1
|
Chun-peng ZHANG, Tian CAO, Xue YANG. Pharmacological mechanisms of Taohe Chengqi decoction in diabetic cardiovascular complications: A systematic review, network pharmacology and molecular docking. Heliyon 2024; 10:e33308. [PMID: 39044965 PMCID: PMC11263673 DOI: 10.1016/j.heliyon.2024.e33308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Background Diabetic cardiovascular complications are the leading cause of diabetes-related deaths. These complications place an enormous and growing burden on global health systems and economies. The objective of this study was to conduct a systematic review on the therapeutic mechanisms of Taohe Chengqi Decoction (THCQD) in the treatment of diabetic cardiovascular complications. To predict the potential mechanisms of action of THCQD on diabetic cardiovascular complications using network pharmacology, and to validate these predictions through molecular docking analysis. Methods To collect relevant animal experiments, we searched a total of 6 databases. Eligibility for the study was determined based on inclusion and exclusion criteria. Data extraction was then performed on the literature. Methodological quality of animal studies was assessed using SYRCLE criteria. Based on network pharmacology, intersecting genes for THCQD and diabetic cardiovascular complications were obtained using Venny, PPI analysis and topology analysis of intersecting genes were performed; GO and KEGG were used for enrichment analysis and prediction of new targets of action. Molecular docking techniques were employed to model the interactions between drug components and target genes, thereby validating the results of network pharmacology predictions. Results A total of 16 studies were finally identified that fit the direction of this review. Included 6 studies of the myocardium, 1 study of the aortic arch, 5 studies of the femoral artery, 4 studies of the thoracic aorta. THCQD exhibited anti-inflammatory, anti-fibrotic and anti-atherosclerotic effects on cardiovascular complications in diabetic rats. Network pharmacology results showed that C0363 (Resveratrol), C0041 (Emodin), and C1114 (Baicalein) were the key components in the treatment of diabetic cardiovascular complications by THCQD. PPI results showed that INS, AKT1, TNF, ALB, IL6, IL1B as the genes that interact with the top 6. KEGG enrichment analysis identified the AGE-RAGE signaling pathway in diabetic complications as the most prominent pathway enriched by THCQD for diabetic cardiovascular complications genes. The results of molecular docking showed that the key active components demonstrated favorable interactions with their corresponding target genes. Conclusion In conclusion, the results of both basic and web-based pharmacological studies support the beneficial effects of the natural herbal formulation THCQD on diabetic cardiovascular complications. This decoction has anti-inflammatory and antifibrotic properties and is effective in ameliorating diabetic cardiovascular disease. The network pharmacology results further support these ideas and identify the AGE-RAGE signaling pathway in diabetic complications as possibly the most relevant pathway for THCQD in the treatment of diabetic cardiovascular complications. The extent of the therapeutic potential of all-natural herbal components in the treatment of diabetic cardiovascular disease merits further investigation.
Collapse
Affiliation(s)
- ZHANG Chun-peng
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - CAO Tian
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - YANG Xue
- Department of Traditional Chinese Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| |
Collapse
|
2
|
Zhou M, Hanschmann EM, Römer A, Linn T, Petry SF. The significance of glutaredoxins for diabetes mellitus and its complications. Redox Biol 2024; 71:103043. [PMID: 38377787 PMCID: PMC10891345 DOI: 10.1016/j.redox.2024.103043] [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: 12/09/2023] [Accepted: 01/13/2024] [Indexed: 02/22/2024] Open
Abstract
Diabetes mellitus is a non-communicable metabolic disease hallmarked by chronic hyperglycemia caused by beta-cell failure. Diabetic complications affect the vasculature and result in macro- and microangiopathies, which account for a significantly increased morbidity and mortality. The rising incidence and prevalence of diabetes is a major global health burden. There are no feasible strategies for beta-cell preservation available in daily clinical practice. Therefore, patients rely on antidiabetic drugs or the application of exogenous insulin. Glutaredoxins (Grxs) are ubiquitously expressed and highly conserved members of the thioredoxin family of proteins. They have specific functions in redox-mediated signal transduction, iron homeostasis and biosynthesis of iron-sulfur (FeS) proteins, and the regulation of cell proliferation, survival, and function. The involvement of Grxs in chronic diseases has been a topic of research for several decades, suggesting them as therapeutic targets. Little is known about their role in diabetes and its complications. Therefore, this review summarizes the available literature on the significance of Grxs in diabetes and its complications. In conclusion, Grxs are differentially expressed in the endocrine pancreas and in tissues affected by diabetic complications, such as the heart, the kidneys, the eye, and the vasculature. They are involved in several pathways essential for insulin signaling, metabolic inflammation, glucose and fatty acid uptake and processing, cell survival, and iron and mitochondrial metabolism. Most studies describe significant changes in glutaredoxin expression and/or activity in response to the diabetic metabolism. In general, mitigated levels of Grxs are associated with oxidative distress, cell damage, and even cell death. The induced overexpression is considered a potential part of the cellular stress-response, counteracting oxidative distress and exerting beneficial impact on cell function such as insulin secretion, cytokine expression, and enzyme activity.
Collapse
Affiliation(s)
- Mengmeng Zhou
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Eva-Maria Hanschmann
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Axel Römer
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Sebastian Friedrich Petry
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
3
|
Goyal P, Maurer MS, Roh J. Aging in Heart Failure: Embracing Biology Over Chronology: JACC Family Series. JACC. HEART FAILURE 2024; 12:795-809. [PMID: 38597865 PMCID: PMC11331491 DOI: 10.1016/j.jchf.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 04/11/2024]
Abstract
Age is among the most potent risk factors for developing heart failure and is strongly associated with adverse outcomes. As the global population continues to age and the prevalence of heart failure rises, understanding the role of aging in the development and progression of this chronic disease is essential. Although chronologic age is on a fixed course, biological aging is more variable and potentially modifiable in patients with heart failure. This review describes the current knowledge on mechanisms of biological aging that contribute to the pathogenesis of heart failure. The discussion focuses on 3 hallmarks of aging-impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing-that are currently being targeted in therapeutic development for older adults with heart failure. In assessing existing and emerging therapeutic strategies, the review also enumerates the importance of incorporating geriatric conditions into the management of older adults with heart failure and in ongoing clinical trials.
Collapse
Affiliation(s)
- Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Mathew S Maurer
- Department of Medicine, Columbia University Medical Center, New York, New York, USA.
| | - Jason Roh
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Liu H, Yan W, Ma C, Zhang K, Li K, Jin R, Xu H, Xu R, Tong J, Yang Z, Guo Y. Early detection of cardiac fibrosis in diabetic mice by targeting myocardiopathy and matrix metalloproteinase 2. Acta Biomater 2024; 176:367-378. [PMID: 38244659 DOI: 10.1016/j.actbio.2024.01.017] [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: 09/27/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/22/2024]
Abstract
Early detection of myocardial fibrosis in diabetic cardiomyopathy (DCM) has significant clinical implications for diabetes management. In this study, we identified matrix metalloproteinase 2 (MMP2) as a potential biomarker for early fibrosis detection. Based on this finding, we designed a dual-targeting nanoparticle CHP-SPIO-ab MMP2 to specifically target myocardiopathy and MMP2, enabling sensitive fibrosis detection using magnetic resonance imaging (MRI). Our results demonstrate that collagen hyperplasia (early fiber formation) begins to develop in diabetic mice at 12 weeks old, with observable fibrosis occurring at 16 weeks old. Additionally, MMP2 expression significantly up-regulates around collagen starting from 12 weeks of age. T2 MRI analysis revealed significant T2% enhancement in the hearts of 12-week-old diabetic mice following administration of the CHP-SPIO-ab MMP2 probe, indicating noninvasive detection of fiber formation. Furthermore, after fibrosis treatment, a reduction in T2% signal was observed in the hearts of 16-week-old diabetic mice. These findings were supported by Sirius red and Prussian blue staining techniques. Overall, our study presents a promising strategy for early identification of myocardial fibrosis. STATEMENT OF SIGNIFICANCE: Myocardial damage typically exhibits irreversibility, underscoring the paramount importance of early fibrosis diagnosis. However, the clinical used T1 mapping for fibrosis detection still exhibits limitations in terms of sensitivity. Therefore, it is imperative to develop highly sensitive strategies for early cardiac fibrosis detection. Here, we investigated the development of myocardial fibrosis in diabetic mice, and designed a highly sensitive probe that specifically targets cardiomyopathy and high expression of MMP2 for the early diagnosis of fibrosis. The probe enables non-invasive detection of abnormalities through MRI imaging as soon as fiber deposition appear, which can be detected earlier than T1 mapping. This advancement holds great potential for clinical diagnosis of myocardial fibrosis using cardiac magnetic resonance.
Collapse
Affiliation(s)
- Hanrui Liu
- Key Laboratory of Obstet & Gynecol & Pediat Dis, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weifeng Yan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengyong Ma
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Zhang
- Key Laboratory of Obstet & Gynecol & Pediat Dis, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kuan Li
- Key Laboratory of Obstet & Gynecol & Pediat Dis, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Huayan Xu
- Key Laboratory of Obstet & Gynecol & Pediat Dis, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rong Xu
- Key Laboratory of Obstet & Gynecol & Pediat Dis, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiyu Tong
- Department of Pediatrics and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, China
| | - Zhigang Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yingkun Guo
- Key Laboratory of Obstet & Gynecol & Pediat Dis, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
5
|
Li Y, Xian H, Xu Y, Li W, Guo J, Wan K, Wang J, Xu Z, Zhang Q, Han Y, Sun J, Chen Y. The impact of type 2 diabetes mellitus on the clinical profile, myocardial fibrosis, and prognosis in non-ischemic dilated cardiomyopathy: a prospective cohort study. Cardiovasc Diabetol 2024; 23:48. [PMID: 38302987 PMCID: PMC10835902 DOI: 10.1186/s12933-024-02134-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND The impact of the coexistence of type 2 diabetes mellitus (T2DM) in patients with non-ischemic dilated cardiomyopathy (DCM) on clinical profiles, myocardial fibrosis, and outcomes remain incompletely understood. METHOD A total of 1152 patients diagnosed with non-ischemic DCM were prospectively enrolled from June 2012 to October 2021 and categorized into T2DM and non-T2DM groups. Clinical characteristics, cardiac function, and myocardial fibrosis evaluated by CMR were compared between the two groups. The primary endpoint included both all-cause mortality and heart transplantation. Cause of mortality was classified into heart failure death, sudden cardiac death, and non-cardiac death. Cox regression analysis and Kaplan-Meier analysis were performed to identify the association between T2DM and clinical outcomes. Propensity score matching (PSM) cohort including 438 patients was analyzed to reduce the bias from confounding covariates. RESULTS Among the 1152 included DCM patients, 155 (13%) patients had T2DM. Patients with T2DM were older (55 ± 12 vs. 47 ± 14 years, P < 0.001), had higher New York Heart Association (NYHA) functional class (P = 0.003), higher prevalence of hypertension (37% vs. 21%, P < 0.001), atrial fibrillation (31% vs. 16%, P < 0.001), lower left ventricular (LV) ejection fraction (EF) (23 ± 9% vs. 27 ± 12%, P < 0.001), higher late gadolinium enhancement (LGE) presence (55% vs. 45%, P = 0.02), and significantly elevated native T1 (1323 ± 81ms vs. 1305 ± 73ms, P = 0.01) and extracellular volume fraction (ECV) (32.7 ± 6.3% vs. 31.3 ± 5.9%, P = 0.01) values. After a median follow-up of 38 months (interquartile range: 20-57 months), 239 patients reached primary endpoint. Kaplan-Meier analysis showed that patients with T2DM had worse clinical outcomes compared with those without T2DM in the overall cohort (annual events rate: 10.2% vs. 5.7%, P < 0.001). T2DM was independently associated with an increased risk of primary endpoint in the overall (Hazard ratio [HR]: 1.61, 95% CI: 1.13-2.33, P = 0.01) and PSM (HR: 1.54, 95% CI: 1.05-2.24, P = 0.02) cohorts. Furthermore, T2DM was associated with a higher risk of heart failure death (P = 0.006) and non-cardiac death (P = 0.02), but not sudden cardiac death (P = 0.16). CONCLUSIONS Patients with T2DM represented a more severe clinical profile and experienced more adverse outcomes compared to those without T2DM in a large DCM cohort. TRIAL REGISTRATION Trial registration number: ChiCTR1800017058; URL: https://www. CLINICALTRIALS gov .
Collapse
Affiliation(s)
- Yangjie Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Hong Xian
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuanwei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Weihao Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Jiajun Guo
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Ke Wan
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jie Wang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Ziqian Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Qing Zhang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China
| | - Yuchi Han
- Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jiayu Sun
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yucheng Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, P. R. China.
| |
Collapse
|
6
|
Lou X, Zhang Y, Guo J, Gao L, Ding Y, Zhuo X, Lei Q, Bian J, Lei R, Gong W, Zhang X, Jiao Q. What is the impact of ferroptosis on diabetic cardiomyopathy: a systematic review. Heart Fail Rev 2024; 29:1-11. [PMID: 37555989 DOI: 10.1007/s10741-023-10336-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Iron overload increases the production of harmful reactive oxygen species in the Fenton reaction, which causes oxidative stress in the body and lipid peroxidation in the cell membrane, and eventually leads to ferroptosis. Diabetes is associated with increased intracellular oxidative stress, inflammation, autophagy, microRNA alterations, and advanced glycation end products (AGEs), which cause cardiac remodeling and cardiac diastolic contractile dysfunction, leading to the development of diabetic cardiomyopathy (DCM). While these factors are also closely associated with ferroptosis, more and more studies have shown that iron-mediated ferroptosis is an important causative factor in DCM. In order to gain fresh insights into the functions of ferroptosis in DCM, this review methodically summarizes the traits and mechanisms connected with ferroptosis and DCM.
Collapse
Affiliation(s)
- Xiaokun Lou
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Yuanyuan Zhang
- Department of Cardiovascular Ultrasonic Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Junfeng Guo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Lina Gao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Yingying Ding
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Xinyu Zhuo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Qingqing Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Jing Bian
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Rumei Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China
| | - Wenyan Gong
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China.
- Hangzhou Institute of Cardiovascular Disease, Hangzhou, 310000, China.
| | - Xingwei Zhang
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China.
- Hangzhou Institute of Cardiovascular Disease, Hangzhou, 310000, China.
| | - Qibin Jiao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, China.
| |
Collapse
|
7
|
Hsieh HH, Chu PA, Lin YH, Kao YCJ, Chung YH, Hsu ST, Mo JM, Wu CY, Peng SL. Imaging diabetic cardiomyopathy in a type 1 diabetic rat model using 18F-FEPPA PET. Nucl Med Biol 2024; 128-129:108878. [PMID: 38324923 DOI: 10.1016/j.nucmedbio.2024.108878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE Diabetic patients often experience chronic inflammation and fibrosis in their cardiac tissues, highlighting the pressing need for the development of sensitive diagnostic methods for longitudinal assessment of diabetic cardiomyopathy. This study aims to evaluate the significance of an inflammatory marker known as translocator protein (TSPO) in a positron emission tomography (PET) protocol for longitudinally monitoring cardiac dysfunction in a diabetic animal model. Additionally, we compared the commonly used radiotracer, 18F-fluoro-2-deoxy-d-glucose (18F-FDG). METHODS Fourteen 7-week-old female Sprague-Dawley rats were used in this study. Longitudinal PET experiments were conducted using 18F-N-2-(2-fluoroethoxy)benzyl)-N-(4-phenoxypyridin-3-yl)acetamide (18F-FEPPA) (n = 3), the TSPO radiotracer, and 18F-FDG (n = 3), both before and after the onset of diabetes. Histological and immunohistochemical staining assays were also conducted in both the control (n = 4) and diabetes (n = 4) groups. RESULTS Results indicated a significant increase in cardiac tissue uptake of 18F-FEPPA after the onset of diabetes (P < 0.05), aligning with elevated TSPO levels observed in diabetic animals according to histological data. Conversely, the uptake of 18F-FDG in cardiac tissue significantly decreased after the onset of diabetes (P < 0.05). CONCLUSION These findings suggest that 18F-FEPPA can function as a sensitive probe for detecting chronic inflammation and fibrosis in the cardiac tissues of diabetic animals.
Collapse
Affiliation(s)
- Hsin-Hua Hsieh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan
| | - Pei-An Chu
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Chieh Jill Kao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan
| | - Yi-Hsiu Chung
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Ting Hsu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jia-Min Mo
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei, Taiwan.
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan; Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
8
|
Vishwakarma VK, Shah S, Kaur T, Singh AP, Arava SK, Kumar N, Yadav RK, Yadav S, Arora T, Yadav HN. Effect of vinpocetine alone and in combination with enalapril in experimental model of diabetic cardiomyopathy in rats: possible involvement of PDE-1/TGF-β/ Smad 2/3 signalling pathways. J Pharm Pharmacol 2023; 75:1198-1211. [PMID: 37229596 DOI: 10.1093/jpp/rgad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Diabetic cardiomyopathy (DC) is one of the severe secondary complications of diabetes mellitus in humans. Vinpocetine is an alkaloid having pleiotropic pharmacological effects. The present study is designed to investigate the effect of vinpocetine in DC in rats. METHODS Rats were fed a high-fat diet for nine weeks along with single dose of streptozotocin after the second week to induce DC. The haemodynamic evaluation was performed to assess the functional status of rats using the Biopac system. Cardiac echocardiography, biochemical, oxidative stress parameters and inflammatory cytokine level were analysed in addition to haematoxylin-eosin and Masson's trichome staining to study histological changes, cardiomyocyte diameter and fibrosis, respectively. Phosphodiesterase-1 (PDE-1), transforming growth factor-β (TGF-β) and p-Smad 2/3 expression in cardiac tissues were quantified using western blot/RT-PCR. KEY FINDING Vinpocetine treatment and its combination with enalapril decreased the glucose levels compared to diabetic rats. Vinpocetine improved the echocardiographic parameters and cardiac functional status of rats. Vinpocetine decreased the cardiac biochemical parameters, oxidative stress, inflammatory cytokine levels, cardiomyocyte diameter and fibrosis in rats. Interestingly, expressions of PDE-1, TGF-β and p-Smad 2/3 were ameliorated by vinpocetine alone and in combination with enalapril. CONCLUSIONS Vinpocetine is a well-known inhibitor of PDE-1 and the protective effect of vinpocetine in DC is exerted by inhibition of PDE-1 and subsequent inhibition of the expression of TGF-β/Smad 2/3.
Collapse
Affiliation(s)
| | - Sadia Shah
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Tajpreet Kaur
- Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Sudheer Kumar Arava
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Niraj Kumar
- Department of Neuroanesthesiogy and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Raj Kanwar Yadav
- Department Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Sushma Yadav
- Department of Obstetrics and Gynaecology, SHKM Government Medical College, Nuh, Haryana, India
| | - Taruna Arora
- RBMCH, ICMR-Head Quarter's Ansari Nagar, New Delhi, India
| | | |
Collapse
|
9
|
Salvador D, Bano A, Wehrli F, Gonzalez-Jaramillo V, Laimer M, Hunziker L, Muka T. Impact of type 2 diabetes on life expectancy and role of kidney disease among inpatients with heart failure in Switzerland: an ambispective cohort study. Cardiovasc Diabetol 2023; 22:174. [PMID: 37438747 DOI: 10.1186/s12933-023-01903-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is expected to worsen the prognosis of inpatients with heart failure (HF) but the evidence from observational studies is inconsistent. We aimed to compare mortality outcomes and life expectancy among inpatients with HF with or without T2D and explored whether chronic kidney disease (CKD) influenced these associations. METHODS We collected hospital and civil registry records of consecutive inpatients from a tertiary hospital in Switzerland with a diagnosis of HF from the year 2015 to 2019. We evaluated the association of T2D with mortality risk using Cox regression and adjusted for confounders. RESULTS Our final cohort consisted of 10,532 patients with HF of whom 27% had T2D. The median age (interquartile range [IQR]) was 75 [68 to 82] and 78 [68 to 86] for the diabetes and non-diabetes groups, respectively. Over a median follow-up [IQR] of 4.5 years [3.3 to 5.6], 5,347 (51%) of patients died. T2D patients had higher risk of all-cause mortality (hazard ratio [HR] 1.21, 95% confidence interval [CI] 1.14 to 1.29). Compared to control (i.e. no T2D nor CKD), average life expectancy (95% CI) among T2D patients, CKD, or both was shorter by 5.4 months (95% CI 1.1 to 9.7), 9.0 months (95% CI 8.4 to 9.6), or 14.8 months (95% CI 12.4 to 17.2), respectively. No difference by sex or ejection fraction category was observed. CONCLUSIONS T2D is associated with a significantly higher risk of all-cause mortality and shorter life expectancy compared to those without among middle-aged and elderly inpatients with HF; presence of CKD may further increase these risks.
Collapse
Affiliation(s)
- Dante Salvador
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Arjola Bano
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Faina Wehrli
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Valentina Gonzalez-Jaramillo
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Markus Laimer
- Department of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Inselspital, University of Bern, Bern, Switzerland
| | - Lukas Hunziker
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.
- Epistudia, Bern, Switzerland.
| |
Collapse
|
10
|
Saleem F, Mansour H, Vichare R, Ayalasomayajula Y, Yassine J, Hesaraghatta A, Panguluri SK. Influence of Age on Hyperoxia-Induced Cardiac Pathophysiology in Type 1 Diabetes Mellitus (T1DM) Mouse Model. Cells 2023; 12:1457. [PMID: 37296578 PMCID: PMC10252211 DOI: 10.3390/cells12111457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Mechanical ventilation often results in hyperoxia, a condition characterized by excess SpO2 levels (>96%). Hyperoxia results in changes in the physiological parameters, severe cardiac remodeling, arrhythmia development, and alteration of cardiac ion channels, all of which can point toward a gradual increase in the risk of developing cardiovascular disease (CVD). This study extends the analysis of our prior work in young Akita mice, which demonstrated that exposure to hyperoxia worsens cardiac outcomes in a type 1 diabetic murine model as compared to wild-type (WT) mice. Age is an independent risk factor, and when present with a major comorbidity, such as type 1 diabetes (T1D), it can further exacerbate cardiac outcomes. Thus, this research subjected aged T1D Akita mice to clinical hyperoxia and analyzed the cardiac outcomes. Overall, aged Akita mice (60 to 68 weeks) had preexisting cardiac challenges compared to young Akita mice. Aged mice were overweight, had an increased cardiac cross-sectional area, and showed prolonged QTc and JT intervals, which are proposed as major risk factors for CVD like intraventricular arrhythmias. Additionally, exposure to hyperoxia resulted in severe cardiac remodeling and a decrease in Kv 4.2 and KChIP2 cardiac potassium channels in these rodents. Based on sex-specific differences, aged male Akita mice had a higher risk of poor cardiac outcomes than aged females. Aged male Akita mice had prolonged RR, QTc, and JT intervals even at baseline normoxic exposure. Moreover, they were not protected against hyperoxic stress through adaptive cardiac hypertrophy, which, at least to some extent, is due to reduced cardiac androgen receptors. This study in aged Akita mice aims to draw attention to the clinically important yet understudied subject of the effect of hyperoxia on cardiac parameters in the presence of preexisting comorbidities. The findings would help revise the provision of care for older T1D patients admitted to ICUs.
Collapse
Affiliation(s)
- Faizan Saleem
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Hussein Mansour
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Riddhi Vichare
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Yashwant Ayalasomayajula
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Jenna Yassine
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Anagha Hesaraghatta
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Siva Kumar Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
- Cell Biology, Microbiology and Molecular Biology, College of Arts and Sciences, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| |
Collapse
|
11
|
Wang S, Tian C, Gao Z, Zhang B, Zhao L. Research status and trends of the diabetic cardiomyopathy in the past 10 years (2012–2021): A bibliometric analysis. Front Cardiovasc Med 2022; 9:1018841. [PMID: 36337893 PMCID: PMC9630656 DOI: 10.3389/fcvm.2022.1018841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022] Open
Abstract
Background Diabetic cardiomyopathy is one of the most life-threatening diabetic complications. However, the previous studies only discuss a particular aspect or characteristic of DCM, the current state and trends were explored by limited research. We aimed to perform a systemically bibliometric study of DCM research progress status in the past decade, visualize the internal conceptual structure and potential associations, and further explore the prospective study trends. Methods Articles related to DCM published from January 2012 to December 2021 were collected in the Web of Science core collection (WoSCC) database on June 24, 2022. We exported all bibliographic records, including titles, abstracts, keywords, authorship, institutions, addresses, publishing sources, references, citation times, and year of publication. In addition, the journal Impact Factor and Hirsch index were obtained from the Journal Citation Report. We conducted the data screening, statistical analysis, and visualization via the Bibliometrix R package. VOS viewer software was employed to generate the collaboration network map among countries and institutions for better performance in visualization. Results In total, 1,887 original research articles from 2012 to 2021 were identified. The number of annual publications rapidly increased from 107 to 278, and a drastic increase in citation times was observed in 2017–2019. As for global contributions, the United States was the most influential country with the highest international collaboration, while China was the most productive country. Professor Cai Lu was the most prolific author. Shandong University published the most articles. Cardiovascular Diabetology journal released the most DCM-related articles. “Metabolic Stress-induced Activation of FoxO1 Triggers Diabetic Cardiomyopathy in Mice” Battiprolu PK et al., J Clin Invest, 2012. was the most top-cited article regarding local citations. The top three keywords in terms of frequency were apoptosis, oxidative stress, and fibrosis. The analysis of future topic trends indicated that “Forkhead box protein O1,” “Heart failure with preserved ejection fraction,” “Dapagliflozin,” “Thioredoxin,” “Mitochondria dysfunction,” “Glucose,” “Pyroptosis,” “Cardiac fibroblast” and “Long non-coding RNA” could be promising hotspots. Conclusion This study provides meaningful insights into DCM, which is expected to assist cardiologists and endocrinologists in exploring frontiers and future research directions in the domain through a refined and concise summary.
Collapse
Affiliation(s)
- Sicheng Wang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanxi Tian
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zezheng Gao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Boxun Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Boxun Zhang,
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Linhua Zhao,
| |
Collapse
|
12
|
Rupee S, Rupee K, Singh RB, Hanoman C, Ismail AMA, Smail M, Singh J. Diabetes-induced chronic heart failure is due to defects in calcium transporting and regulatory contractile proteins: cellular and molecular evidence. Heart Fail Rev 2022; 28:627-644. [PMID: 36107271 DOI: 10.1007/s10741-022-10271-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 11/04/2022]
Abstract
Heart failure (HF) is a major deteriorating disease of the myocardium due to weak myocardial muscles. As such, the heart is unable to pump blood efficiently around the body to meet its constant demand. HF is a major global health problem with more than 7 million deaths annually worldwide, with some patients dying suddenly due to sudden cardiac death (SCD). There are several risk factors which are associated with HF and SCD which can negatively affect the heart synergistically. One major risk factor is diabetes mellitus (DM) which can cause an elevation in blood glucose level or hyperglycaemia (HG) which, in turn, has an insulting effect on the myocardium. This review attempted to explain the subcellular, cellular and molecular mechanisms and to a lesser extent, the genetic factors associated with the development of diabetes- induced cardiomyopathy due to the HG which can subsequently lead to chronic heart failure (CHF) and SCD. The study first explained the structure and function of the myocardium and then focussed mainly on the excitation-contraction coupling (ECC) processes highlighting the defects of calcium transporting (SERCA, NCX, RyR and connexin) and contractile regulatory (myosin, actin, titin and troponin) proteins. The study also highlighted new therapies and those under development, as well as preventative strategies to either treat or prevent diabetic cardiomyopathy (DCM). It is postulated that prevention is better than cure.
Collapse
|
13
|
Diabetes and Myocardial Fibrosis: A Systematic Review and Meta-Analysis. JACC. CARDIOVASCULAR IMAGING 2022; 15:796-808. [PMID: 35512952 DOI: 10.1016/j.jcmg.2021.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/23/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVES This systematic review and meta-analysis investigated the association of diabetes and glycemic control with myocardial fibrosis (MF). BACKGROUND MF is associated with an increased risk of heart failure, coronary artery disease, arrhythmias, and death. Diabetes may influence the development of MF, but evidence is inconsistent. METHODS The authors searched EMBASE, Medline Ovid, Cochrane CENTRAL, Web of Science, and Google Scholar for observational and interventional studies investigating the association of diabetes, glycemic control, and antidiabetic medication with MF assessed by histology and cardiac magnetic resonance (ie, extracellular volume fraction [ECV%] and T1 time). RESULTS A total of 32 studies (88% exclusively on type 2 diabetes) involving 5,053 participants were included in the systematic review. Meta-analyses showed that diabetes was associated with a higher degree of MF assessed by histological collagen volume fraction (n = 6 studies; mean difference: 5.80; 95% CI: 2.00-9.59) and ECV% (13 studies; mean difference: 2.09; 95% CI: 0.92-3.27), but not by native or postcontrast T1 time. Higher glycosylated hemoglobin levels were associated with higher degrees of MF. CONCLUSIONS Diabetes is associated with higher degree of MF assessed by histology and ECV% but not by T1 time. In patients with diabetes, worse glycemic control was associated with higher MF degrees. These findings mostly apply to type 2 diabetes and warrant further investigation into whether these associations are causal and which medications could attenuate MF in patients with diabetes.
Collapse
|
14
|
Dugbartey GJ, Wonje QL, Alornyo KK, Robertson L, Adams I, Boima V, Mensah SD. Combination Therapy of Alpha-Lipoic Acid, Gliclazide and Ramipril Protects Against Development of Diabetic Cardiomyopathy via Inhibition of TGF-β/Smad Pathway. Front Pharmacol 2022; 13:850542. [PMID: 35401218 PMCID: PMC8988231 DOI: 10.3389/fphar.2022.850542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/21/2022] [Indexed: 12/17/2022] Open
Abstract
Background: Diabetic cardiomyopathy (DCM) is a major long-term complication of diabetes mellitus, accounting for over 20% of annual mortality rate of diabetic patients globally. Although several existing anti-diabetic drugs have improved glycemic status in diabetic patients, prevalence of DCM is still high. This study investigates cardiac effect of alpha-lipoic acid (ALA) supplementation of anti-diabetic therapy in experimental DCM. Methods: Following 12 h of overnight fasting, 44 male Sprague Dawley rats were randomly assigned to two groups of healthy control (n = 7) and diabetic (n = 37) groups, and fasting blood glucose was measured. Type 2 diabetes mellitus (T2DM) was induced in diabetic group by intraperitoneal (i.p.) administration of nicotinamide (110 mg/kg) and streptozotocin (55 mg/kg). After confirmation of T2DM on day 3, diabetic rats received monotherapies with ALA (60 mg/kg; n = 7), gliclazide (15 mg/kg; n = 7), ramipril (10 mg/kg; n = 7) or combination of the three drugs (n = 7) for 6 weeks while untreated diabetic rats received distilled water and were used as diabetic control (n = 9). Rats were then sacrificed, and blood, pancreas and heart tissues were harvested for analyses using standard methods. Results: T2DM induction caused pancreatic islet destruction, hyperglycemia, weight loss, high relative heart weight, and development of DCM, which was characterized by myocardial degeneration and vacuolation, cardiac fibrosis, elevated cardiac damage markers (plasma and cardiac creatine kinase-myocardial band, brain natriuretic peptide and cardiac troponin I). Triple combination therapy of ALA, gliclazide and ramipril preserved islet structure, maintained body weight and blood glucose level, and prevented DCM development compared to diabetic control (p < 0.001). In addition, the combination therapy markedly reduced plasma levels of inflammatory markers (IL-1β, IL-6 and TNF-α), plasma and cardiac tissue malondialdehyde, triglycerides and total cholesterol while significantly increasing cardiac glutathione and superoxide dismutase activity and high-density lipoprotein-cholesterol compared to diabetic control (p < 0.001). Mechanistically, induction of T2DM upregulated cardiac expression of TGF-β1, phosphorylated Smad2 and Smad3 proteins, which were downregulated following triple combination therapy (p < 0.001). Conclusion: Triple combination therapy of ALA, gliclazide and ramipril prevented DCM development by inhibiting TGF-β1/Smad pathway. Our findings can be extrapolated to the human heart, which would provide effective additional pharmacological therapy against DCM in T2DM patients.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Quinsker L Wonje
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Karl K Alornyo
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Louis Robertson
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ismaila Adams
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Vincent Boima
- Department of Medicine and Therapeutics, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Samuel D Mensah
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
15
|
Xiao Y, Su C, Zhang G, Liang L, Jin T, Bradley J, Ornato JP, Tang W. Vitamin C Improves the Outcomes of Cardiopulmonary Resuscitation and Alters Shedding of Syndecan-1 and p38/MAPK Phosphorylation in a Rat Model. J Am Heart Assoc 2022; 11:e023787. [PMID: 35289183 PMCID: PMC9075447 DOI: 10.1161/jaha.121.023787] [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] [Indexed: 11/24/2022]
Abstract
Background Post‐resuscitation syndrome, involves a severe inflammatory response following successful cardiopulmonary resuscitation. The potential mechanism of Vitamin C (VitC) after cardiopulmonary resuscitation on myocardial and cerebral function, duration of survival is undefined. Methods and Results A first set of experiments were done in 18 male Sprague‐Dawley rats for the investigation of short‐term follow‐up, randomized into 3 groups: (1) sham; (2) controls; (3) VitC. Ventricular fibrillation was electrically induced and untreated for 6 minutes. Cardiopulmonary resuscitation including chest compression and mechanical ventilation were then initiated and continued for 8 minutes followed by defibrillation. At 5 minutes after return of spontaneous circulation, either VitC (200 mg/kg) or placebo was administered by intravenous infusion with a syringe pump for half an hour. There were significant improvements in myocardial function and buccal microcirculation in rats treated with VitC after return of spontaneous circulation 4 hours compared with controls. VitC inhibited proinflammatory cytokines (interleukin‐6 and tumor necrosis factor‐α), SDC‐1 (Syndecan‐1), and hyaluronic acid in plasma compared with controls (P<0.01). VitC decreased reactive oxygen species production and inhibited p38/MAPK (mitogen‐activated protein kinase) pathway phosphorylation. A second set with 20 animals was used for assessing the neurological deficit score after return of spontaneous circulation 72 hours, randomized into 2 groups: 1) controls; 2) VitC. The survival rate and neurological deficit score after return of spontaneous circulation 72 hours were improved in VitC‐treated animals compared with those of the control group. Conclusions VitC reduces the severity of post‐resuscitation myocardial and cerebral dysfunction and improves the survival. The mechanisms may involve inhibiting transcription of inflammatory cytokines and oxidative stress, thus protecting the integrity of the vascular endothelium. Meanwhile VitC reduces shedding of SDC‐1 and alters p38/MAPK phosphorylation and microcirculation.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Emergency and Critical Care Medicine The Second Affiliated Hospital of Soochow University Soochow China.,Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA
| | - Chenglei Su
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA.,Department of Emergency Medicine Center the Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu China
| | - Guozhen Zhang
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA
| | - Lian Liang
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA
| | - Tao Jin
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA
| | - Jennifer Bradley
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA
| | - Joseph P Ornato
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA.,Department of Emergency Medicine Virginia Commonwealth University Health System Richmond VA
| | - Wanchun Tang
- Weil Institute of Emergency and Critical Care ResearchVirginia Commonwealth University Richmond VA.,Department of Emergency Medicine Virginia Commonwealth University Health System Richmond VA
| |
Collapse
|
16
|
Gomes KP, Jadli AS, de Almeida LGN, Ballasy NN, Edalat P, Shandilya R, Young D, Belke D, Shearer J, Dufour A, Patel VB. Proteomic Analysis Suggests Altered Mitochondrial Metabolic Profile Associated With Diabetic Cardiomyopathy. Front Cardiovasc Med 2022; 9:791700. [PMID: 35310970 PMCID: PMC8924072 DOI: 10.3389/fcvm.2022.791700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Diabetic cardiomyopathy (DbCM) occurs independently of cardiovascular diseases or hypertension, leading to heart failure and increased risk for death in diabetic patients. To investigate the molecular mechanisms involved in DbCM, we performed a quantitative proteomic profiling analysis in the left ventricle (LV) of type 2 diabetic mice. Six-month-old C57BL/6J-lepr/lepr (db/db) mice exhibited DbCM associated with diastolic dysfunction and cardiac hypertrophy. Using quantitative shotgun proteomic analysis, we identified 53 differentially expressed proteins in the LVs of db/db mice, majorly associated with the regulation of energy metabolism. The subunits of ATP synthase that form the F1 domain, and Cytochrome c1, a catalytic core subunit of the complex III primarily responsible for electron transfer to Cytochrome c, were upregulated in diabetic LVs. Upregulation of these key proteins may represent an adaptive mechanism by diabetic heart, resulting in increased electron transfer and thereby enhancement of mitochondrial ATP production. Conversely, diabetic LVs also showed a decrease in peptide levels of NADH dehydrogenase 1β subcomplex subunit 11, a subunit of complex I that catalyzes the transfer of electrons to ubiquinone. Moreover, the atypical kinase COQ8A, an essential lipid-soluble electron transporter involved in the biosynthesis of ubiquinone, was also downregulated in diabetic LVs. Our study indicates that despite attempts by hearts from diabetic mice to augment mitochondrial ATP energetics, decreased levels of key components of the electron transport chain may contribute to impaired mitochondrial ATP production. Preserved basal mitochondrial respiration along with the markedly reduced maximal respiratory capacity in the LVs of db/db mice corroborate the association between altered mitochondrial metabolic profile and cardiac dysfunction in DbCM.
Collapse
Affiliation(s)
- Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Luiz G. N. de Almeida
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Pariya Edalat
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Ruchita Shandilya
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Daniel Young
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, Calgary, AB, Canada
- Department of Cardiac Sciences, Cumming School of Medicine, Calgary, AB, Canada
| | - Jane Shearer
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
- *Correspondence: Vaibhav B. Patel ;
| |
Collapse
|
17
|
Ding H, Yao J, Xie H, Wang C, Chen J, Wei K, Ji Y, Liu L. MicroRNA-195-5p Downregulation Inhibits Endothelial Mesenchymal Transition and Myocardial Fibrosis in Diabetic Cardiomyopathy by Targeting Smad7 and Inhibiting Transforming Growth Factor Beta 1-Smads-Snail Pathway. Front Physiol 2021; 12:709123. [PMID: 34658906 PMCID: PMC8514870 DOI: 10.3389/fphys.2021.709123] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a complication of diabetes mellitus, which is associated with fibrosis and microRNAs (miRs). This study estimated the mechanism of miR-195-5p in endothelial mesenchymal transition (EndMT) and myocardial fibrosis in DCM. After the establishment of DCM rat models, miR-195-5p was silenced by miR-195-5p antagomir. The cardiac function-related indexes diastolic left ventricular anterior wall (LVAW, d), systolic LVAW (d), diastolic left ventricular posterior wall (LVPW, d), systolic LVPW (d), left ventricular ejection fraction (LVEF), and fractional shortening (FS) were measured and miR-195-5p expression in myocardial tissue was detected. Myocardial fibrosis, collagen deposition, and levels of fibrosis markers were detected. Human umbilical vein endothelial cells (HUVECs) were exposed to high glucose (HG) and miR-195-5p was silenced. The levels of fibrosis proteins, endothelial markers, fibrosis markers, EndMT markers, and transforming growth factor beta 1 (TGF-β1)/Smads pathway-related proteins were measured in HUVECs. The interaction between miR-195-5p and Smad7 was verified. In vivo, miR-195-5p was highly expressed in the myocardium of DCM rats. Diastolic and systolic LVAW, diastolic and systolic LVPW were increased and LVEF and FS were decreased. Inhibition of miR-195-5p reduced cardiac dysfunction, myocardial fibrosis, collagen deposition, and EndMT, promoted CD31 and VE-cadehrin expressions, and inhibited α-SMA and vimentin expressions. In vitro, HG-induced high expression of miR-195-5p and the expression changes of endothelial markers CD31, VE-cadehrin and fibrosis markers α-SMA and vimentin were consistent with those in vivo after silencing miR-195-5p. In mechanism, miR-195-5p downregulation blocked EndMT by inhibiting TGF-β1-smads pathway. Smad7 was the direct target of miR-195-5p and silencing miR-195-5p inhibited EndMT by promoting Smad7 expression. Collectively, silencing miR-195-5p inhibits TGF-β1-smads-snail pathway by targeting Smad7, thus inhibiting EndMT and alleviating myocardial fibrosis in DCM.
Collapse
Affiliation(s)
- Huaisheng Ding
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Jianhui Yao
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Hongxiang Xie
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Chengyu Wang
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Jing Chen
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Kaiyong Wei
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Yangyang Ji
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| | - Lihong Liu
- Cardiovascular Department, Meishan People's Hospital, Meishan, China
| |
Collapse
|
18
|
El-Sayed N, Mostafa YM, AboGresha NM, Ahmed AAM, Mahmoud IZ, El-Sayed NM. Dapagliflozin attenuates diabetic cardiomyopathy through erythropoietin up-regulation of AKT/JAK/MAPK pathways in streptozotocin-induced diabetic rats. Chem Biol Interact 2021; 347:109617. [PMID: 34391751 DOI: 10.1016/j.cbi.2021.109617] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE This study was designed to investigate the mechanism of Dapagliflozin (Dapa) cardioprotection against diabetic cardiomyopathy (DCM). Structural and functional changes in the heart as well as decrease of erythropoietin (EPO) levels were reported in DCM. EPO simultaneously activates three pathways: the Janus-activated kinase-signal transducer and activator of transcription (JAK2/STAT5), phosphatidylinositol-3-kinase-Akt (PI3K/Akt), and extracellular signal-related kinase (ERK/MAPK) cascades, that result in proliferation and differentiation of cardiac cells. METHODS AND RESULTS DCM was induced by a high fat diet for 10 weeks followed by administration of streptozotocin. After confirmation of diabetes, rats were divided randomly to 5 groups: Group 1; normal control group, Group 2; untreated diabetic group and Groups (3-5); diabetic groups received Dapa daily (0.75 mg, 1.5 or 3 mg/Kg, p.o) respectively for a month. At the end of the experiment, full anaesthesia was induced in all rats using ether inhalation and ECG was recorded. Blood samples were collected then rats were sacrificed and their heart were dissected out and processed for biochemical and histopathological studies. Untreated diabetic rats showed abnormal ECG pattern, elevation of serum cardiac enzymes, decrease EPO levels, downregulation of P-Akt, P-JAK2 and pMAPK pathways, abnormal histological structure of the heart and increase immunostaining intensity of P53 and TNF α in the cardiomyocytes. Dapa in a dose dependent manner attenuated the alterations in the previously mentioned parameters. CONCLUSION The cardioprotective effect of Dapa could be mediated by increasing EPO levels and activation of P-Akt, P-JAK2 and pMAPK signalling cascades which in turn decrease apoptosis.
Collapse
Affiliation(s)
- Nora El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University, Badr, Egypt
| | - Noha M AboGresha
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Islam Z Mahmoud
- Department of Cardiovascular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
19
|
Târtea GC, Florescu DR, Mihailovici AR, Donoiu I, Istrătoaie O. Alpha-lipoic acid and vitamin B complex slow down the changes in mice diabetic cardiomyopathy. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 61:521-528. [PMID: 33544804 PMCID: PMC7864294 DOI: 10.47162/rjme.61.2.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aim: The aim of our study was to assess histologically and by cardiac ultrasound the effects of alpha-lipoic acid (ALA) and vitamin B complex, as pathogenic therapies, in diabetic cardiomyopathy (DCM) in mice. Materials and Methods: We performed an experimental animal study, in which we analyzed from a structural and functional point of view the changes produced in DCM. To produce DCM, we induced diabetes mellitus (DM) in C57BL/6 mice by intraperitoneal injection of a single 150 mg/kg body weight dose of streptozotocin (STZ). We formed a sham group (animals without DM), a control group (animals with DM but without treatment, DM_Control) and a group of animals with DM that were treated with ALA and vitamin B complex (DM_Treated). Results: At six weeks after STZ administration, there was no decrease in left ventricular ejection fraction (LVEF) in the sham group, while in the control group there was a significant decrease in LVEF, about 43.75±3.37%, compared to the group that received treatment with ALA and vitamin B complex, in which LVEF decreased to 49.6±5.02% (p=0.0432). Also, the degree of interstitial myocardial fibrosis was higher in animals with DM compared to animals without DM, but the applied therapeutic protocol considerably improved the accumulation of interstitial collagen. The same observation was maintained regarding the evaluation of polysaccharide deposits. Conclusions: We can say that the administration of ALA and vitamin B complex in mice with STZ-induced DM, improves the degree of myocardial fibrosis, the accumulation of polysaccharides, and prevents severe deterioration of systolic and diastolic function of the heart.
Collapse
|
20
|
Tuleta I, Frangogiannis NG. Fibrosis of the diabetic heart: Clinical significance, molecular mechanisms, and therapeutic opportunities. Adv Drug Deliv Rev 2021; 176:113904. [PMID: 34331987 PMCID: PMC8444077 DOI: 10.1016/j.addr.2021.113904] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/02/2023]
Abstract
In patients with diabetes, myocardial fibrosis may contribute to the pathogenesis of heart failure and arrhythmogenesis, increasing ventricular stiffness and delaying conduction. Diabetic myocardial fibrosis involves effects of hyperglycemia, lipotoxicity and insulin resistance on cardiac fibroblasts, directly resulting in increased matrix secretion, and activation of paracrine signaling in cardiomyocytes, immune and vascular cells, that release fibroblast-activating mediators. Neurohumoral pathways, cytokines, growth factors, oxidative stress, advanced glycation end-products (AGEs), and matricellular proteins have been implicated in diabetic fibrosis; however, the molecular links between the metabolic perturbations and activation of a fibrogenic program remain poorly understood. Although existing therapies using glucose- and lipid-lowering agents and neurohumoral inhibition may act in part by attenuating myocardial collagen deposition, specific therapies targeting the fibrotic response are lacking. This review manuscript discusses the clinical significance, molecular mechanisms and cell biology of diabetic cardiac fibrosis and proposes therapeutic targets that may attenuate the fibrotic response, preventing heart failure progression.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA.
| |
Collapse
|
21
|
Ding W, Feng H, Li WJ, Liao HH, Zhang N, Zhou ZY, Mou SQ, Lin Z, Xia-He NZ, Xia H, Tang QZ. Apocynin attenuates diabetic cardiomyopathy by suppressing ASK1-p38/JNK signaling. Eur J Pharmacol 2021; 909:174402. [PMID: 34348125 DOI: 10.1016/j.ejphar.2021.174402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022]
Abstract
Diabetic cardiomyopathy (DCM) significantly increased the morbidity of heart failure in diabetic patients. Long-time oxidative stress is an indisputable contributor for DCM development. Apocynin (APO) has been suggested to be a potential drug against oxidative stress. The study aims to find out the effects of APO on DCM and the related mechanisms. Mice were randomly divided into four groups: control (CON), APO, DCM and DCM + APO. Echocardiography analyses, histological analyses, Western blot and RT-PCR were used to explore the roles and mechanisms of APO in DCM. Isolated neonatal rat cardiomyocytes (NRCMs) and cardiac fibroblasts (CFs) were used for further confirming the APO treatment effects in vitro. Deteriorated cardiac function, enlarged cardiomyocytes, excess cardiac fibrosis and significant cardiac oxidative stress were observed in DCM group. However, APO treatment successfully improved cardiac function, decreased cardiac hypertrophy and fibrosis, and depressed oxidative stress. Mechanistically, APO treatment markedly suppressed apoptosis signal regulating kinase 1(ASK1)-p38/c-jun N-terminal kinase (JNK) signaling and reduced apoptosis. It also inhibited NRCM apoptosis and CF activation via depressing ASK1-p38/JNK signaling in vitro. Moreover, adenovirus-mediated ASK1 overexpression completely removed the protection of APO in vitro. In conclusion, APO treatment could effectively attenuate DCM-associated injuries in vivo and protect against high glucose-induced NRCM and CF injuries in vitro via suppressing ASK1-p38/JNK signaling. APO might be a potential ASK1 inhibitor for treating DCM.
Collapse
Affiliation(s)
- Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Hong Feng
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen-Jing Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Hai-Han Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zi-Ying Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Shan-Qi Mou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zheng Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Na-Zi Xia-He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| |
Collapse
|
22
|
MARSH SPENCER, RAUDAT MADELINE, LEFEBER BETHANY, HERNDON LAURABETH, HERBERT HOWARD, MCCALLUM LAURA, SIMIONESCU AGNETA. DYNAMIC BIOREACTOR MODEL TO MIMIC EARLY CARDIAC FIBROSIS IN DIABETES. J MECH MED BIOL 2021. [DOI: 10.1142/s0219519421500470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In clinical diabetic cardiomyopathy, hyperglycemia and dyslipidemia induce tissue injury, activation of cardiac fibroblasts and interstitial and perivascular fibrosis. Myofibroblasts repair the injured tissue by increasing collagen deposition in the cardiac interstitium and suppressing the activity of matrix metalloproteinases. The goal of this study was to find an ideal model to mimic the effect of high glucose concentration on human cardiac fibroblast activation. The profibrotic role of the transforming growth factor-[Formula: see text] (TGF-[Formula: see text]) and the protective modulation of nitric oxide were examined in two-dimensional and three-dimensional cell culture models, as well as tissue engineering models, that involved the use of cardiac fibroblasts cultured within myocardial matrix scaffolds mounted in a bioreactor that delivered biochemical and mechanical stimuli. Results showed that high glucose levels were potent pro-fibrotic stimuli. In addition, high glucose levels in concert with TGF-[Formula: see text] constituted very strong signals that induced human cardiac fibroblast activation. Cardiac fibroblasts cultured within decellularized myocardial scaffolds and exposed to biochemical and mechanical stimuli represented an adequate model for this pathology. In conclusion, the bioreactor platform was instrumental in establishing an in vitro model of early fibrosis; this platform could be used to test the effects of various agents targeted to mitigate the fibrotic processes.
Collapse
Affiliation(s)
- SPENCER MARSH
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| | - MADELINE RAUDAT
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| | - BETHANY LEFEBER
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| | - LAURA BETH HERNDON
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| | - HOWARD HERBERT
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| | - LAURA MCCALLUM
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| | - AGNETA SIMIONESCU
- Department of Bioengineering, Clemson University, 507 Rhodes Research Center, Clemson, SC 29634, USA
| |
Collapse
|
23
|
Gallego M, Zayas-Arrabal J, Alquiza A, Apellaniz B, Casis O. Electrical Features of the Diabetic Myocardium. Arrhythmic and Cardiovascular Safety Considerations in Diabetes. Front Pharmacol 2021; 12:687256. [PMID: 34305599 PMCID: PMC8295895 DOI: 10.3389/fphar.2021.687256] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetes is a chronic metabolic disease characterized by hyperglycemia in the absence of treatment. Among the diabetes-associated complications, cardiovascular disease is the major cause of mortality and morbidity in diabetic patients. Diabetes causes a complex myocardial dysfunction, referred as diabetic cardiomyopathy, which even in the absence of other cardiac risk factors results in abnormal diastolic and systolic function. Besides mechanical abnormalities, altered electrical function is another major feature of the diabetic myocardium. Both type 1 and type 2 diabetic patients often show cardiac electrical remodeling, mainly a prolonged ventricular repolarization visible in the electrocardiogram as a lengthening of the QT interval duration. The underlying mechanisms at the cellular level involve alterations on the expression and activity of several cardiac ion channels and their associated regulatory proteins. Consequent changes in sodium, calcium and potassium currents collectively lead to a delay in repolarization that can increase the risk of developing life-threatening ventricular arrhythmias and sudden death. QT duration correlates strongly with the risk of developing torsade de pointes, a form of ventricular tachycardia that can degenerate into ventricular fibrillation. Therefore, QT prolongation is a qualitative marker of proarrhythmic risk, and analysis of ventricular repolarization is therefore required for the approval of new drugs. To that end, the Thorough QT/QTc analysis evaluates QT interval prolongation to assess potential proarrhythmic effects. In addition, since diabetic patients have a higher risk to die from cardiovascular causes than individuals without diabetes, cardiovascular safety of the new antidiabetic drugs must be carefully evaluated in type 2 diabetic patients. These cardiovascular outcome trials reveal that some glucose-lowering drugs actually reduce cardiovascular risk. The mechanism of cardioprotection might involve a reduction of the risk of developing arrhythmia.
Collapse
Affiliation(s)
- Mónica Gallego
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Julián Zayas-Arrabal
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Amaia Alquiza
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Beatriz Apellaniz
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Oscar Casis
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| |
Collapse
|
24
|
Hussein AM, Eid EA, Bin-Jaliah I, Taha M, Lashin LS. Exercise and Stevia Rebaudiana (R) Extracts Attenuate Diabetic Cardiomyopathy in Type 2 Diabetic Rats: Possible Underlying Mechanisms. Endocr Metab Immune Disord Drug Targets 2021; 20:1117-1132. [PMID: 32310054 DOI: 10.2174/1871530320666200420084444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/31/2020] [Accepted: 02/19/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS In the current work, we studied the effects of exercise and stevia rebaudiana (R) extracts on diabetic cardiomyopathy (DCM) in type 2 diabetic rats and their possible underlying mechanisms. METHODS Thirty-two male Sprague Dawley rats were randomly allocated into 4 equal groups; a) normal control group, b) DM group, type 2 diabetic rats received 2 ml oral saline daily for 4 weeks, c) DM+ Exercise, type 2 diabetic rats were treated with exercise for 4 weeks and d) DM+ stevia R extracts: type 2 diabetic rats received methanolic stevia R extracts. By the end of the experiment, serum blood glucose, HOMA-IR, insulin and cardiac enzymes (LDH, CK-MB), cardiac histopathology, oxidative stress markers (MDA, GSH and CAT), myocardial fibrosis by Masson trichrome, the expression of p53, caspase-3, α-SMA and tyrosine hydroxylase (TH) by immunostaining in myocardial tissues were measured. RESULTS T2DM caused a significant increase in blood glucose, HOMA-IR index, serum CK-MB and LDH, myocardial damage and fibrosis, myocardial MDA, myocardial α-SMA, p53, caspase-3, Nrf2 and TH density with a significant decrease in serum insulin and myocardial GSH and CAT (p< 0.05). On the other hand, treatment with either exercise or stevia R extracts significantly improved all studied parameters (p< 0.05). Moreover, the effects of stevia R was more significant than exercise (p< 0.05). CONCLUSION Both exercise and methanolic stevia R extracts showed cardioprotective effects against DCM and Stevia R offered more cardioprotective than exercise. This cardioprotective effect of these lines of treatment might be due to attenuation of oxidative stress, apoptosis, sympathetic nerve density and fibrosis and upregulation of the antioxidant transcription factor, Nrf2.
Collapse
Affiliation(s)
- Abdelaziz M Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Elsayed A Eid
- Department of Internal Medicine and Endocrinology, Delta University for Science and Technology, Gamasa, Egypt
| | - Ismaeel Bin-Jaliah
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Medhat Taha
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Lashin S Lashin
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
25
|
Byrne NJ, Rajasekaran NS, Abel ED, Bugger H. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radic Biol Med 2021; 169:317-342. [PMID: 33910093 PMCID: PMC8285002 DOI: 10.1016/j.freeradbiomed.2021.03.046] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Even in the absence of coronary artery disease and hypertension, diabetes mellitus (DM) may increase the risk for heart failure development. This risk evolves from functional and structural alterations induced by diabetes in the heart, a cardiac entity termed diabetic cardiomyopathy (DbCM). Oxidative stress, defined as the imbalance of reactive oxygen species (ROS) has been increasingly proposed to contribute to the development of DbCM. There are several sources of ROS production including the mitochondria, NAD(P)H oxidase, xanthine oxidase, and uncoupled nitric oxide synthase. Overproduction of ROS in DbCM is thought to be counterbalanced by elevated antioxidant defense enzymes such as catalase and superoxide dismutase. Excess ROS in the cardiomyocyte results in further ROS production, mitochondrial DNA damage, lipid peroxidation, post-translational modifications of proteins and ultimately cell death and cardiac dysfunction. Furthermore, ROS modulates transcription factors responsible for expression of antioxidant enzymes. Lastly, evidence exists that several pharmacological agents may convey cardiovascular benefit by antioxidant mechanisms. As such, increasing our understanding of the pathways that lead to increased ROS production and impaired antioxidant defense may enable the development of therapeutic strategies against the progression of DbCM. Herein, we review the current knowledge about causes and consequences of ROS in DbCM, as well as the therapeutic potential and strategies of targeting oxidative stress in the diabetic heart.
Collapse
Affiliation(s)
- Nikole J Byrne
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
26
|
Barteková M, Adameová A, Görbe A, Ferenczyová K, Pecháňová O, Lazou A, Dhalla NS, Ferdinandy P, Giricz Z. Natural and synthetic antioxidants targeting cardiac oxidative stress and redox signaling in cardiometabolic diseases. Free Radic Biol Med 2021; 169:446-477. [PMID: 33905865 DOI: 10.1016/j.freeradbiomed.2021.03.045] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiometabolic diseases (CMDs) are metabolic diseases (e.g., obesity, diabetes, atherosclerosis, rare genetic metabolic diseases, etc.) associated with cardiac pathologies. Pathophysiology of most CMDs involves increased production of reactive oxygen species and impaired antioxidant defense systems, resulting in cardiac oxidative stress (OxS). To alleviate OxS, various antioxidants have been investigated in several diseases with conflicting results. Here we review the effect of CMDs on cardiac redox homeostasis, the role of OxS in cardiac pathologies, as well as experimental and clinical data on the therapeutic potential of natural antioxidants (including resveratrol, quercetin, curcumin, vitamins A, C, and E, coenzyme Q10, etc.), synthetic antioxidants (including N-acetylcysteine, SOD mimetics, mitoTEMPO, SkQ1, etc.), and promoters of antioxidant enzymes in CMDs. As no antioxidant indicated for the prevention and/or treatment of CMDs has reached the market despite the large number of preclinical and clinical studies, a sizeable translational gap is evident in this field. Thus, we also highlight potential underlying factors that may contribute to the failure of translation of antioxidant therapies in CMDs.
Collapse
Affiliation(s)
- Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia.
| | - Adriana Adameová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Kristína Ferenczyová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Oľga Pecháňová
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, 81371 Bratislava, Slovakia
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, And Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
27
|
DiNicolantonio JJ, McCarty MF, Barroso-Aranda J, Assanga S, Lujan LML, O'Keefe JH. A nutraceutical strategy for downregulating TGFβ signalling: prospects for prevention of fibrotic disorders, including post-COVID-19 pulmonary fibrosis. Open Heart 2021; 8:openhrt-2021-001663. [PMID: 33879509 PMCID: PMC8061562 DOI: 10.1136/openhrt-2021-001663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- James J DiNicolantonio
- Preventive Cardiology, Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | | | - Simon Assanga
- Department of Research and Postgraduate Studies in Food, University of Sonora, Sonora, Mexico
| | | | - James H O'Keefe
- University of Missouri-Kansas City, Saint Lukes Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
28
|
Thakur V, Alcoreza N, Delgado M, Joddar B, Chattopadhyay M. Cardioprotective Effect of Glycyrrhizin on Myocardial Remodeling in Diabetic Rats. Biomolecules 2021; 11:569. [PMID: 33924458 PMCID: PMC8069839 DOI: 10.3390/biom11040569] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 01/31/2023] Open
Abstract
Myocardial fibrosis is one of the major complications of long-term diabetes. Hyperglycemia induced cardiomyocyte atrophy is a frequent pathophysiological indicator of diabetic heart. The objective of this study was to investigate the cardioprotective effect of glycyrrhizin (GLC) on myocardial damage in diabetic rats and assess the anti-inflammatory and anti-fibrotic effect of GLC. Our study demonstrates that hyperglycemia can elevate cardiac atrophy in diabetic animals. Type 2 diabetic fatty and the lean control rats were evaluated for cardiac damage and inflammation at 8-12 weeks after the development of diabetes. Western blot and immunohistochemical studies revealed that gap junction protein connexin-43 (CX43), cardiac injury marker troponin I, cardiac muscle specific voltage gated sodium channel NaV1.5 were significantly altered in the diabetic heart. Furthermore, oxidative stress mediator receptor for advanced glycation end-products (RAGE), as well as inflammatory mediator phospho-p38 MAPK and chemokine receptor CXCR4 were increased in the diabetic heart whereas the expression of nuclear factor erythroid-2-related factor 2 (Nrf2), the antioxidant proteins that protect against oxidative damage was reduced. We also observed an increase in the expression of the pleiotropic cytokine, transforming growth factor beta (TGF-β) in the diabetic heart. GLC treatment exhibited a decrease in the expression of phospho-p38 MAPK, RAGE, NaV1.5 and TGF-β and it also altered the expression of CX43, CXCR4, Nrf2 and troponin I. These observations suggest that GLC possesses cardioprotective effects in diabetic cardiac atrophy and that these effects could be mediated through activation of Nrf2 and inhibition of CXCR4/SDF1 as well as TGF-β/p38MAPK signaling pathway.
Collapse
Affiliation(s)
- Vikram Thakur
- Center of Emphasis in Diabetes and Metabolism, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA;
| | - Narah Alcoreza
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA;
| | - Monica Delgado
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.D.); (B.J.)
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.D.); (B.J.)
| | - Munmun Chattopadhyay
- Center of Emphasis in Diabetes and Metabolism, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA;
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA;
| |
Collapse
|
29
|
Vivar R, Anfossi R, Humeres C, Catalán M, Reyes C, Cárdenas S, Contreras A, Aránguiz P, González F, Diaz-Araya G. FoxO1 is required for high glucose-dependent cardiac fibroblasts into myofibroblast phenoconversion. Cell Signal 2021; 83:109978. [PMID: 33722671 DOI: 10.1016/j.cellsig.2021.109978] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022]
Abstract
In the normal heart, cardiac fibroblasts (CFs) maintain extracellular matrix (ECM) homeostasis, whereas in pathological conditions, such as diabetes mellitus (DM), CFs converse into cardiac myofibroblasts (CMFs) and this CFs phenoconversion increase the synthesis and secretion of ECM proteins, promoting cardiac fibrosis and heart dysfunction. High glucose (HG) conditions increase TGF-β1 expression and FoxO1 activity, whereas FoxO1 is crucial to CFs phenoconversion induced by TGF-β1. In addition, FoxO1 increases CTGF expression, whereas CTGF plays an active role in the fibrotic process induced by hyperglycemia. However, the role of FoxO1 and CTGF in CFs phenoconversion induced by HG is not clear. In this study, we investigated the effects of FoxO1 pharmacological inhibition on CFs phenoconversion in both in vitro and ex vivo models of DM. Our results demonstrate that HG induces CFs phenoconversion and FoxO1 activation. Moreover, AS1842856, a pharmacological inhibitor of FoxO1 activity, prevents CFs phenoconversion and CTGF expression increase induced by HG, whereas these results were corroborated by FoxO1 silencing. Additionally, K252a, a pharmacological blocker of CTGF receptor, prevents HG-induced CFs phenoconversion, which was corroborated with CTGF expression knockdown. Furthermore, through CFs isolation from heart of diabetic rats, we showed that hyperglycemia induces FoxO1 activation, the increase of CTGF expression and CFs phenoconversion, whereas the FoxO1 activity inhibition reverses the effects induced by hyperglycemia on CFs. Altogether, our results demonstrate that FoxO1 and CTGF are necessary for CFs phenoconversion induced by HG and suggest that both proteins are likely to become a potential targeted drug for fibrotic response induced by hyperglycemic conditions.
Collapse
Affiliation(s)
- Raúl Vivar
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Renatto Anfossi
- Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudio Humeres
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Christopher Reyes
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Simone Cárdenas
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Contreras
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, 2520000 Viña del Mar, Chile
| | - Fabiola González
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Guillermo Diaz-Araya
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.; Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
30
|
Shao M, Lu L, Wang Q, Ma L, Tian X, Li C, Li C, Guo D, Wang Q, Wang W, Wang Y. The multi-faceted role of retinoid X receptor in cardiovascular diseases. Biomed Pharmacother 2021; 137:111264. [PMID: 33761589 DOI: 10.1016/j.biopha.2021.111264] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 01/14/2023] Open
Abstract
Retinoid X receptors (RXRs) are members of ligand-dependent transcription factors whose effects on a diversity of cellular processes, including cellular proliferation, the immune response, and lipid and glucose metabolism. Knock out of RXRα causes a hypoplasia of the myocardium which is lethal during fetal life. In addition, the heart maintains a well-orchestrated balances in utilizing fatty acids (FAs) and other substrates to meet the high energy requirements. As the master transcriptional regulators of lipid metabolism, RXRs become particularly important for the energy needs of the heart. Accumulating evidence suggested that RXRs may exert direct beneficial effects in the heart both through heterodimerization with other nuclear receptors (NRs) and homodimerization, thus standing as suitable targets for treating in cardiovascular diseases. Although compounds that target RXRs are promising drugs, their use is limited by toxicity. A better understanding of the structural biology of RXRs in cardiovascular disease should enable the rational design of more selective nuclear receptor modulators to overcome these problems. Here, this review summarizes a brief overview of RXRs structure and versatility of RXR action in the control of cardiovascular diseases. And we also discussed the therapeutic potential of RXR ligand.
Collapse
Affiliation(s)
- Mingyan Shao
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Linghui Lu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lin Ma
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue Tian
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Changxiang Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chun Li
- Modern Research Center of Traditional Chinese Medicine, School of Traditional Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dongqing Guo
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiyan Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yong Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China; College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
31
|
Forskolin Protected against Streptozotocin-Induced Diabetic Cardiomyopathy via Inhibition of Oxidative Stress and Cardiac Fibrosis in Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8881843. [PMID: 33564685 PMCID: PMC7867442 DOI: 10.1155/2021/8881843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 12/22/2022]
Abstract
Background Diabetic cardiomyopathy is one of the cardiac complications in diabetes patients, eventually resulting in heart failure and increasing morbidity and mortality. Oxidative stress is a critical pathological feature in diabetic hearts, contributing to the development of DCM. Forskolin (FSK) was shown to reduce oxidative stress. This study was aimed at investigating the effects of FSK on diabetic hearts and the relevant molecular mechanisms. Methods Streptozotocin- (STZ-) induced diabetes in mice was treated with FSK through intraperitoneal injection. Cardiac functions were evaluated by echocardiography. Hematoxylin-eosin and Masson trichrome staining was employed to determine heart morphological changes and cardiac fibrosis, respectively. Cardiac fibrosis-related markers were detected by western blot. Superoxide dismutase activity, reduced/oxidized glutathione ratio, and malondialdehyde concentration in left ventricles were determined using respective commercial kits. Results Abnormal cardiac diastolic dysfunction and cardiac fibrosis were observed in diabetic hearts. FSK treatment significantly improved the cardiac diastolic function and attenuated the abnormal morphological change in diabetic hearts. Moreover, FSK treatment in diabetic mice decreased the expression of fibronectin, collagen I, TGF-β, and α-SMA and reduced myocardial fibrosis. Furthermore, we observed that FSK significantly blocked oxidative stress in diabetic hearts. Conclusions Our study demonstrates that FSK protects against the development of DCM in STZ-induced diabetes in mice. Our study suggests that FSK might be a potential target for drug development in treating DCM.
Collapse
|
32
|
Identification and analysis of circulating long non-coding RNAs with high significance in diabetic cardiomyopathy. Sci Rep 2021; 11:2571. [PMID: 33510471 PMCID: PMC7843621 DOI: 10.1038/s41598-021-82345-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/19/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) lacks diagnostic biomarkers. Circulating long non-coding RNAs (lncRNAs) can serve as valuable diagnostic biomarkers in cardiovascular disease. To seek potential lncRNAs as a diagnostic biomarker for DCM, we investigated the genome-wide expression profiling of circulating lncRNAs and mRNAs in type 2 diabetic db/db mice with and without DCM and performed bioinformatic analyses of the deregulated lncRNA-mRNA co-expression network. Db/db mice had obesity and hyperglycemia with normal cardiac function at 6 weeks of age (diabetes without DCM) but with an impaired cardiac function at 20 weeks of age (DCM) on an isolated Langendorff apparatus. Compared with the age-matched controls, 152 circulating lncRNAs, 127 mRNAs and 3355 lncRNAs, 2580 mRNAs were deregulated in db/db mice without and with DCM, respectively. The lncRNA-mRNA co-expression network analysis showed that five deregulated lncRNAs, XLOC015617, AK035192, Gm10435, TCR-α chain, and MouselincRNA0135, have the maximum connections with differentially expressed mRNAs. Bioinformatic analysis revealed that these five lncRNAs were highly associated with the development and motion of myofilaments, regulation of inflammatory and immune responses, and apoptosis. This finding was validated by the ultrastructural examination of myocardial samples from the db/db mice with DCM using electron microscopy and changes in the expression of myocardial tumor necrosis factor-α and phosphorylated p38 mitogen-activated protein kinase in db/db mice with DCM. These results indicate that XLOC015617, AK035192, Gm10435, TCR-α chain, and MouselincRNA0135 are crucial circulating lncRNAs in the pathogenesis of DCM. These five circulating lncRNAs may have high potential as a diagnostic biomarker for DCM.
Collapse
|
33
|
Al-Ani B, Alzamil NM, Hewett PW, Al-Hashem F, Bin-Jaliah I, Shatoor AS, Kamar SS, Latif NSA, Haidara MA, Dawood AF. Metformin ameliorates ROS-p53-collagen axis of fibrosis and dyslipidemia in type 2 diabetes mellitus-induced left ventricular injury. Arch Physiol Biochem 2021; 129:734-740. [PMID: 33439743 DOI: 10.1080/13813455.2020.1869265] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The link between oxidative stress (ROS), apoptosis (p53) and fibrosis (collagen) in type 2 diabetes mellitus (T2DM)-induced cardiac injury in the presence and absence of the antidiabetic drug, metformin has not been investigated before. MATERIAL AND METHODS T2DM was induced in rats by a combination of high carbohydrate and fat diets (HCFD) and streptozotocin (50 mg/kg) injection. The protection group started metformin (200 mg/kg) treatment 14 days prior to the induction of diabetes and continued on metformin and HCFD until being sacrificed at week 12. RESULTS Diabetes significantly induced blood levels of ROS and left ventricular p53 and collagen expression that was inhibited by metformin. Metformin also significantly reduced glycated haemoglobin and dyslipidemia induced by diabetes. In addition, a significant correlation between ROS-p53-collagen axis and glycaemia and hyperlipidaemia was observed. CONCLUSIONS These findings show that metformin provides substantial protection against diabetic cardiomyopathy-induced ROS-p53 mediated fibrosis and dyslipidemia.
Collapse
Affiliation(s)
- Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Norah M Alzamil
- Department of Clinical Science, Family Medicine, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Peter W Hewett
- Institute of Cardiovascular Sciences, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Fahaid Al-Hashem
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ismaeel Bin-Jaliah
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abdullah S Shatoor
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Samaa S Kamar
- Departments of Medical Histology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noha S Abdel Latif
- Department of Medical Pharmacology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed A Haidara
- Department of Physiology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Amal F Dawood
- Department of Physiology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Li Y, Cao Z, Li Q, Wang C, Zhou Z. Effects of Dendrobium Polysaccharides on the Functions of Human Skin Fibroblasts and Expression of Matrix Metalloproteinase-2 under High-Glucose Conditions. Int J Endocrinol 2021; 2021:1092975. [PMID: 33777140 PMCID: PMC7969111 DOI: 10.1155/2021/1092975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
The effects of Dendrobium polysaccharides (PDC) on the functions of human skin fibroblasts (HSFs) and expression of matrix metalloproteinase-2 under high-glucose conditions and exploration of the underlying mechanism remain unclear. We used the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) analysis and flow cytometry to evaluate the cell viability and apoptosis. The collagen levels were determined by the Sircol™ Collagen Assay. Real-time quantitative polymerase chain reaction (RT-PCR) was used to detect the expression of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase inhibitor (TIMP-2) mRNA. We found the following: (1) under the high-glucose condition, the HSF cell viability, the expression of TIMP-2 mRNA, and the collagen levels were reduced, while the apoptosis rate and the expression of MMP-2 mRNA increased (P < 0.05). (2) In the high-glucose + PDC group, the PDC reversed the changes in the collagen level, viability, and apoptosis rate of the HSF cells caused by high glucose, with the expression of protein and TIMP-2 mRNA increased and the level of MMP-2 mRNA decreased (P < 0.05). This is the first time attempting to reveal that PDC can exhibit protective effects on HSF under high-glucose conditions, which may be related to the upregulation of the TIMP-2 expression and inhibition of the MMP-2 expression.
Collapse
Affiliation(s)
- Yajia Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqin Cao
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiangxiang Li
- National Clinical Research Center for Geriatric Disorders of Xiangya Hospital, Central South University (Sub-Center of Ningxia), Yinchuan, Ningxia Hui Autonomous Region 750001, China
- Ningxia Geriatric Disease Clinical Research Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, China
- Hunan People's Hospital, Department of Hunan Institute of Geriatrics, Changsha 410002, China
| | - Chenxu Wang
- Hunan People's Hospital, Department of Hunan Institute of Geriatrics, Changsha 410002, China
| | - Zhuo Zhou
- Hunan People's Hospital, Department of Hunan Institute of Geriatrics, Changsha 410002, China
| |
Collapse
|
35
|
Hong T, Wei Y, Xue X, Li Y, Dong H, Guo X, Shi X, He B. A Novel Anti-Coagulative Nanocomplex in Delivering miRNA-1 Inhibitor Against Microvascular Obstruction of Myocardial Infarction. Adv Healthc Mater 2020; 9:e1901783. [PMID: 32338452 DOI: 10.1002/adhm.201901783] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/09/2020] [Indexed: 12/17/2022]
Abstract
Great progress has been made in miRNA nanodelivery for the treatment of myocardial infarction (MI). However, miRNA nanodelivery within the infarct is impeded by microvascular obstruction as a local circulatory disorder caused by microthrombus formation in microvessels. Knowing that low molecular weight heparin (LMWH) can effectively prevent microthrombus formation in microcirculation, it is hypothesized whether surface modification of the nanocarrier with LMWH can overcome microvascular obstruction in the infarct area for better miRNA delivery. Herein, a novel nanocomlex consisting of dendrigraft poly-l-lysine (DGL)-loaded miR-1 inhibitor as the core to decrease apoptosis of cardiomyocytes and LMWH as the shell to overcome microvascular obstruction of the infarct area is developed. The results show that this anti-coagulative nanocomlex is able to reduce microthrombus formation in microvessels and inhibit blood-coagulation factor Xa, thereby overcoming microvascular obstruction in the infarct area. In addition, it further enhances the uptake of miR-1 inhibitor within the infarct and decreases myocardiocyte apoptosis, thus improving the cardiac function and attenuating the myocardial fibrosis. In conclusion, modification of DGL-loaded miR-1 inhibitor with LMWH helps overcome microvascular obstruction in delivering the drug to the infarct area, thus providing a promising therapeutic strategy for achieving a better therapeutic outcome of MI.
Collapse
Affiliation(s)
- Ting Hong
- Department of Anesthesiology and SICUXinhua HospitalSchool of Medicine Shanghai Jiao Tong University Shanghai 200092 China
| | - Yazhong Wei
- Department of Anesthesiology and SICUXinhua HospitalSchool of Medicine Shanghai Jiao Tong University Shanghai 200092 China
| | - Xiaomei Xue
- Department of Anesthesiology and SICUXinhua HospitalSchool of Medicine Shanghai Jiao Tong University Shanghai 200092 China
| | - Yongyong Li
- Institute for Biomedical Engineering & Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Haiqing Dong
- Institute for Biomedical Engineering & Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Xiaoyu Guo
- Department of Anesthesiology and SICUXinhua HospitalSchool of Medicine Shanghai Jiao Tong University Shanghai 200092 China
| | - Xueyin Shi
- Department of Anesthesiology and SICUXinhua HospitalSchool of Medicine Shanghai Jiao Tong University Shanghai 200092 China
| | - Bin He
- Department of Anesthesiology and SICUXinhua HospitalSchool of Medicine Shanghai Jiao Tong University Shanghai 200092 China
| |
Collapse
|
36
|
Diabetic cardiomyopathy: molecular mechanisms, detrimental effects of conventional treatment, and beneficial effects of natural therapy. Heart Fail Rev 2020; 24:279-299. [PMID: 30349977 DOI: 10.1007/s10741-018-9749-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTARCT Diabetic complications are among the largely exigent health problems currently. Cardiovascular complications, including diabetic cardiomyopathy (DCM), account for more than 80% of diabetic deaths. Investigators are exploring new therapeutic targets to slow or abate diabetes because of the growing occurrence and augmented risk of deaths due to its complications. Research on rodent models of type 1 and type 2 diabetes mellitus, and the use of genetic engineering techniques in mice and rats have significantly sophisticated for our understanding of the molecular mechanisms in human DCM. DCM is featured by pathophysiological mechanisms that are hyperglycemia, insulin resistance, oxidative stress, left ventricular hypertrophy, damaged left ventricular systolic and diastolic functions, myocardial fibrosis, endothelial dysfunction, myocyte cell death, autophagy, and endoplasmic reticulum stress. A number of molecular and cellular pathways, such as cardiac ubiquitin proteasome system, FoxO transcription factors, hexosamine biosynthetic pathway, polyol pathway, protein kinase C signaling, NF-κB signaling, peroxisome proliferator-activated receptor signaling, Nrf2 pathway, mitogen-activated protein kinase pathway, and micro RNAs, play a major role in DCM. Currently, there are a few drugs for the management of DCM and some of them have considerable adverse effects. So, researchers are focusing on the natural products to ameliorate it. Hence, in this review, we discuss the pathogical, molecular, and cellular mechanisms of DCM; the current diagnostic methods and treatments; adverse effects of conventional treatment; and beneficial effects of natural product-based therapeutics, which may pave the way to new treatment strategies. Graphical Abstract.
Collapse
|
37
|
The Beneficial Effects of Alpha Lipoic Acid Supplementation on Lp-PLA2 Mass and Its Distribution between HDL and apoB-Containing Lipoproteins in Type 2 Diabetic Patients: A Randomized, Double-Blind, Placebo-Controlled Trial. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5850865. [PMID: 32256955 PMCID: PMC7085885 DOI: 10.1155/2020/5850865] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/11/2020] [Indexed: 02/06/2023]
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) is a new specific vascular inflammation biomarker that is carried by the lipoproteins in the blood and plays a prominent role in the pathogenesis of atherosclerosis. Increased Lp-PLA2 levels and impaired Lp-PLA2 distribution across high-density lipoprotein (HDL) and non-HDL lipoproteins have been reported in diabetic patients, which is associated with the increase in cardiovascular disease (CVD) risk. This study is aimed at investigating the effect of alpha lipoic acid (ALA), as an antioxidant with potential cardioprotective properties, on the Lp-PLA2 mass and its distribution in diabetic patients. In a double-blind, randomized, placebo-controlled clinical trial, seventy diabetic patients were randomly allocated to ALA (1200 mg ALA as two 600 mg capsules/day) and placebo (two maltodextrin capsules/day) groups. The serum levels of total Lp-PLA2 mass, HDL-Lp-PLA2, oxidized low-density lipoproteins (ox-LDL), apolipoprotein A1 (apo A1), lipid profiles, fasting blood sugar (FBS), and insulin were measured, and apolipoprotein B- (apoB-) associated Lp-PLA2 and homeostasis model of assessment index (HOMA-IR) were calculated at the baseline and after 8 weeks of intervention. ALA significantly decreased the ox-LDL, total Lp-PLA2 mass, apoB-associated Lp-PLA2, and percent of apoB-associated Lp-PLA2 and triglyceride and increased the percent of HDL-Lp-PLA2 compared with the placebo group but had no significant effect on HDL-Lp-PLA2 mass, apo A1, lipid profiles, and glycemic indices. There was a positive correlation between the reduction in the ox-LDL level and total Lp-PLA2 mass in the ALA group. In conclusion, ALA may decrease the CVD risk by reducing the ox-LDL and Lp-PLA2 mass and improving the Lp-PLA2 distribution among lipoproteins in type 2 diabetic patients.
Collapse
|
38
|
Comparative Study of the Effects of GLP1 Analog and SGLT2 Inhibitor against Diabetic Cardiomyopathy in Type 2 Diabetic Rats: Possible Underlying Mechanisms. Biomedicines 2020; 8:biomedicines8030043. [PMID: 32106580 PMCID: PMC7175346 DOI: 10.3390/biomedicines8030043] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/27/2020] [Accepted: 01/31/2020] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the possible cardioprotective effects of GLP1 and SGLT2i against diabetic cardiomyopathy (DCM) in type 2 diabetic rats and the possible underlying mechanisms. METHODS Thirty-two male Sprague Dawley rats were randomly subdivided into 4 equal groups: a) control group, b) DM group, type 2 diabetic rats with saline daily for 4 weeks, c) DM+ GLP1, as DM group with GLP1 analogue (liraglutide) at a dose of 75 µg/kg for 4 weeks, and d) DM+ SGLT2i as DM group with SGLT2 inhibitor (dapagliflozin) at a dose of 1mg/kg for 4 weeks. By the end of treatment (4 weeks), serum blood glucose, homeostasis model assessment insulin resistance (HOMA-IR), insulin, and cardiac enzymes (LDH, CK-MB) were measured. Also, the cardiac histopathology, myocardial oxidative stress markers (malondialdehyde (MDA), glutathione (GSH) and CAT) and norepinephrine (NE), myocardial fibrosis, the expression of caspase-3, TGF-β, TNF-α, and tyrosine hydroxylase (TH) in myocardial tissues were measured. RESULTS T2DM caused significant increase in serum glucose, HOMA-IR, serum CK-MB, and LDH (p < 0.05). Also, DM caused significant myocardial damage and fibrosis; elevation of myocardial MDA; NE with upregulation of myocardial caspase-3, TNF-α, TGF-β, and TH; and significant decrease in serum insulin and myocardial GSH and CAT (p < 0.05). Administration of either GLP1 analog or SGLT2i caused a significant improvement in all studied parameters (p < 0.05). CONCLUSION We concluded that both GLP1 and SGLT2i exhibited cardioprotective effects against DCM in T2DM, with the upper hand for SGLT2i. This might be due to attenuation of fibrosis, oxidative stress, apoptosis (caspase-3), sympathetic nerve activity, and inflammatory cytokines (TNF-α and TGF-β).
Collapse
|
39
|
Li XL, Yu F, Li BY, Fu CL, Yu X, Xu M, Cheng M, Gao HQ. The protective effects of grape seed procyanidin B2 against asporin mediates glycated low-density lipoprotein induced-cardiomyocyte apoptosis and fibrosis. Cell Biol Int 2020; 44:268-277. [PMID: 31498521 DOI: 10.1002/cbin.11229] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/31/2019] [Indexed: 01/24/2023]
Abstract
The progression of diabetic cardiomyopathy is related to cardiomyocyte dysfunction and apoptosis. Our previous studies showed that asporin (ASPN) was significantly increased in the myocardium of db/db mice through proteomics, and grape seed procyanidin B2 (GSPB2) significantly inhibited the expression of ASPN in the heart of db/db mice. We report here that ASPN played a critical role in glycated low-density lipoproteins (gly-LDL) induced-cardiomyocyte apoptosis. We found that gly-LDL upregulated ASPN expression. ASPN increased H9C2 cardiomyocyte apoptosis with down-regulation of Bcl-2, upregulation of transforming growth factor-β1, Bax, collagen III, fibronectin, and phosphorylation of smad2 and smad3. However, GSPB2 treatment reversed ASPN-induced impairments in H9C2 cardiomyocytes. These results provide evidence for the cardioprotective action of GSPB2 against ASPN injury, and thus suggest a new target for fighting against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Drug Purchase and Supply, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Fei Yu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Bao-Ying Li
- Department of Geriatric Medicine, Bai-Ren Hospital of Weinan, Middle Section of Letian Street, Weinan, Shanxi Province, 714000, China
| | - Chun-Li Fu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Xin Yu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Mei Xu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Mei Cheng
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| | - Hai-Qing Gao
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong Province, 250012, China
| |
Collapse
|
40
|
Bilska-Wilkosz A, Kotańska M, Górny M, Filipek B, Iciek M. Can Lipoic Acid Attenuate Cardiovascular Disturbances Induced by Ethanol and Disulfiram Administration Separately or Jointly in Rats? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1974982. [PMID: 31885774 PMCID: PMC6893278 DOI: 10.1155/2019/1974982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/24/2019] [Accepted: 11/02/2019] [Indexed: 11/27/2022]
Abstract
The exogenous lipoic acid (LA) is successfully used as a drug in the treatment of many diseases. It is assumed that after administration, LA is transported to the intracellular compartments and reduced to dihydrolipoic acid (DHLA) which is catalyzed by NAD(P)H-dependent enzymes. The purpose of this study was to investigate whether LA can attenuate cardiovascular disturbances induced by ethanol (EtOH) and disulfiram (DSF) administration separately or jointly in rats. For this purpose, we measured systolic and diastolic blood pressure, recorded electrocardiogram (ECG), and estimated mortality of rats. We also studied the activity of aldehyde dehydrogenase (ALDH) in the rat liver. It was shown for the first time that LA partially attenuated the cardiac arrhythmia (extrasystoles and atrioventricular blocks) induced by EtOH and reduced the EtOH-induced mortality of animals, which suggests that LA may have a potential for use in cardiac disturbance in conditions of acute EtOH intoxication. The administration of EtOH, LA, and DSF separately or jointly affected the ALDH activity in the rat liver since a significant decrease in the activity of the enzyme was observed in all treatment groups. The results indicating that LA is an inhibitor of ALDH activity are very surprising.
Collapse
Affiliation(s)
- Anna Bilska-Wilkosz
- Chair of Medical Biochemistry, Jagiellonian University, Medical College, 7 Kopernika Street, PL 31-034 Kraków, Poland
| | - Magdalena Kotańska
- Department of Pharmacodynamics, Jagiellonian University, Medical College, 9 Medyczna Street, PL 30-688 Kraków, Poland
| | - Magdalena Górny
- Chair of Medical Biochemistry, Jagiellonian University, Medical College, 7 Kopernika Street, PL 31-034 Kraków, Poland
| | - Barbara Filipek
- Department of Pharmacodynamics, Jagiellonian University, Medical College, 9 Medyczna Street, PL 30-688 Kraków, Poland
| | - Małgorzata Iciek
- Chair of Medical Biochemistry, Jagiellonian University, Medical College, 7 Kopernika Street, PL 31-034 Kraków, Poland
| |
Collapse
|
41
|
Pınar N, Çakırca G, Hakverdi S, Kaplan M. Protective effect of alpha lipoic acid on cisplatin induced hepatotoxicity in rats. Biotech Histochem 2019; 95:219-224. [DOI: 10.1080/10520295.2019.1667025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Neslihan Pınar
- Department of Medical Pharmacology, School of Medicine, Mustafa Kemal University, Hatay, Turkey
| | - Gökhan Çakırca
- Department of Biochemistry, Sanliurfa Mehmet Akif Inan Training and Research Hospital, Sanliurfa, Turkey
| | - Sibel Hakverdi
- Department of Pathology, School of Medicine, Mustafa Kemal University, Hatay, Turkey
| | - Mahir Kaplan
- Department of Medical Pharmacology, School of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
42
|
Wang SQ, Li D, Yuan Y. Long-term moderate intensity exercise alleviates myocardial fibrosis in type 2 diabetic rats via inhibitions of oxidative stress and TGF-β1/Smad pathway. J Physiol Sci 2019; 69:861-873. [PMID: 31392590 PMCID: PMC10716963 DOI: 10.1007/s12576-019-00696-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/15/2019] [Indexed: 12/25/2022]
Abstract
Exercise has an effect on the reduction of myocardial fibrosis in diabetic rats as previously reported, in which oxidative stress and the TGF-β1/Smad signaling pathway may play key roles. There is little direct experimental evidence that exercise alleviates myocardial fibrosis in type 2 diabetes mellitus (T2DM). Here we established a type 2 diabetic model by using streptozotocin and a high-fat diet. Rats were divided into groups of normal control (NC), T2DM and T2DM plus exercise (T2DME). The T2DME group received further treadmill training at moderate intensity for 8 weeks. Histological and biochemical methods were used to detect the benefits of exercise to T2DM. Results showed that the weight of rats in the T2DM group dropped dramatically, along with significant increases in blood glucose, myocardial fibrosis and oxidative stress, associated with upregulated expression of factors of myocardial fibrosis, except Smad7. Exercise largely reversed T2DM-induced alterations in factors of myocardial fibrosis, including suppressing expression of MMP-2, CTGF, TGF-β1, p-Smad2 and p-Smad3, and increased expression of TIMP-1 and Smad7. Therefore, exercise might be considered an alternative therapeutic remedy for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shi-Qiang Wang
- Physical Education College, Hunan University of Technology, Zhuzhou, Hunan, China
| | - Dan Li
- Physical Education College, Hunan University of Technology, Zhuzhou, Hunan, China
| | - Yang Yuan
- School of Physical Education, Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, China.
| |
Collapse
|
43
|
Li H, Shi Y, Wang X, Li P, Zhang S, Wu T, Yan Y, Zhan Y, Ren Y, Rong X, Xia T, Chu M, Wu R. Piceatannol alleviates inflammation and oxidative stress via modulation of the Nrf2/HO-1 and NF-κB pathways in diabetic cardiomyopathy. Chem Biol Interact 2019; 310:108754. [PMID: 31323227 DOI: 10.1016/j.cbi.2019.108754] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/04/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
Diabetic cardiomyopathy (DCM) is one of the leading causes of morbidity and mortality in diabetic patients. Piceatannol (PIC) has protective effects against cardiovascular disease; however, it remains unknown whether it also protects against DCM. A Cell Counting Kit-8 (CCK-8) assay was used to evaluate the effects of PIC on the viability of high glucose (HG)-induced H9C2 cells. Protein expression and mRNA levels were detected by western blotting and real-time polymerase chain reaction (RT-PCR), respectively. In vivo, physical and biochemical analyses, together with transthoracic echocardiography and hemodynamic measurements, were used to detect the effects of PIC treatment on cardiac function in DCM rats. Reactive oxygen species production was determined using an ELISA kit, and inflammatory cytokines were detected by RT-PCR. Pathological changes were assessed by hematoxylin-eosin staining, immunohistochemical staining, and TUNEL staining. According to the results, PIC treatment improved cell viability and inhibited cell apoptosis in HG-induced H9C2 cardiac myoblasts. In addition, PIC not only attenuated the over-production of interleukin-6 (IL-6) (P < 0.05) and tumor necrosis factor alpha (TNF-α) (P < 0.05), but also improved the expression of nuclear factor E2-related factor 2 (Nrf2) (P < 0.05) and heme oxygenase-1 (HO-1) (P < 0.01). Importantly, knockdown of Nrf2 suppressed PIC-mediated activation of the Nrf2/HO-1 pathway and abolished its anti-inflammatory effects. In vivo, oral administration of PIC suppressed STZ-induced inflammation, oxidative stress hypertrophy, fibrosis(myocardial collagen volume fraction in 5 mg/kg and 10 mg/kg PIC group was decreased 25.83% and 55.61% compared with the DM group), and apoptosis(Caspase-3 level in 5 mg/kg and 10 mg/kg PIC group was decreased 13.21% and 33.91% compared with the DM group), thereby relieving cardiac dysfunction and improving both fibrosis and pathological changes in cardiac tissues of diabetic rats. These findings define for the first time that the effects of PIC against DCM can be attributed to its role in inflammation and oxidative stress inhibition.
Collapse
Affiliation(s)
- Hao Li
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Youyang Shi
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Xuliang Wang
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Ping Li
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Songyue Zhang
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Yaoyao Yan
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Yi Zhan
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Yue Ren
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Xing Rong
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Tianhe Xia
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China
| | - Maoping Chu
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China.
| | - Rongzhou Wu
- Children's Heart Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325027, China.
| |
Collapse
|
44
|
Kronlage M, Dewenter M, Grosso J, Fleming T, Oehl U, Lehmann LH, Falcão-Pires I, Leite-Moreira AF, Volk N, Gröne HJ, Müller OJ, Sickmann A, Katus HA, Backs J. O-GlcNAcylation of Histone Deacetylase 4 Protects the Diabetic Heart From Failure. Circulation 2019; 140:580-594. [PMID: 31195810 DOI: 10.1161/circulationaha.117.031942] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Worldwide, diabetes mellitus and heart failure represent frequent comorbidities with high socioeconomic impact and steadily growing incidence, calling for a better understanding of how diabetic metabolism promotes cardiac dysfunction. Paradoxically, some glucose-lowering drugs have been shown to worsen heart failure, raising the question of how glucose mediates protective versus detrimental cardiac signaling. Here, we identified a histone deacetylase 4 (HDAC4) subdomain as a molecular checkpoint of adaptive and maladaptive signaling in the diabetic heart. METHODS A conditional HDAC4 allele was used to delete HDAC4 specifically in cardiomyocytes (HDAC4-knockout). Mice were subjected to diabetes mellitus either by streptozotocin injections (type 1 diabetes mellitus model) or by crossing into mice carrying a leptin receptor mutation (db/db; type 2 diabetes mellitus model) and monitored for remodeling and cardiac function. Effects of glucose and the posttranslational modification by β-linked N-acetylglucosamine (O-GlcNAc) on HDAC4 were investigated in vivo and in vitro by biochemical and cellular assays. RESULTS We show that the cardio-protective N-terminal proteolytic fragment of HDAC4 is enhanced in vivo in patients with diabetes mellitus and mouse models, as well as in vitro under high-glucose and high-O-GlcNAc conditions. HDAC4-knockout mice develop heart failure in models of type 1 and type 2 diabetes mellitus, whereas wild-type mice do not develop clear signs of heart failure, indicating that HDAC4 protects the diabetic heart. Reexpression of the N-terminal fragment of HDAC4 prevents HDAC4-dependent diabetic cardiomyopathy. Mechanistically, the posttranslational modification of HDAC4 at serine (Ser)-642 by O-GlcNAcylation is an essential step for production of the N-terminal fragment of HDAC4, which was attenuated by Ca2+/calmodulin-dependent protein kinase II-mediated phosphorylation at Ser-632. Preventing O-GlcNAcylation at Ser-642 not only entirely precluded production of the N-terminal fragment of HDAC4 but also promoted Ca2+/calmodulin-dependent protein kinase II-mediated phosphorylation at Ser-632, pointing to a mutual posttranslational modification cross talk of (cardio-detrimental) phosphorylation at Ser-632 and (cardio-protective) O-GlcNAcylation at Ser-642. CONCLUSIONS In this study, we found that O-GlcNAcylation of HDAC4 at Ser-642 is cardio-protective in diabetes mellitus and counteracts pathological Ca2+/calmodulin-dependent protein kinase II signaling. We introduce a molecular model explaining how diabetic metabolism possesses important cardio-protective features besides its known detrimental effects. A deeper understanding of the here-described posttranslational modification cross talk may lay the groundwork for the development of specific therapeutic concepts to treat heart failure in the context of diabetes mellitus.
Collapse
Affiliation(s)
- Mariya Kronlage
- Institute of Experimental Cardiology (M.K., M.D., J.G., U.O., L.H.L., J.B.), Heidelberg University, Germany.,Department of Cardiology (M.K., L.H.L., O.J.M., H.A.K.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Matthias Dewenter
- Institute of Experimental Cardiology (M.K., M.D., J.G., U.O., L.H.L., J.B.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Johannes Grosso
- Institute of Experimental Cardiology (M.K., M.D., J.G., U.O., L.H.L., J.B.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Thomas Fleming
- Department of Internal Medicine I (T.F.), Heidelberg University, Germany
| | - Ulrike Oehl
- Institute of Experimental Cardiology (M.K., M.D., J.G., U.O., L.H.L., J.B.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Lorenz H Lehmann
- Institute of Experimental Cardiology (M.K., M.D., J.G., U.O., L.H.L., J.B.), Heidelberg University, Germany.,Department of Cardiology (M.K., L.H.L., O.J.M., H.A.K.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Inês Falcão-Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal (I.F.-P., A.F.L.-M.)
| | - Adelino F Leite-Moreira
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal (I.F.-P., A.F.L.-M.)
| | - Nadine Volk
- Tissue Bank of the National Center for Tumor Diseases, Heidelberg, Germany (N.V.)
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg (H.-J.G.).,Institute of Pathology, University of Marburg, Germany (H.-J.G.)
| | - Oliver J Müller
- Department of Cardiology (M.K., L.H.L., O.J.M., H.A.K.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Albert Sickmann
- Leibniz Institute for Analysical Sciences (ISAS), Dortmund, Germany (A.S.).,Medical Faculty, Medical Proteomics Center, Ruhr-University Bochum, Germany (A.S.).,Department of Chemistry, College of Physical Sciences, University of Aberdeen, United Kingdom (A.S.). Dr Müller is currently at the Department of Internal Medicine III, University of Kiel, Germany
| | - Hugo A Katus
- Department of Cardiology (M.K., L.H.L., O.J.M., H.A.K.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| | - Johannes Backs
- Institute of Experimental Cardiology (M.K., M.D., J.G., U.O., L.H.L., J.B.), Heidelberg University, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (M.K., M.D., J.G., U.O., L.H.L., J.B., L.H.L., O.J.M., H.A.K.)
| |
Collapse
|
45
|
Yang R, Jia Q, Ma SF, Wang Y, Mehmood S, Chen Y. Exogenous H2S mitigates myocardial fibrosis in diabetic rats through suppression of the canonical Wnt pathway. Int J Mol Med 2019; 44:549-558. [PMID: 31198980 PMCID: PMC6605697 DOI: 10.3892/ijmm.2019.4237] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023] Open
Abstract
Hydrogen sulfide (H2S) has antifibrotic activity in the kidneys, heart, lungs, and other organs. The present study investigated the protective activity of exogenous H2S against myocardial fibrosis in a rat model of diabetes. Animals were assigned to normal control, diabetes mellitus (DM), DM + sodium hydrosulfide (NaHS; DM + NaHS) and NaHS groups. Fasting blood glucose (FBG), cardiac function and hydroxyproline were monitored. Heart histomorphology and ultrastructure were additionally evaluated. Wnt1-inducible signaling pathway protein (WISP)-1 protein expression in the myocardium was determined by immunohistochemical staining. Matrix metalloprotease (MMP)-2, tissue inhibitor of metalloproteinase (TIMP)-2, collagens, and canonical Wnt and transforming growth factor (TGF)-β1/SMAD family member 3 (Smad3) pathway-related proteins were assessed by western blotting. Cardiac function was decreased, and myocardial injury, hypertrophy and fibrosis were increased in the diabetes model rats. MMP-2 expression was decreased, and the expressions of WISP-1, TIMP-2, collagens, and canonical Wnt and TGF-β1/Smad3 pathway-related proteins were increased in the myocardia of the diabetes model rats. The present results indicated that the canonical Wnt pathway promoted diabetic myocardial fibrosis by upregulating the TGF-β1/Smad3 pathway. Except for FBG, exogenous H2S ameliorated the changes in diabetes-associated indices in rats in the DM + NaHS group. The results are consistent with H2S protection of streptozotocin-induced myocardial fibrosis in the diabetes model rats by downregulation of the canonical Wnt and TGF-β1/Smad3 pathway and decreased myocardial collagen deposition.
Collapse
Affiliation(s)
- Rui Yang
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, P.R. China
| | - Qiang Jia
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Shan-Feng Ma
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Ya Wang
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, P.R. China
| | - Shomaila Mehmood
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, P.R. China
| | - Yan Chen
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
46
|
Insights on alpha lipoic and dihydrolipoic acids as promising scavengers of oxidative stress and possible chelators in mercury toxicology. J Inorg Biochem 2019; 195:111-119. [DOI: 10.1016/j.jinorgbio.2019.03.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022]
|
47
|
Shen J, Xing W, Gong F, Wang W, Yan Y, Zhang Y, Xie C, Fu S. MiR-150-5p retards the progression of myocardial fibrosis by targeting EGR1. Cell Cycle 2019; 18:1335-1348. [PMID: 31122130 DOI: 10.1080/15384101.2019.1617614] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
To investigate the differential expression of microRNA-150-5p (miR-150-5p) and early growth response 1 (EGR1) in myocardial fibrosis (MF) cells, and determine the effect between miR-150-5p and EGR1 on MF. Human MF cells were generated via Trypanosoma cruzi (T. cruzi) infection, a mouse model of MF was generated via angiotensin II. The expression levels of miR-150-5p and EGR1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assay. The correlation between miR-150-5p and EGR1 was confirmed by a luciferase reporter assay. The viability, proliferation, and apoptotic rate were detected by cell counting kit-8 (CCK-8), colony-formation and flow cytometry assays. Hematoxylin-eosin (HE) staining and Masson staining visualized the degree of MF. Echocardiography was performed to obtain the levels of left ventricle fractional shortening (LVFS) and left ventricle ejection fraction (LVEF), computer algorithms and a videographics program were used to obtain the levels of left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP) and ±left ventricular dp/dt maximum (LV dp/dtmax). We found that the expression of miR-150-5p in MF cells was lower than normal cardiomyocytes, while the expression level of EGR1 in MF cells were higher than normal cardiomyocytes. Cell experiments demonstrated that EGR1 and miR-150-5p could influence the development of MF, and the expression of EGR1 in cardiomyocytes was regulated by miR-150-5p directly. Lastly, we confirmed that sh-Egr1 would decrease the severity of MF, while miR-150-5p antagomir could aggravate MF. Our results illustrate the mechanism of MF development, and provide a potential target for MF treatment.
Collapse
Affiliation(s)
- Jie Shen
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Wanhong Xing
- b Children's Heart Center , Sichuan Provincial Hospital for Women and Children , Chengdu , Sichuan , PR China
| | - Fangqi Gong
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Wei Wang
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Yufeng Yan
- c Trainning Center of Medical Experiments, School of Basic Medical Sciences , Fudan University , Shanghai , PR China
| | - Yiying Zhang
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Chunhong Xie
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Songling Fu
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| |
Collapse
|
48
|
Drankowska J, Kos M, Kościuk A, Marzęda P, Boguszewska-Czubara A, Tylus M, Święch-Zubilewicz A. MMP targeting in the battle for vision: Recent developments and future prospects in the treatment of diabetic retinopathy. Life Sci 2019; 229:149-156. [PMID: 31100326 DOI: 10.1016/j.lfs.2019.05.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/15/2022]
Abstract
Matrix metalloproteinases (MMPs) are enzymes capable of degrading nearly all types of extracellular matrix. They perform a wide range of roles in physiological processes, which is the reason for their strict regulation by numerous mechanisms including natural tissue inhibitors of metalloproteinases (TIMP). Research only started to shed light on more troublesome aspects of MMPs function, like cancer progression, Alzheimer's disease, atherosclerosis, ageing. Moreover, their profound role in diabetes is being carefully investigated including one of its most debilitating complications - diabetic retinopathy (DR), the leading cause of acquired blindness worldwide. Traditional treatment of this condition seems to be only mildly satisfactory, which elicited substantial interest in the field of new therapeutic methods including MMP targeting. So far, significant roles of MMP-2 and MMP-9 in the development of retinopathy have been established, with special attention given to the process of blood-retinal barrier impairment. Further exploration revealed MMP-10 and MMP-14 involvement as well as changes in MMP/TIMP ratio. In this review, we provide insight into MMPs role in diabetic retinopathy with a clarification of various mechanisms regulating MMP activity in the light of the recent studies. We conclude with an overview of novel DR therapies targeting MMPs and point to the need of further examination of their usefulness in clinical setting, with an eye towards future research.
Collapse
Affiliation(s)
- Justyna Drankowska
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4a, PL 20-093 Lublin, Poland.
| | - Michał Kos
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4a, PL 20-093 Lublin, Poland.
| | - Andrzej Kościuk
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4a, PL 20-093 Lublin, Poland
| | - Paweł Marzęda
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4a, PL 20-093 Lublin, Poland
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4a, PL 20-093 Lublin, Poland
| | - Magdalena Tylus
- Department of Retinal and Vitreal Surgery, Medical University of Lublin, Chmielna 1, PL 20-079 Lublin, Poland
| | - Anna Święch-Zubilewicz
- Department of Retinal and Vitreal Surgery, Medical University of Lublin, Chmielna 1, PL 20-079 Lublin, Poland
| |
Collapse
|
49
|
Zuo GF, Ren XM, Ge Q, Luo J, Ye P, Wang F, Wu W, Chao YL, Gu Y, Gao XF, Ge Z, Gao HB, Hu ZY, Zhang JJ, Chen SL. Activation of the PP2A catalytic subunit by ivabradine attenuates the development of diabetic cardiomyopathy. J Mol Cell Cardiol 2019; 130:170-183. [PMID: 30998977 DOI: 10.1016/j.yjmcc.2019.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/30/2022]
Abstract
Hyperglycemia-induced apoptosis plays a critical role in the pathogenesis of diabetic cardiomyopathy (DCM). Our previous study demonstrated that ivabradine, a selective If current antagonist, significantly attenuated myocardial apoptosis in diabetic mice, but the underlying mechanisms remained unknown. This study investigated the underlying mechanisms by which ivabradine exerts anti-apoptotic effects in experimental DCM. Pretreatment with ivabradine, but not ZD7288 (an established If current blocker), profoundly inhibited high glucose-induced apoptosis via inactivation of nuclear factor (NF)-κB signaling in neonatal rat cardiomyocytes. The effect was abolished by transfection of an siRNA targeting protein phosphatase 2A catalytic subunit (PP2Ac). In streptozotocin-induced diabetic mice, ivabradine treatment significantly inhibited left ventricular hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) and HCN4 (major components of the If current), activated PP2Ac, and attenuated NF-κB signaling activation and apoptosis, in line with improved histological abnormalities, fibrosis, and cardiac dysfunction without affecting hyperglycemia. These effects were not observed in diabetic mice with virus-mediated knockdown of HCN2 or HCN4 after myocardial injection, but were alleviated by knockdown of PP2Acα. Molecular docking and phosphatase activity assay confirmed direct binding of ivabradine to, and activation of, PP2Ac. In conclusion, ivabradine may directly activate PP2Ac, leading to inhibition of NF-κB signaling activation, myocardial apoptosis, and fibrosis, and eventually improving cardiac function in experimental DCM. Taken together, the present findings suggest that ivabradine may be a promising drug for treatment of DCM.
Collapse
Affiliation(s)
- Guang-Feng Zuo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Min Ren
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qing Ge
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Ye
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Feng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wen Wu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue-Lin Chao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Xiao-Fei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhen Ge
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Han-Bin Gao
- The First People's Hospital of Taicang, Soochow University, Suzhou, China
| | - Zuo-Ying Hu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Jun-Jie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
50
|
Murtaza G, Virk HUH, Khalid M, Lavie CJ, Ventura H, Mukherjee D, Ramu V, Bhogal S, Kumar G, Shanmugasundaram M, Paul TK. Diabetic cardiomyopathy - A comprehensive updated review. Prog Cardiovasc Dis 2019; 62:315-326. [PMID: 30922976 DOI: 10.1016/j.pcad.2019.03.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/21/2019] [Indexed: 01/04/2023]
Abstract
Diabetes causes cardiomyopathy and increases the risk of heart failure independent of hypertension and coronary heart disease. This condition called "Diabetic Cardiomyopathy" (DCM) is becoming a well- known clinical entity. Recently, there has been substantial research exploring its molecular mechanisms, structural and functional changes, and possible development of therapeutic approaches for the prevention and treatment of DCM. This review summarizes the recent advancements to better understand fundamental molecular abnormalities that promote this cardiomyopathy and novel therapies for future research. Additionally, different diagnostic modalities, up to date screening tests to guide clinicians with early diagnosis and available current treatment options has been outlined.
Collapse
Affiliation(s)
- Ghulam Murtaza
- Department of Internal Medicine, Division of Cardiology, East Tennessee State University, Johnson City, TN, USA
| | | | - Muhammad Khalid
- Department of Internal Medicine, Division of Cardiology, East Tennessee State University, Johnson City, TN, USA
| | - Carl J Lavie
- Department of Cardiology, Ochsner Clinic, New Orleans, LA, USA
| | - Hector Ventura
- Department of Cardiology, Ochsner Clinic, New Orleans, LA, USA
| | - Debabrata Mukherjee
- Division of Cardiology, Department of Internal Medicine, Texas Tech University, TX, USA
| | - Vijay Ramu
- Department of Internal Medicine, Division of Cardiology, East Tennessee State University, Johnson City, TN, USA
| | - Sukhdeep Bhogal
- Department of Internal Medicine, Division of Cardiology, East Tennessee State University, Johnson City, TN, USA
| | - Gautam Kumar
- Emory University School of Medicine, Atlanta VA Medical Center, Atlanta, GA, USA
| | | | - Timir K Paul
- Department of Internal Medicine, Division of Cardiology, East Tennessee State University, Johnson City, TN, USA.
| |
Collapse
|