time analysis, and transcription factor analysis, a novel mechanism of cardiac vascular endothelial damage in diabetes is identified. Specifically, a single-cell transcription map of cardiac vascular endothelial cells is presented in db/db mice. Diverse cellular clusters are found to play vital roles under diabetes-induced damage, highlighting crucial transcription factors involved in their regulation. In addition, the essential transcription factor Ets1 is found to protect against vascular endothelial injury in db/db mice. In summary, the work provides a comprehensive understanding of the development of diabetic cardiac vascular endothelial damage and the heterogeneity of the cells involved. These findings offer valuable insights into potential treatments and assessments of diabetic cardiovascular endothelial damage.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen Ge
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
34
|
Li X, He Y, Wang D, Momeni MR. Chronobiological disruptions: unravelling the interplay of shift work, circadian rhythms, and vascular health in the context of stroke risk. Clin Exp Med 2024; 25:6. [PMID: 39541048 PMCID: PMC11564290 DOI: 10.1007/s10238-024-01514-w] [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: 06/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Shift work, particularly night shifts, disrupts circadian rhythms and increases stroke risk. This manuscript explores the mechanisms connecting shift work with stroke, focusing on circadian rhythms, hypertension, and diabetes. The circadian system, controlled by different mechanisms including central and peripheral clock genes, suprachiasmatic nuclei (SCN), and pineal gland (through melatonin production), regulates body functions and responds to environmental signals. Disruptions in this system affect endothelial cells, leading to blood pressure issues. Type 2 diabetes mellitus (T2DM) is significantly associated with night shifts, with circadian disturbances affecting glucose metabolism, insulin sensitivity, and hormone regulation. The manuscript examines the relationship between melatonin, insulin, and glucose balance, highlighting pathways that link T2DM to stroke risk. Additionally, dyslipidemia, particularly reduced HDL-c levels, results from shift work and contributes to stroke development. High lipid levels cause oxidative stress, inflammation, and endothelial dysfunction, increasing cerebrovascular risks. The manuscript details the effects of dyslipidemia on brain functions, including disruptions in blood flow, blood-brain barrier integrity, and neural cell death. This comprehensive analysis emphasizes the complex interplay of circadian disruption, hypertension, diabetes, and dyslipidemia in increasing stroke risk among shift workers. Understanding these mechanisms is essential for developing targeted interventions to reduce stroke susceptibility and improve cerebrovascular health in this vulnerable population.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanjin He
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Dawu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | | |
Collapse
|
35
|
Sieńko D, Szabłowska-Gadomska I, Nowak-Szwed A, Rudziński S, Gofron M, Zygmunciak P, Lewandowska-Szumieł M, Zgliczyński WS, Czupryniak L, Mrozikiewicz-Rakowska B. The Potential of Mesenchymal Stem/Stromal Cells in Diabetic Wounds and Future Directions for Research and Therapy-Is It Time for Use in Everyday Practice? Int J Mol Sci 2024; 25:12171. [PMID: 39596237 PMCID: PMC11594847 DOI: 10.3390/ijms252212171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The treatment of diabetic wounds is impaired by the intricate nature of diabetes and its associated complications, necessitating novel strategies. The utilization of mesenchymal stem/stromal cells (MSCs) as a therapeutic modality for chronic and recalcitrant wounds in diabetic patients is an active area of investigation aimed at enhancing its therapeutic potential covering tissue regeneration. The threat posed to the patient and their environment by the presence of a diabetic foot ulcer (DFU) is so significant that any additional therapeutic approach that opens new pathways to halt the progression of local changes, which subsequently lead to a generalized inflammatory process, offers a chance to reduce the risk of amputation or even death. This article explores the potential of MSCs in diabetic foot ulcer treatment, examining their mechanisms of action, clinical application challenges, and future directions for research and therapy.
Collapse
Affiliation(s)
- Damian Sieńko
- Department of Diabetology and Internal Diseases, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (A.N.-S.); (L.C.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Ilona Szabłowska-Gadomska
- Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097 Warsaw, Poland; (I.S.-G.); (S.R.); (M.L.-S.)
| | - Anna Nowak-Szwed
- Department of Diabetology and Internal Diseases, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (A.N.-S.); (L.C.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Stefan Rudziński
- Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097 Warsaw, Poland; (I.S.-G.); (S.R.); (M.L.-S.)
| | - Maksymilian Gofron
- Department of Urology, Municipal Complex Hospital, 42-200 Czestochowa, Poland;
| | - Przemysław Zygmunciak
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland; (P.Z.); (W.S.Z.)
| | - Małgorzata Lewandowska-Szumieł
- Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097 Warsaw, Poland; (I.S.-G.); (S.R.); (M.L.-S.)
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Wojciech Stanisław Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland; (P.Z.); (W.S.Z.)
| | - Leszek Czupryniak
- Department of Diabetology and Internal Diseases, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (A.N.-S.); (L.C.)
| | - Beata Mrozikiewicz-Rakowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland; (P.Z.); (W.S.Z.)
| |
Collapse
|
36
|
Xu M, Zhong S, Zhu N, Wang S, Wang J, Li X, Ren X, Kong H. Oxidative and endoplasmic reticulum stress in diabetes-related hearing loss: Protective effects of thioredoxin. Life Sci 2024; 359:123223. [PMID: 39515416 DOI: 10.1016/j.lfs.2024.123223] [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: 08/19/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Diabetes mellitus (DM) induces complex physiological changes in the inner ear environment. This study investigates the roles of oxidative stress (OS) and endoplasmic reticulum stress (ERS) in diabetes-related hearing loss (DRHL) and explores the potential of thioredoxin (Trx) in regulating OS, ERS, and apoptosis-related factors to mitigate the progression of hearing impairment. We conducted auditory and serological assessments in 63 patients with type 2 diabetes and 30 healthy controls. Type 2 diabetes models were induced in wild-type and Trx transgenic (Tg) mice, with auditory brainstem response (ABR) used to evaluate hearing changes. Cochlear tissues were isolated to analyse markers of apoptosis, OS, and ERS. Both patients with diabetes and mouse models exhibited hearing loss, alongside increased serum levels of Trx1, TXNIP, and AOPP, indicating oxidative damage. H&E and succinate dehydrogenase (SDH) staining revealed varying degrees of hair cell loss from the base to the apex of the cochlea in diabetic mice, with decreased expression of the hair cell protein prestin gene. Notably, Tg mice showed significant delay in hearing loss progression. In vitro, advanced glycation end-products (AGEs) induced OS and ERS in cochlear-like HEI-OC1 cells, while Trx overexpression enhanced Nrf2 activity, alleviating AGE-induced cellular stress. In conclusion, Trx exhibits protective effects against DRHL, potentially by enhancing Nrf2/HO-1/SOD2 function to reduce OS and ERS.
Collapse
Affiliation(s)
- Meng Xu
- Department of Otorhinolaryngology of the Second Hospital, Dalian Medical University, Dalian 116023, LiaoNing Province, China
| | - Shiwen Zhong
- Department of Otorhinolaryngology of the Second Hospital, Dalian Medical University, Dalian 116023, LiaoNing Province, China
| | - Na Zhu
- Department of Otorhinolaryngology of the Second Hospital, Dalian Medical University, Dalian 116023, LiaoNing Province, China
| | - Sifan Wang
- Department of Otorhinolaryngology of the Second Hospital, Dalian Medical University, Dalian 116023, LiaoNing Province, China
| | - Jingyi Wang
- Department of Histology and Embryology, Dalian Medical University, Dalian 116044, LiaoNing Province, China
| | - Xiang Li
- Department of Otorhinolaryngology of the Second Hospital, Dalian Medical University, Dalian 116023, LiaoNing Province, China
| | - Xiang Ren
- Department of Histology and Embryology, Dalian Medical University, Dalian 116044, LiaoNing Province, China.
| | - Hui Kong
- Department of Otorhinolaryngology of the Second Hospital, Dalian Medical University, Dalian 116023, LiaoNing Province, China.
| |
Collapse
|
37
|
Taguchi K, Kondo H, Matsumoto T, Kobayashi T. Effect of esaxerenone on the onset of aortic endothelial dysfunction and circulating microparticles in type 1 diabetic male mice. Sci Rep 2024; 14:26266. [PMID: 39487333 PMCID: PMC11530539 DOI: 10.1038/s41598-024-78321-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024] Open
Abstract
Endothelial dysfunction exacerbates hypertension and other vascular complications in diabetes mellitus (DM). Circulating microparticles (MPs) and extracellular vesicles released in patients with DM have emerged as novel regulators of endothelial dysfunction. The obstruction of mineralocorticoid receptors (MRs) is a potential therapeutic approach to reduce cardiovascular complications. Their impact on the obstruction of MRs on circulating MPs and endothelial dysfunction in DM remains unclear. DM was induced in mice through a single intravenous dose of streptozotocin (STZ; 200 mg/kg). Esaxerenone (ESAX; 3 mg/kg/day), a MR blocker was administered via diet for 8 weeks. In this study, the aortas of the DM group showed the endothelial dysfunction and the administration of ESAX ameliorated the endothelial-dependent responses. Moreover, ESAX influences the impaired endothelial-dependent responses of DM-derived MPs. Interestingly, MP levels increased in DM whereas decreased after ESAX administration. In the aorta, the DM-derived MPs increased the expression of intercellular adhesion molecule-1 (ICAM-1). ESAX inhibited the adhesion of DM-derived MPs. Moreover, the ICAM-1 inhibitor A205804 shows similar effects as ESAX. These results indicate that the release and adhesion properties of MPs can be partially obstructed by ESAX via the ICAM-1 signaling pathway, which clarifies the other functions beyond the anti-hypertensive effects of ESAX.
Collapse
MESH Headings
- Animals
- Cell-Derived Microparticles/metabolism
- Cell-Derived Microparticles/drug effects
- Male
- Mice
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Aorta/drug effects
- Aorta/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/complications
- Pyrroles/pharmacology
- Sulfones/pharmacology
- Mice, Inbred C57BL
- Mineralocorticoid Receptor Antagonists/pharmacology
- Receptors, Mineralocorticoid/metabolism
Collapse
Affiliation(s)
- Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Hiroyuki Kondo
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, 1714-1 Yoshino-cho, Nobeoka, 882-8508, Miyazaki, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan.
| |
Collapse
|
38
|
Zhang JJ, Ni P, Song Y, Gao MJ, Guo XY, Zhao BQ. Effective protective mechanisms of HO-1 in diabetic complications: a narrative review. Cell Death Discov 2024; 10:433. [PMID: 39389941 PMCID: PMC11466965 DOI: 10.1038/s41420-024-02205-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Diabetes mellitus is a metabolic disorder with persistent hyperglycemia caused by a variety of underlying factors. Chronic hyperglycemia can lead to diverse serious consequences and diversified complications, which pose a serious threat to patients. Among the major complications are cardiovascular disease, kidney disease, diabetic foot ulcers, diabetic retinopathy, and neurological disorders. Heme oxygenase 1 (HO-1) is a protective enzyme with antioxidant, anti-inflammatory and anti-apoptotic effects, which has been intensively studied and plays an important role in diabetic complications. By inducing the expression and activity of HO-1, it can enhance the antioxidant, anti-inflammatory, and anti-apoptotic capacity of tissues, and thus reduce the degree of damage in diabetic complications. The present study aims to review the relationship between HO-1 and the pathogenesis of diabetes and its complications. HO-1 is involved in the regulation of macrophage polarization and promotes the M1 state (pro-inflammatory) towards to the M2 state (anti-inflammatory). Induction of HO-1 expression in dendritic cells inhibits them maturation and secretion of pro-inflammatory cytokines and promotes regulatory T cell (Treg cell) responses. The induction of HO-1 can reduce the production of reactive oxygen species, thereby reducing oxidative stress and inflammation. Besides, HO-1 also has an important effect in novel programmed cell death such as pyroptosis and ferroptosis, thereby playing a protective role against diabetes. In conclusion, HO-1 plays a significant role in the occurrence and development of diabetic complications and is closely associated with a variety of complications. HO-1 is anticipated to serve as a novel target for addressing diabetic complications, and it holds promise as a potential therapeutic agent for diabetes and its associated complications. We hope to provide inspiration and ideas for future studies in the mechanism and targets of HO-1 through this review.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Medicine Research Institute & Hubei Key Laboratory of Diabetes and Angiopathy, Xianning, Hubei, China
- Schools of Pharmacy and Hubei University of Science and Technology, Xianning, China
| | - Ping Ni
- Clinical Medicine, Hubei University of Science and Technology, Xianning, China
| | - Yi Song
- Schools of Pharmacy and Hubei University of Science and Technology, Xianning, China
| | - Man-Jun Gao
- Schools of Pharmacy and Hubei University of Science and Technology, Xianning, China
| | - Xi-Ying Guo
- Medicine Research Institute & Hubei Key Laboratory of Diabetes and Angiopathy, Xianning, Hubei, China.
| | - Bao-Qing Zhao
- Medicine Research Institute & Hubei Key Laboratory of Diabetes and Angiopathy, Xianning, Hubei, China.
| |
Collapse
|
39
|
Shi L, Xu Y, Zhao C, Qu G, Hao M. Liraglutide ameliorates high glucose-induced vascular endothelial injury through TRIB3/NF-κB signaling pathway. In Vitro Cell Dev Biol Anim 2024; 60:1046-1057. [PMID: 39039329 DOI: 10.1007/s11626-024-00947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/26/2024] [Indexed: 07/24/2024]
Abstract
As one of the most commonly used antidiabetic medications clinically, liraglutide is involved in the protection of vascular endothelium, and whether it can relieve high glucose-induced vascular endothelial damage was unknown. This study aims to address the response of liraglutide (LIRA) on human umbilical vein endothelial cells, as well as to elucidate its possible underlying mechanism. We established a vascular endothelial cell injury model by exposing human umbilical vein endothelial cells (HUVECs) to high glucose, and used LIRA pretreatment before HG treatment to address the endothelial protective effect of LIRA. Our results suggest that LIRA prevented HG-induced HUVEC apoptosis, oxidative stress, inflammasome activation, and pyroptosis. Furthermore, silencing of tribbles homolog 3 (TRIB3) could markedly reduce HG-induced HUVEC apoptosis, ROS level, the expressions of TXNIP, cleaved caspase3, NLRP3, and caspase1, indicating TRIB3 inhibition protected HUVECs against HG-induced vascular endothelial injury. In addition, LIRA restrained NF-κB/IκB-α signaling pathway activation in HUVECs. Thus, LIRA appears to mitigate HG-induced apoptosis, oxidative stress, inflammasome activation, and pyroptosis in HUVECs via regulating the TRIB3/NF-κB/IκB-α signaling pathway. Our study provides new insight into the mechanisms underlying the protective activity of LIRA against the vascular endothelial injury in diabetic vascular complication.
Collapse
Affiliation(s)
- Lili Shi
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yingying Xu
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Chao Zhao
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Guangjin Qu
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ming Hao
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
40
|
Dantas IV, Perrier-Melo RJ, Costa MDC, Brito ADF, Brito-Gomes JLD, Vancea DMM. Acute endothelial, blood pressure, and glycemic responses after aerobic sessions in type-2 diabetic with hypertension: A double-blinded randomized study. J Bodyw Mov Ther 2024; 40:1932-1938. [PMID: 39593547 DOI: 10.1016/j.jbmt.2024.10.064] [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: 01/18/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVES To compare the acute response of moderate intensity continuous exercise (MICE) vs moderate intensity interval exercise (MIIE) on endothelial function, post-exercise hypotension (PEH), and glycemia in type 2 diabetes (T2DM) with hypertension. METHODS Twelve T2DM (aged 52.8 ± 3.8 years old) patients with hypertension underwent a randomized cross-over study following isocaloric (200 kcal) protocols: (i) MICE: walk-jogging at 50% of VO2peak, (ii) MIIE session: walking - jogging (stimulus/recovery - 3:3 min each) at 60% of VO2peak followed by recovery at 40% of VO2peak, and (iii) control (CON): lying quietly in a supine position for 30 min. A generalized estimating equation was utilized to verify possible differences over time × session. RESULTS The %FMD (Baseline: 3.59 ± 1.58 vs. 30 min: 6.73 ± 4.34) and the absolute FMD changed after MIIE (Baseline: 0.16 ± 0.10 vs. 30 min: 0.17 ± 0.14). Only absolute FMD changed after MIIE (Baseline: 0.14 ± 0.07 vs. 30 min: 0.16 ± 0.07). Besides, MIIE at 30 min provides higher absolute FMD values when compared to baseline from MICE and baseline, 30 min, and 60 min from control. Regarding blood pressure, no PEH statistical main effect was found. Finally, the glycemia changed at baseline vs. 30 min and 60 min after MIIE (210.5 ± 9.4 vs. 127.6 ± 10.0 and 120 ± 8.9), MICE (219.5 ± 12.7 vs. 125.2 ± 12.0 and 118.2 ± 11.6), and control sessions (215.9 ± 11.8 vs. 187.4 ± 11.2 and 172.6 ± 11.3 mg/dL, p < 0.05). However, MIIE and MICE showed higher decreases compared to the control. CONCLUSIONS MIIE and MICE sessions are similar and effective exercise strategies to induce changes in endothelial function and glycemic responses in type-2 diabetics with hypertension.
Collapse
|
41
|
Geng XF, Shang WY, Qi ZW, Zhang C, Li WX, Yan ZP, Fan XB, Zhang JP. The mechanism and promising therapeutic strategy of diabetic cardiomyopathy dysfunctions: Focus on pyroptosis. J Diabetes Complications 2024; 38:108848. [PMID: 39178624 DOI: 10.1016/j.jdiacomp.2024.108848] [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: 07/05/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
Diabetes is a major risk factor for cardiovascular diseases, and myocardial damage caused by hyperglycemia is the main cause of heart failure. However, there is still a lack of systematic understanding of myocardial damage caused by diabetes. At present, we believe that the cellular inflammatory damage caused by hyperglycemia is one of the causes of diabetic cardiomyopathy. Pyroptosis, as a proinflammatory form of cell death, is closely related to the occurrence and development of diabetic cardiomyopathy. Therefore, this paper focuses on the important role of inflammation in the occurrence and development of diabetic cardiomyopathy. From the perspective of pyroptosis, we summarize the pyroptosis of different types of cells in diabetic cardiomyopathy and its related signaling pathways. It also summarizes the treatment of diabetic cardiomyopathy, hoping to provide methods for the prevention and treatment of diabetic cardiomyopathy by inhibiting pyroptosis.
Collapse
Affiliation(s)
- Xiao-Fei Geng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Wen-Yu Shang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhong-Wen Qi
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Chi Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Wen-Xiu Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhi-Peng Yan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xin-Biao Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Jun-Ping Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
42
|
Zhang T, Jiang D, Zhang X, Chen L, Jiang J, Zhang C, Li S, Li Q. The role of nonmyocardial cells in the development of diabetic cardiomyopathy and the protective effects of FGF21: a current understanding. Cell Commun Signal 2024; 22:446. [PMID: 39327594 PMCID: PMC11426003 DOI: 10.1186/s12964-024-01842-0] [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: 06/05/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) represents a unique myocardial disease originating from diabetic metabolic disturbances that is characterized by myocardial fibrosis and diastolic dysfunction. While recent research regarding the pathogenesis and treatment of DCM has focused primarily on myocardial cells, nonmyocardial cells-including fibroblasts, vascular smooth muscle cells (VSMCs), endothelial cells (ECs), and immune cells-also contribute significantly to the pathogenesis of DCM. Among various therapeutic targets, fibroblast growth factor 21 (FGF21) has been identified as a promising agent because of its cardioprotective effects that extend to nonmyocardial cells. In this review, we aim to elucidate the role of nonmyocardial cells in DCM and underscore the potential of FGF21 as a therapeutic strategy for these cells.
Collapse
Affiliation(s)
- Tianyi Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Donghui Jiang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiao Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Shengbiao Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Qiuhong Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
43
|
Hoffmann M, Poschenrieder J, Incudini M, Baier S, Fritz A, Maier A, Hartung M, Hoffmann C, Trummer N, Adamowicz K, Picciani M, Scheibling E, Harl M, Lesch I, Frey H, Kayser S, Wissenberg P, Schwartz L, Hafner L, Acharya A, Hackl L, Grabert G, Lee SG, Cho G, Cloward M, Jankowski J, Lee H, Tsoy O, Wenke N, Pedersen A, Bønnelykke K, Mandarino A, Melograna F, Schulz L, Climente-González H, Wilhelm M, Iapichino L, Wienbrandt L, Ellinghaus D, Van Steen K, Grossi M, Furth P, Hennighausen L, Di Pierro A, Baumbach J, Kacprowski T, List M, Blumenthal D. Network medicine-based epistasis detection in complex diseases: ready for quantum computing. Nucleic Acids Res 2024; 52:10144-10160. [PMID: 39175109 PMCID: PMC11417373 DOI: 10.1093/nar/gkae697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
Most heritable diseases are polygenic. To comprehend the underlying genetic architecture, it is crucial to discover the clinically relevant epistatic interactions (EIs) between genomic single nucleotide polymorphisms (SNPs) (1-3). Existing statistical computational methods for EI detection are mostly limited to pairs of SNPs due to the combinatorial explosion of higher-order EIs. With NeEDL (network-based epistasis detection via local search), we leverage network medicine to inform the selection of EIs that are an order of magnitude more statistically significant compared to existing tools and consist, on average, of five SNPs. We further show that this computationally demanding task can be substantially accelerated once quantum computing hardware becomes available. We apply NeEDL to eight different diseases and discover genes (affected by EIs of SNPs) that are partly known to affect the disease, additionally, these results are reproducible across independent cohorts. EIs for these eight diseases can be interactively explored in the Epistasis Disease Atlas (https://epistasis-disease-atlas.com). In summary, NeEDL demonstrates the potential of seamlessly integrated quantum computing techniques to accelerate biomedical research. Our network medicine approach detects higher-order EIs with unprecedented statistical and biological evidence, yielding unique insights into polygenic diseases and providing a basis for the development of improved risk scores and combination therapies.
Collapse
Affiliation(s)
- Markus Hoffmann
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Advanced Study (Lichtenbergstrasse 2 a) Technical University of Munich, D-85748 Garching, Germany
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Julian M Poschenrieder
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Massimiliano Incudini
- Dipartimento di Informatica, Universit‘a di Verona, Strada le Grazie 15 - 34137 Verona, Italy
| | - Sylvie Baier
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Amelie Fritz
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, DTU, 2800 Kgs. Lyngby, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Michael Hartung
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Christian Hoffmann
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Nico Trummer
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Klaudia Adamowicz
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Mario Picciani
- Computational Mass Spectrometry, Technical University of Munich, Freising, Germany
| | - Evelyn Scheibling
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Maximilian V Harl
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Ingmar Lesch
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hunor Frey
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Simon Kayser
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Paul Wissenberg
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Leon Schwartz
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Leon Hafner
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Advanced Study (Lichtenbergstrasse 2 a) Technical University of Munich, D-85748 Garching, Germany
| | - Aakriti Acharya
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technische Universität Braunschweig and Hannover Medical School, Rebenring 56, 38106 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Lena Hackl
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Gordon Grabert
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technische Universität Braunschweig and Hannover Medical School, Rebenring 56, 38106 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Sung-Gwon Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Gyuhyeok Cho
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Korea
| | | | - Jakub Jankowski
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Hye Kyung Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Olga Tsoy
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Nina Wenke
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Anders Gorm Pedersen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, DTU, 2800 Kgs. Lyngby, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Antonio Mandarino
- International Centre for Theory of Quantum Technologies, University of Gdańsk, 80-309 Gdańsk, Poland
| | - Federico Melograna
- BIO3 - Systems Genetics; GIGA-R Medical Genomics, University of Liège, Liège, Belgium
- BIO3 - Systems Medicine; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Laura Schulz
- Leibniz Supercomputing Centre of the Bavarian Academy of Sciences and Humanities (LRZ), Garching b. München, Germany
| | | | - Mathias Wilhelm
- Computational Mass Spectrometry, Technical University of Munich, Freising, Germany
- Munich Data Science Institute (MDSI), Technical University of Munich, Garching, Germany
| | - Luigi Iapichino
- Leibniz Supercomputing Centre of the Bavarian Academy of Sciences and Humanities (LRZ), Garching b. München, Germany
| | - Lars Wienbrandt
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Kristel Van Steen
- BIO3 - Systems Genetics; GIGA-R Medical Genomics, University of Liège, Liège, Belgium
- BIO3 - Systems Medicine; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Michele Grossi
- European Organization for Nuclear Research (CERN), Geneva1211, Switzerland
| | - Priscilla A Furth
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA
| | - Lothar Hennighausen
- Institute for Advanced Study (Lichtenbergstrasse 2 a) Technical University of Munich, D-85748 Garching, Germany
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Alessandra Di Pierro
- Dipartimento di Informatica, Universit‘a di Verona, Strada le Grazie 15 - 34137 Verona, Italy
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Germany
- Computational BioMedicine Lab, University of Southern Denmark, Denmark
| | - Tim Kacprowski
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technische Universität Braunschweig and Hannover Medical School, Rebenring 56, 38106 Braunschweig, Germany
| | - Markus List
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
44
|
Park J, Song H, Moon S, Kim Y, Cho S, Han K, Park CY, Cho SW, Oh CM. Cardiometabolic benefits of fenofibrate in heart failure related to obesity and diabetes. Cardiovasc Diabetol 2024; 23:343. [PMID: 39285303 PMCID: PMC11406805 DOI: 10.1186/s12933-024-02417-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a serious and common condition affecting millions of people worldwide, with obesity being a major cause of metabolic disorders such as diabetes and cardiovascular disease. This study aimed to investigate the effects of fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARα) agonist, on the obese- and diabetes-related cardiomyopathy. METHODS AND RESULTS We used db/db mice and high fat diet-streptozotocin induced diabetic mice to investigate the underlying mechanisms of fenofibrate's beneficial effects on heart function. Fenofibrate reduced fibrosis, and lipid accumulation, and suppressed inflammatory and immunological responses in the heart via TNF signaling. In addition, we investigated the beneficial effects of fenofibrate on HF hospitalization. The Korean National Health Insurance database was used to identify 427,154 fenofibrate users and 427,154 non-users for comparison. During the 4.22-year follow-up, fenofibrate use significantly reduced the risk of HF hospitalization (hazard ratio, 0.907; 95% CI 0.824-0.998). CONCLUSIONS The findings suggest that fenofibrate may be a useful therapeutic agent for obesity- and diabetes-related cardiomyopathy.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hangyul Song
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Shinje Moon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Sungsoo Cho
- Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Cheol-Young Park
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje Univeristy Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Gyeonggi-Do, Korea.
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea.
| |
Collapse
|
45
|
Zhang Y, Chai S, Dai H, Chen X, Meng Z, Ying X. Vascular endothelial function and its response to moderate-intensity aerobic exercise in trained and untrained healthy young men. Sci Rep 2024; 14:20450. [PMID: 39242762 PMCID: PMC11379850 DOI: 10.1038/s41598-024-71471-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024] Open
Abstract
30 min of moderate-intensity aerobic exercise per day is recommended, but the response and adaptation of endothelial function (EF) to this exercise remains controversial. The purpose of this study was to determine the changes in EF in endurance trained and untrained individuals before and after this exercise and to compare the differences between trained and untrained individuals. Twelve endurance-trained male college athletes (trained group) and 12 untrained male college students (untrained group) performed a 30-min run at an intensity of 60% VO2max. Brachial artery flow-mediated dilation (FMD) was measured before exercise, 30 min and 60 min after exercise, and the following morning. Resting diameter and maximum diameter showed large time effects (p < 0.001, η2 = 0.533; p < 0.001, η2 = 0.502). Resting diameters at 30 and 60 min after exercise were higher than before exercise in both the untrained and trained groups (p < 0.05), and maximum diameters at 30 min after exercise were higher than before exercise in both the untrained and trained groups (p < 0.01). Resting diameter and maximum diameter also exhibited some group effects (p = 0.055, η2 = 0.157; p = 0.041, η2 = 0.176). Resting diameters and maximum diameters were higher in the trained group than in the untrained group before exercise (p < 0.05). FMD (%) showed no time, group, or time-group interaction effects. 30 min of moderate-intensity aerobic exercise can increase resting and maximal arterial diameters in both trained and untrained young men, but has no effect on FMD. Long-term endurance training has the potential to increase resting and maximal arterial diameters in young men, but not necessarily FMD.
Collapse
Affiliation(s)
- Yong Zhang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Shiyi Chai
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Hailun Dai
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Xiaofei Chen
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Zhaofeng Meng
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China.
| | - Xiaofang Ying
- School of Business, Shaoxing University, Shaoxing, China.
| |
Collapse
|
46
|
Hu SY, Xue CD, Li YJ, Li S, Gao ZN, Qin KR. Microfluidic investigation for shear-stress-mediated repair of dysglycemia-induced endothelial cell damage. MECHANOBIOLOGY IN MEDICINE 2024; 2:100069. [PMID: 40395495 PMCID: PMC12082321 DOI: 10.1016/j.mbm.2024.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/29/2024] [Accepted: 04/19/2024] [Indexed: 01/03/2025]
Abstract
Dysglycemia causes arterial endothelial damage, which is an early critical event in vascular complications for diabetes patients. Physiologically, moderate shear stress (SS) helps maintain endothelial cell health and normal function. Reactive oxygen species (ROS) and calcium ions (Ca2+) signals are involved in dysglycemia-induced endothelial dysfunction and are also implicated in SS-mediated regulation of endothelial cell function. Therefore, it is urgent to establish in vitro models for studying endothelial biomechanics and mechanobiology, aiming to seek interventions that utilize appropriate SS to delay or reverse endothelial dysfunction. Microfluidic technology, as a novel approach, makes it possible to replicate blood glucose environment and accurate pulsatile SS in vitro. Here, we reviewed the progress of microfluidic systems used for SS-mediated repair of dysglycemia-induced endothelial cell damage (ECD), revealing the crucial roles of ROS and Ca2+ during the processes. It holds significant implications for finding appropriate mechanical intervention methods, such as exercise training, to prevent and treat cardiovascular complications in diabetes.
Collapse
Affiliation(s)
- Si-Yu Hu
- School of Mechanical Engineering, Dalian University of Technology, No. 2, Linggong Rd., Dalian 116024, Liaoning Province, China
| | - Chun-Dong Xue
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, No. 2, Linggong Rd., Dalian 116024, Liaoning Province, China
| | - Yong-Jiang Li
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, No. 2, Linggong Rd., Dalian 116024, Liaoning Province, China
| | - Shen Li
- Central Hospital of Dalian University of Technology, No. 826, Xinan Rd., Dalian 116033, Liaoning Province, China
| | - Zheng-Nan Gao
- Central Hospital of Dalian University of Technology, No. 826, Xinan Rd., Dalian 116033, Liaoning Province, China
| | - Kai-Rong Qin
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, No. 2, Linggong Rd., Dalian 116024, Liaoning Province, China
- Central Hospital of Dalian University of Technology, No. 826, Xinan Rd., Dalian 116033, Liaoning Province, China
| |
Collapse
|
47
|
Mao A, Li Z, Shi X, Zhang K, Kan H, Geng L, He D. Complement Factor C1q Mediates Vascular Endothelial Dysfunction in STZ-Induced Diabetic Mice. Diabetes 2024; 73:1527-1536. [PMID: 38869460 DOI: 10.2337/db23-0981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Diabetes is a significant global public health issue with implications for vascular endothelial cells (ECs) dysfunction and the subsequent development and advancement of diabetes complications. This study aims to compare the cellular and molecular properties of the aorta in normal and streptozotocin (STZ)-induced diabetic mice, with a focus on elucidating potential mechanism underlying EC dysfunction. Here, we performed a single-cell RNA sequencing survey of 32,573 cells from the aorta of normal and STZ-induced diabetic mice. We found a compendium of 10 distinct cell types, mainly ECs, smooth muscle cells, fibroblast, pericyte, immune cells, and stromal cells. As the diabetes condition progressed, we observed a subpopulation of aortic ECs that exhibited significantly elevated expression of complement (C) molecule C1qa compared with their healthy counterparts. This increased expression of C1qa was found to induce reactive oxygen species (ROS) production, facilitate EC migration and increased permeability, and impair the vasodilation within the aortic segment of mice. Furthermore, AAV-Tie2-shRNA-C1qa was administered into diabetic mice by tail vein injection, showing that inhibition of C1qa in the endothelium led to a reduction in ROS production, decreased vascular permeability, and improved vasodilation. Collectively, these findings highlight the crucial involvement of C1qa in endothelial dysfunction associated with diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zicheng Li
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoming Shi
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongxu He
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
48
|
Guo L, Zong Y, Yang W, Lin Y, Feng Q, Yu C, Liu X, Li C, Zhang W, Wang R, Li L, Pei Y, Wang H, Liu D, Niu H, Nie L. DCBLD2 deletion increases hyperglycemia and induces vascular remodeling by inhibiting insulin receptor recycling in endothelial cells. FEBS J 2024; 291:4076-4095. [PMID: 38872483 DOI: 10.1111/febs.17198] [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: 09/22/2023] [Revised: 04/02/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Discoidin, CUB, LCCL domain-containing 2 (DCBLD2) is a type I transmembrane protein with a similar structure to neuropilin, which acts as a co-receptor for certain receptor tyrosine kinases (RTKs). The insulin receptor is an RTK and plays a critical role in endothelial cell function and glycolysis. However, how and whether DCBLD2 regulates insulin receptor activity in endothelial cells is poorly understood. Diabetes was induced through treatment of Dcbld2 global-genome knockout mice and endothelium-specific knockout mice with streptozotocin. Vascular ultrasound, vascular tension test, and hematoxylin and eosin staining were performed to assess endothelial function and aortic remodeling. Glycolytic rate assays, real-time PCR and western blotting were used to investigate the effects of DCBLD2 on glycolytic activity and insulin receptor (InsR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway in endothelial cells. Co-immunoprecipitation was used to assess the effects of DCBLD2 on insulin receptor endocytosis and recycling. Membrane and cytoplasmic proteins were isolated to determine whether DCBLD2 could affect the localization of the insulin receptor. We found that Dcbld2 deletion exacerbated endothelial dysfunction and vascular remodeling in diabetic mice. Both Dcbld2 knockdown and Dcbld2 deletion inhibited glycolysis and the InsR/PI3K/Akt signaling pathway in endothelial cells. Furthermore, Dcbld2 deletion inhibited insulin receptor recycling. Taken together, Dcbld2 deficiency exacerbated diabetic endothelial dysfunction and vascular remodeling by inhibiting the InsR/PI3K/Akt pathway in endothelial cells through the inhibition of Rab11-dependent insulin receptor recycling. Our data suggest that DCBLD2 is a potential therapeutic target for diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
- Lingling Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yanhong Zong
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Weiwei Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yanling Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Qi Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Chao Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Xiaoning Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Chenyang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Wenjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Runtao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Lijing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Yunli Pei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Huifang Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| | - Demin Liu
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Honglin Niu
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
- School of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Lei Nie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, China
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
49
|
Aluksanasuwan S, Somsuan K, Chiangjong W, Rongjumnong A, Jaidee W, Rujanapun N, Chutipongtanate S, Laphookhieo S, Charoensup R. SWATH-proteomics reveals Mathurameha, a traditional anti-diabetic herbal formula, attenuates high glucose-induced endothelial dysfunction through the EGF/NO/IL-1β regulatory axis. J Proteomics 2024; 306:105263. [PMID: 39047940 DOI: 10.1016/j.jprot.2024.105263] [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: 05/25/2024] [Revised: 07/02/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Mathurameha is a traditional Thai herbal formula with a clinically proven effect of blood sugar reduction in patients with diabetes mellitus, but its anti-diabetic complication potential is largely unknown. This study aimed to elucidate the effects of Mathurameha and its underlying mechanisms against high glucose-induced endothelial dysfunction in human endothelial EA.hy926 cells. After confirming no cytotoxic effects, the cells were treated with normal glucose (NG), high glucose (HG), or high glucose plus Mathurameha (HG + M) for 24 h. A quantitative label-free proteomic analysis using the sequential window acquisition of all theoretical mass spectra (SWATH-MS) approach identified 24 differentially altered proteins among the three groups: 7 between HG and NG, 9 between HG + M and NG, and 13 between HG + M and HG. Bioinformatic analyses suggested a potential anti-diabetic action through the epidermal growth factor (EGF) pathway. Subsequent functional validations demonstrated that Mathurameha reduced the EGF secretion and the intracellular reactive oxygen species (ROS) level in high glucose-treated cells. Mathurameha also exhibited a stimulatory effect on nitric oxide (NO) production while significantly reducing the secretion of endothelin-1 (ET-1) and interleukin-1β (IL-1β) in high glucose-treated cells. In conclusion, our findings demonstrated that Mathurameha attenuated high glucose-induced endothelial dysfunction through the EGF/NO/IL-1β regulatory axis. SIGNIFICANCE: This study reveals the potential of Mathurameha, a traditional Thai herbal formula, in mitigating high glucose-induced endothelial dysfunction, a common complication in diabetes mellitus. Using proteomics and bioinformatic analyses followed by functional validations, the present study highlights the protective effects of Mathurameha through the EGF/NO/IL-1β regulatory axis. These findings support its potential use as a therapeutic intervention for diabetic vascular complications and provide valuable information for developing more effective anti-diabetic drugs.
Collapse
Affiliation(s)
- Siripat Aluksanasuwan
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand.
| | - Keerakarn Somsuan
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Artitaya Rongjumnong
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Wuttichai Jaidee
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Narawadee Rujanapun
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Somchai Chutipongtanate
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0056, USA
| | - Surat Laphookhieo
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai 57100, Thailand; Center of Chemical Innovation for Sustainability (CIS), School of Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Rawiwan Charoensup
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai 57100, Thailand; School of Integrative Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand.
| |
Collapse
|
50
|
Usatiuc LO, Pârvu M, Pop RM, Uifălean A, Vălean D, Szabo CE, Țicolea M, Cătoi FA, Ranga F, Pârvu AE. Phytochemical Profile and Antidiabetic, Antioxidant, and Anti-Inflammatory Activities of Gypsophila paniculata Ethanol Extract in Rat Streptozotocin-Induced Diabetes Mellitus. Antioxidants (Basel) 2024; 13:1029. [PMID: 39334688 PMCID: PMC11428820 DOI: 10.3390/antiox13091029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/17/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
The present study aimed to investigate the effects of the Gypsophila paniculata ethanol extract (GPEE) on oxidative stress, inflammation, and metabolic markers in a rat model of streptozotocin-induced diabetes mellitus (DM). Phytochemical analysis using high-performance liquid chromatography coupled with mass spectrometry was performed to measure the total phenolic and flavonoid contents. In vitro antioxidant activity was evaluated through DPPH, FRAP, H2O2, and NO scavenging tests, and the in vivo effects of the GPEE were assessed in streptozotocin-induced DM rats. Treatments with the GPEE, metformin, and Trolox were administrated by gavage for 10 days. On day 11, blood was collected, and serum oxidative stress (total oxidative status, oxidative stress index, malondialdehyde, advanced oxidation protein products, 8-hydroxydeoxyguanosine, nitric oxide, 3-nitrotyrosine, advanced glycation end-products, total antioxidant reactivity, total thiols), inflammatory (IL-1β, NF-κB, IL-18, and gasdermin D), metabolic (fasting glucose, total cholesterol, triglycerides, and triglyceride-glucose index), and liver injury (AST, ALT, and AST:ALT ratio) markers were measured. The GPEE was found to have a significant polyphenols content and a moderate in vitro antioxidant effect. In vivo, the GPEE lowered oxidants and increased antioxidants, decreased inflammatory markers and blood glucose, and improved lipid profiles and transaminases in a dose-dependent manner, with higher doses having a better effect, being comparable to those of metformin and Trolox.
Collapse
Affiliation(s)
- Lia-Oxana Usatiuc
- Pathophysiology, Department 1-Morphofunctional Sciences, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Marcel Pârvu
- Department of Taxonomy, Faculty of Biology and Geology, "Babes-Bolyai" University, 400012 Cluj-Napoca, Romania
| | - Raluca Maria Pop
- Pharmacology, Toxicology and Clinical Pharmacology, Department 1-Morphofunctional Sciences, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ana Uifălean
- Pathophysiology, Department 1-Morphofunctional Sciences, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Dan Vălean
- Surgery Department, "Iuliu Haţieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Csilla-Eniko Szabo
- Pediatric Clinic 1, Department of Mother and Child, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Mădălina Țicolea
- Pathophysiology, Department 1-Morphofunctional Sciences, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Florinela Adriana Cătoi
- Pathophysiology, Department 1-Morphofunctional Sciences, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Floricuța Ranga
- Food Science and Technology, Department of Food Science, University of Agricultural Science and Veterinary Medicine Cluj-Napoca, Calea Mănăștur, No 3-5, 400372 Cluj-Napoca, Romania
| | - Alina Elena Pârvu
- Pathophysiology, Department 1-Morphofunctional Sciences, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|