1
|
Liu Z, Wang H, Dai L, Zeng H, Zhong X. Y-box binding protein 1: A critical target for understanding and treating cardiovascular disease. Cell Signal 2025; 132:111797. [PMID: 40204098 DOI: 10.1016/j.cellsig.2025.111797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Cardiovascular diseases (CVDs) remain a significant public health burden, characterized by escalating morbidity and mortality rates and demanding novel therapeutic approaches. Cold shock protein Y-box binding protein 1 (YB-1), a highly conserved RNA/DNA-binding protein, has emerged as a pivotal regulator in various pathophysiological processes, including CVDs. YB-1 exerts pleiotropic functions by modulating gene transcription, pre-mRNA splicing, mRNA translation, and stability. The expression and function of YB-1 are intricately regulated by its subcellular localization, post-translational modifications, upstream regulatory signals. YB-1 plays a multifaceted role in CVDs, influencing inflammation, oxidative stress, cell proliferation, apoptosis, phenotypic switching of smooth muscle cells, and mitochondrial dysfunction. However, the regulation of YB-1 expression and function in CVDs is complex and context-dependent, exhibiting divergent effects even in the same disease across different cell types or at disease stages. This review comprehensively explores the structure, regulation, and functional significance of YB-1 in CVDs. We delve into the transcriptional and translational control mechanisms of YB-1, as well as its post-translational modifications. Furthermore, we elucidate the upstream signaling pathways that influence YB-1 expression, with a particular emphasis on non-coding RNAs and specific upstream molecules. Finally, we systematically examine the role of YB-1 in CVDs, summarizing its expression patterns, regulatory mechanisms, and therapeutic potential as a promising target for novel therapeutic interventions. By providing a comprehensive overview of YB-1's involvement in CVDs, this review aims to stimulate further research and facilitate the development of targeted therapies to improve cardiovascular health.
Collapse
Affiliation(s)
- Zixuan 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
| | - Hongjie Wang
- 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
| | - Lei Dai
- 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
| | - Hesong Zeng
- 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.
| | - Xiaodan Zhong
- 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
|
2
|
Wang M, Cai H, Wang M, Gao H, Zhao X, Rong R, Zhang J, Zhang Z, Liu G, Yuan T, Xia F, Zhu L, Yan Z, Kong X, Qin W. Lonicerin targets ADRA1D and RSPO3 to ameliorate diabetes-induced vascular injury through Ca 2+/Calcineurin/NFAT1-dependent anti-EndMT pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156832. [PMID: 40398181 DOI: 10.1016/j.phymed.2025.156832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/11/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Cardiovascular complications are the primary cause of mortality and disability among diabetic patients. Lonicerin, a major bioactive compound in Lonicera japonica Thunb., has unclear effects and mechanisms on diabetic vascular injury. PURPOSE The research aims to investigate the therapeutic effects of lonicerin on diabetic vascular injury and elucidate its underlying molecular mechanisms. METHODS Streptozotocin (STZ)-induced mice and high glucose-treated human aortic endothelial cells (HAECs) were employed as animal and cellular models of diabetes. Endothelial-to-mesenchymal transition (EndMT) was validated by examining EndMT-related markers and endothelial/mesenchymal functions. RNA sequencing was used to identify potential mechanisms through which lonicerin alleviates EndMT. The relationship between lonicerin and its targets was investigated using RNAi, plasmid overexpression, western blot, qRT-PCR, immunofluorescence, flow cytometry, and calcineurin activity assays. RESULTS Lonicerin dose-dependently alleviates diabetes-induced vascular injury (intimal damage, inflammation, calcification, and fibrosis) and EndMT. Lonicerin produces anti-EndMT effect by inhibiting cytoplasmic Ca²⁺ levels. Further analysis reveals that adrenoceptor alpha 1D (ADRA1D) and R-spondin 3 (RSPO3) are targets of lonicerin. Lonicerin reduces high glucose-induced upregulation of ADRA1D and RSPO3, leading to decreased cytoplasmic Ca²⁺ levels. This reduction inhibits calcineurin activity, promotes nuclear factor of activated T cells 1 (NFAT1) phosphorylation, and prevents its nuclear translocation, ultimately exerting an anti-EndMT effect. EC-specific overexpression of ADRA1D or RSPO3 negates the inhibitory effects of lonicerin on EndMT and its therapeutic impact on diabetic vascular injury. CONCLUSION Lonicerin targets ADRA1D and RSPO3 to ameliorate diabetes-induced vascular injury through the Ca2+/Calcineurin/NFAT1-dependent anti-EndMT pathway. Thus, this study provides the first evidence that lonicerin is a potential novel therapeutic agent for diabetic vasculopathy.
Collapse
Affiliation(s)
- Minghui Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, PR China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Huiying Cai
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, PR China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Manman Wang
- Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment), Affiliated Hospital of Jining Medical University, Jining 272000, Shandong, PR China
| | - Honggang Gao
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Xiaona Zhao
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China; School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, PR China
| | - Ruixue Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, PR China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Jing Zhang
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Zejin Zhang
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China; School of Pharmacy, Binzhou Medical University, Yantai 264000, Shandong, PR China
| | - Guoqing Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, PR China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Tao Yuan
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China; School of Pharmacy, Shandong First Medical University, Jinan 250000, Shandong, PR China
| | - Fangjie Xia
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China; School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, PR China
| | - Lin Zhu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, PR China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Zhenzhen Yan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, PR China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China
| | - Xinxin Kong
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China; School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, PR China
| | - Wei Qin
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, PR China; Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment), Affiliated Hospital of Jining Medical University, Jining 272000, Shandong, PR China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Jinan 250000, Shandong, PR China.
| |
Collapse
|
3
|
Yin J, Song Z, Zhang L, Cong J. Methylophiopogonanone A alleviates diabetic cardiomyopathy via inhibiting JNK1 signaling. Cell Signal 2025; 131:111762. [PMID: 40139620 DOI: 10.1016/j.cellsig.2025.111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Diabetic cardiomyopathy (DCM) is a common complication of type 2 diabetes mellitus (T2DM). The effects of methylophiopogonanone A (MO-A), a natural homoisoflavonoid with anti-inflammatory effects, on DCM and its underlying mechanisms were investigated in this study. METHODS The T2DM mouse model was induced by intraperitoneal injection of 30 mg/kg streptozotocin for 7 consecutive days and fed with a high-fat diet for 12 weeks. T2DM mice received MO-A (2.5, 5, or 10 mg/kg) treatment for two weeks. Cardiac function, hypertrophy, fibrosis, and inflammation were evaluated. The binding energy between MO-A and JNK1 was analyzed using molecular docking. The underlying mechanism was further investigated in high glucose (HG)-induced H9C2 cells. The cytotoxic effects, cardiomyocyte hypertrophy, fibrosis, inflammation, and relevant signaling proteins were assessed. RESULTS MO-A treatment alleviated cardiac function and histopathological changes in DCM mice. Moreover, MO-A treatment significantly decreased COLI, TGF-β1, MYH7, and ANP expression levels in DCM mice. Furthermore, TNF-α, IL-6, and IL-1β expression levels were notably downregulated after treatment with MO-A in DCM mice. Similar results were also observed in vitro. Mechanistically, MO-A targets JNK1 and downregulates its phosphorylation levels in DCM mice. The protective properties of MO-A were reversed by JNK1 overexpression in HG-induced H9C2 cells. CONCLUSION Our results revealed that MO-A could alleviate cardiac function, hypertrophy, fibrosis, and inflammation in DCM via inhibiting JNK1 signaling.
Collapse
Affiliation(s)
- Jing Yin
- Department of Traditional Chinese Medicine, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Zhicheng Song
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong, China
| | - Lijun Zhang
- Department of Endocrinology, Longkou Traditional Chinese Medicine Hospital, Yantai 265701, Shandong, China
| | - Jialin Cong
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong, China.
| |
Collapse
|
4
|
Yang SJ. Dose-dependent L-arginine cardiotoxicity in diabetic cardiomyopathy: Mechanisms and clinical implications. World J Diabetes 2025; 16:104851. [DOI: 10.4239/wjd.v16.i6.104851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 06/13/2025] Open
Abstract
This letter addresses the study in recent publication in the World Journal of Diabetes. The study provides valuable insights and a comprehensive analysis of the effects of L-arginine (L-Arg) supplementation on cardiac health in diabetic rats, highlighting the potential risks associated with high doses of L-Arg. The findings suggest that while low doses of L-Arg may offer some benefits, higher doses can exacerbate myocardial injury through increased oxidative stress, inflammation, and structural disruption. This finding challenges the prevailing point of L-Arg as a universally beneficial supplement and emphasizes the need for further research in its clinical application, particularly in diabetic patients.
Collapse
Affiliation(s)
- Si-Jun Yang
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi 435000, Hubei Province, China
| |
Collapse
|
5
|
Shen S, Sugai-Munson A, Xiao X, Zhang Y, Kang J, Khetarpal SA, Jonas ER, Ambardekar AV, Bristow MR, Li H. Exercise Mitigates Diabetic Cardiomyopathy by Suppressing Ferroptosis via N-Acetyltransferase 10. Circ Res 2025. [PMID: 40492308 DOI: 10.1161/circresaha.125.326303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
Affiliation(s)
- Siman Shen
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
| | - Aya Sugai-Munson
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
| | - Xiao Xiao
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
| | - Yajing Zhang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., S.A.K., H.L.)
| | - Jiayi Kang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University, New York, NY (J.K.)
| | - Sumeet A Khetarpal
- Cardiovascular Research Center, Massachusetts General Hospital, Boston. (S.A.K.)
- Harvard Medical School, Boston, MA (Y.Z., S.A.K., H.L.)
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA (S.A.K.)
| | - Eric R Jonas
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Amrut V Ambardekar
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Michael R Bristow
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Haobo Li
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., S.A.K., H.L.)
| |
Collapse
|
6
|
Dennis KM, Gopal K, Montes Aparicio CN, Zhang JA, Castro-Guarda M, Nicol T, Devereux RM, Carter RD, Azizi SA, Lan T, Purnama U, Carr CA, Simsek G, Gill EK, Swietach P, Sorop O, Heinonen IH, Schianchi F, Luiken JJ, Aksentijevic D, Duncker DJ, Dickinson BC, De Val S, Ussher JR, Fuller W, Heather LC. FoxO1-zDHHC4-CD36 S-Acylation Axis Drives Metabolic Dysfunction in Diabetes. Circ Res 2025; 136:1545-1560. [PMID: 40357580 PMCID: PMC12136392 DOI: 10.1161/circresaha.124.325918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND The fatty acid (FA) transporter CD36 (FA translocase/cluster of differentiation 36) is the gatekeeper of cardiac FA metabolism. Preferential localisation of CD36 to the sarcolemma is one of the initiating cellular responses in the development of muscle insulin resistance and in the type 2 diabetic heart. Post-translational S-acylation controls protein trafficking, and in this study we hypothesised that increased CD36 S-acylation may underpin the preferential sarcolemmal localisation of CD36, driving metabolic and contractile dysfunction in diabetes. METHODS Type 2 diabetes was induced in the rat using high fat diet and a low dose of streptozotocin. Forkhead box O1 (FoxO1) transcriptional regulation of zDHHC4 (zinc finger DHHC-type palmitoyltransferase 4) and subsequent S-acylation of CD36 was assessed using chromatin immunoprecipitation (ChIP) sequencing, ChIP-quantitative polymerase chain reaction, luciferase assays, siRNA (small interfering RNA) and shRNA silencing. RESULTS Type 2 diabetes increased cardiac CD36 S-acylation, CD36 sarcolemmal localisation, FA oxidation rates and triglyceride storage in the diabetic heart. CD36 S-acylation was increased in diabetic rats, db/db mice, diabetic pigs and insulin-resistant human iPSC-derived cardiomyocytes, demonstrating conservation between species. The enzyme responsible for S-acylating CD36, zDHHC4, was transcriptionally upregulated in the diabetic heart, and genetic silencing of zDHHC4 decreased CD36 S-acylation. We identified that zDHHC4 expression is under the regulation of the transcription factor FoxO1. Diabetic mice with cardiomyocyte-specific FoxO1 deletion had decreased cardiac zDHHC4 expression and decreased CD36 S-acylation, which was further confirmed using diabetic mice treated with the FoxO1 inhibitor AS1842856. Pharmacological inhibition of zDHHC enzymes in diabetic hearts decreased CD36 S-acylation, sarcolemmal CD36 content, FA oxidation rates and triglyceride storage, culminating in improved cardiac function in diabetes. Conversely, inhibiting the de-acylating enzymes in control hearts increased CD36 S-acylation, sarcolemmal CD36 content and FA metabolic rates in control hearts, recapitulating the metabolic phenotype seen in diabetic hearts. CONCLUSIONS Activation of the FoxO1-zDHHC4-CD36 S-acylation axis drives metabolic and contractile dysfunction in the type 2 diabetic heart.
Collapse
Affiliation(s)
- Kaitlyn M.J.H. Dennis
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada (K.G., J.R.U.)
| | - Claudia N. Montes Aparicio
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Jiashuo Aaron Zhang
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Marcos Castro-Guarda
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Thomas Nicol
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Ríona M. Devereux
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
- Department of Chemistry (R.M.D.), University of Oxford, United Kingdom
| | - Ryan D. Carter
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Saara-Anne Azizi
- Department of Chemistry, University of Chicago, IL (S.-A.A., T.L., B.C.D.)
| | - Tong Lan
- Department of Chemistry, University of Chicago, IL (S.-A.A., T.L., B.C.D.)
| | - Ujang Purnama
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Gul Simsek
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Eleanor K. Gill
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands (O.S., I.H.A.H., D.J.D.)
| | - Ilkka H.A. Heinonen
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands (O.S., I.H.A.H., D.J.D.)
- Turku PET Centre, University of Turku and Turku University Hospital, Finland (I.H.A.H.)
| | - Francesco Schianchi
- Faculty of Health, Medicine and Life Sciences, Department of Genetics and Cell Biology, Maastricht University, the Netherlands (F.S., J.J.F.P.L.)
| | - Joost J.F.P. Luiken
- Faculty of Health, Medicine and Life Sciences, Department of Genetics and Cell Biology, Maastricht University, the Netherlands (F.S., J.J.F.P.L.)
| | - Dunja Aksentijevic
- Barts and the London Faculty of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, United Kingdom (D.A.)
| | - Dirk J. Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, the Netherlands (O.S., I.H.A.H., D.J.D.)
| | - Bryan C. Dickinson
- Department of Chemistry, University of Chicago, IL (S.-A.A., T.L., B.C.D.)
| | - Sarah De Val
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| | - John R. Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada (K.G., J.R.U.)
| | - William Fuller
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (W.F.)
| | - Lisa C. Heather
- Department of Physiology, Anatomy and Genetics (K.M.J.H.D., C.N.M.A., J.A.Z., M.C.-G., T.N., R.M.D., R.D.C., U.P., C.A.C., G.S., E.K.G., P.S., S.D.V., L.C.H.), University of Oxford, United Kingdom
| |
Collapse
|
7
|
Alcover S, López S, Ramos-Regalado L, Muñoz-García N, Gallinat A, Suades R, Badimon L, Vilahur G. Cardioprotection During Myocardial Infarction in Diabetic Cardiomyopathy. Diabetes 2025; 74:1021-1032. [PMID: 40080393 PMCID: PMC12097457 DOI: 10.2337/db24-0510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Patients with diabetes are at an increased risk of diabetic cardiomyopathy (DCM) and acute myocardial infarction (AMI). Protecting the heart against AMI is more challenging in DCM than in nondiabetic hearts. We investigated whether intravenous (i.v.) atorvastatin administration during AMI exerts cardioprotection in DCM as seen in nondiabetic hearts. Sprague-Dawley rats were divided into streptozotocin-induced DCM and normoglycemic control groups. Our model of DCM rats exhibited interstitial fibrosis and cardiac dysfunction at 5 weeks. At this time point, all animals underwent AMI induction (coronary ligation for 45 min), receiving i.v. atorvastatin or vehicle during ischemia. Animals were reperfused and sacrificed 24 h later for myocardial infarct size analysis and cardiac tissue sampling. Echocardiography was performed. DCM vehicle rats had larger infarcts than normoglycemic vehicle-treated animals at a comparable area-at-risk. Intravenous atorvastatin reduced infarct size and preserved systolic function in both groups. Compared with vehicle animals, i.v. atorvastatin inhibited RhoA membrane translocation, induced AMPK phosphorylation, prevented apoptosis execution, and improved cardiac remodelling in the infarcted heart of both groups, whereas innate immune cell infiltration was further reduced in i.v. atorvastatin-treated DCM animals. The proven cardioprotective effectiveness of this i.v. statin formulation in the presence of DCM warrants its further development into a clinically therapeutic option. ARTICLE HIGHLIGHTS Diabetic cardiomyopathy (DCM) significantly increases the risk of acute myocardial infarction and attenuates or abolishes the cardioprotective effects of several therapeutic approaches. Whether intravenous atorvastatin administration during ongoing acute myocardial infarction retains its cardioprotective potential in the presence of DCM was investigated. Intravenous atorvastatin during ischemia reduces infarct size and preserves cardiac function in DCM rats. The efficacy of this intravenous statin formulation in DCM supports its development as a viable therapeutic option for clinical use.
Collapse
Affiliation(s)
| | - Sergi López
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | | | | | - Alex Gallinat
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | - Rosa Suades
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | - Lina Badimon
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
- CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
- CiberCV, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Weeks KL, Bernardo BC, Bell JR, Delbridge LMD, Mellor KM. New insights into diabetes-induced cardiac pathology. J Mol Cell Cardiol 2025; 203:76-81. [PMID: 40262687 DOI: 10.1016/j.yjmcc.2025.04.008] [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: 11/15/2024] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Individuals with diabetes have an elevated risk of heart disease, and there is a significant clinical need for evidence-based treatments. Heart disease in diabetes manifests as a distinct cardiopathology, with cardiac structural and functional remodeling underlying increased susceptibility to cardiac dysfunction and arrhythmias. An understanding of the mechanisms associated with cardiac vulnerability in diabetes is incomplete, but recent studies have advanced new insights into the roles of metabolic disturbances, gene dysregulation and epicardial adipose influence. This perspective article highlights these three promising new developments in proposed mechanisms, and discusses exciting advances in cardiac-targeting for potential treatment of diabetic heart disease.
Collapse
Affiliation(s)
- K L Weeks
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia; Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - B C Bernardo
- Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia; Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Victoria, Australia
| | - J R Bell
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Microbiology, Anatomy, Physiology & Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - L M D Delbridge
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - K M Mellor
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Cai Z, Sun F, Wang Q, Li S, Wang L, Li H, Su Y, Yang H, Dong B. Icariin alleviates cardiomyocyte pyroptosis through AMPK-NLRP3 pathway to ameliorates diabetic cardiomyopathy. Int Immunopharmacol 2025; 156:114690. [PMID: 40262250 DOI: 10.1016/j.intimp.2025.114690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Among the multitude of pressing global health concerns, diabetes mellitus stands out as a significant issue. An alarming consequence of this condition is diabetic cardiomyopathy (DCM), which represents a critical contributor to mortality in individuals with diabetes. Recently, research has unveiled the pivotal role that pyroptosis plays in the progression of myocardial fibrosis associated with DCM. An epimedial flavonoid monomer, Icariin (ICA), primarily sourced from Epimedium genus plants, has shown a safeguarding influence on cardiac health through various means, encompassing anti-inflammatory actions and its capacity against oxidative stress. Our research endeavor focuses on elucidating the beneficial impacts alongside the underlying physiological processes triggered by ICA within the context of DCM. An animal model representative of DCM was developed through intraperitoneal administration of streptozotocin (STZ). In parallel, in vitro experiments utilized H9C2 cardiomyocytes to mimic hyperglycemic environments relevant to disease states. In vivo experiments found that ICA improved cardiac function, alleviated myocardial fibrosis, and reduced NLRP3-mediated pyroptosis in heart tissue of DCM mice. Under in vitro settings characterized by elevated glucose concentrations, there was a notable elevation in both NLRP3 pyroptosis-associated proteins and oxidative stress markers within the heart muscle cells. ICA treatment attenuated pyroptosis and oxidative stress caused by high glucose in cardiomyocytes. Further studies revealed that when treated with an AMPK inhibitor, the shielding benefits conferred by ICA on cardiomyocytes were negated, suggesting that the regulatory effects of ICA on cardiomyocyte pyroptosis may be achieved through the AMPK-NLRP3 pathway. In conclusion, ICA exerts protective effects in DCM by inhibiting cardiomyocyte pyroptosis, alleviating myocardial fibrosis, and improving cardiac function via the AMPK-NLRP3 pathway.
Collapse
Affiliation(s)
- Zhenhao Cai
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China; Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxiao Sun
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingyan Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shengnan Li
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lanlan Wang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Huixin Li
- Binhai New Area Hospital of TCM, Tianjin, China
| | - Yudong Su
- Tongnan District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Hongbo Yang
- Weifang Rehabilitation Hospital, Weifang, China
| | - Bo Dong
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
10
|
Lee CJM, Kosyakovsky LB, Khan MS, Wu F, Chen G, Hill JA, Ho JE, Foo RSY, Zannad F. Cardiovascular, Kidney, Liver, and Metabolic Interactions in Heart Failure: Breaking Down Silos. Circ Res 2025; 136:1170-1207. [PMID: 40403106 PMCID: PMC12125648 DOI: 10.1161/circresaha.125.325602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 05/24/2025]
Abstract
Over the past few decades, the rising burden of metabolic disease, including type 2 diabetes, prediabetes, obesity, and metabolic dysfunction-associated steatotic liver disease, has corresponded with fundamental shifts in the landscape of heart failure (HF) epidemiology, including the rising prevalence of HF with preserved ejection fraction. It has become increasingly important to understand the role of extracardiac contributors and interorgan communication in the pathophysiology and phenotypic heterogeneity of HF. Whereas traditional epidemiological strategies have separately examined individual contributions of specific comorbidities to HF risk, these approaches may not capture the shared mechanisms and more complex, bidirectional relationships between cardiac and noncardiac comorbidities. In this review, we highlight the cardiac, kidney, liver, and metabolism multiorgan interactions and pathways that complicate HF development and progression and propose research strategies to further understand HF in the context of multiple organ disease. This includes evolving epidemiological approaches such as multiomics and machine learning which may better capture common underlying mechanisms and interorgan crosstalk. We review existing preclinical models of HF and how they have enhanced our understanding of the role of multiorgan disease in the development of HF subtypes. We suggest recommendations as to how clinical practice across multiple specialties should screen for and manage multiorgan involvement in HF. Finally, recognizing the advent of novel combinatorial therapeutic agents that may have multiple indications across the cardiac-kidney-liver metabolism continuum, we review the current clinical trials landscape. We specifically highlight a pressing need for the design of more inclusive trials that examine the contributions of multimorbidity and incorporate multiorgan end points, which we propose may lead to outcomes that are evermore clinically relevant today.
Collapse
Affiliation(s)
- Chang Jie Mick Lee
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STaR), 61 Biopolis Drive, Singapore 138673, Singapore
| | - Leah B. Kosyakovsky
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Muhammad Shahzeb Khan
- Baylor Scott and White Research Institute, Dallas, TX, USA
- The Heart Hospital, Plano, TX, USA
| | - Feng Wu
- Division of Cardiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Guo Chen
- Division of Cardiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A. Hill
- Division of Cardiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jennifer E. Ho
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Roger S-Y. Foo
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STaR), 61 Biopolis Drive, Singapore 138673, Singapore
| | - Faiez Zannad
- Université de Lorraine, CHRU, Inserm Clinical Investigation Center 1433, Nancy, France
| |
Collapse
|
11
|
Xiao L, Gu Y, Guo S, Liu Y, Cai X, Ji X, Zheng Z, Li Y, Du Y, Wang X, Gao L. STRA13 exacerbates T2DM-induced diabetic cardiomyopathy by regulating the RXRα/UCP-1 signaling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167903. [PMID: 40412731 DOI: 10.1016/j.bbadis.2025.167903] [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: 01/18/2025] [Revised: 05/05/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
STRA13, a basic helix-loop-helix protein superfamily member, is a CLOCK gene. Previous studies have reported the role of STRA13 in regulating blood pressure. However, the role of STRA13 in diabetic cardiomyopathy (DCM) has not been fully elucidated. In this study, STRA13 full knockout mice were subjected to a high-fat diet (HFD) to induce DCM. We found that STRA13 was upregulated in both heart tissue and cardiomyocytes undergoing metabolic disorders. STRA13 knockout ameliorated HFD-induced cardiac dysfunction, fibrosis, mitochondrial dysfunction and cell apoptosis. STRA13 deficiency also protected against HFD-induced glucose and lipid metabolism disorders. STRA13 overexpression in mice worsened HFD-induced cardiac dysfunction, fibrosis, and injury. STRA13 overexpression in cardiomyocytes worsened high glucose-induced cell injury, mitochondrial dysfunction and oxidative stress. STRA13 silencing in cardiomyocytes protected against the high glucose (HG)-induced alterations described above. Moreover, STRA13 was found to downregulate retinoid X receptor alpha (RXRα), resulting in reduced expression of uncoupling protein 1 (UCP-1). Co-IP confirmed that STRA13 interacted with RXRα. A luciferase assay confirmed that RXRα regulated the transcription of UCP-1. Silencing of STRA13 did not protect cardiomyocytes from HG-induced injury caused by RXRα or UCP-1 knockdown. Cardiac overexpression of UCP-1 also blunted the deteriorating effects of STRA13. However, STRA13 inhibited RXRα nuclear expression, which hampered the protective effects of RXRα overexpression in vivo. Taken together, our findings demonstrate that STRA13 exacerbates diabetic cardiomyopathy by impairing mitochondrial function through the disruption of RXRα-UCP-1 signaling. Therefore, targeting the STRA13-RXRα-UCP-1 axis may represent a promising therapeutic strategy for mitigating mitochondrial dysfunction and cardiac injury in the context of DCM.
Collapse
Affiliation(s)
- Lili Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yang Gu
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Sen Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xintong Cai
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaoyang Ji
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhe Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Youyou Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
12
|
Li J, Ma C, Wang X, Li J, Liu P, Zhu M. Development and validation of a novel glucolipid metabolism-related nomogram to enhance the predictive performance for osteoporosis complications in prediabetic and diabetic patients. Lipids Health Dis 2025; 24:183. [PMID: 40399876 PMCID: PMC12093595 DOI: 10.1186/s12944-025-02602-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] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Diabetes is the most prevalent metabolic disorder worldwide, imposing a significant economic burden on society. Prediabetes has not received as much attention as diabetes, and among its complications, osteoporosis has been relatively under-researched compared to cardiovascular disease. Recent studies have identified nine indices related to glucose and lipid metabolism that may enhance osteoporosis risk assessment in diabetic and prediabetic individuals. The research examined the osteoporosis risk prediction potential of these indices and developed a nomogram to enhance predictive performance. METHODS 2,735 prediabetic and diabetic subjects were derived from National Health and Nutrition Examination Survey (NHANES) dataset collected between 2011 and 2020, then randomly assigned to development and validation cohorts in 7:3. The predictive capacity of glucolipid metabolism-related indices for osteoporosis risk was evaluated using receiver operating characteristic (ROC) curve analysis. The least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression were used to identify predictors for constructing the risk model, which was visualized using a nomogram. The model's performance was further validated. RESULTS All the glucolipid metabolism-related indices showed predictive ability, and the best-performing index was metabolic score for insulin resistance (METS-IR). Multivariate logistic regression identified 5 predictors [Triglyceride-glucose index (TyG), age, METS-IR, TyG-waist circumference, and TyG-body mass index] with good predictive performance. These predictors were selected to establish the nomogram. ROC curve, calibration plot, as well as decision curve analysis (DCA) collectively demonstrated fairly good predictive ability of the nomogram. CONCLUSIONS Glucolipid metabolism-related indices are the predictors of osteoporosis risk. This newly developed nomogram based on glucolipid metabolism indices demonstrates optimal predictive accuracy for assessing combined osteoporosis risk in individuals with prediabetes and diabetes.
Collapse
Affiliation(s)
- Junhong Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Cong Ma
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xinran Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianwen Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ping Liu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China.
| | - Meipeng Zhu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
13
|
Wang P, Wang Z, Jin X, Zhang M, Shen M, Li D. Oral Sulforaphane Intervention Protects Against Diabetic Cardiomyopathy in db/db Mice: Focus on Cardiac Lipotoxicity and Substrate Metabolism. Antioxidants (Basel) 2025; 14:603. [PMID: 40427484 PMCID: PMC12109042 DOI: 10.3390/antiox14050603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The protective effect of cruciferae-derived sulforaphane (SFN) on diabetic cardiomyopathy (DCM) has garnered increasing attention. However, no studies have specifically explored its mechanistic involvement in cardiac substrate metabolism and mitochondrial function. To address this gap, Type 2 diabetes mellitus (T2DM) db/db mice were orally gavaged with vehicle or 10 mg/kg body weight SFN every other day for 16 weeks, with vehicle-treated wild-type mice as controls. SFN intervention (SFN-I) alleviated hyperglycemia, dyslipidemia, HOMA-IR, serum MDA levels, and liver inflammation. Furthermore, SFN-I improved the lipotoxicity-related phenotype of T2DM cardiomyopathy, manifested as attenuation of diastolic dysfunction, cardiac injury, fibrosis, lipid accumulation and peroxidation, ROS generation, and decreased mitochondrial complex I and II activities and ATP content, despite having no effect on ceramide abnormalities. Protein expression data revealed that the model mice exhibited upregulated cardiac CD36, H-FABP, FATP4, CPT1B, PPARα, and PDK4 but downregulated GLUT4, with unchanged MPC1 and MPC2. Notably, SFN-I significantly attenuated the increase in CD36, H-FABP, CPT1B, and PPARα. These results suggest that chronic oral SFN-I protects against DCM by mitigating overall metabolic dysregulation and inhibiting cardiolipotoxicity. The latter might involve controlling cardiac fatty acid metabolism and improving mitochondrial function, rather than promoting glucose metabolism.
Collapse
Affiliation(s)
- Pan Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (P.W.); (Z.W.); (X.J.); (M.Z.); (M.S.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Ziling Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (P.W.); (Z.W.); (X.J.); (M.Z.); (M.S.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Xinyuan Jin
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (P.W.); (Z.W.); (X.J.); (M.Z.); (M.S.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Mengdi Zhang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (P.W.); (Z.W.); (X.J.); (M.Z.); (M.S.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Mengfan Shen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (P.W.); (Z.W.); (X.J.); (M.Z.); (M.S.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China; (P.W.); (Z.W.); (X.J.); (M.Z.); (M.S.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| |
Collapse
|
14
|
Wang C, Shen S, Kang J, Sugai-Munson A, Xiao X, Zhang Y, Zhu J, Liu Z, McKay TB, Akeju O, Jonas ER, Ambarar AV, Bristow MR, Yao W, Li H. METTL3 Is Essential for Exercise Benefits in Diabetic Cardiomyopathy. Circulation 2025. [PMID: 40357551 DOI: 10.1161/circulationaha.124.070279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 04/18/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Exercise improves functional outcomes in patients with diabetic cardiomyopathy (DiaCM). The molecular mechanism underlying cardiac benefits of exercise in DiaCM remains incompletely understood. N6-methyladenosine (m6A) is the most common form of messenger RNA modification in eukaryotes and has been implicated in cardiac development and disease. However, the role of m6A in DiaCM and in the mitigating effects of exercise on this disease are unclear. METHODS Cardiomyocyte-specific N6-adenosine-methyltransferase-like 3 (METTL3, an m6A methyltransferase) knockout mice and their wild-type littermates were subjected to either chow diet or high-fat diet feeding and injection of streptozotocin to induce DiaCM, followed by an 8-week exercise training and assessment of cardiac function. Some of the mice were injected with adeno-associated viral vector encoding METTL3 to overexpress METTL3 in cardiomyocytes. Cardiac METTL3 expressions were assessed in patients with nonischemic primary dilated cardiomyopathies without or with diabetes. Potential METTL3 downstream effector YBX1 (Y-box binding protein 1) was identified through RNA sequencing. The functional role of YBX1 was examined through adeno-associated viral vector overexpression or knockdown in cardiomyocytes in DiaCM mice. RESULTS We showed that cardiac METTL3 protein expression and m6A level were downregulated in patient with dilated cardiomyopathy and further downregulated in patients with dilated cardiomyopathy and diabetes. Consistently, cardiac METTL3 and m6A were downregulated in mouse with DiaCM, whereas they were upregulated by exercise. Cardiomyocyte-specific METTL3 knockout eliminated the cardiac benefits of exercise on DiaCM. Conversely, cardiomyocyte-specific METTL3 overexpression improved systolic and diastolic function in 2 DiaCM mouse models. We demonstrated that exercise enhanced cardiac METTL3 expression in DiaCM through signal transducer and activator of transcription 3. Moreover, METTL3 attenuated DiaCM through m6A-depdendent YBX1 upregulation and the subsequent activation of Nrf2. Cardiomyocyte-specific YBX1 overexpression promoted Nrf2 activation and attenuated oxidative stress, resulting in an improvement in cardiac function in DiaCM. In contrast, cardiomyocyte-specific YBX1 gene knockdown abolished the effect of METTL3 on cardiac improvement in mice with DiaCM. Further, pharmacological activation of METTL3 using a small molecule attenuated cardiac dysfunction in DiaCM. CONCLUSIONS These studies reveal an essential role of METTL3 in the cardiac benefits of exercise and identify METTL3 and YBX1 as promising therapeutic targets for treating DiaCM.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| | - Siman Shen
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| | - Jiayi Kang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| | - Aya Sugai-Munson
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| | - Xiao Xiao
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| | - Yajing Zhang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., T.B.M., O.A.)
| | - Junyuan Zhu
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| | - Zipeng Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (Z.L.)
| | - Tina B McKay
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., T.B.M., O.A.)
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., T.B.M., O.A.)
| | - Eric R Jonas
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Amrut V Ambarar
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Michael R Bristow
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Weifeng Yao
- Department of Anesthesiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (W.Y.)
| | - Haobo Li
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston (C.W., S.S., J.K., A.S.-M., X.X., Y.Z., J.Z., T.B.M., O.A., H.L.)
| |
Collapse
|
15
|
Tabatabaei Dakhili SA, Yang K, Stenlund MJ, Ussher JR. The multifaceted roles of ketones in physiology. Exp Physiol 2025. [PMID: 40349316 DOI: 10.1113/ep092243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/16/2025] [Indexed: 05/14/2025]
Abstract
The production of ketones, referred to as ketogenesis, plays an essential role in maintaining energy homeostasis during prolonged fasting/starvation, which primarily stems from its ability to serve as a fuel source to support neuronal ATP production, thereby limiting muscle wasting. Over the years, the field has come to appreciate that ketones are much more than just a fuel source supporting neuronal metabolism, as many other oxidative organs, such as the heart and skeletal muscle, are capable of metabolizing ketones. Furthermore, ketones appear to be an important fuel source for exercising muscle. Beyond supporting ATP production, it is also becoming widely recognized that ketones are powerful signalling molecules, as they serve as ligands for G-protein coupled receptors and can even modify gene expression via regulating DNA post-translational modifications. As they play a key role in supporting whole-body physiology, it is not surprising that perturbations in ketone metabolism can contribute to various pathologies, particularly in relation to cardiometabolic diseases. Some of the strongest evidence supporting the aforementioned statement is seen for both heart failure and type 2 diabetes. Accordingly, we will review herein the multifaceted roles of ketones in supporting whole-body physiology, while interrogating the evidence to suggest whether modifying ketone metabolism may have a therapeutic role in the management of heart failure and type 2 diabetes.
Collapse
Affiliation(s)
- Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kunyan Yang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Magnus J Stenlund
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
16
|
Kang HL, Várkonyi Á, Csonka Á, Szász A, Várkonyi T, Pósa A, Kupai K. Endothelial-Mesenchymal Transition and Possible Role of Cytokines in Streptozotocin-Induced Diabetic Heart. Biomedicines 2025; 13:1148. [PMID: 40426976 PMCID: PMC12109261 DOI: 10.3390/biomedicines13051148] [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: 02/10/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Although endothelial mesenchymal transition (EndMT) has been characterized as a basic process in embryogenesis, EndMT is the mechanism that accelerates the development of cardiovascular diseases, including heart failure, aging, and complications of diabetes or hypertension as well. Endothelial cells lose their distinct markers and take on a mesenchymal phenotype during EndMT, expressing distinct products. Methods: In this study, type 1 Diabetes mellitus (T1DM) was induced in rats with streptozotocin (STZ) by intraperitoneal injection at a 60 mg/kg dose. Diabetic rats were randomly divided into two groups, namely, control and diabetic rats, for 4 weeks. Heart, aorta, and plasma samples were collected at the end of 4 weeks. Sequentially, biochemical parameters, cytokines, reactive oxygen species (ROS), protein expression of EndMT markers (Chemokine C-X-C motif ligand-1 (CXCL-1), vimentin, citrullinated histone H3 (H3Cit), α-smooth muscle actin (α-SMA), and transforming growth factor beta (TGF-β) and versican), components of the extracellular matrix (matrix metalloproteinase 2 (MMP-2), tissue inhibitor of metalloproteinase-1(TIMP-1), and discoidin domain tyrosine kinase receptor 2 (DDR-2)) were detected by ELISA or Western blot, respectively. Results: Cytokines and ROS were increased in diabetic hearts, which induced partial EndMT. Among EndMT markers, histone citrullination, α-SMA, and CXCL-1 were increased; vimentin was decreased in DM. The endothelial marker endothelin-1 was significantly higher in the aortas of DM rats. Interestingly, TGF-β showed a significant decrease in the diabetic heart, plasma, and aorta. Additionally, MMP-2/TIMP-1 levels also decreased in DM. Conclusions: To sum up, the identification of molecules and regulatory pathways involved in EndMT provided novel therapeutic approaches for cardiac pathophysiological conditions.
Collapse
Affiliation(s)
- Hsu Lin Kang
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Ákos Várkonyi
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Ákos Csonka
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary; (Á.C.); (T.V.)
| | - András Szász
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Tamás Várkonyi
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary; (Á.C.); (T.V.)
| | - Anikó Pósa
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Krisztina Kupai
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary; (Á.C.); (T.V.)
| |
Collapse
|
17
|
Xiong Z, Liao Y, Zhang Z, Wan Z, Liang S, Guo J. Molecular Insights into Oxidative-Stress-Mediated Cardiomyopathy and Potential Therapeutic Strategies. Biomolecules 2025; 15:670. [PMID: 40427563 PMCID: PMC12108637 DOI: 10.3390/biom15050670] [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: 03/27/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiomyopathies comprise a heterogeneous group of cardiac disorders characterized by structural and functional abnormalities in the absence of significant coronary artery disease, hypertension, valvular disease, or congenital defects. Major subtypes include hypertrophic, dilated, arrhythmogenic, and stress-induced cardiomyopathies. Oxidative stress (OS), resulting from an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has emerged as a key contributor to the pathogenesis of these conditions. ROS-mediated injury drives inflammation, protease activation, mitochondrial dysfunction, and cardiomyocyte damage, thereby promoting cardiac remodeling and functional decline. Although numerous studies implicate OS in cardiomyopathy progression, the precise molecular mechanisms remain incompletely defined. This review provides an updated synthesis of current findings on OS-related signaling pathways across cardiomyopathy subtypes, emphasizing emerging therapeutic targets within redox-regulatory networks. A deeper understanding of these mechanisms may guide the development of targeted antioxidant strategies to improve clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Zhenyu Xiong
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Yuanpeng Liao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhengdong Wan
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
| | - Sijia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
18
|
Karatzia L, Cullen F, Young M, Lam SH, Morales V, Bianchi K, Henson SM, Aksentijevic D. New Model of Experimental Diabetic Cardiomyopathy Using Combination of Multiple Doses of Anomer-Equilibrated Streptozotocin and High-Fat Diet: Sex Matters. Diabetes 2025; 74:760-772. [PMID: 39836371 PMCID: PMC12015147 DOI: 10.2337/db24-0385] [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: 05/08/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Diabetes leads to a more rapid development of diabetic cardiomyopathy (dbCM) and progression to heart failure in women than in men. Combination of high-fat diet (HFD) and freshly injected streptozotocin (STZ) has been widely used for diabetes induction; however, emerging data show that anomer-equilibrated STZ produces an early-onset and robust diabetes model. We designed a novel protocol using a combination of multiple doses of anomer-equilibrated STZ injections and HFD to develop a stable murine diabetes model featuring dbCM analogous to that in humans. Furthermore, we examined the effect of biological sex on the evolution of cardiometabolic dysfunction in diabetes. Our study included six experimental protocols (8 weeks) in male and female C57BL/6J mice (N = 109): fresh STZ + HFD, anomer-equilibrated STZ + HFD, HFD, fresh STZ, anomer-equilibrated STZ, and control diet + vehicle. Animals were characterized by extensive phenotyping in vivo and ex vivo. Anomer-equilibrated STZ + HFD led to induction of stable experimental murine diabetes characterized by impaired glucose homeostasis, cardiometabolic dysfunction, and altered metabolome of liver, skeletal muscle, kidney, and plasma. dbCM was more severe in female mice, including systolic dysfunction and reduced cardiac energy reserve. This study establishes a novel robust model of inducible murine diabetes and emphasizes the impact of biological sex on diabetes progression and severity. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Loucia Karatzia
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Fenn Cullen
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Megan Young
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Shing Hei Lam
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Valle Morales
- Barts Cancer Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Katiuscia Bianchi
- Barts Cancer Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Sian M. Henson
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Dunja Aksentijevic
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, U.K
| |
Collapse
|
19
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
20
|
Xing Y, Zhao R, Zhang X, Jin J, Yang Y, Wang H, Wang Y, Zhong C, Chen L, Zhang Y. Predictive Value of Left Atrial Structure and Function Parameters in Left Ventricular Remodeling in Patients With Type 2 Diabetes Mellitus. Echocardiography 2025; 42:e70196. [PMID: 40367362 DOI: 10.1111/echo.70196] [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: 05/16/2025] Open
Abstract
BACKGROUND The 4-dimensional automated left atrial quantification (4D Auto LAQ) technology for the left atrium was recently available. We aimed to evaluate LA function using 4D Auto LAQ in patients with type 2 diabetes mellitus (T2DM) and investigate its value in predicting left ventricular remodeling (LVR). METHODS A total of 106 T2DM patients (56 with left ventricular [LV] remodeling and 50 with normal geometry) and 46 age- and sex-matched controls were enrolled. LA total emptying fraction (LATEF), LA active emptying fraction (LAAEF), LA passive emptying fraction (LAPEF), and strain parameters, including LA reservoir longitudinal/circumferential strain (LASr/LASr-c), LA conduit longitudinal/circumferential strain (LAScd/LAScd-c), and LA contraction longitudinal/circumferential strain (LASct/LASct-c), were assessed with 4D Auto LAQ. RESULTS Compared to controls, LASr, LAScd, and LAPEF significantly decreased in both groups of T2DM patients (p < 0.001). T2DM patients with LVR had significantly lower LASr and LAScd than those with normal geometry (p < 0.001). LATEF was also reduced in T2DM patients with LVR compared to the control group (p < 0.05). Among the 4D-LAQ parameters, only LASr (odds ratio [OR]: 0.860, p = 0.016) was associated with LV remodeling (LVR) in multivariate analysis. Receiver operating characteristic (ROC) curves identified a LASr value of ≤22.5% as the optimal cutoff point to predict LVR in the T2DM cohort (sensitivity, 86.0%; specificity, 64.3%; area under the curve [AUC], 0.770; p < 0.001). In addition, LASr was found to be negatively correlated with both LV mass index (LVMI) and relative wall thickness (RWT) but positively correlated with the absolute value of the LV global longitudinal strain (LVGLS). CONCLUSIONS Impairment of LA reservoir and conduit functions can be observed in patients with T2DM, particularly in those with LVR. LASr may serve as a predictor of LVR in patients with T2DM.
Collapse
Affiliation(s)
- Yumeng Xing
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| | - Rui Zhao
- Department of Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, Illinois, USA
| | - Xiaoli Zhang
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Jiamei Jin
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| | - Yiming Yang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Haier Wang
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhong
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| | - Lin Chen
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| | - Yinjia Zhang
- Department of Ultrasound, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Li C, Cai D, Yuan W, Cai R, Qiu X, Qin Y, Feng Y, Zhu Q, Liu Y, Chen Y, Yuan X, Jiang W, Hou N. The canonical Wnt/β-catenin signaling pathway upregulates carbonic anhydrase 2 via transcription factor 7-like 2 to promote cardiomyopathy in type 2 diabetic mice. Life Sci 2025; 368:123506. [PMID: 40010634 DOI: 10.1016/j.lfs.2025.123506] [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: 10/15/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
Excessive activation of the canonical Wnt/β-catenin pathway contributes to the development of diabetic cardiomyopathy (DCM). Transcription factor 7-like 2 (TCF7L2) is the main β-catenin partner of the TCF family in adult human hearts. Carbonic anhydrase 2 (CA2) is implicated in various hypertrophic cardiomyopathy. In this study, we aimed to investigate the role of the Wnt/β-catenin/TCF7L2 signaling and CA2 in the development of DCM. Streptozotocin (STZ)/high-fat diet (HFD)-induced diabetic mice and high glucose-stimulated neonatal rat cardiomyocytes (NRCMs) were used as in-vivo and in-vitro models of Type 2 diabetes (T2DM), respectively. Histopathological changes in the mouse myocardium were assessed with hematoxylin-eosin (HE) or Masson's trichrome staining. Cardiac function was evaluated with echocardiography. TCF7L2, β-catenin, and CA2 expression was determined with RT-qPCR, western blotting, and immunohistochemistry. Immunoprecipitation (IP) was used to evaluate the formation of the β-catenin/TCF7L2 bipartite. The regulatory relationship between the β-catenin/TCF7L2 bipartite and CA2 was investigated with chromatin immunoprecipitation (ChIP) and a luciferase reporter assay. Compared with the control mice, the T2DM mice exhibited increased myocardial β-catenin and TCF7L2 expression that was concentrated in the nucleus. Treatment of diabetic mice with the β-catenin/TCF7L2 bipartite inhibitor iCRT14 prevented myocardial remodeling and improved cardiac dysfunction. iCRT14 also prevented high glucose-induced hypertrophy in NRCMs, while the β-catenin stabilizer SKL2001 worsened hypertrophy. IP experiments confirmed the formation of the β-catenin/TCF7L2 bipartite in the control and T2DM mouse cardiomyocytes. Moreover, based on the results of RNA-sequencing analysis, CA2 was upregulated in T2DM cardiomyocytes in vitro and in vivo. TCF7L2 overexpression upregulated CA2, while iCRT14 treatment or TCF7L2 knockdown downregulated CA2. CA2 knockdown ameliorated NRCM hypertrophy induced by high glucose and SKL2001. The ChIP experiments revealed an increased interaction between β-catenin/TCF7L2 and the transcription initiation region of CA2 in the heart tissue of T2DM mice. The luciferase reporter assay confirmed that CA2 is directly regulated by the β-catenin/TCF7L2 bipartite. The results indicate that the canonical Wnt/β-catenin pathway upregulates CA2 via TCF7L2 to promote DCM. This research sheds new light on the pathogenesis of DCM and presents new potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Conglin Li
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China; NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China; Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, PR China
| | - Daofeng Cai
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China; NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wenchang Yuan
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Rui Cai
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xiaoxia Qiu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yuan Qin
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yaofeng Feng
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China; KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qiulian Zhu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yun Liu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yilin Chen
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xun Yuan
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Wenyue Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China.
| | - Ning Hou
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China; NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
22
|
Panwar A, Malik SO, Adib M, Lopaschuk GD. Cardiac energy metabolism in diabetes: emerging therapeutic targets and clinical implications. Am J Physiol Heart Circ Physiol 2025; 328:H1089-H1112. [PMID: 40192025 DOI: 10.1152/ajpheart.00615.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/15/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Patients with diabetes are at an increased risk for developing diabetic cardiomyopathy and other cardiovascular complications. Alterations in cardiac energy metabolism in patients with diabetes, including an increase in mitochondrial fatty acid oxidation and a decrease in glucose oxidation, are important contributing factors to this increase in cardiovascular disease. A switch from glucose oxidation to fatty acid oxidation not only decreases cardiac efficiency due to increased oxygen consumption but it can also increase reactive oxygen species production, increase lipotoxicity, and redirect glucose into other metabolic pathways that, combined, can lead to heart dysfunction. Currently, there is a lack of therapeutics available to treat diabetes-induced heart failure that specifically target cardiac energy metabolism. However, it is becoming apparent that part of the benefit of existing agents such as GLP-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors may be related to their effects on cardiac energy metabolism. In addition, direct approaches aimed at inhibiting cardiac fatty acid oxidation or increasing glucose oxidation hold future promise as potential therapeutic approaches to treat diabetes-induced cardiovascular disease.
Collapse
Affiliation(s)
- Archee Panwar
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sufyan O Malik
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Muhtasim Adib
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
23
|
Roberts TD, Hutchinson DS, Wootten D, De Blasio MJ, Ritchie RH. Advances in incretin therapies for targeting cardiovascular disease in diabetes. J Mol Cell Cardiol 2025; 202:102-115. [PMID: 40086589 DOI: 10.1016/j.yjmcc.2025.03.007] [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: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
The global prevalence of obesity is skyrocketing at an alarming rate, with recent data estimating that one-in-eight people are now living with the disease. Obesity is a chronic metabolic disorder that shares underlying pathophysiology with other metabolically-linked diseases such as type 2 diabetes mellitus, cardiovascular disease and diabetic cardiomyopathy. There is a distinct correlation between type 2 diabetes status and the likelihood of heart failure. Of note, there is an apparent sexual dimorphism, with women disproportionately affected with respect to the degree of severity of the cardiac phenotype of diabetic cardiomyopathy that results from diabetes. The current pharmacotherapies available for the attenuation of hyperglycaemia in type 2 diabetes are not always effective, and have varying degrees of efficacy in the setting of heart failure. Insulin can worsen heart failure prognosis whereas metformin, sodium-glucose cotransporter 2 inhibitors (SGLT2i) and more recently, glucagon-like peptide-1 receptor agonists (GLP-1RAs), have demonstrated cardioprotection with their administration. This review will highlight the advancement of incretin therapies for individuals with diabetes and heart failure and explore newly-reported evidence of the clinical usefulness of GLP-1R agonists in this distinct phenotype of heart failure.
Collapse
Affiliation(s)
- Timothy D Roberts
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Dana S Hutchinson
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Denise Wootten
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| |
Collapse
|
24
|
Xu Q, Zhang H, Ruan N, Jing J, Li Y, Zhao J, Fang Z. HMOX1 as a Novel Biomarker for Glucose-Lipid Metabolism Disorder and T2DM: Systematic Bioinformatics Investigation and Experimental Verification. ACS OMEGA 2025; 10:16123-16137. [PMID: 40321502 PMCID: PMC12044504 DOI: 10.1021/acsomega.4c09662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/11/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025]
Abstract
Type 2 diabetes mellitus (T2DM) has led to a considerable increase in morbidity and mortality worldwide. Current treatments control blood glucose but cannot reverse the disease, making it important to identify biomarkers that predict T2DM onset and progression. This study explores heme oxygenase 1(HMOX1) as a novel biomarker for T2DM through bioinformatics and experimental validation. Core differentially expressed genes (DEGs) were identified using the Gene Expression Omnibus database, with Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Enrichment Analysis analyses revealing notable pathways, including Toll-like receptor signaling and cytokine receptor interactions. A Nomogram model and receiver operating characteristic curves demonstrated strong diagnostic effectiveness for these core DEGs. The CIBERSORT algorithm assessed the relation between core DEGs and immune cell infiltration, showing substantial associations with several immune cell types, particularly highlighting HMOX1's correlation with eight immune cells (p < 0.05). In a mouse model, db/db mice displayed typical diabetic characteristics and lower serum HMOX1 levels compared to db/m controls (p < 0.01). Histological analysis confirmed liver damage and decreased expression of NFE2L2 and HMOX1 in diabetic mice tissues (p < 0.05). HMOX1 is identified as a promising biomarker for T2DM, with its downregulation confirmed through bioinformatics and experimental methods.
Collapse
Affiliation(s)
- Qi Xu
- The
First Affiliated Hospital, Anhui University
of Chinese Medicine, Hefei, Anhui 230038, China
- School
of Chinese Medicine, Anhui University of
Chinese Medicine, Hefei, Anhui 230012, China
| | - Hongrong Zhang
- School
of Medical Informatics Engineering, Anhui
University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Nuobing Ruan
- The
First Affiliated Hospital, Anhui University
of Chinese Medicine, Hefei, Anhui 230038, China
| | - Jiawen Jing
- The
First Affiliated Hospital, Anhui University
of Chinese Medicine, Hefei, Anhui 230038, China
| | - Yufan Li
- The
First Affiliated Hospital, Anhui University
of Chinese Medicine, Hefei, Anhui 230038, China
| | - Jindong Zhao
- The
First Affiliated Hospital, Anhui University
of Chinese Medicine, Hefei, Anhui 230038, China
- School
of Chinese Medicine, Anhui University of
Chinese Medicine, Hefei, Anhui 230012, China
| | - Zhaohui Fang
- The
First Affiliated Hospital, Anhui University
of Chinese Medicine, Hefei, Anhui 230038, China
| |
Collapse
|
25
|
Zhang X, Chen Z, Cao J, Zhou P, Zhang Z, Sun X, Liu Y, Li T, Cheng HJ, Cheng CP. Enhanced negative modulation of urotensin II on cardiac function and [Ca 2+] i regulation in a diabetic rat model: Insights into molecular and cellular mechanisms. J Pharmacol Exp Ther 2025; 392:103594. [PMID: 40403578 DOI: 10.1016/j.jpet.2025.103594] [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: 01/06/2025] [Revised: 03/29/2025] [Accepted: 04/21/2025] [Indexed: 05/24/2025] Open
Abstract
The direct cardiac effects of urotensin II (UII) in normal and diabetic subjects remain controversial. The alteration and functional significance of cardiac UII/UII receptor (UT) in diabetes are still unclear. We assessed the hypothesis that in diabetes, the cardiomyocyte UII/UT system is increased. This augmentation is proposed to exacerbate the dysfunctional [Ca2+]i regulation, enhance inhibitions of left ventricle (LV) and myocyte contraction and relaxation, leading to worsening cardiac dysfunction. We compared LV myocyte UII and UT expression, LV and myocyte contractile, [Ca2+]i transient ([Ca2+]iT) and calcium current (ICa,L) responses to UII stimulation in male Sprague-Dawley rats (12/group) with streptozotocin-induced diabetes mellitus and controls. We found that UII and UT protein levels were significantly greater in diabetic myocytes than in control myocytes. Compared with control rats, UII (400 pmol/kg, i.p.) administration produced greater decreases in LV contractility of EES (diabetes mellitus: 32% vs C: 13%) and MSW with significantly increased LV time constant relaxation in diabetes. In response to UII (10-5 M) superfusion, diabetic myocytes had much greater decreases in the velocity of shortening and relengthening accompanied by significantly larger decreases in the peak systolic [Ca2+]iT and ICa,L (29% vs 15%). These responses were abolished by pretreatment of diabetic myocytes with urantide, pertussis toxin, or dibutyryl-cAMP, respectively. We conclude that UII has direct negative inotropic and lusitropic cardiac effects in both normal and diabetic rats. In diabetes, cardiac UII/UT is upregulated, enhancing UII-caused negative modulation on cardiac function and [Ca2+]i regulation. This may contribute to the progression of cardiac dysfunction in diabetes and diabetic cardiomyopathy. SIGNIFICANCE STATEMENT: Urotensin II (UII) has direct negative inotropic and lusitropic cardiac effects in both normal and diabetic rats. Compared with normal rats, cardiac UII/UII receptors (UT) were upregulated in diabetic rats, resulting in significantly greater decreases in [Ca2+]iT and ICa,L and increased inhibitions of left ventricle and myocyte contraction and relaxation. These effects are coupled with UT and mediated by Gi proteins. These data provide new insights and evidence that upregulation of cardiomyocyte UII/UT may promote the progressive cardiac dysfunction in diabetes and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Zhe Chen
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jing Cao
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Peng Zhou
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Internal Medicine, Ming De Hospital, Beijing, China
| | - Zhi Zhang
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (originally named "Shanghai First People's Hospital"), Shanghai, China
| | - Xiaoqiang Sun
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Yixi Liu
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tiankai Li
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Heng-Jie Cheng
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Che Ping Cheng
- Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
26
|
Yu H, Yang C, Lv J, Zhao Y, Wang G, Wang X. The association between monocyte-to-high-density lipoprotein cholesterol ratio and type 2 diabetes mellitus: a cross-sectional study. Front Med (Lausanne) 2025; 12:1521342. [PMID: 40357304 PMCID: PMC12066528 DOI: 10.3389/fmed.2025.1521342] [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: 11/01/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Objective Type 2 diabetes mellitus (T2DM) is a prevalent chronic condition often associated with low-grade inflammation. Previous studies have indicated that the monocyte-to-high-density lipoprotein cholesterol ratio (MHR) may serve as a novel inflammatory biomarker with potential predictive value for various metabolic diseases. This study aims to investigate the association between the MHR and the prevalence of T2DM in a general population, using data from the National Health and Nutrition Examination Survey (NHANES). Methods We conducted a cross-sectional study analyzing data from five NHANES cycles spanning 2007-2016. We excluded individuals aged under 20 years, those with missing data on monocytes, HDL-C, diabetes status, or other key covariates, and extreme MHR outliers. Statistical analyses were performed using SPSS 26.0, EmpowerStats 4.1, Stata 16, and DecisionLinnc1.0. We employed weighted logistic regression models, subgroup analyses, restricted cubic splines (RCS), and threshold analyses were used to assess the MHR-T2DM association. Results A total of 10,066 participants met the inclusion criteria, of whom 1,792 were diagnosed with T2DM. The MHR levels in the T2DM group were significantly higher than those in the non-T2DM group. After adjusting for potential confounders, elevated MHR levels were significantly associated with an increased prevalence of T2DM (p < 0.001, OR = 2.80, 95% CI: 1.823-4.287). Subgroup analyses revealed a significant interaction between MHR and T2DM with respect to gender (P for interaction < 0.05), with a stronger association in women. No significant interactions were observed for age, race, education level, poverty income ratio (PIR), body mass index (BMI), smoking status, physical activity, alcohol consumption, or hypertension (P for interaction > 0.05). RCS analysis indicated a significant nonlinear relationship between MHR and T2DM, with a threshold point for MHR identified at 0.51. Above this threshold, the risk of T2DM increased significantly. Conclusion Our findings suggest that elevated MHR levels, particularly above the threshold of 0.51, are significantly associated with an increased prevalence of T2DM. The gender-specific interaction further highlights that women may be more susceptible to the impact of elevated MHR on T2DM risk. These findings suggest MHR as a potential biomarker for early T2DM screening and highlight gender-specific risk factors.
Collapse
Affiliation(s)
- Honghai Yu
- Department of Traditional Chinese Medicine, Hunan University of Medicine General Hospital, Huaihua, China
| | - Cunqing Yang
- Department of Dermatology, Guang'Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Postdoctoral Research Station, Guang'Anmen Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Jiao Lv
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yunyun Zhao
- Department of Endocrinology, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Guoqiang Wang
- Department of Endocrinology, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xiuge Wang
- Department of Endocrinology, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
27
|
Kwast S, Lässing J, Falz R, Hoffmann J, Pökel C, Schulze A, Schröter T, Borger M, Busse M. Altered cardiac contractility and aerobic muscular capacity markers during exercise in patients with obesity and DMT II. BMC Sports Sci Med Rehabil 2025; 17:100. [PMID: 40296039 PMCID: PMC12036242 DOI: 10.1186/s13102-025-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Impaired exercise capacity influences obesity and diabetes disease progression and vice versa. The primary objective of this prospective, observational, real-world study was to characterize exercise capacity in patients with obesity or type II diabetes mellitus and healthy controls by cardiac capacity (cardiac output (CO), cardiac power output (CPO)) and peripheral muscle capacity (peak power output (Pmax) and arterio-venous oxygen difference (avDO2)). The effects of an exercise and lifestyle intervention on these cardiac and peripheral muscular markers in obese and diabetic patient groups were additionally evaluated. METHODS At a university sports medicine outpatient clinic, 24 obese (OB) and 38 diabetes mellitus type II (DM) patients and 20 healthy controls (HE) were investigated in a cross-sectional analysis. OB and DM were reexamined after a standard of care exercise intervention. Parameters were assessed at rest and during a cardiopulmonary exercise test (CPET). Blood pressure, impedance cardiography, and respiratory gas analysis were continuously recorded during CPET. RESULTS At Pmax, CO and CPO were lower in DM compared to obese (CO 16.26 l/min vs. 18.13 l/min, p < 0.04; CPO 5.67 W vs. 4.81 W, p < 0.01). HE did not differ in CO (18.19 l/min)) or CPO (5.27 W) from OB and DM. Maximum CPO in OB and DM was based on higher stroke volume and blood pressure, while HE had higher heart rates. Pmax was higher (p < 0.01) in HE (268 W) compared to OB (108 W) and DM (89 W), mainly caused by a higher (p < 0.01) avDO2 (HE 18.22 ml/dl, OB 10.45 ml/dl, DM 9.65 ml/dl). Exercise intervention improved Pmax in both groups of patients (+ 16 W in OB, + 12 W in DM), which was attributed to increased avDO2, but not to cardiac parameters. CONCLUSIONS Obese patients had higher cardiac power outputs and were primarily limited by muscular performance, while diabetic patients showed both muscular and cardiac limitations. Healthy subjects had comparable cardiac power outputs with significantly lower pressure-volume loads. Resistance training improved the alteration of our patient groups in exercise capacity. Future research is needed to interpret our findings regarding clinical endpoints, such as mortality and hospitalization. TRIAL REGISTRATION The study was retrograde registered in the German Clinical Trial Register (DRKS00032545, 24.08.2023).
Collapse
Affiliation(s)
- Stefan Kwast
- Institute of Sports Medicine and Prevention, University Leipzig, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany.
| | - Johannes Lässing
- Department of Exercise Science and Sports Medicine, University Halle-Wittenberg, Von-Seckendorff-Platz 2, 06120, Halle (Saale), Germany
| | - Roberto Falz
- Institute of Sports Medicine and Prevention, University Leipzig, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany
| | - Jana Hoffmann
- University Leipzig, Sports Medicine Outpatient Clinic, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany
| | - Christoph Pökel
- Institute of Sports Medicine and Prevention, University Leipzig, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany
| | - Antina Schulze
- Institute of Sports Medicine and Prevention, University Leipzig, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany
- University Leipzig, Sports Medicine Outpatient Clinic, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany
| | - Thomas Schröter
- Leipzig Heart Center, Department of Cardiac Surgery, University Leipzig, Strümpelstr. 39, 04289, Leipzig, Germany
| | - Michael Borger
- Leipzig Heart Center, Department of Cardiac Surgery, University Leipzig, Strümpelstr. 39, 04289, Leipzig, Germany
| | - Martin Busse
- University Leipzig, Sports Medicine Outpatient Clinic, Rosa-Luxemburg-Str. 30, 04103, Leipzig, Germany
| |
Collapse
|
28
|
Sayer M, Hamano H, Nagasaka M, Lee BJ, Doh J, Patel PM, Zamami Y, Ozaki AF. Time dependent predictors of cardiac inflammatory adverse events in cancer patients receiving immune checkpoint inhibitors. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:40. [PMID: 40296103 PMCID: PMC12036232 DOI: 10.1186/s40959-025-00331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Cardio-inflammatory immune related adverse events (irAEs) while receiving immune checkpoint inhibitor (ICI) therapy are particularly consequential due to their associations with poorer treatment outcomes. Evaluation of predictive factors of these serious irAEs with a time dependent approach allows better understanding of patients most at risk. OBJECTIVE To identify different elements of patient data that are significant predictors of early and late-onset or delayed cardio-inflammatory irAEs through various predictive modeling strategies. METHODS A cohort of patients receiving ICI therapy from January 1, 2010 to May 1, 2022 was identified from TriNetX meeting inclusion/exclusion criteria. Patient data collected included occurrence of early and later cardio-inflammatory irAEs, patient survival time, patient demographic information, ICI therapies, comorbidities, and medication histories. Predictive and statistical modeling approaches identified unique risk factors for early and later developing cardio-inflammatory irAEs. RESULTS A cohort of 66,068 patients on ICI therapy were identified in the TriNetX platform; 193 (0.30%) experienced early cardio-inflammatory irAEs and 175 (0.26%) experienced later cardio-inflammatory irAEs. Significant predictors for early irAEs included: anti-PD-1 therapy at index, combination ICI therapy at index, and history of peripheral vascular disease. Significant predictors for later irAEs included: a history of myocarditis and/or pericarditis, cerebrovascular disease, and history of non-steroidal anti-inflammatory medication use. CONCLUSIONS Cardio-inflammatory irAEs can be divided into clinically meaningful categories of early and late based on time since initiation of ICI therapy. Considering distinct risk factors for early-onset and late-onset events may allow for more effective patient monitoring and risk assessment.
Collapse
Affiliation(s)
- Michael Sayer
- School of Pharmacy & Pharmaceutical Sciences, University of California, 802 W Peltason Dr, Room 106A, Irvine, CA, 92617, USA
| | - Hirofumi Hamano
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Misako Nagasaka
- Division of Hematology and Oncology, University of California, Irvine, CA, USA
| | - Benjamin J Lee
- Department of Pharmacy, University of California Irvine Health, Orange, CA, USA
| | - Jean Doh
- Department of Pharmacy, University of California Irvine Health, Orange, CA, USA
| | - Pranav M Patel
- Division of Cardiology, Department of Medicine, University of California, Irvine, CA, USA
| | - Yoshito Zamami
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Aya F Ozaki
- School of Pharmacy & Pharmaceutical Sciences, University of California, 802 W Peltason Dr, Room 106A, Irvine, CA, 92617, USA.
| |
Collapse
|
29
|
Wang X, Tan X, Zhang T, Xu S, Zeng Y, Xu A, Li X, Zhang G, Jiang Y, Jiang H, Fan J, Bo X, Fan H, Zhou Y. Modeling diabetic cardiomyopathy using human cardiac organoids: Effects of high glucose and lipid conditions. Chem Biol Interact 2025; 411:111421. [PMID: 39984109 DOI: 10.1016/j.cbi.2025.111421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a complex metabolic disorder resulting from chronic hyperglycemia and lipid toxicity, which leads to cardiac dysfunction, fibrosis, inflammation, and mitochondrial impairment. Traditional two-dimensional (2D) cell cultures and animal models have limitations in replicating human cardiac physiology and pathophysiology. In this study, we successfully developed a three-dimensional (3D) model of DCM using cardiac organoids generated from human induced pluripotent stem cells (hiPSCs). These organoids were treated with varying concentrations of glucose and sodium palmitate to mimic the high-glucose and high-lipid environment associated with diabetes. At lower concentrations, glucose and sodium palmitate enhanced cell viability, while higher concentrations induced significant cardiotoxic effects, including apoptosis, oxidative stress, and mitochondrial dysfunction. The cardiac organoids also exhibited increased expression of cardiac injury markers, fibrosis-related genes, and inflammatory cytokines under high-glucose and high-lipid conditions. Treatment with metformin, a widely used antidiabetic drug, mitigated these adverse effects, indicating the model's potential for drug testing and evaluation. Our findings demonstrate that this human-derived 3D cardiac organoid model provides a more physiologically relevant platform for studying DCM and can effectively complement traditional models. This model holds promise for advancing the understanding of diabetic heart disease and for assessing the efficacy of potential therapeutic interventions.
Collapse
Affiliation(s)
- Xiangyu Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Xin Tan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Ting Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China; Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Ahhui Medical University, Hefei, 230011, China
| | - Shuai Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Yiyao Zeng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Anchen Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Xian Li
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Hezi Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Jili Fan
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Xiaohong Bo
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Huimin Fan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou Dushu Lake Hospital, Suzhou, 215028, China.
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Institute for Hypertension, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
30
|
Liu B, Jin Q, Sun YK, Yang ZM, Meng P, Zhang X, Chen Q, Gan P, Zhao T, He JJ, He GP, Xue Q. From bench to bedside: targeting ferroptosis and mitochondrial damage in the treatment of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2025; 16:1563362. [PMID: 40352456 PMCID: PMC12061709 DOI: 10.3389/fendo.2025.1563362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common and fatal cardiac complication caused by diabetes, with its pathogenesis involving various forms of cell death and mitochondrial dysfunction, particularly ferroptosis and mitochondrial injury. Recent studies have indicated that ferroptosis and mitochondrial damage play crucial roles in the onset and progression of DCM, though their precise regulatory mechanisms remain unclear. Of particular interest is the interaction between ferroptosis and mitochondrial damage, as well as their synergistic effects, which are not fully understood. This review summarizes the roles of ferroptosis and mitochondrial injury in the progression of DCM and explores the molecular mechanisms involved, with an emphasis on the interplay between these two processes. Additionally, the article offers an overview of targeted drugs shown to be effective in cellular experiments, animal models, and clinical trials, analyzing their mechanisms of action and potential side effects. The goal is to provide insights for future drug development and clinical applications. Moreover, the review explores the challenges and prospects of multi-target combination therapies and personalized medicine interventions in clinical practice to offer strategic guidance for the comprehensive prevention and management of DCM.
Collapse
Affiliation(s)
- Bin Liu
- Department of Cardiology, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu People’s Hospital, Gejiu, Yunnan, China
| | - Qing Jin
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Yi Kang Sun
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Zhi Ming Yang
- Department of Cardiology, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu People’s Hospital, Gejiu, Yunnan, China
| | - Ping Meng
- Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Xi Zhang
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Qiu Chen
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
- Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Pin Gan
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Tao Zhao
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Jia Ji He
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Gui Ping He
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Qiang Xue
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| |
Collapse
|
31
|
Li M, Sun X, Zeng L, Sun A, Ge J. Metabolic Homeostasis of Immune Cells Modulates Cardiovascular Diseases. RESEARCH (WASHINGTON, D.C.) 2025; 8:0679. [PMID: 40270694 PMCID: PMC12015101 DOI: 10.34133/research.0679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025]
Abstract
Recent investigations into the mechanisms underlying inflammation have highlighted the pivotal role of immune cells in regulating cardiac pathophysiology. Notably, these immune cells modulate cardiac processes through alternations in intracellular metabolism, including glycolysis and oxidative phosphorylation, whereas the extracellular metabolic environment is changed during cardiovascular disease, influencing function of immune cells. This dynamic interaction between immune cells and their metabolic environment has given rise to the novel concept of "immune metabolism". Consequently, both the extracellular and intracellular metabolic environment modulate the equilibrium between anti- and pro-inflammatory responses. This regulatory mechanism subsequently influences the processes of myocardial ischemia, cardiac fibrosis, and cardiac remodeling, ultimately leading to a series of cardiovascular events. This review examines how local microenvironmental and systemic environmental changes induce metabolic reprogramming in immune cells and explores the subsequent effects of aberrant activation or polarization of immune cells in the progression of cardiovascular disease. Finally, we discuss potential therapeutic strategies targeting metabolism to counteract abnormal immune activation.
Collapse
Affiliation(s)
- Mohan Li
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Linqi Zeng
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| |
Collapse
|
32
|
Alemu F, Beza L, Jiru T, Endale D. Assessment of Knowledge on Cardiovascular Disease Risk Factors Among Diabetes Mellitus Patients in Tikur Anbessa Specialized Hospital, Ethiopia 2021: A Cross-Sectional Study. Nurs Res Pract 2025; 2025:7737392. [PMID: 40290112 PMCID: PMC12033066 DOI: 10.1155/nrp/7737392] [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: 02/22/2023] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction: Cardiovascular diseases (CVDs) are a growing problem with increasing global prevalence and the most common cause of mortality worldwide. Knowledge about the disease and risk factors reduces exposure to modifiable risk factors and, as a result, contributes to prevention. As diabetes is a prevalent disease and there is limited research about CVD risk factors in Ethiopia, we conducted a study to assess this knowledge. Methods: A cross-sectional study was conducted on diabetes mellitus patients on follow-up at Tikur Anbessa Specialized Hospital from April 11 to May 16, 2021. The participants were selected using a consecutive sampling method. Knowledge was measured using a heart disease fact questionnaire, and a score of less than 70% was defined as suboptimal. Data were analyzed using SPSS Version 26.0. Associations between dependent and independent variables were identified based on AOR, with 95% CI and a p value less than or equal to 0.05. Result: A total of 404 patients with a mean age of 52.03 ± 14.39 participated in the study, and more than half, 217 (53.7%), of patients were females. About half of the patients (52%) had good knowledge of CVD risk factors. In multivariable logistic regression, urban residency (AOR, 3.335; 95% CI [1.134-9.809]), higher educational level (AOR, 4.016; 95% CI [1.78-9.061]), being employed (AOR, 1.942; 95% CI [1.058-3.566]), and hearing information about CVD risk factors (AOR, 2.492; 95% CI [1.573-3.949]) are associated with knowledge of CVD risk factors. Conclusion: This study revealed that almost half of diabetes mellitus patients had suboptimal knowledge about CVD risk factors. Urban residence, higher education level, employment, and information about CVD risk factors are positively associated with good knowledge of CVD risk factors. Health education is needed to improve their knowledge.
Collapse
Affiliation(s)
- Filipos Alemu
- Department of Nursing, College of Health Science, Dilla University, Dilla, Ethiopia
| | - Lemlem Beza
- Department of Emergency Medicine, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tilahun Jiru
- Department of Emergency Medicine, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Dereje Endale
- Department of Nursing, College of Health Science, Dilla University, Dilla, Ethiopia
| |
Collapse
|
33
|
Bai X, Zhang Z, Zhang M, Xu J, Dong K, Du Q, Chen L, Ma P, Yang J. α-Mangostin prevents diabetic cardiomyopathy by inhibiting oxidative damage and lipotoxicity through the AKT-FOXO1-CD36 pathway. Front Pharmacol 2025; 16:1566311. [PMID: 40313619 PMCID: PMC12043880 DOI: 10.3389/fphar.2025.1566311] [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: 01/24/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction Diabetic cardiomyopathy (DCM), a cardiac complication of diabetes, is the main cause of the high prevalence of heart failure and associated mortality in diabetic patients. Oxidative stress and lipid metabolism disorder-induced myocardial cell damage are part of the pathogenesis of DCM. In this study, we investigated the effects of alpha-mangostin (A-MG), a natural antioxidant extracted from mangosteen peel, on in vitro and in vivo DCM models. Methods H9C2 rat cardiomyocytes were treated with high glucose (HG) and palmitic acid (PA) for 24 h to establish an in vitro DCM cell model. Cell viability and cytotoxicity were evaluated after treatment with varying concentrations of A-MG (0.3, 1, 3, 9, or 27 μM) using Cell Counting Kit-8 (CCK8) and lactate dehydrogenase (LDH) assays. Flow cytometry assessment was used to detect apoptosis. Molecular mechanisms were investigated through transcriptome analysis, quantitative PCR (RT-qPCR), and Western blotting. Type 2 diabetic (T2D) mice, induced by feeding a high-fat diet (HFD) combined with low-dose streptozotocin (STZ), received either vehicle, low-dose A-MG (100 mg/kg/d), or high-dose A-MG (200 mg/kg/d) for 6 weeks. Cardiac function was assessed by echocardiography. H&E and Masson's staining were used to evaluate cardiac tissue structure and fibrosis, and Western blotting was used to evaluate myocardial protein expression. Results In HG/F-induced H9C2 cells, A-MG (1 and 3 μM) significantly increased cell viability (p < 0.01) and reduced LDH release (p < 0.05). A-MG (3 μM) attenuated lipid accumulation (p < 0.05), normalized mitochondrial membrane potential (p < 0.01), and inhibited reactive oxygen species (ROS) generation (p < 0.05), malondialdehyde (MDA) production (p < 0.01), and apoptosis (p < 0.05). A-MG also inhibited the nuclear translocation of Forkhead box class O1 (FOXO1) (p < 0.05); reduced the expression of CD36 (p < 0.05), PPARα (p < 0.01), and CPT1β (p < 0.05) proteins; enhanced superoxide dismutase (SOD) activity (p < 0.05); and upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) (p < 0.01), HO-1 (p < 0.05), and SOD2 (p < 0.05) protein expression levels. Further investigation in HG/F-induced H9C2 cells indicated that A-MG inhibits the uptake of fatty acids (FAs) by regulating the AKT/FOXO1/CD36 signaling pathway, reduces excessive β-oxidation of FAs mediated by PPARα/CPT1β through the inhibition of FOXO1 nuclear translocation, and stimulates the AKT/Nrf2/HO-1 signaling pathway to increase the cellular antioxidant capacity. In diabetic mice, low-dose A-MG treatment increased anti-oxidative stress capacity, decreased myocardial lipid accumulation, reduced fibrosis and cardiomyocyte apoptosis, and improved left ventricular contractile function. Conclusion Using both in vitro and in vivo DCM models, our study demonstrates that A-MG reduces lipid accumulation and excessive mitochondrial β-oxidation while enhancing antioxidant capacity. These results suggest that A-MG may be a novel therapeutic option for DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jianhong Yang
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
34
|
Wu J, Hu H, Li X. Spinal neuron-glial crosstalk and ion channel dysregulation in diabetic neuropathic pain. Front Immunol 2025; 16:1480534. [PMID: 40264787 PMCID: PMC12011621 DOI: 10.3389/fimmu.2025.1480534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Diabetic neuropathic pain (DNP) is one of the most prevalent complications of diabetes, characterized by a high global prevalence and a substantial affected population with limited effective therapeutic options. Although DNP is closely associated with hyperglycemia, an increasing body of research suggests that elevated blood glucose levels are not the sole inducers of DNP. The pathogenesis of DNP is intricate, involving the release of inflammatory mediators, alterations in synaptic plasticity, demyelination of nerve fibers, and ectopic impulse generation, yet the precise mechanisms remain to be elucidated. The spinal dorsal horn coordinates dynamic interactions between peripheral and central pain pathways, wherein dorsal horn neurons, microglia, and astrocytes synergize with Schwann cell-derived signals to process nociceptive information flow. Abnormally activated neurons can alter signal transduction by modifying the local microenvironment, compromising myelin integrity, and diminishing trophic support, leading to neuronal sensitization and an amplifying effect on peripheral pain signals, which in turn triggers neuropathic pain. Ion channels play a pivotal role in signal conduction, with the modulation of sodium, potassium, and calcium channels being particularly crucial for the regulation of pain signals. In light of the rising incidence of diabetes and the current scarcity of effective DNP treatments, a thorough investigation into the interactions between neurons and glial cells, especially the mechanisms of ion channel function in DNP, is imperative for identifying potential drug targets, developing novel therapeutic strategies, and thereby enhancing the prospects for DNP management.
Collapse
Affiliation(s)
- Jie Wu
- Department of Anesthesiology, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi, China
| | - Haijun Hu
- Department of Anesthesiology, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi, China
| | - Xi Li
- Department of Anesthesiology, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi, China
| |
Collapse
|
35
|
Song M, Dai H, Zhou Q, Meng X. The immunology of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2025; 16:1542208. [PMID: 40260277 PMCID: PMC12009709 DOI: 10.3389/fendo.2025.1542208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Diabetic cardiomyopathy is a notable microvascular complication of diabetes, characterized primarily by myocardial fibrosis and functional abnormalities. Long-term hyperglycemia induces excessive activation and recruitment of immune cells and triggers the cascade of inflammatory responses, resulting in systemic and local cardiac inflammation. Emerging evidence highlights the significant roles of immunology in modulating the pathology of diabetic cardiomyopathy. As the primary effectors of inflammatory reactions, immune cells are consistently present in cardiac tissue and can be recruited under pathological hyperglycemia circumstances. A disproportionate favor to proinflammatory types of immune cells and the increased proinflammatory cytokine levels mediate fibroblast proliferation, phenotypic transformation, and collagen synthesis and ultimately rise to cardiac fibrosis and hypertrophy. Meanwhile, the severity of cardiac fibrosis is also strongly associated with the diverse phenotypes and phenotypic alterations of the immune cells, including macrophages, dendritic cells, mast cells, neutrophils, and natural killer cells in innate immunity and CD4+ T lymphocytes, CD8+ T lymphocytes, and B lymphocytes in adaptive immunity. In this review, we synthesized the current analysis of the critical role played by the immune system and its components in the progression of diabetic cardiomyopathy. Finally, we highlight preclinical and clinical immune targeting strategies and translational implications.
Collapse
Affiliation(s)
| | | | | | - Xiao Meng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
36
|
Luong TVT, Yang S, Kim J. Lipotoxicity as a therapeutic target in the type 2 diabetic heart. J Mol Cell Cardiol 2025; 201:105-121. [PMID: 40020774 DOI: 10.1016/j.yjmcc.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Cardiac lipotoxicity, characterized by excessive lipid accumulation in the cardiac tissue, is a critical contributor to the pathogenesis of diabetic heart. Recent research has highlighted the key mechanisms underlying lipotoxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammation, and cell apoptosis, which ultimately impair the cardiac function. Various therapeutic interventions have been developed to target these pathways, mitigate lipotoxicity, and improve cardiovascular outcomes in diabetic patients. Given the global escalation in the prevalence of diabetes and the urgent demand for effective therapeutic approaches, this review focuses on how targeting cardiac lipotoxicity may be a promising avenue for treating diabetes.
Collapse
Affiliation(s)
- Trang Van T Luong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Seonbu Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
37
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
38
|
Guo L, Du Y, Li H, He T, Yao L, Yang G, Yang X. Metabolites-mediated posttranslational modifications in cardiac metabolic remodeling: Implications for disease pathology and therapeutic potential. Metabolism 2025; 165:156144. [PMID: 39864796 DOI: 10.1016/j.metabol.2025.156144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
The nonenergy - producing functions of metabolism are attracting increasing attention, as metabolic changes are involved in discrete pathways modulating enzyme activity and gene expression. Substantial evidence suggests that myocardial metabolic remodeling occurring during diabetic cardiomyopathy, heart failure, and cardiac pathological stress (e.g., myocardial ischemia, pressure overload) contributes to the progression of pathology. Within the rewired metabolic network, metabolic intermediates and end-products can directly alter protein function and/or regulate epigenetic modifications by providing acyl groups for posttranslational modifications, thereby affecting the overall cardiac stress response and providing a direct link between cellular metabolism and cardiac pathology. This review provides a comprehensive overview of the functional diversity and mechanistic roles of several types of metabolite-mediated histone and nonhistone acylation, namely O-GlcNAcylation, lactylation, crotonylation, β-hydroxybutyrylation, and succinylation, as well as fatty acid-mediated modifications, in regulating physiological processes and contributing to the progression of heart disease. Furthermore, it explores the potential of these modifications as therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Lifei Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; Cadet Team 6 of School of Basic Medicine, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Yuting Du
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Heng Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Ting He
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Li Yao
- Department of Pathology, Xi' an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi' an 710018, China
| | - Guodong Yang
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China.
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China.
| |
Collapse
|
39
|
Ramos S. Protective Effects of Flavonoids in Diabetic Cardiomyopathy: A Comprehensive Review on the Mechanistic Insights. Mol Nutr Food Res 2025:e70038. [PMID: 40159847 DOI: 10.1002/mnfr.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Diabetic cardiomyopathy (DCM) is one of the leading causes of mortality among diabetic patients. Flavonoid are the most abundant group of phytochemicals in fruits and vegetables, and have received increasing interest as potential chemopreventive antidiabetic agents. Flavonoids might contribute to prevent or delay DCM by regulating the cardiac metabolism, insulin signaling, oxidative stress, apoptosis, autophagy, and inflammation. Among other effects, flavonoids have been proved to enhance glucose uptake, decrease cellular lipid accumulation, or suppress oxidative stress. However, the mechanistic basis of these effects is not fully understood, and many points remain to be clarified. This review provides insight into the molecular mechanisms of flavonoid chemopreventive activity by summarizing cell culture and animal model studies.
Collapse
Affiliation(s)
- Sonia Ramos
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Ciudad Universitaria, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
40
|
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
|
41
|
Saki H, Nazem F, Khaiyat O, Fariba F. Effects of 12-week combined interval running and resistance training on cardiac structure and performance in patients with type 1 diabetes. Ther Adv Endocrinol Metab 2025; 16:20420188251325148. [PMID: 40162367 PMCID: PMC11954380 DOI: 10.1177/20420188251325148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/29/2025] [Indexed: 04/02/2025] Open
Abstract
Background Exercise has been suggested to effectively improve cardiac performance in children with type 1 diabetes (T1D) by enhancing the glycemic control. The purpose of this study was to investigate (1) effects of a 12-week combined interval running and resistance training (CIRRT) and (2) 1 month of detraining on cardiac structure and myocardial performance in adolescent males with T1D. Methods A total of 72 participants, including 48 adolescent males with T1D (fasting blood glucose (FBG): 274.67 ± 52.99 mg/dL, age: 15.20 ± 1.78 years) and 24 healthy adolescents (FBG: 90.75 ± 5.47 mg/dL, age: 15.08 ± 1.67 years), were recruited to the study. Participants were allocated into diabetes exercise (DE), diabetes control (DC), and healthy controls (HC) groups. The DE group performed 12 weeks of a CIRRT program three times per week. Blood glucose profile, echocardiography (ECHO) indices, and peak oxygen consumption (VO2peak) were measured pre- and post-intervention and following 1-month detraining period. Repeated measures ANOVA was used for pre- and post-intervention comparisons within the DE group and across the three study groups. Significance level was set at p < 0.05. Results Exercise intervention resulted in decreased hemoglobin A1c (HbA1c% = Pre: 10.44 ± 2.03, Post: 9.38 ± 1.66, p < 0.05), FBG, left ventricular (LV) internal diameter, and both tricuspid and mitral deceleration time (DT) in the DE group. VO2peak, ejection fraction (EF% = Pre: 62.38 ± 1.6, Post: 64.08 ± 1.18, p < 0.05), fractional shortening, early tricuspid diastolic inflow E velocity, and tricuspid velocity during atrial contraction were also increased following the exercise training. HbA1c (Pre vs Follow-up: 9.83 ± 1.73, p < 0.05), EF (Pre vs Follow-up: 62.97 ± 1.56, p < 0.05), LV, and DT tricuspid remained significantly improved after detraining period compared to the baseline. In the baseline, the glycemic index and ECHO variable significantly differed in the DE and DC groups with the HC group (p < 0.05). However, after the intervention, the DC and HC groups did not change significantly (p > 0.05). Conclusion The CIRRT intervention was associated with improved cardiac structure and performance in male adolescents with T1D potentially due to exercise-induced adaptations. Meanwhile, the results indicate that most cardiac morphological and functional changes are reversible following periods of inactivity in patients with T1D.
Collapse
Affiliation(s)
- Hossein Saki
- Department of Exercise Physiology, Sports Science Faculty, Bu-Ali Sina University, Hamadan, Iran
| | - Farzad Nazem
- Department of Exercise Physiology, Sports Science Faculty, Bu-Ali Sina University, Hamadan 65174, Iran
| | - Omid Khaiyat
- School of Health and Sport Sciences, Liverpool Hope University, Liverpool, UK
| | - Farnaz Fariba
- Department of Cardiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
42
|
Niu W, Liu X, Deng B, Hong T, Wang C, Yan Y, Liu J, Jiang Y, Li J. Piezo1 deletion mitigates diabetic cardiomyopathy by maintaining mitochondrial dynamics via ERK/Drp1 pathway. Cardiovasc Diabetol 2025; 24:127. [PMID: 40114130 PMCID: PMC11927149 DOI: 10.1186/s12933-025-02625-8] [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: 12/16/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE Increasing evidence highlights the critical role of Piezo1 in cardiovascular diseases, with its expression upregulated in diabetic heart. However, the involvement of Piezo1 in the pathogenesis of diabetic cardiomyopathy (DCM) remains unclear. This study aims to elucidate the regulatory role of Piezo1 in mitochondrial dynamics within the context of DCM and to investigate the underlying mechanisms. METHODS We constructed cardiac-specific knockout of Piezo1 (Piezo1∆Myh6) mice. Type 1 diabetes was induced using streptozotocin (STZ) injection while type 2 diabetes was established through a high-fat diet combined with STZ. Echocardiography assessed left ventricular function, histological evaluations used HE and Masson staining to examine cardiac pathology in Piezo1fl/fl controls, Piezo1∆Myh6 controls, Piezo1fl/fl diabetic and Piezo1∆Myh6 diabetic mice. Mitochondrial function including oxygen species level, mitochondrial morphology, and respiration rate were also assessed. RESULTS Our findings revealed that Piezo1 expression was upregulated in the myocardium of diabetic mice and in high-glucose-treated cells. Cardiac-specific knockout of Piezo1 improved cardiac dysfunction and ameliorated cardiac fibrosis in diabetic mice. Moreover, Piezo1 deficiency also attenuated mitochondrial impairment. Piezo1fl/fl diabetic mice exhibited increased calpain activity and excessive mitochondrial fission mediated by Drp1 and obvious reduced fusion; however, Piezo1 deficiency restored calpain levels and mitochondrial dysfunction. These observations were also corroborated in H9C2 cells and neonatal mouse cardiomyocytes. Cardiac-specific knockout of Piezo1 increased phosphorylation of Drp1 and ERK1/2 in vivo and in vitro. Piezo1 knockout or treatment with inhibitor improved mitochondrial function. CONCLUSIONS This study provides the first evidence that Piezo1 is elevated in DCM through the modulation of mitochondrial dynamics, which is reversed by Piezo1 deficiency. Thus, Piezo1 inhibition may provide a promising therapeutic strategy for the treatment of DCM.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/prevention & control
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/etiology
- Mitochondrial Dynamics
- Dynamins/metabolism
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/chemically induced
- Ion Channels/genetics
- Ion Channels/deficiency
- Ion Channels/metabolism
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Male
- Fibrosis
- Mice, Inbred C57BL
- Phosphorylation
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/complications
- Ventricular Function, Left
- Extracellular Signal-Regulated MAP Kinases/metabolism
- MAP Kinase Signaling System
- Rats
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/complications
- Mice
Collapse
Affiliation(s)
- Weipin Niu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, China
| | - Xin Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Shandong Institute of Commerce and Technology, Jinan, 250103, China
| | - Bo Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Tianying Hong
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Cuifen Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yameng Yan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jiali Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yuehua Jiang
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
43
|
Li D, Ma Q. Ubiquitin-specific protease: an emerging key player in cardiomyopathy. Cell Commun Signal 2025; 23:143. [PMID: 40102846 PMCID: PMC11921692 DOI: 10.1186/s12964-025-02123-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Protein quality control (PQC) plays a vital role in maintaining normal heart function, as cardiomyocytes are relatively sensitive to misfolded or damaged proteins, which tend to accumulate under pathological conditions. Ubiquitin-specific protease (USP) is the largest deubiquitinating enzyme family and a key component of the ubiquitin proteasome system (UPS), which is a non-lysosomal protein degradation machinery to mediate PQC in cells. USPs regulate the stability or activity of the target proteins that involve intracellular signaling, transcriptional control of inflammation, antioxidation, and cell growth. Recent studies demonstrate that the USPs can regulate fibrosis, lipid metabolism, glucose homeostasis, hypertrophic response, post-ischemic recovery and cell death such as apoptosis and ferroptosis in cardiomyocytes. Since myocardial cell loss is an important component of cardiomyopathy, therefore, these findings suggest that the UPSs play emerging roles in cardiomyopathy. This review briefly summarizes recent literature on the regulatory roles of USPs in the occurrence and development of cardiomyopathy, giving us new insights into the molecular mechanisms of USPs in different cardiomyopathy and potential preventive strategies for cardiomyopathy.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Qilin Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China.
| |
Collapse
|
44
|
Abdullah AR, Seliem MA, Khidr EG, Sobhy AM, El-Shiekh RA, Hafeez MSAE, El-Husseiny AA. A comprehensive review on diabetic cardiomyopathy (DCM): histological spectrum, diagnosis, pathogenesis, and management with conventional treatments and natural compounds. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03980-9. [PMID: 40100371 DOI: 10.1007/s00210-025-03980-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/22/2025] [Indexed: 03/20/2025]
Abstract
Diabetic complications are among the most pressing health issues currently. Cardiovascular problems, particularly diabetic cardiomyopathy (DCM), are responsible for almost 80% of diabetic deaths. Because of the increasing prevalence of diabetes and the increased threat of death from its consequences, researchers are searching for new pharmaceutical targets to delay or cure it. Currently, there are a few medicines available for the treatment of DCM, some of which have serious side effects. To address this issue, researchers are focusing on natural products. Thus, in this review, we discuss the prevalence, incidence, risk factors, histological spectrum, diagnosis, pathogenic pathways of DCM, genetic and epigenetic mechanisms involved in DCM, the current treatments, and the beneficial effects of natural product-based therapeutics. Natural treatments range from single doses to continuous regimens lasting weeks or months. Flavonoids are the largest class of natural compounds reported for the treatment of DCM. Natural regimens may cover the way for new treatment strategies for DCM for being multi-target agents in the treatment of DCM, with the ability to play a variety of functions via distinct signaling pathways.
Collapse
Affiliation(s)
- Ahmed R Abdullah
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11231, Egypt
| | - Mahmoud A Seliem
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11231, Egypt
| | - Ayah M Sobhy
- Pharmacognosy Department, Faculty of Pharmacy, Badr University in Assiut, Assiut, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| | - Mohamed S Abd El Hafeez
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, 11829, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11231, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt
| |
Collapse
|
45
|
Ogurtsova E, Arefieva T, Filatova A, Radyukhina N, Ovchinnikov A. Cardiometabolic Phenotype in HFpEF: Insights from Murine Models. Biomedicines 2025; 13:744. [PMID: 40149720 PMCID: PMC11940576 DOI: 10.3390/biomedicines13030744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) remains a significant challenge in modern healthcare. It accounts for the majority of heart failure cases and their number worldwide is steadily increasing. With its high prevalence and substantial clinical impact, therapeutic strategies for HFpEF are still inadequate. This review focuses on the cardiometabolic phenotype of HFpEF which is characterised by such conditions as obesity, type 2 diabetes mellitus, and hypertension. Various murine models that mimic this phenotype are discussed. Each model's pathophysiological aspects, namely inflammation, oxidative stress, endothelial dysfunction, changes in cardiomyocyte protein function, and myocardial metabolism alterations are examined in detail. Understanding these models can provide insight into the mechanisms underlying HFpEF and aid in the development of effective therapeutic interventions.
Collapse
Affiliation(s)
- Ekaterina Ogurtsova
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Prospekt, 27/1, 117192 Moscow, Russia
| | - Tatiana Arefieva
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Prospekt, 27/1, 117192 Moscow, Russia
| | - Anastasiia Filatova
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
| | - Natalya Radyukhina
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
| | - Artem Ovchinnikov
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
- Department of Clinical Functional Diagnostics, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, 127473 Moscow, Russia
| |
Collapse
|
46
|
Tang Q, Ji Y, Xia Z, Zhang Y, Dong C, Sun Q, Lei S. Identification and validation of endoplasmic reticulum stress-related diagnostic biomarkers for type 1 diabetic cardiomyopathy based on bioinformatics and machine learning. Front Endocrinol (Lausanne) 2025; 16:1478139. [PMID: 40171194 PMCID: PMC11959167 DOI: 10.3389/fendo.2025.1478139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Background Diabetic cardiomyopathy (DC) is a serious complication in patients with type 1 diabetes mellitus and has become a growing public health problem worldwide. There is evidence that endoplasmic reticulum stress (ERS) is involved in the pathogenesis of DC, and related diagnostic markers have not been well-studied. Therefore, this study aimed to screen ERS-related genes (ERGs) with potential diagnostic value in DC. Methods Gene expression data on DC were downloaded from the GEO database, and ERGs were obtained from The Gene Ontology knowledgebase. Limma package analyzed differentially expressed genes (DEGs) in the DC and control groups, and then integrated with ERGs to identify ERS-related DEGs (ERDEGs). The ERDEGs diagnostic model was developed based on a combination of LASSO and Random Forest approaches, and the diagnostic performance was evaluated by the area under the receiver operating characteristic curve (ROC-AUC) and validated against external datasets. In addition, the association of the signature genes with immune infiltration was analyzed using the CIBERSORT algorithm and the Spearman correlation test. Results Gene expression data on DC were downloaded from the GEO database and ERGs were obtained from the Gene Ontology Knowledgebase. Limma package analysis identified 3100 DEGs between DC and control groups and then integrated with ERGs to identify 65 ERDEGs. Four diagnostic markers, Npm1, Jkamp, Get4, and Lpcat3, were obtained based on the combination of LASSO and random forest approach, and their ROC-AUCs were 0.9112, 0.9349, 0.8994, and 0.8639, respectively, which proved their diagnostic potential in DC. Meanwhile, Npm1, Jkamp, Get4, and Lpcat3 were validated by external datasets and a mouse model of type 1 DC. In addition, Npm1 was significantly negatively correlated with plasma cells, activated natural killer cells, or quiescent mast cells, whereas Get4 was significantly positively correlated with quiescent natural killer cells and significantly negatively correlated with activated natural killer cells (P < 0.05). Conclusions This study provides novel diagnostic biomarkers (Npm1, Jkamp, Get4, and Lpcat3) for DC from the perspective of ERS, which provides new insights into the development of new targets for individualized treatment of type 1 diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Qiao Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanwei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuxi Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chong Dong
- Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| | - Qian Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
47
|
El-Afify D, El Amrousy D. Cardioprotective Effect of Nigella sativa in Pediatric Patients with Type 1 Diabetes Mellitus: A Randomized Controlled Study. Paediatr Drugs 2025. [DOI: 10.1007/s40272-025-00687-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 04/02/2025]
|
48
|
Fang T, Wang CL, Zhang Y, Zhu R, Lin Y, Dai QX, Cai S, Chen LW, Wen XL. Evaluation of silicosis combined with type 2 diabetes mellitus based on the quantitative CT measured parameters. Quant Imaging Med Surg 2025; 15:2258-2269. [PMID: 40160656 PMCID: PMC11948391 DOI: 10.21037/qims-24-1748] [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: 08/22/2024] [Accepted: 01/16/2025] [Indexed: 04/02/2025]
Abstract
Background Type 2 diabetes mellitus (T2DM) and silicosis both have significant impacts on cardiopulmonary health. However, there is a lack of research investigating whether the presence of T2DM causes additional damage to the heart and lungs of patients with silicosis. This study aims to assess the alterations in pulmonary and cardiac structures, as well as lung function, in patients with silicosis who also have T2DM, and to explore the factors influencing lung function. Methods We included 30 silicosis patients with T2DM and 30 silicosis patients without diabetes. The two groups were matched by age, silicosis stage, smoking history, and dust exposure duration. Demographic details, occupational history, hematological results, computed tomography (CT)-measured cardiac and lung parameters, and pulmonary function test (PFT) results were collected. We compared these parameters between the two groups, evaluated the correlation between lung function and CT parameters in the diabetic group, and analyzed factors affecting lung function in this group. Results The silicosis combined with T2DM group showed significantly higher values for body mass index (BMI), the longest diameters from left to right of the left ventricle (LVLR), blood glucose, and triglycerides compared to the silicosis but without T2DM group (all P<0.05). The silicosis combined with T2DM group showed significantly lower total lung mass, and the ratio of forced expiratory volume in one second to forced vital capacity (FEV1/FVC) compared to the silicosis but without T2DM group (all P<0.05). In the silicosis with T2DM group, FEV1/FVC ratio showed significant correlations with total lung mass, the longest diameters from left to right of the left atrium, LVLR, and the longest diameters from left to right of the right atrium (r values were 0.51, 0.47, 0.40, and 0.44, respectively; all P<0.05). Multivariate analysis revealed that BMI and the LVLR were independent determinants of FEV1/FVC ratio in the silicosis with T2DM group (t values were -3.367 and 2.471, respectively; all P<0.05). Conclusions Diabetes mellitus induces structural changes in the lungs and heart of patients with silicosis and exacerbates the impairment of lung function. BMI and LVLR are key determinants of the FEV1/FVC ratio highlighting the need for enhanced comprehensive management strategies.
Collapse
Affiliation(s)
- Ting Fang
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Cheng-Long Wang
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Rui Zhu
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yue Lin
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Qin-Xi Dai
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Shu Cai
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lai-Wei Chen
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiao-Ling Wen
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Zeng JQ, Zhou HF, Du HX, Wu YJ, Mao QP, Yin JJ, Wan HT, Yang JH. Tongmai Hypoglycemic Capsule Attenuates Myocardial Oxidative Stress and Fibrosis in the Development of Diabetic Cardiomyopathy in Rats. Chin J Integr Med 2025; 31:251-260. [PMID: 39644459 DOI: 10.1007/s11655-024-4002-3] [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] [Accepted: 07/03/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE To investigate the effect of Tongmai Hypoglycemic Capsule (THC) on myocardium injury in diabetic cardiomyopathy (DCM) rats. METHODS A total of 24 Sprague Dawley rats were fed for 4 weeks with high-fat and high-sugar food and then injected with streptozotocin intraperitoneally for the establishment of the DCM model. In addition, 6 rats with normal diets were used as the control group. After modeling, 24 DCM rats were randomly divided into the model, L-THC, M-THC, and H-THC groups by computer generated random numbers, and 0, 0.16, 0.32, 0.64 g/kg of THC were adopted respectively by gavage, with 6 rats in each group. After 12 weeks of THC administration, echocardiography, histopathological staining, biochemical analysis, and Western blot were used to detect the changes in myocardial structure, oxidative stress (OS), biochemical indexes, protein expressions of myocardial fibrosis, and nuclear factor erythroid 2-related faactor 2 (Nrf2) element, respectively. RESULTS Treatment with THC significantly decreased cardiac markers such as creatine kinase, lactate dehydrogenase, and creatine kinase-MB, etc., (P<0.01); enhanced cardiac function indicators including heart rate, ejection fraction, cardiac output, interventricular septal thickness at diastole, and others (P<0.05 or P<0.01); decreased levels of biochemical indicators such as fasting blood glucose, total cholesterol, triglycerides, low-density lipoprotein cholesterol, aspartate transaminase, (P<0.05 or P<0.01); and decreased the levels of myocardial fibrosis markers α-smooth muscle actin (α-SMA), and collagen I (Col-1) protein (P<0.01), improved myocardial morphology and the status of myocardial interstitial fibrosis. THC significantly reduced malondialdehyde levels in model rats (P<0.01), increased levels of catalase, superoxide dismutase, and glutathione (P<0.01), and significantly increased the expression of Nrf2, NAD(P)H:quinone oxidoreductase 1, heme oxygenase-1, and superoxide dismutase 2 proteins in the left ventricle of rats (P<0.01). CONCLUSION THC activates the Nrf2 signaling pathway and plays a protective role in reducing OS injury and cardiac fibrosis in DCM rats.
Collapse
Affiliation(s)
- Jie-Qiong Zeng
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Hui-Fen Zhou
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Hai-Xia Du
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Yu-Jia Wu
- College of Life Science, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Qian-Ping Mao
- College of Life Science, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Jun-Jun Yin
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Hai-Tong Wan
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Jie-Hong Yang
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China.
| |
Collapse
|
50
|
Zhang M, Sun L, Wu X, Qin Y, Lin M, Ding X, Zhu W, Jiang Z, Jin S, Leng C, Wang J, Lv X, Cai Q. Effects of 3-month dapagliflozin on left atrial function in treatment-naïve patients with type 2 diabetes mellitus: Assessment using 4-dimensional echocardiography. Hellenic J Cardiol 2025; 82:43-53. [PMID: 38092177 DOI: 10.1016/j.hjc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/21/2023] [Accepted: 12/09/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND The sodium-glucose transporter-2 (SGLT-2) inhibitor dapagliflozin can improve left ventricular (LV) performance in patients with type 2 diabetes mellitus (T2DM). However, the effects on left atrial (LA) function in treatment-naïve T2DM patients remain unclear. The aim of our study was 1) to investigate the effects of 3-month treatment with dapagliflozin on LA function in treatment-naïve patients with T2DM using 4-dimensional automated LA quantification (4D Auto LAQ) and 2) to explore linked covariation patterns of changes in clinical and LA echocardiographic variables. METHODS 4D Auto LAQ was used to evaluate LA volumes, longitudinal and circumferential strains in treatment-naïve T2DM patients at baseline, at follow-up, and in healthy control (HC). Sparse canonical correlation analysis (sCCA) was performed to capture the linked covariation patterns between changes in clinical and LA echocardiographic variables within the treatment-naïve T2DM patient group. RESULTS This study finally included 61 treatment-naïve patients with T2DM without cardiovascular disease and 39 healthy controls (HC). Treatment-naïve T2DM patients showed reduced LA reservoir and conduit function at baseline compared to HC, independent of age, sex, BMI, and blood pressure (LASr: 21.11 ± 5.39 vs. 27.08 ± 5.31 %, padjusted = 0.017; LAScd: -11.51 ± 4.48 vs. -16.74 ± 4.51 %, padjusted = 0.013). After 3-month treatment with dapagliflozin, T2DM patients had significant improvements in LA reservoir and conduit function independent of BMI and blood pressure changes (LASr: 21.11 ± 5.39 vs. 23.84 ± 5.74 %, padjusted < 0.001; LAScd: -11.51 ± 4.48 vs. -12.75 ± 4.70 %, padjusted < 0.001). The clinical and LA echocardiographic parameters showed significant covariation (r = 0.562, p = 0.039). In the clinical dataset, changes in heart rate, insulin, and BMI were most associated with the LA echocardiographic variate. In the LA echocardiographic dataset, changes in LAScd, LASr, and LASr_c were most associated with the clinical variate. CONCLUSION Compared with HC, treatment-naïve patients with T2DM had lower LA function, and these patients benefited from dapagliflozin administration, particularly in LA function.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Lanlan Sun
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaopeng Wu
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yunyun Qin
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Mingming Lin
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xueyan Ding
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Weiwei Zhu
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhe Jiang
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Shan Jin
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Chenlei Leng
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China
| | | | - Xiuzhang Lv
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Qizhe Cai
- Department of Ultrasound Medicine, Beijing Chao Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|