1
|
Kakkar C, Sharma V, Mannan A, Gupta G, Singh S, Kumar P, Dua K, Kaur A, Singh S, Dhiman S, Singh TG. Diabetic Cardiomyopathy: An Update on Emerging Pathological Mechanisms. Curr Cardiol Rev 2025; 21:88-107. [PMID: 39501954 DOI: 10.2174/011573403x331870241025094307] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 04/25/2025] Open
Abstract
Diabetic Cardiomyopathy (DCM) is a notable consequence of diabetes mellitus, distinguished by cardiac dysfunction that occurs separately from coronary artery disease or hypertension. A recent study has revealed an intricate interaction of pathogenic processes that contribute to DCM. Important aspects involve the dysregulation of glucose metabolism, resulting in heightened oxidative stress and impaired mitochondrial function. In addition, persistent high blood sugar levels stimulate inflammatory pathways, which contribute to the development of heart fibrosis and remodelling. Additionally, changes in the way calcium is managed and the presence of insulin resistance are crucial factors in the formation and advancement of DCM. This may be due to the involvement of many molecular mechanistic pathways such as NLRP3, NF-κB, PKC, and MAPK with their downstream associated signaling pathways. Gaining a comprehensive understanding of these newly identified pathogenic pathways is crucial in order to design precise therapy approaches that can enhance the results for individuals suffering from diabetes. In addition, this review offers an in-depth review of not just pathogenic pathways and molecular mechanistic pathways but also diagnostic methods, treatment options, and clinical trials.
Collapse
Affiliation(s)
- Chirag Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Gaurav Gupta
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, 346, United Arab Emirates
| | - Sachin Singh
- Lovely Institute of Technology (Pharmacy), Lovely Professional University, Phagwara, Punjab, India
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Broadway, P.O. Box 123, Ultimo, NSW, 2007, Australia
| | - Puneet Kumar
- Department of Pharmacology, School of Pharmaceutical Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Broadway, P.O. Box 123, Ultimo, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
2
|
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
|
3
|
Cummins ML, Delmonte G, Wechsler S, Schlesinger JJ. Alleviating mitochondrial dysfunction in diabetic cardiomyopathy through the Adipsin and Irak2 pathways. Mil Med Res 2024; 11:11. [PMID: 38303084 PMCID: PMC10832134 DOI: 10.1186/s40779-024-00513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Affiliation(s)
- Mabel L Cummins
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA.
| | - Grace Delmonte
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Skylar Wechsler
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| | - Joseph J Schlesinger
- Division of Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, 37212, USA
| |
Collapse
|
4
|
Jiang MY, Man WR, Zhang XB, Zhang XH, Duan Y, Lin J, Zhang Y, Cao Y, Wu DX, Shu XF, Xin L, Wang H, Zhang X, Li CY, Gu XM, Zhang X, Sun DD. Adipsin inhibits Irak2 mitochondrial translocation and improves fatty acid β-oxidation to alleviate diabetic cardiomyopathy. Mil Med Res 2023; 10:63. [PMID: 38072993 PMCID: PMC10712050 DOI: 10.1186/s40779-023-00493-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) causes the myocardium to rely on fatty acid β-oxidation for energy. The accumulation of intracellular lipids and fatty acids in the myocardium usually results in lipotoxicity, which impairs myocardial function. Adipsin may play an important protective role in the pathogenesis of DCM. The aim of this study is to investigate the regulatory effect of Adipsin on DCM lipotoxicity and its molecular mechanism. METHODS A high-fat diet (HFD)-induced type 2 diabetes mellitus model was constructed in mice with adipose tissue-specific overexpression of Adipsin (Adipsin-Tg). Liquid chromatography-tandem mass spectrometry (LC-MS/MS), glutathione-S-transferase (GST) pull-down technique, Co-immunoprecipitation (Co-IP) and immunofluorescence colocalization analyses were used to investigate the molecules which can directly interact with Adipsin. The immunocolloidal gold method was also used to detect the interaction between Adipsin and its downstream modulator. RESULTS The expression of Adipsin was significantly downregulated in the HFD-induced DCM model (P < 0.05). Adipose tissue-specific overexpression of Adipsin significantly improved cardiac function and alleviated cardiac remodeling in DCM (P < 0.05). Adipsin overexpression also alleviated mitochondrial oxidative phosphorylation function in diabetic stress (P < 0.05). LC-MS/MS analysis, GST pull-down technique and Co-IP studies revealed that interleukin-1 receptor-associated kinase-like 2 (Irak2) was a downstream regulator of Adipsin. Immunofluorescence analysis also revealed that Adipsin was co-localized with Irak2 in cardiomyocytes. Immunocolloidal gold electron microscopy and Western blotting analysis indicated that Adipsin inhibited the mitochondrial translocation of Irak2 in DCM, thus dampening the interaction between Irak2 and prohibitin (Phb)-optic atrophy protein 1 (Opa1) on mitochondria and improving the structural integrity and function of mitochondria (P < 0.05). Interestingly, in the presence of Irak2 knockdown, Adipsin overexpression did not further alleviate myocardial mitochondrial destruction and cardiac dysfunction, suggesting a downstream role of Irak2 in Adipsin-induced responses (P < 0.05). Consistent with these findings, overexpression of Adipsin after Irak2 knockdown did not further reduce the accumulation of lipids and their metabolites in the cardiac myocardium, nor did it enhance the oxidation capacity of cardiomyocytes expose to palmitate (PA) (P < 0.05). These results indicated that Irak2 may be a downstream regulator of Adipsin. CONCLUSIONS Adipsin improves fatty acid β-oxidation and alleviates mitochondrial injury in DCM. The mechanism is related to Irak2 interaction and inhibition of Irak2 mitochondrial translocation.
Collapse
Affiliation(s)
- Meng-Yuan Jiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Wan-Rong Man
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xue-Bin Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Hua Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yang Cao
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - De-Xi Wu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Fei Shu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Lei Xin
- Department of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Hao Wang
- Department of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Cong-Ye Li
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Ming Gu
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, 710032, China
| | - Xuan Zhang
- Institute for Hospital Management Research, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Dong-Dong Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
5
|
Farrag EAE, Hammad MO, Safwat SM, Hamed S, Hellal D. Artemisinin attenuates type 2 diabetic cardiomyopathy in rats through modulation of AGE-RAGE/HMGB-1 signaling pathway. Sci Rep 2023; 13:11043. [PMID: 37422477 PMCID: PMC10329689 DOI: 10.1038/s41598-023-37678-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/26/2023] [Indexed: 07/10/2023] Open
Abstract
Diabetes mellitus is a common metabolic disorder. About two-thirds of diabetic patients develop diabetic cardiomyopathy (DCM), which becomes a challenging issue as it severely threatens the patient's life. Hyperglycemia and the resulting advanced glycated end products (AGE) and their receptor (RAGE)/High Mobility Group Box-1 (HMGB-1) molecular pathway are thought to be key players. Recently, artemisinin (ART) has gained more attention owing to its potent biological activities beyond its antimalarial effect. Herein, we aim to evaluate the effect of ART on DCM and the possible underlying mechanisms. Twenty-four male Sprague-Dawley rats were divided into: control, ART, type 2 diabetic and type 2 diabetic treated with ART groups. At the end of the research, the ECG was recorded, then the heart weight to body weight (HW/BW) ratio, fasting blood glucose, serum insulin and HOMA-IR were evaluated. Cardiac biomarkers (CK-MB and LDH), oxidative stress markers, IL-1β, AGE, RAGE and HMGB-1 expression were also measured. The heart specimens were stained for H&E as well as Masson's trichrome. DCM induced disturbances in all studied parameters; contrary to this, ART improved these insults. Our study concluded that ART could improve DCM through modulation of the AGE-RAGE/HMGB-1 signaling pathway, with subsequent impacts on oxidative stress, inflammation and fibrosis. ART could therefore be a promising therapy for the management of DCM.
Collapse
Affiliation(s)
- Eman A E Farrag
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Maha O Hammad
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Sally M Safwat
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shereen Hamed
- Department of Medical Histology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Hellal
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
6
|
Jin B, Chen Y, Wang J, Chen Y, Zhang M, Huang J, Wang Y. Costunolide alleviates hyperglycaemia-induced diabetic cardiomyopathy via inhibiting inflammatory responses and oxidative stress. J Cell Mol Med 2023; 27:831-845. [PMID: 36810875 PMCID: PMC10002915 DOI: 10.1111/jcmm.17686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 02/24/2023] Open
Abstract
Hyperglycaemia-induced myocardial injury promotes the induction of heart failure in diabetic patients. Impaired antioxidant capability and sustained chronic inflammation play a vital role in the progression of diabetic cardiomyopathy (DCM). Costunolide (Cos), a natural compound with anti-inflammatory and antioxidant properties, has exhibited therapeutic effects in various inflammatory diseases. However, the role of Cos in diabetes-induced myocardial injury remains poorly understood. In this study, we investigated the effect of Cos on DCM and explored the potential mechanisms. C57BL/6 mice were administered intraperitoneal streptozotocin for DCM induction. Cos-mediated anti-inflammatory and antioxidation activities were examined in heart tissues of diabetic mice and high glucose (HG)-stimulated cardiomyocytes. Cos markedly inhibited HG-induced fibrotic responses in diabetic mice and H9c2 cells, respectively. The cardioprotective effects of Cos could be correlated to the reduced expression of inflammatory cytokines and decreased oxidative stress. Further investigations demonstrated Cos reversed diabetes-induced nuclear factor-κB (NF-κB) activation and alleviated impaired antioxidant defence system, principally via activation of nuclear factor-erythroid 2 p45-related factor-2 (Nrf-2). Cos alleviated cardiac damage and improved cardiac function in diabetic mice by inhibiting NF-κB-mediated inflammatory responses and activating the Nrf-2-mediated antioxidant effects. Therefore, Cos could be a potential candidate for the treatment of DCM.
Collapse
Affiliation(s)
- Bo Jin
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiong Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yue Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mengpei Zhang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Jianxiong Huang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Yi Wang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Sadeghi S, Delphan M, Shams M, Esmaeili F, Shanaki-Bavarsad M, Shanaki M. The high-intensity interval training (HIIT) and curcumin supplementation can positively regulate the autophagy pathway in myocardial cells of STZ-induced diabetic rats. BMC Res Notes 2023; 16:21. [PMID: 36841820 PMCID: PMC9960211 DOI: 10.1186/s13104-023-06295-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
OBJECTIVE Targeting autophagy is a new therapeutic strategy for the complications of diabetes,such as diabetic cardiomyopathy (DCM). During diabetes, increased or insufficient autophagic activity causes aberrations in cellular homeostasis. Regarding the conflicting and unclear results regarding the effect of HIIT and curcumin supplementation on the expression of genes associated to autophagy, this study aimed to assess whether 4-week high-intensity interval training (HIIT) and curcumin supplementation are able to influence the expression of autophagy-related genes in myocardial cells of diabetic rats. METHODS In an experimental design, 24 male Wistar rats were randomly divided into 4 groups: non-diabetic control (NC), diabetic control (DC), diabetes + HIIT (D + HIIT), and diabetes + curcumin (D + CU). After HIIT program and curcumin treatment, the genes expression of autophagy pathway were assessed in the myocardium by real-time PCR Tanique. RESULTS The results indicated that the expression levels of ATG1, Beclin1, ATG5, and LAMP-2 genes were significantly reduced in the DC group compared to the NC group (p < 0.001). Following 4-week HIIT, the expression of Beclin1, ATG-5, and LAMP-2 improved considerably compared to the DC group (p < 0.001, p < 0.001, and p < 0.05, respectively). In addition, after 4 weeks of curcumin supplementation, the expression levels of ATG-5 and Beclin-1 were significantly improved compared to the DC group (p < 0.001, p < 0.05, respectively). It seems HIIT and curcumin supplementation can be an effective approach for inducing autophagy and improving cardiac function in DCM rats.However, HIIT seems more effective than curcumin in this regard.
Collapse
Affiliation(s)
- Samira Sadeghi
- grid.411600.2Department of Medical Laboratory Sciences, School of Allied Medical Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Delphan
- grid.411354.60000 0001 0097 6984Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Alzahra University, Tehran, Iran
| | - Masoumeh Shams
- grid.411600.2Department of Medical Laboratory Sciences, School of Allied Medical Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fataneh Esmaeili
- grid.411705.60000 0001 0166 0922Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Shanaki-Bavarsad
- grid.266102.10000 0001 2297 6811Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA USA
| | - Mehrnoosh Shanaki
- Department of Medical Laboratory Sciences, School of Allied Medical Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Involvement of circRNAs in the Development of Heart Failure. Int J Mol Sci 2022; 23:ijms232214129. [PMID: 36430607 PMCID: PMC9697219 DOI: 10.3390/ijms232214129] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/05/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
In recent years, interest in non-coding RNAs as important physiological regulators has grown significantly. Their participation in the pathophysiology of cardiovascular diseases is extremely important. Circular RNA (circRNA) has been shown to be important in the development of heart failure. CircRNA is a closed circular structure of non-coding RNA fragments. They are formed in the nucleus, from where they are transported to the cytoplasm in a still unclear mechanism. They are mainly located in the cytoplasm or contained in exosomes. CircRNA expression varies according to the type of tissue. In the brain, almost 12% of genes produce circRNA, while in the heart it is only 9%. Recent studies indicate a key role of circRNA in cardiomyocyte hypertrophy, fibrosis, autophagy and apoptosis. CircRNAs act mainly by interacting with miRNAs through a "sponge effect" mechanism. The involvement of circRNA in the development of heart failure leads to the suggestion that they may be promising biomarkers and useful targets in the treatment of cardiovascular diseases. In this review, we will provide a brief introduction to circRNA and up-to-date understanding of their role in the mechanisms leading to the development of heart failure.
Collapse
|
9
|
郑 冬, 陈 琳, 韦 其, 朱 梓, 刘 子, 金 琳, 杨 观, 谢 曦. [Fucoxanthin regulates Nrf2/Keap1 signaling to alleviate myocardial hypertrophy in diabetic rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:752-759. [PMID: 35673921 PMCID: PMC9178635 DOI: 10.12122/j.issn.1673-4254.2022.05.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the protective effect of fucoxanthin (FX) against diabetic cardiomyopathy and explore the underlying mechanism. METHODS Rat models of diabetes mellitus (DM) induced by intraperitoneal injection of streptozotocin (60 mg/kg) were randomized into DM model group, fucoxanthin treatment (DM+FX) group and metformin treatment (DM+ Met) group, and normal rats with normal feeding served as the control group. In the two treatment groups, fucoxanthin and metformin were administered after modeling by gavage at the daily dose of 200 mg/kg and 230 mg/kg, respectively for 12 weeks, and the rats in the DM model group were given saline only. HE staining was used to examine the area of cardiac myocyte hypertrophy in each group. The expression levels of fibrotic proteins TGF-β1 and FN proteins in rat hearts were detected with Western blotting. In the cell experiment, the effect of 1 μmol/L FX on H9C2 cell hypertrophy induced by exposure to high glucose (HG, 45 mmol/L) was evaluated using FITC-labeled phalloidin. The mRNA expression levels of the hypertrophic factors ANP, BNP and β-MHC in H9C2 cells were detected using qRT-PCR. The protein expressions of Nrf2, Keap1, HO-1 and SOD1 proteins in rat heart tissues and H9C2 cells were determined using Western blotting. The DCFH-DA probe was used to detect the intracellular production of reactive oxygen species (ROS). RESULTS In the diabetic rats, fucoxanthin treatment obviously alleviated cardiomyocyte hypertrophy and myocardial fibrosis, increased the protein expressions of Nrf2 and HO-1, and decreased the protein expressions of Keap1 in the heart tissue (P < 0.05). In H9C2 cells with HG exposure, fucoxanthin significantly inhibited the enlargement of cell surface area, lowered the mRNA expression levels of ANP, BNP and β-MHC (P < 0.05), promoted Nrf2 translocation from the cytoplasm to the nucleus, and up-regulated the protein expressions its downstream targets SOD1 and HO-1 (P < 0.05) to enhance cellular antioxidant capacity and reduce intracellular ROS production. CONCLUSION Fucoxanthin possesses strong inhibitory activities against diabetic cardiomyocyte hypertrophy and myocardial fibrosis and is capable of up-regulating Nrf2 signaling to promote the expression of its downstream antioxidant proteins SOD1 and HO-1 to reduce the level of ROS.
Collapse
Affiliation(s)
- 冬晓 郑
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 琳琳 陈
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 其慧 韦
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 梓然 朱
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 子略 刘
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 琳 金
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 观玉 杨
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - 曦 谢
- />海南大学药学院,海南 海口 570228School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| |
Collapse
|
10
|
El-Azab MF, Wakiel AE, Nafea YK, Youssef ME. Role of cannabinoids and the endocannabinoid system in modulation of diabetic cardiomyopathy. World J Diabetes 2022; 13:387-407. [PMID: 35664549 PMCID: PMC9134026 DOI: 10.4239/wjd.v13.i5.387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/18/2021] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetic complications, chiefly seen in long-term situations, are persistently deleterious to a large extent, requiring multi-factorial risk reduction strategies beyond glycemic control. Diabetic cardiomyopathy is one of the most common deleterious diabetic complications, being the leading cause of mortality among diabetic patients. The mechanisms of diabetic cardiomyopathy are multi-factorial, involving increased oxidative stress, accumulation of advanced glycation end products (AGEs), activation of various pro-inflammatory and cell death signaling pathways, and changes in the composition of extracellular matrix with enhanced cardiac fibrosis. The novel lipid signaling system, the endocannabinoid system, has been implicated in the pathogenesis of diabetes and its complications through its two main receptors: Cannabinoid receptor type 1 and cannabinoid receptor type 2, alongside other components. However, the role of the endocannabinoid system in diabetic cardiomyopathy has not been fully investigated. This review aims to elucidate the possible mechanisms through which cannabinoids and the endocannabinoid system could interact with the pathogenesis and the development of diabetic cardiomyopathy. These mechanisms include oxidative/ nitrative stress, inflammation, accumulation of AGEs, cardiac remodeling, and autophagy. A better understanding of the role of cannabinoids and the endocannabinoid system in diabetic cardiomyopathy may provide novel strategies to manipulate such a serious diabetic complication.
Collapse
Affiliation(s)
- Mona F El-Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed E Wakiel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Yossef K Nafea
- Program of Biochemistry, McMaster University, Hamilton L8S 4L8, Ontario, Canada
| | - Mahmoud E Youssef
- Department of Pharmacology and Biochemistry, Delta University for Science and Technology, Mansoura 35511, New Cairo, Egypt
| |
Collapse
|
11
|
Kleissl-Muir S, Rasmussen B, Owen A, Zinn C, Driscoll A. Low Carbohydrate Diets for Diabetic Cardiomyopathy: A Hypothesis. Front Nutr 2022; 9:865489. [PMID: 35529461 PMCID: PMC9069235 DOI: 10.3389/fnut.2022.865489] [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: 01/29/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Elevated blood glucose levels, insulin resistance (IR), hyperinsulinemia and dyslipidemia the key aspects of type 2 diabetes mellitus (T2DM), contribute to the development of a certain form of cardiomyopathy. This cardiomyopathy, also known as diabetic cardiomyopathy (DMCM), typically occurs in the absence of overt coronary artery disease (CAD), hypertension or valvular disease. DMCM encompasses a variety of pathophysiological processes impacting the myocardium, hence increasing the risk for heart failure (HF) and significantly worsening outcomes in this population. Low fat (LF), calorie-restricted diets have been suggested as the preferred eating pattern for patients with HF. However, LF diets are naturally higher in carbohydrates (CHO). We argue that in an insulin resistant state, such as in DMCM, LF diets may worsen glycaemic control and promote further insulin resistance (IR), contributing to a physiological and functional decline in DMCM. We postulate that CHO restriction targeting hyperinsulinemia may be able to improve tissue and systemic IR. In recent years low carbohydrate diets (LC) including ketogenic diets (KD), have emerged as a safe and effective tool for the management of various clinical conditions such as T2DM and other metabolic disorders. CHO restriction achieves sustained glycaemic control, lower insulin levels and successfully reverses IR. In addition to this, its pleiotropic effects may present a metabolic stress defense and facilitate improvement to cardiac function in patients with HF. We therefore hypothesize that patients who adopt a LC diet may require less medications and experience improvements in HF-related symptom burden.
Collapse
Affiliation(s)
| | - Bodil Rasmussen
- School of Nursing and Midwifery, Deakin University, Geelong, VIC, Australia
- Centre for Quality and Patient Safety, School of Nursing and Midwifery, Institute for Health Transformation, Deakin University, Geelong, VIC, Australia
- The Centre for Quality and Patient Safety, Institute of Health Transformation -Western Health Partnership, Western Health, St Albans, VIC, Australia
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark and Steno Diabetes Centre, Odense, Denmark
| | - Alice Owen
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Caryn Zinn
- Human Potential Centre, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Andrea Driscoll
- School of Nursing and Midwifery, Deakin University, Geelong, VIC, Australia
- Centre for Quality and Patient Safety, School of Nursing and Midwifery, Institute for Health Transformation, Deakin University, Geelong, VIC, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiology, Austin Health, Heidelberg, VIC, Australia
| |
Collapse
|
12
|
Zhao X, Liu S, Wang X, Chen Y, Pang P, Yang Q, Lin J, Deng S, Wu S, Fan G, Wang B. Diabetic cardiomyopathy: Clinical phenotype and practice. Front Endocrinol (Lausanne) 2022; 13:1032268. [PMID: 36568097 PMCID: PMC9767955 DOI: 10.3389/fendo.2022.1032268] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition of cardiac structure and function changes in diabetic patients without coronary artery disease, hypertension, and other types of heart diseases. DCM is not uncommon in people with diabetes, which increases the risk of heart failure. However, the treatment is scarce, and the prognosis is poor. Since 1972, one clinical study after another on DCM has been conducted. However, the complex phenotype of DCM still has not been fully revealed. This dilemma hinders the pace of understanding the essence of DCM and makes it difficult to carry out penetrating clinical or basic research. This review summarizes the literature on DCM over the last 40 years and discusses the overall perspective of DCM, phase of progression, potential clinical indicators, diagnostic and screening criteria, and related randomized controlled trials to understand DCM better.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Shengwang Liu
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Xiao Wang
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Yibing Chen
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Pai Pang
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Qianjing Yang
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Jingyi Lin
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Shuaishuai Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Shentao Wu
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Guanwei Fan
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| | - Bin Wang
- Department of Endocrine and Metabolic Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Xiqing, Tianjin, China
| |
Collapse
|
13
|
Kambis TN, Tofilau HMN, Gawargi FI, Chandra S, Mishra PK. Regulating Polyamine Metabolism by miRNAs in Diabetic Cardiomyopathy. Curr Diab Rep 2021; 21:52. [PMID: 34902085 PMCID: PMC8668854 DOI: 10.1007/s11892-021-01429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 11/08/2022]
Abstract
PURPOSE OF REVIEW Insulin is at the heart of diabetes mellitus (DM). DM alters cardiac metabolism causing cardiomyopathy, ultimately leading to heart failure. Polyamines, organic compounds synthesized by cardiomyocytes, have an insulin-like activity and effect on glucose metabolism, making them metabolites of interest in the DM heart. This review sheds light on the disrupted microRNA network in the DM heart in relation to developing novel therapeutics targeting polyamine biosynthesis to prevent/mitigate diabetic cardiomyopathy. RECENT FINDINGS Polyamines prevent DM-induced upregulation of glucose and ketone body levels similar to insulin. Polyamines also enhance mitochondrial respiration and thereby regulate all major metabolic pathways. Non-coding microRNAs regulate a majority of the biological pathways in our body by modulating gene expression via mRNA degradation or translational repression. However, the role of miRNA in polyamine biosynthesis in the DM heart remains unclear. This review discusses the regulation of polyamine synthesis and metabolism, and its impact on cardiac metabolism and circulating levels of glucose, insulin, and ketone bodies. We provide insights on potential roles of polyamines in diabetic cardiomyopathy and putative miRNAs that could regulate polyamine biosynthesis in the DM heart. Future studies will unravel the regulatory roles these miRNAs play in polyamine biosynthesis and will open new doors in the prevention/treatment of adverse cardiac remodeling in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Tyler N Kambis
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | | | - Flobater I Gawargi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surabhi Chandra
- Department of Biology, University of Nebraska-Kearney, Kearney, NE, 68845, USA
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
14
|
Oka S, Kai T, Hoshino K, Watanabe K, Nakamura J, Abe M, Watanabe A. Effects of empagliflozin in different phases of diabetes mellitus-related cardiomyopathy: a prospective observational study. BMC Cardiovasc Disord 2021; 21:217. [PMID: 33926386 PMCID: PMC8086321 DOI: 10.1186/s12872-021-02024-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/19/2021] [Indexed: 12/31/2022] Open
Abstract
Background Diabetes mellitus-related cardiomyopathy (DMCMP), defined as left ventricular (LV) dysfunction caused by hyperglycemia in the absence of coronary artery disease, leads to heart failure (HF). Previous studies have shown that treatment with sodium-glucose co-transporter 2 inhibitor (SGLT2i) reduces the risk of exacerbation of HF. The beneficial effects of SGLT2i on HF depend not only on indirect actions such as osmotic diuresis but also on direct actions on the myocardium, leading to improvements in LV function. However, it remains unclear whether SGLT2i treatment is equally effective in any phase of DMCMP. The aim of this observational study was to compare the efficacy of SGLT2i treatment on LV dysfunction between early and advanced DMCMP. Methods Thirty-five symptomatic non-ischemic HF patients with LV ejection fraction > 40% and type 2 diabetes mellitus (T2DM) treated with empagliflozin (EMPA group) and 20 controls treated without SGLT2i were enrolled. According to the myocardial extracellular volume fraction (ECV), a reliable marker of cardiac fibrosis quantified by cardiac magnetic resonance, the EMPA group was further divided into early DMCMP (n = 16, ECV ≤ 30%) and advanced DMCMP (n = 19, ECV > 30%) groups and followed up prospectively. Echocardiography was performed at baseline and after 12 months. LV function assessed as LV global longitudinal strain (LVGLS) and the ratio of early diastolic mitral inflow velocity to early diastolic mitral annular velocity (E/e′) were compared. Results ECV was strongly correlated with T2DM duration (r2 = 0.65, p < 0.001). At baseline, each group had a similar background. After 12 months, the EMPA group, especially the early DMCMP group, showed remarkable improvements in LVGLS (ΔLVGLS: 2.9 ± 3.0% (EMPA) vs. 0.6 ± 2.2% (controls), p = 0.005, and 4.6 ± 1.5% (early DMCMP) vs. 1.6 ± 3.3% (advanced DMCMP), p = 0.003) and E/e′ (ΔE/e′: − 1.5 ± 4.7 vs. − 0.3 ± 3.0, p = 0.253, and − 3.4 ± 5.5 vs. − 0.1 ± 3.5, p = 0.043). Conclusions The positive effects of empagliflozin on LV dysfunction were more remarkable in early than in advanced DMCMP. Early intervention of SGLT2i for DMCMP may be preferable.
Collapse
Affiliation(s)
- Satoshi Oka
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan.
| | - Takahiko Kai
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan
| | - Katsuomi Hoshino
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan
| | - Kazunori Watanabe
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan
| | - Jun Nakamura
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan
| | - Makoto Abe
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan
| | - Akinori Watanabe
- Department of Cardiology, Fujieda Municipal General Hospital, Surugadai 4-1-11, Fujieda, Shizuoka, 426-8677, Japan
| |
Collapse
|
15
|
Generating Ins2 +/-/miR-133aTg Mice to Model miRNA-Driven Cardioprotection of Human Diabetic Heart. Methods Mol Biol 2021; 2224:113-121. [PMID: 33606210 DOI: 10.1007/978-1-0716-1008-4_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Diabetes mellitus (DM) is caused either due to insulin deficiency (T1DM) or insulin resistance (T2DM). DM increases the risk of heart failure by diabetic cardiomyopathy (DMCM), a cardiac muscle disorder that leads to a progressive decline in diastolic function, and ultimately systolic dysfunction. Mouse models of T1DM and T2DM exhibit clinical signs of DMCM. Growing evidence implicates microRNA (miRNA), an endogenous, non-coding, regulatory RNA, in the pathogenesis and signaling of DMCM. Therefore, inhibiting deleterious miRNAs and mimicking cardioprotective miRNAs could provide a potential therapeutic intervention for DMCM. miRNA-133a (miR-133a) is a highly abundant miRNA in the human heart. It is a cardioprotective miRNA, which is downregulated in the DM heart. It has anti-hypertrophic and anti-fibrotic effects. miR-133a mimic treatment after the onset of early DMCM can reverse histological and clinical signs of the disease in mice. We hypothesized that overexpression of cardiac-specific miR-133a in Ins2+/- Akita (T1DM) mice can prevent progression of DMCM. Here, we describe a method to create and validate cardiac-specific Ins2+/-/miR-133aTg mice to determine whether cardiac-specific miR-133a overexpression prevents development of DMCM. These strategies demonstrate the value of genetic modeling of human disease such as DMCM and evaluate the potential of miRNA as a therapeutic intervention.
Collapse
|
16
|
Abstract
Coronavirus disease 2019 (COVID-19) and diabetes outcomes (CORONADO) trial revealed that 10.6% of patients with diabetes mellitus hospitalized for COVID-19 (COVID-19) die within 7 days. Several studies from New York, Italy, and China confirm that patients with diabetes are at a much higher risk for mortality due to COVID-19. Besides respiratory illness, COVID-19 increases cardiac injury and diabetic ketoacidosis. In the absence of specific guidelines for the prevention and treatment of COVID-19 for patients with diabetes, they remain at higher risk and are more susceptible to COVID-19. Furthermore, there is a scarcity of basic knowledge on how diabetes affects pathogenesis of severe acute respiratory coronavirus (SARS-CoV-2) infection. In patients with diabetes, impaired glucose use alters metabolic and consequently biological processes instigating pathological remodeling, which has detrimental effects on cardiovascular systems. A majority of biological processes are regulated by noncoding microRNAs (miRNAs), which have emerged as a promising therapeutic candidate for several diseases. In consideration of the higher risk of mortality in patients with diabetes and COVID-19, novel diagnostic test and treatment strategy are urgently warranted in post-COVID-19 era. Here, we describe potential roles of miRNA as a biomarker and therapeutic candidate, especially for heart failure, in patients with diabetes and COVID-19.
Collapse
Affiliation(s)
- Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ritesh Tandon
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
17
|
Induction of caveolin-3/eNOS complex by nitroxyl (HNO) ameliorates diabetic cardiomyopathy. Redox Biol 2020; 32:101493. [PMID: 32182574 PMCID: PMC7078438 DOI: 10.1016/j.redox.2020.101493] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Nitroxyl (HNO), one-electron reduced and protonated sibling of nitric oxide (NO), is a potential regulator of cardiovascular functions. It produces positive inotropic, lusitropic, myocardial anti-hypertrophic and vasodilator properties. Despite of these favorable actions, the significance and the possible mechanisms of HNO in diabetic hearts have yet to be fully elucidated. H9c2 cells or primary neonatal mouse cardiomyocytes were incubated with normal glucose (NG) or high glucose (HG). Male C57BL/6 mice received intraperitoneal injection of streptozotocin (STZ) to induce diabetes. Here, we demonstrated that the baseline fluorescence signals of HNO in H9c2 cells were reinforced by both HNO donor Angeli's salt (AS), and the mixture of hydrogen sulfide (H2S) donor sodium hydrogen sulfide (NaHS) and NO donor sodium nitroprusside (SNP), but decreased by HG. Pretreatment with AS significantly reduced HG-induced cell vitality injury, apoptosis, reactive oxygen species (ROS) generation, and hypertrophy in H9c2 cells. This effect was mediated by induction of caveolin-3 (Cav-3)/endothelial nitric oxide (NO) synthase (eNOS) complex. Disruption of Cav-3/eNOS by pharmacological manipulation or small interfering RNA (siRNA) abolished the protective effects of AS in HG-incubated H9c2 cells. In STZ-induced diabetic mice, administration of AS ameliorated the development of diabetic cardiomyopathy, as evidenced by improved cardiac function and reduced cardiac hypertrophy, apoptosis, oxidative stress and myocardial fibrosis without affecting hyperglycemia. This study shed light on how interaction of NO and H2S regulates cardiac pathology and provide new route to treat diabetic cardiomyopathy with HNO.
Collapse
|
18
|
Grigorescu ED, Lacatusu CM, Floria M, Mihai BM, Cretu I, Sorodoc L. Left Ventricular Diastolic Dysfunction in Type 2 Diabetes-Progress and Perspectives. Diagnostics (Basel) 2019; 9:diagnostics9030121. [PMID: 31533216 PMCID: PMC6787758 DOI: 10.3390/diagnostics9030121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 12/17/2022] Open
Abstract
In-depth understanding of early cardiovascular manifestations in diabetes is high on international research and prevention agendas given that cardiovascular events are the leading cause of death for diabetic patients. Our aim was to review recent developments in the echocardiographic assessment of left ventricular diastolic dysfunction (LVDD) as a telltale pre-clinical disturbance preceding diabetic cardiomyopathy. We analyzed papers in which patients had been comprehensively assessed echocardiographically according to the latest LVDD guidelines (2016), and those affording comparisons with previous, widely used recommendations (2009). We found that the updated algorithm for LVDD is more effective in predicting adverse cardiovascular events in patients with established LVDD, and less specific in grading other patients (labelled "indeterminate"). This may prove instrumental for recruiting "indeterminate" LVDD cases among patients with type 2 diabetes mellitus (T2DM) in future screening programs. As an interesting consideration, the elevated values of the index E/e' can point to early diastolic impairment, foretelling diabetic cardiomyopathy. Identifying subclinical signs early makes clinical sense, but the complex nature of T2DM calls for further research. Specifically, longitudinal studies on rigorously selected cohorts of diabetic patients are needed to better understand and predict the subtle, slow onset of cardiac manifestations with T2DM as a complicating backdrop.
Collapse
Affiliation(s)
- Elena-Daniela Grigorescu
- Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania (B.-M.M.)
| | - Cristina-Mihaela Lacatusu
- Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania (B.-M.M.)
- “Sf. Spiridon” Emergency Hospital, 700111 Iași, Romania; (M.F.); (L.S.)
- Correspondence: ; Tel.: +40-72-321-1116
| | - Mariana Floria
- “Sf. Spiridon” Emergency Hospital, 700111 Iași, Romania; (M.F.); (L.S.)
- Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Bogdan-Mircea Mihai
- Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania (B.-M.M.)
- “Sf. Spiridon” Emergency Hospital, 700111 Iași, Romania; (M.F.); (L.S.)
| | - Ioana Cretu
- Department Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
| | - Laurentiu Sorodoc
- “Sf. Spiridon” Emergency Hospital, 700111 Iași, Romania; (M.F.); (L.S.)
- Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
19
|
The microRNA in ventricular remodeling: the miR-30 family. Biosci Rep 2019; 39:BSR20190788. [PMID: 31320543 PMCID: PMC6680373 DOI: 10.1042/bsr20190788] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/07/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
Ventricular remodeling (VR) is a complex pathological process of cardiomyocyte apoptosis, cardiac hypertrophy, and myocardial fibrosis, which is often caused by various cardiovascular diseases (CVDs) such as hypertension, acute myocardial infarction, heart failure (HF), etc. It is also an independent risk factor for a variety of CVDs, which will eventually to damage the heart function, promote cardiovascular events, and lead to an increase in mortality. MicroRNAs (miRNAs) can participate in a variety of CVDs through post-transcriptional regulation of target gene proteins. Among them, microRNA-30 (miR-30) is one of the most abundant miRNAs in the heart. In recent years, the study found that the miR-30 family can participate in VR through a variety of mechanisms, including autophagy, apoptosis, oxidative stress, and inflammation. VR is commonly found in ischemic heart disease (IHD), hypertensive heart disease (HHD), diabetic cardiomyopathy (DCM), antineoplastic drug cardiotoxicity (CTX), and other CVDs. Therefore, we will review the relevant mechanisms of the miR-30 in VR induced by various diseases.
Collapse
|
20
|
Zhang S, Wang H, Li L, Chang X, Ma H, Zhang M, Qing X, Zhang L, Zhang Z. Qishen Yiqi Drop Pill, a novel compound Chinese traditional medicine protects against high glucose-induced injury in cardiomyocytes. J Cell Mol Med 2019; 23:6393-6402. [PMID: 31278860 PMCID: PMC6714141 DOI: 10.1111/jcmm.14527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 05/17/2019] [Accepted: 06/19/2019] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Qishen Yiqi Drop Pill (QSYQ) has been recognized as a potential protective agent for various cardiovascular diseases. However, the effect of QSYQ in cardiac complications associated with diabetes is not clear currently. In this study, we investigate whether QSYQ could exert cardiac protective effects against high glucose-induced injuries in cardiac H9c2 cells. METHODS H9c2 cells were exposed to 24 hours of high glucose in presence or absence of QSYQ and LY294002. Cell cytotoxicity, apoptosis, reactive oxygen species (ROS) generation, mitochondrial membrane potential and mitochondrial permeability transition pore (mPTP) opening were determined. Levels of bax, bcl-2, p53, cleaved caspase-3, PI3K and Akt were evaluated by Western blot. RESULTS Our data indicated that QSYQ significantly increased the cell viability and decreased cytotoxicity. By analysing the apoptotic rate as well as the expression levels of cytoapoptosis-related factors including cleaved caspase-3, bax, bcl-2, and p53, we found that QSYQ could remarkably suppress apoptosis of cardiomyoblasts caused by high glucose. In addition, it also showed that QSYQ reduced the generation of ROS. We further found that QSYQ treatment could inhibit the loss of mitochondrial membrane potential and mPTP opening. Moreover, Western blot analysis showed enhanced phosphorylation of PI3K/Akt. The specific inhibitor of PI3K, LY294002 not only inhibited QSYQ induced PI3K/Akt signalling pathway activation, but alleviated its protective effects. CONCLUSIONS In summary, these findings demonstrated that QSYQ effectively protected H9c2 cells against the series injuries due to high glucose at least partially by activating the PI3K/Akt signalling pathway.
Collapse
Affiliation(s)
- Shouyan Zhang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Hao Wang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Lixia Li
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Xuewei Chang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Huifang Ma
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Mingming Zhang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Xiaochun Qing
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Lijun Zhang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| | - Zhuo Zhang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang Institute of Cardio-cerebrovascular Diseases, Luoyang Key Laboratory of Cardiac-cerebro Tissue Injury and Repair, Luoyang, China
| |
Collapse
|
21
|
Long non-coding RNA MEG3 serves as a ceRNA for microRNA-145 to induce apoptosis of AC16 cardiomyocytes under high glucose condition. Biosci Rep 2019; 39:BSR20190444. [PMID: 31085717 PMCID: PMC6554216 DOI: 10.1042/bsr20190444] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the most serious complications of diabetes, but its pathogenesis remains largely unclear. In the present study, we aimed to explore the potential role of long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3) and to investigate the underlying mechanisms in human AC16 cardiomyocytes under high glucose (HG) condition. The results demonstrated that MEG3 was overexpressed in HG-treated AC16 cells, and MEG3 knockdown suppressed the HG-induced apoptosis in AC16 cells. Mechanistically, MEG3 directly binds to miR-145 in AC16 cells, thereby up-regulating the expression of PDCD4. Rescue experiments showed that the role of MEG3 in HG-treated AC16 cells was partly dependent on its suppression on miR-145. In summary, our findings suggested that the role of MEG3 in HG-treated human cardiomyocytes is to serve as a competing endogenous RNA (ceRNA), which negatively regulates miR-145. These findings may provide a valuable and promising therapeutic target for the treatment of DCM in the future.
Collapse
|
22
|
He A, Fang W, Zhao K, Wang Y, Li J, Yang C, Benadjaoud F, Shi GP. Mast cell-deficiency protects mice from streptozotocin-induced diabetic cardiomyopathy. Transl Res 2019; 208:1-14. [PMID: 30738862 PMCID: PMC6527494 DOI: 10.1016/j.trsl.2019.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/20/2018] [Accepted: 01/17/2019] [Indexed: 01/01/2023]
Abstract
Mast cells (MCs) have been implicated in the pathogenesis of cardiometabolic diseases by releasing pro-inflammatory mediators. Patients and animals with diabetic cardiomyopathy (DCM) also show inflammatory cell accumulation in the heart. Here, we detected MCs in mouse heart after streptozotocin (STZ)-induced DCM. DCM production caused significant systole and diastole interventricular septum and left ventricular (LV) posterior wall thinning, and systolic LV internal dilation in wild-type (WT) mice. DCM production also led to significant reductions of fractional shortening percentage, heart rate, body weight, heart weight, and significant increases of kidney, pancreas, and lung weight to body weight ratios, and blood hemoglobin HbA1c and glucose levels in WT mice. All these changes were improved or disappeared in MC-deficient KitW-sh/W-sh mice. In the myocardium from WT DCM mice, we detected significant decrease of cardiac cell proliferation and increases of cardiac cell death, chemokine expression, macrophage infiltration, inflammatory cytokine expression, and collagen deposition. These changes were also improved or disappeared in KitW-sh/W-sh DCM mice. Adoptive transfer of bone marrow-derived MCs (BMMCs) from WT mice fully or partially reversed these cardiac functional and morphologic changes in KitW-sh/W-sh DCM recipient mice. Yet, adoptive transfer of BMMCs from Il6-/- and Tnf-/- mice failed to make these corrections or at much less extent than the WT BMMCs. Mechanistic studies demonstrated a role of MC and MC-derived IL6 and TNF-α in promoting cardiomyocyte death and cardiac fibroblast TGF-β signaling, and collagen synthesis and deposition. Therefore, MC inhibition may have therapeutic potential in attenuating DCM progression.
Collapse
Affiliation(s)
- Aina He
- Department of Oncology, Affiliated Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China; Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wenqian Fang
- Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kun Zhao
- Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yajun Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jie Li
- Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Chongzhe Yang
- Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Feriel Benadjaoud
- Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Guo-Ping Shi
- Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
23
|
Sciacqua A, Perticone M, Miceli S, Pinto A, Cassano V, Succurro E, Andreozzi F, Hribal ML, Sesti G, Perticone F. Elevated 1-h post-load plasma glucose is associated with right ventricular morphofunctional parameters in hypertensive patients. Endocrine 2019; 64:525-535. [PMID: 30790176 DOI: 10.1007/s12020-019-01873-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Emerging data demonstrate that type 2 diabetes mellitus (T2DM) is associated with right ventricular (RV) dysfunction. A cutoff point of 155 mg/dL for the 1-hour (h) post-load plasma glucose, during oral glucose tolerance test (OGTT), identifies patients with normal glucose tolerance (NGT) at high risk to develop T2DM and cardiovascular (CV) disease. We investigated if 1-h post-load glucose may affect RV geometry and function in a group of never-treated hypertensive individuals. METHODS We enrolled 446 Caucasian newly diagnosed hypertensive outpatients. All patients underwent an OGTT and a standard echocardiography. The tricuspid annular plane systolic excursion (TAPSE) and the RV fractional area change (RVFAC) were measured together with systolic pulmonary arterial pressure (s-PAP) and pulmonary vascular resistances (PVR). Insulin sensitivity was evaluated using the Matsuda index. RESULTS Among all partecipants, 296 had NGT, 100 impaired glucose tolerance (IGT), and 50 T2DM. Considering the cutoff point of 155 mg/dl for 1-h glucose, NGT subjects were stratified into two groups: NGT < 155 (n = 207), NGT ≥ 155 (n = 89). Subjects NGT ≥ 155 presented a worse metabolic and inflammatory profile than NGT < 155. RV functional parameters (TAPSE, RVFAC, TAPSE/s-PAP, and TAPSE/PVR) were significantly reduced in NGT ≥ 155 subjects compared with NGT < 155 patients. On the contrary, s-PAP and PVR were significantly higher. At multiple regression analysis, 1-h glucose was the strongest predictor of TAPSE in NGT ≥ 155, IGT, and T2DM. CONCLUSIONS The presence of RV impairment in hypertensive NGT ≥ 155 subjects further complicates their CV burden and it may, at least in part, justify the worse clinical outcome in this setting of patients.
Collapse
Affiliation(s)
- Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy.
| | - Maria Perticone
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Sofia Miceli
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Angelina Pinto
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Velia Cassano
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Elena Succurro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Marta Letizia Hribal
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
24
|
Kambis TN, Shahshahan HR, Kar S, Yadav SK, Mishra PK. Transgenic Expression of miR-133a in the Diabetic Akita Heart Prevents Cardiac Remodeling and Cardiomyopathy. Front Cardiovasc Med 2019; 6:45. [PMID: 31069235 PMCID: PMC6491745 DOI: 10.3389/fcvm.2019.00045] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/25/2019] [Indexed: 01/22/2023] Open
Abstract
Advanced diabetes mellitus (DM) may have both insulin resistance and deficiency (double DM) that accelerates diabetic cardiomyopathy (DMCM), a cardiac muscle disorder. Reduced cardiac miR-133a, a cardioprotective miRNA, is associated with DMCM. However, it is unclear whether increasing miR-133a levels in the double DM heart could prevent DMCM. We hypothesized that increasing cardiac levels of miR-133a could prevent DMCM in Akita, a mouse model of double DM. To test the hypothesis, we created Akita/miR-133aTg mice, a new strain of Akita where miR-133a is overexpressed in the heart, by crossbreeding male Akita with female cardiac-specific miR-133a transgenic mice. We validated Akita/miR-133aTg mice by genotyping and phenotyping (miR-133a levels in the heart). To determine whether miR-133a overexpression could prevent cardiac remodeling and cardiomyopathy, we evaluated cardiac fibrosis, hypertrophy, and dysfunction (P-V loop) in 13-15 week male WT, Akita, Akita/miR-133aTg, and miR-133aTg mice. Our results revealed that miR-133a overexpression in the Akita heart prevents DM-induced cardiac fibrosis (reduced collagen deposition), hypertrophy (decreased beta-myosin heavy chain), and impaired contractility (downregulated calcium handling protein sarco-endoplasmic reticulum-ATPase-2a). These results demonstrate that increased levels of miR-133a in the DM heart could prevent cardiac remodeling. Our P-V loop analysis showed a trend of decreased cardiac output, stroke volume, and ± dp/dt in Akita, which were blunted in Akita/miR-133aTg heart. These findings suggest that 13-15 week Akita heart undergoes adverse remodeling toward cardiomyopathy, which is prevented by miR-133a overexpression. In addition, increased cardiac miR-133a in the Akita heart did not change blood glucose levels but decreased lipid accumulation in the heart, suggesting inhibition of metabolic remodeling in the heart. Thus, miR-133a could be a promising therapeutic candidate to prevent DMCM.
Collapse
Affiliation(s)
- Tyler N Kambis
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hamid R Shahshahan
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sumit Kar
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Santosh K Yadav
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
25
|
Nandi SS, Shahshahan HR, Shang Q, Kutty S, Boska M, Mishra PK. MiR-133a Mimic Alleviates T1DM-Induced Systolic Dysfunction in Akita: An MRI-Based Study. Front Physiol 2018; 9:1275. [PMID: 30364155 PMCID: PMC6192327 DOI: 10.3389/fphys.2018.01275] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 08/21/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetic cardiomyopathy is a leading cause of heart failure. Developing a novel therapeutic strategy for diabetic cardiomyopathy and characterizing animal models used for diabetes mellitus (DM) are important. Insulin 2 mutant (Ins2+/-) Akita is a spontaneous, genetic, mouse model for T1DM, which is relevant to humans. There are contrasting reports on systolic dysfunction and pathological remodeling (hypertrophy and fibrosis) in Akita heart. Here, we used magnetic resonance imaging (MRI) approach, a gold standard reference for evaluating cardiac function, to measure ejection fraction (indicator of systolic dysfunction) in Akita. Moreover, we performed Wheat Germ Agglutinin (WGA) and hematoxylin and Eosin stainings to determine cardiac hypertrophy, and Masson's Trichrome and picrosirius red stainings to determine cardiac fibrosis in Akita. MiR-133a, an anti-hypertrophy and anti-fibrosis miRNA, is downregulated in Akita heart. We determined if miR-133a mimic treatment could mitigate systolic dysfunction and remodeling in Akita heart. Our MRI results revealed decreased ejection fraction in Akita as compared to WT and increased ejection fraction in miR-133a mimic-treated Akita. We also found that miR-133a mimic treatment mitigates T1DM-induced cardiac hypertrophy and fibrosis in Akita. We conclude that Akita shows cardiac hypertrophy, fibrosis and systolic dysfunction and miR-133a mimic treatment to Akita could ameliorate them.
Collapse
Affiliation(s)
- Shyam Sundar Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hamid Reza Shahshahan
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Quanliang Shang
- Department of Pediatric Cardiology, Children's Hospital, Omaha, NE, United States
| | - Shelby Kutty
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pediatric Cardiology, Children's Hospital, Omaha, NE, United States
| | - Michael Boska
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paras Kumar Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
26
|
Soga F, Tanaka H, Tatsumi K, Mochizuki Y, Sano H, Toki H, Matsumoto K, Shite J, Takaoka H, Doi T, Hirata KI. Impact of dapagliflozin on left ventricular diastolic function of patients with type 2 diabetic mellitus with chronic heart failure. Cardiovasc Diabetol 2018; 17:132. [PMID: 30296931 PMCID: PMC6174555 DOI: 10.1186/s12933-018-0775-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
Background The objective of this study was to investigate the impact of sodium glucose cotransporter type 2 (SGLT2) inhibitors on left ventricular (LV) diastolic function of type 2 diabetes mellitus (T2DM) patients with heart failure (HF). Methods This trial was a prospective multicenter study of 58 T2DM patients with stable HF at five institutions in Japan. Patients who had been taking at least one antidiabetic drugs other than SGLT2 inhibitors started the administration of 5 mg/day of dapagliflozin. The physical examinations, blood tests, and echocardiography were performed at baseline and 6 months after administration of dapagliflozin. The primary endpoint was defined as a change in mitral inflow E and mitral e′ annular velocities (E/e′) between baseline and 6 months after the administration of dapagliflozin. The secondary end points consisted of a change in brain natriuretic peptide (BNP), LV mass index (LVMI) and left atrial volume index (LAVI). Results E/e′ significantly decreased from 9.3 to 8.5 cm/s (p = 0.020) 6 months after administration of dapagliflozin. LAVI and LVMI significantly decreased from 31 to 26 mL/m2 (p = 0.001), and from 75.0 to 67.0 g/m2 (p < 0.001), respectively, 6 months after administration of dapagliflozin. No significant change was observed in BNP (from 27.9 to 28.9 pg/mL; p = 0.132) 6 months after administration of dapagliflozin, except for a significant decrease from 168.8 to 114.3 pg/mL (p = 0.012) in patients with BNP ≥ 100 pg/mL. Conclusion This prospective multicenter trial showed the beneficial effect of SGLT2 inhibitors on LV diastolic functional parameters for T2DM patients with HF. Our findings may thus offer a new insight into the management of T2DM patients. Trial registration UMIN000019789, Registered 28 September 2014, Date of registration: 11/14/2015, Date of enrolment of the first participant to the trial: 6/15/2016, Date of enrolment of the last participant to the trial: 12/9/2017
Collapse
Affiliation(s)
- Fumitaka Soga
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hidekazu Tanaka
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kazuhiro Tatsumi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.,Tatsumi Clinic, Himeji, Japan
| | - Yasuhide Mochizuki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.,Division of Cardiology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Hiroyuki Sano
- Division of Cardiology, Aijinkai Takatsuki Hospital, Takatsuki, Japan
| | - Hiromi Toki
- Division of Cardiology, Kobe Red Cross Hospital, Kobe, Japan
| | - Kensuke Matsumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Junya Shite
- Division of Cardiology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Hideyuki Takaoka
- Division of Cardiology, Aijinkai Takatsuki Hospital, Takatsuki, Japan
| | - Tomofumi Doi
- Division of Cardiology, Kobe Red Cross Hospital, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
27
|
Li H, Li X, Guo J, Wu G, Dong C, Pang Y, Gao S, Wang Y. Identification of biomarkers and mechanisms of diabetic cardiomyopathy using microarray data. Cardiol J 2018; 27:807-816. [PMID: 30246236 DOI: 10.5603/cj.a2018.0113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 09/20/2018] [Accepted: 05/03/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The study aimed to uncover the regulation mechanisms of diabetic cardiomyopathy (DCM) and provide novel prognostic biomarkers. METHODS The dataset GSE62203 downloaded from the Gene Expression Omnibus database was utilized in the present study. After pretreatment using the Affy package, differentially expressed genes (DEGs) were identified by the limma package, followed by functional enrichment analysis and protein- protein interaction (PPI) network analysis. Furthermore, module analysis was conducted using MCODE plug-in of Cytoscape, and functional enrichment analysis was also performed for genes in the modules. RESULTS A set of 560 DEGs were screened, mainly enriched in the metabolic process and cell cycle related process. Hub nodes in the PPI network were LDHA (lactate dehydrogenase A), ALDOC (aldolase C, fructose-bisphosphate) and ABCE1 (ATP Binding Cassette Subfamily E Member 1), which were also highlighted in Module 1 or Module 2 and predominantly enriched in the processes of glycolysis and ribosome biogenesis. Additionally, LDHA were linked with ALDOC in the PPI network. Besides, activating transcription factor 4 (ATF4) was prominent in Module 3; while myosin heavy chain 6 (MYH6) was highlighted in Module 4 and was mainly involved in muscle cells related biological processes. CONCLUSIONS Five potential biomarkers including LDHA, ALDOC, ABCE1, ATF4 and MYH6 were identified for DCM prognosis.
Collapse
Affiliation(s)
- Hui Li
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Xiaoyan Li
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Jian Guo
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Guifu Wu
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Chunping Dong
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Yaling Pang
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Shan Gao
- Department of Endocrinology, Shaanxi Provincial People's Hospital
| | - Yangwei Wang
- Department of Endocrinology, Shaanxi Provincial People's Hospital.
| |
Collapse
|
28
|
Huang L, Yuan P, Yu P, Kong Q, Xu Z, Yan X, Shen Y, Yang J, Wan R, Hong K, Tang Y, Hu J. O-GlcNAc-modified SNAP29 inhibits autophagy-mediated degradation via the disturbed SNAP29-STX17-VAMP8 complex and exacerbates myocardial injury in type I diabetic rats. Int J Mol Med 2018; 42:3278-3290. [PMID: 30221662 PMCID: PMC6202107 DOI: 10.3892/ijmm.2018.3866] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/29/2018] [Indexed: 12/19/2022] Open
Abstract
The O-linked β-N-acetylglucosamine (O-GlcNAc) modification and autophagy are associated with diabetic myocardial injury, however, the molecular mechanisms between the two processes remain to be fully elucidated. The purpose of the present study was to elucidate the molecular regulation of autophagy by O-GlcNAc-modified synaptosomal-associated protein 29 (SNAP29) in diabetic myocardial injury. A rat model of type I diabetes was established via intraperitoneal injection of streptozotocin (STZ; 55 mg/kg). Significant increases in the O-GlcNAc modification and accumulation of the autophagy markers microtubule-associated protein 1 light chain 3α II/I and P62, which suggest that autophagic flux is inhibited, were observed in rats 8 weeks following STZ induction. Subsequently, the selective O-GlcNAcase inhibitor, thiamet G, increased the level of O-GlcNAc modification, which further disrupted autophagic flux; deteriorated cardiac diastolic function, as indicated by an increased left ventricular filling peak velocity/atrial contraction flow peak velocity ratio shown by echocardiography; and exacerbated myocardial abnormalities, as characterized by cardiomyocyte disorganization and fat and interstitial fibrosis accumulation. By contrast, 6-diazo-5-oxo-L-norleucine, an inhibitor of glucosamine fructose-6-phosphate aminotransferase isomerizing 1, acted as an O-GlcNAc antagonist and reduced the level of O-GlcNAc modification, which maintained autophagic flux and improved cardiac diastolic function. In vitro, high glucose (25 mM) was used to stimulate primary neonatal rat cardiomyocytes (NRCMs). Consistent with the myocardium of diabetic rats, it was also shown in the NRCMs that O-GlcNAc modification of SNAP29 negatively regulated autophagic flux. The application of the short hairpin RNA interference lysosome-associated membrane protein (LAMP2) and the autophagy inhibitor 3-methyladenine demonstrated that high glucose inhibited autophagy-mediated degradation rather than affected the initial stage of autophagy. Finally, co-immunoprecipitation was used to determine the role of the O-GlcNAc-modified substrate protein SNAP29, which acted as an SNAP29-syntaxin-17 (STX17)-vesicle-associated membrane protein 8 (VAMP8) complex during disease progression. The present study is the first, to the best of our knowledge, to demonstrate that SNAP29 is an O-GlcNAc substrate and that an increase in O-GlcNAc-modified SNAP29 inhibits SNAP29-STX17-VAMP8 complex formation, thereby inhibiting the degradation of autophagy and exacerbating myocardial injury in type I diabetic rats.
Collapse
Affiliation(s)
- Lin Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Yuan
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Peng Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiling Kong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zixuan Xu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xia Yan
- The Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Shen
- The Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Juesheng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rong Wan
- The Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Kui Hong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanhua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jinzhu Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
29
|
The Chinese Herb Yi-Qi-Huo-Xue Protects Cardiomyocyte Function in Diabetic Cardiomyopathy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:7316840. [PMID: 29853969 PMCID: PMC5960524 DOI: 10.1155/2018/7316840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/15/2018] [Accepted: 01/30/2018] [Indexed: 11/18/2022]
Abstract
Aims. To study the effect of the Chinese herb Yi-qi-huo-xue on cardiomyopathy in diabetic rats. Methods. Rats were fed a high fat and high glucose diet and injected with 50 ml/kg streptozotocin (STZ) to induce diabetic cardiomyopathy (DCM), followed by treatment with Yi-qi-huo-xue for 4 weeks. We measured the rats' heart weight index, observed the myocardial morphology using hematoxylin eosin (HE) staining, and determined the content of collagen types I and III in the myocardium using enzyme-linked immunosorbent assay (ELISA). We determined Bcl-2, Bax, and P53 protein expression by Western blot analysis and the cardiomyocyte apoptosis rate via a flow cytometry assay. Results. Compared with the rats in the control group, the diabetic rats gained weight and had increased blood sugar levels, an enhanced heart weight index, and increased myocardial pathophysiological damage. There was a decrease in their Bcl-2 expression, and their Bax and P53 expression increased. The Bcl-2/Bax ratio was enhanced, and there was an increase in the content of collagen types I and III in the myocardium. After treatment with Yi-qi-huo-xue, all levels listed above returned to normal. Conclusion. The Chinese herb Yi-qi-huo-xue degraded the myocardial interstitial collagen types I and III to protect the myocardium of the diabetic rats, thus delaying the role of myocardial fibrosis. Yi-qi-huo-xue could play an important role in protecting the myocardium of DCM rats by enhancing the expression of the Bcl-2 protein, inhibiting the expression of the Bax and P53 proteins, increasing the ratio of Bcl-2/Bax, and inhibiting the apoptosis of cardiomyocytes.
Collapse
|
30
|
Ofstad AP, Atar D, Gullestad L, Langslet G, Johansen OE. The heart failure burden of type 2 diabetes mellitus-a review of pathophysiology and interventions. Heart Fail Rev 2018; 23:303-323. [PMID: 29516230 PMCID: PMC5937871 DOI: 10.1007/s10741-018-9685-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetes and heart failure (HF) are both global epidemics with tremendous costs on society with increased rates of HF hospitalizations and worsened prognosis when co-existing, making it a significant "deadly duo." The evidence for pharmacological treatment of HF in patients with type 2 diabetes mellitus (T2DM) stems typically from either subgroup analyses of patients that were recruited to randomized controlled trials of HF interventions, usually in patients with reduced ejection fraction (EF), or from subgroup analyses of HF patients recruited to cardiovascular (CV) outcome trials (CVOT) of glucose lowering agents involving patients with T2DM. Studies in patients with HF with preserved EF are sparse. This review summarizes the literature on pathophysiology and interventions aiming to reduce the HF burden in T2DM and includes HF trials of ACEi, digoxin, β-blocker, ARB, If-blocker, MRA, and ARNI involving 38,600 patients, with or without prevalent diabetes, and CV outcome trials in T2DM involving 74,351 patients, with or without prevalent HF. In all HF trials, HF outcomes by prevalent diabetes were reported with an incremental risk of HF and death confessed by prevalent diabetes and a treatment effect similar to those without diabetes. All T2DM CVOTs reported on HF outcomes with heterogeneity between trials with two reporting benefits (empagliflozin and canagliflozin) and two reporting increased risk (saxagliptin, pioglitazone). In vulnerable T2DM patients with concomitant HF, guideline-recommended HF drugs are effective. When choosing glucose-lowering therapy, outcomes from available CVOTs should be considered.
Collapse
Affiliation(s)
- Anne Pernille Ofstad
- Bærum Hospital, Vestre Viken HF, Rud, Norway.
- Medical Department, Boehringer Ingelheim, Asker, Norway.
| | - Dan Atar
- Department of Cardiology B, Oslo University Hospital, Ullevål, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Gisle Langslet
- Rikshospitalet, Lipid Clinic, Oslo University Hospital, Oslo, Norway
| | - Odd Erik Johansen
- Bærum Hospital, Vestre Viken HF, Rud, Norway
- Medical Department, Boehringer Ingelheim, Asker, Norway
| |
Collapse
|
31
|
Hathaway QA, Pinti MV, Durr AJ, Waris S, Shepherd DL, Hollander JM. Regulating microRNA expression: at the heart of diabetes mellitus and the mitochondrion. Am J Physiol Heart Circ Physiol 2018; 314:H293-H310. [PMID: 28986361 PMCID: PMC5867655 DOI: 10.1152/ajpheart.00520.2017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/20/2017] [Accepted: 10/02/2017] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes mellitus is a major risk factor for cardiovascular disease and mortality. Uncontrolled type 2 diabetes mellitus results in a systemic milieu of increased circulating glucose and fatty acids. The development of insulin resistance in cardiac tissue decreases cellular glucose import and enhances mitochondrial fatty acid uptake. While triacylglycerol and cytotoxic lipid species begin to accumulate in the cardiomyocyte, the energy substrate utilization ratio of free fatty acids to glucose changes to almost entirely free fatty acids. Accumulating evidence suggests a role of miRNA in mediating this metabolic transition. Energy substrate metabolism, apoptosis, and the production and response to excess reactive oxygen species are regulated by miRNA expression. The current momentum for understanding the dynamics of miRNA expression is limited by a lack of understanding of how miRNA expression is controlled. While miRNAs are important regulators in both normal and pathological states, an additional layer of complexity is added when regulation of miRNA regulators is considered. miRNA expression is known to be regulated through a number of mechanisms, which include, but are not limited to, epigenetics, exosomal transport, processing, and posttranscriptional sequestration. The purpose of this review is to outline how mitochondrial processes are regulated by miRNAs in the diabetic heart. Furthermore, we will highlight the regulatory mechanisms, such as epigenetics, exosomal transport, miRNA processing, and posttranslational sequestration, that participate as regulators of miRNA expression. Additionally, current and future treatment strategies targeting dysfunctional mitochondrial processes in the diseased myocardium, as well as emerging miRNA-based therapies, will be summarized.
Collapse
Affiliation(s)
- Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
- Toxicology Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Mark V Pinti
- Division of Pharmaceutical and Pharmacological Sciences, West Virginia School of Pharmacy , Morgantown, West Virginia
| | - Andrya J Durr
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Shanawar Waris
- Department of Biomedical Engineering, West Virginia College of Engineering , Morgantown, West Virginia
| | - Danielle L Shepherd
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
- Toxicology Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| |
Collapse
|
32
|
You S, Qian J, Sun C, Zhang H, Ye S, Chen T, Xu Z, Wang J, Huang W, Liang G. An Aza resveratrol-chalcone derivative 6b protects mice against diabetic cardiomyopathy by alleviating inflammation and oxidative stress. J Cell Mol Med 2018; 22:1931-1943. [PMID: 29327811 PMCID: PMC5824376 DOI: 10.1111/jcmm.13477] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/04/2017] [Indexed: 12/14/2022] Open
Abstract
Inflammation and oxidative stress play a crucial role in the development of diabetic cardiomyopathy (DCM). We previously had synthesized an Aza resveratrol–chalcone derivative 6b, of which effectively suppressing lipopolysaccharide (LPS)‐induced inflammatory response in macrophages. This study aimed to investigate the potential protective effect of 6b on DCM and underlying mechanism. In H9c2 myocardial cells, 6b potently decreased high glucose (HG)‐induced cell fibrosis, hypertrophy and apoptosis, alleviating inflammatory response and oxidant stress. In STZ‐induced type 1 diabetic mice (STZ‐DM1), orally administration with 6b for 16 weeks significantly attenuated cardiac hypertrophy, apoptosis and fibrosis. The expression of inflammatory cytokines and oxidative stress biomarkers was also suppressed by 6b distinctly, without affecting blood glucose and body weight. The anti‐inflammatory and antioxidative activities of 6b were mechanistic associated with nuclear factor‐kappa B (NF‐κB) nucleus entry blockage and Nrf2 activation both in vitro and in vivo. The results indicated that 6b can be a promising cardioprotective agent in treatment of DCM via inhibiting inflammation and alleviating oxidative stress. This study also validated the important role of NF‐κB and Nrf2 taken in the pathogenesis of DCM, which could be therapeutic targets for diabetic comorbidities.
Collapse
Affiliation(s)
- Shengban You
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Translational Medicine Center in Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chuchu Sun
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Translational Medicine Center in Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hailing Zhang
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiju Ye
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Taiwei Chen
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheng Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingying Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Translational Medicine Center in Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
33
|
Targeting miRNA for Therapy of Juvenile and Adult Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:47-59. [PMID: 29754174 DOI: 10.1007/978-3-319-74470-4_4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prevalence of diabetes mellitus (DM), a multifactorial disease often diagnosed with high blood glucose levels, is rapidly increasing in the world. Association of DM with multi-organ dysfunction including cardiomyopathy makes it a leading cause of morbidity and mortality. There are two major types of DM: type 1 DM (T1D) and type 2 DM (T2D). T1D is diagnosed by reduced levels of insulin and high levels of glucose in the blood. It is caused due to pancreatic beta cell destruction/loss, and mostly found in juveniles (juvenile DM). T2D is diagnosed by increased levels of insulin and glucose in the blood. It is caused due to insulin receptor dysfunction, and mostly found in the adults (adult DM). Both T1D and T2D impair cardiac muscle function, which is referred to as diabetic cardiomyopathy. We and others have reported that miRNAs, a novel class of tiny non-coding regulatory RNAs, are differentially expressed in the diabetic heart and they contribute to diabetic cardiomyopathy. Here, we elaborated the biogenesis of miRNA, how miRNA regulates a gene, cardioprotective roles of different miRNAs including miRNAs present in exosomes, underlying molecular mechanisms by which miRNA ameliorates diabetic cardiomyopathy, and the role of miRNA as a potential therapeutic target for juvenile and adult diabetic cardiomyopathy.
Collapse
|
34
|
Yang YY, Sun XT, Li ZX, Chen WY, Wang X, Liang ML, Shi H, Yang ZS, Zeng WT. Protective effect of angiotensin-(1-7) against hyperglycaemia-induced injury in H9c2 cardiomyoblast cells via the PI3K̸Akt signaling pathway. Int J Mol Med 2017; 41:1283-1292. [PMID: 29286068 PMCID: PMC5819934 DOI: 10.3892/ijmm.2017.3322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 11/20/2017] [Indexed: 01/17/2023] Open
Abstract
Angiotensin-(1-7) [Ang-(1-7)], a heptapeptide mainly generated from cleavage of AngⅠ and AngⅡ, possesses physiological and pharmacological properties, including anti‑inflammatory and antidiabetic properties. Activation of the phosphoinositide 3-kinase and protein kinase B (PI3K̸Akt) signaling pathway has been confirmed to participate in cardioprotection against hyperglycaemia-induced injury. The aim of the present study was to test the hypothesis that Ang-(1-7) protects H9c2 cardiomyoblast cells against high glucose (HG)-induced injury by activating the PI3K̸Akt pathway. To examine this hypothesis, H9c2 cells were treated with 35 mmol/l (mM) glucose (HG) for 24 h to establish a HG-induced cardiomyocyte injury model. The cells were co-treated with 1 µmol/l (µM) Ang-(1-7) and 35 mM glucose. The findings of the present study demonstrated that exposure of H9c2 cells to HG for 24 h markedly induced injury, as evidenced by an increase in the percentage of apoptotic cells, generation of reactive oxygen species and level of inflammatory cytokines, as well as a decline in cell viability and mitochondrial luminosity. These injuries were significantly attenuated by co-treatment of the cells with Ang-(1-7) and HG. In addition, PI3K̸Akt phosphorylation was suppressed by HG treatment, but this effect was abolished when the H9c2 cells were co-treated with Ang-(1-7) and HG. Furthermore, the cardioprotection of Ang-(1-7) against HG-induced injury in H9c2 cardiomyoblasts was highly attenuated in the presence of either D-Ala7-Ang-(1-7) (A-779, an antagonist of the Mas receptor) or LY294002 (an inhibitor of PI3K̸Akt). In conclusion, the present study provided new evidence that Ang-(1-7) protects H9c2 cardiomyoblasts against HG-induced injury by activating the PI3K̸Akt signaling pathway.
Collapse
Affiliation(s)
- Yi-Ying Yang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiu-Ting Sun
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zheng-Xun Li
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wei-Yan Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Xiang Wang
- Department of Cardiology, Laiwu City People's Hospital, Laiwu, Shandong 27110, P.R. China
| | - Mei-Ling Liang
- Department of Cardiology, Sun Yat-Sen Cardiovascular Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Hui Shi
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhi-Sheng Yang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wu-Tao Zeng
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
35
|
Wei X, Yang Y, Jiang YJ, Lei JM, Guo JW, Xiao H. Relaxin ameliorates high glucose-induced cardiomyocyte hypertrophy and apoptosis via the Notch1 pathway. Exp Ther Med 2017; 15:691-698. [PMID: 29399073 PMCID: PMC5772593 DOI: 10.3892/etm.2017.5448] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the role of relaxin (RLX) on high glucose (HG)-induced cardiomyocyte hypertrophy and apoptosis, as well as the possible molecular mechanism. H9c2 cells were exposed to 33 mmol/l HG with or without RLX (100 nmol/ml). Cell viability, apoptosis, oxidative stress, cell hypertrophy and the levels of Notch1, hairy and enhancer of split 1 (hes1), atrial natriuretic polypeptide (ANP), brain natriuretic peptide (BNP), manganese superoxide dismutase (MnSOD), cytochrome C and caspase-3 were assessed in cardiomyocytes. Compared with the HG group, the viability of H9c2 cells was increased by RLX in a time- and dose-dependent manner, and was accompanied with a significant reduction in apoptosis. Furthermore, RLX significantly suppressed the formation of reactive oxygen species and malondialdehyde, and enhanced the activity of SOD. In addition, the levels of ANP, BNP, cytochrome C and caspase-3 were increased and Notch1, hes1 and MnSOD were inhibited in the HG group compared with those in the normal group. However, the Notch inhibitor DAPT almost abolished the protective effects of RLX. These results suggested that RLX protected cardiomyocytes from HG-induced hypertrophy and apoptosis partly through a Notch1-dependent pathway, which may be associated with reducing oxidative stress.
Collapse
Affiliation(s)
- Xiao Wei
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yuan Yang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yin-Jiu Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jian-Ming Lei
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jing-Wen Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hua Xiao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
36
|
Copier CU, León L, Fernández M, Contador D, Calligaris SD. Circulating miR-19b and miR-181b are potential biomarkers for diabetic cardiomyopathy. Sci Rep 2017; 7:13514. [PMID: 29044172 PMCID: PMC5647433 DOI: 10.1038/s41598-017-13875-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy is characterized by metabolic changes in the myocardium that promote a slow and silent dysfunction of muscle fibers, leading to myocardium remodelling and heart failure, independently of the presence of coronary artery diseases or hypertension. At present, no imaging methods allow an early diagnosis of this disease. Circulating miRNAs in plasma have been proposed as biomarkers in the prognosis of several cardiac diseases. This study aimed to determine whether circulating miRNAs could be potential biomarkers of diabetic cardiomyopathy. Mice that were fed with a high fat diet for 16 months, showed metabolic syndrome manifestations, cardiac hypertrophy (without hypertension) and a progressive cardiac function decline. At 16 months, when maximal degree of cardiac dysfunction was observed, 15 miRNAs from a miRNA microarray screening in myocardium were selected. Then, selected miRNAs expression in myocardium (at 4 and 16 months) and plasma (at 4, 12 and 16 months) were measured by RT-qPCR. Circulating miR-19b-3p and miR-181b-5p levels were associated with myocardium levels during the development of diabetic cardiomyopathy (in terms of cardiac dysfunction), suggesting that these miRNAs could be suitable biomarkers of this disease in asymptomatic diabetic patients.
Collapse
Affiliation(s)
- Camila Uribe Copier
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Las Condes 12.438, Lo Barnechea, Santiago, Chile
| | - Luis León
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Mauricio Fernández
- Departamento de Cardiología, Clínica Alemana de Santiago - Universidad del Desarrollo, Vitacura 5951, Vitacura, Santiago, Chile
| | - David Contador
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Las Condes 12.438, Lo Barnechea, Santiago, Chile
| | - Sebastián D Calligaris
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Las Condes 12.438, Lo Barnechea, Santiago, Chile.
| |
Collapse
|
37
|
Kennedy L, Kaltenbrun E, Greco TM, Temple B, Herring LE, Cristea IM, Conlon FL. Formation of a TBX20-CASZ1 protein complex is protective against dilated cardiomyopathy and critical for cardiac homeostasis. PLoS Genet 2017; 13:e1007011. [PMID: 28945738 PMCID: PMC5629033 DOI: 10.1371/journal.pgen.1007011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/05/2017] [Accepted: 09/07/2017] [Indexed: 01/01/2023] Open
Abstract
By the age of 40, one in five adults without symptoms of cardiovascular disease are at risk for developing congestive heart failure. Within this population, dilated cardiomyopathy (DCM) remains one of the leading causes of disease and death, with nearly half of cases genetically determined. Though genetic and high throughput sequencing-based approaches have identified sporadic and inherited mutations in a multitude of genes implicated in cardiomyopathy, how combinations of asymptomatic mutations lead to cardiac failure remains a mystery. Since a number of studies have implicated mutations of the transcription factor TBX20 in congenital heart diseases, we investigated the underlying mechanisms, using an unbiased systems-based screen to identify novel, cardiac-specific binding partners. We demonstrated that TBX20 physically and genetically interacts with the essential transcription factor CASZ1. This interaction is required for survival, as mice heterozygous for both Tbx20 and Casz1 die post-natally as a result of DCM. A Tbx20 mutation associated with human familial DCM sterically interferes with the TBX20-CASZ1 interaction and provides a physical basis for how this human mutation disrupts normal cardiac function. Finally, we employed quantitative proteomic analyses to define the molecular pathways mis-regulated upon disruption of this novel complex. Collectively, our proteomic, biochemical, genetic, and structural studies suggest that the physical interaction between TBX20 and CASZ1 is required for cardiac homeostasis, and further, that reduction or loss of this critical interaction leads to DCM. This work provides strong evidence that DCM can be inherited through a digenic mechanism. A molecular understanding of cardiomyocyte development is an essential goal for improving clinical approaches to CHD. While TBX20 is an essential transcription factor for heart development and its disease relevance is well established, many fundamental questions remain about the mechanism of TBX20 function. Principle among these is how TBX20 mutations associated with adult dilated cardiomyopathy circumvent (DCM) the essential embryonic requirement for TBX20 in heart development. Here we report using an integrated approach that TBX20 complexes with the cardiac transcription factor CASZ1 in vivo. We confirmed TBX20 and CASZ1 interact biochemically and genetically, and show mice heterozygous for both Tbx20 and Casz1 die, beginning at 4 to 8 weeks post birth, exhibiting hallmarks of DCM. Interestingly, the human mutant TBX20F256I bypasses the early essential requirement for TBX20 but leads to DCM. We report here that TBX20F256I disrupts the TBX20-CASZ1 interaction, ascribing clinical relevance to this protein complex. Further, by using quantitative proteomics we have identified the molecular pathways altered in TBX20-CASZ1-mediated DCM. Together, these results identify a novel interaction between TBX20 and CASZ1 that is essential for maintaining cardiac homeostasis and imply that DCM can be inherited through a digenic mechanism.
Collapse
Affiliation(s)
- Leslie Kennedy
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological & Genome Sciences, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, UNC-Chapel Hill, Chapel Hill, NC, United States of America
| | - Erin Kaltenbrun
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological & Genome Sciences, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, UNC-Chapel Hill, Chapel Hill, NC, United States of America
| | - Todd M. Greco
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States of America
| | - Brenda Temple
- R.L. Juliano Structural Bioinformatics Core, Department of Biochemistry and Biophysics, UNC-Chapel Hill, Chapel Hill, NC, United States of America
| | - Laura E. Herring
- UNC Proteomics Core Facility, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Department of Pharmacology, UNC-Chapel Hill, Chapel Hill, NC, United States of America
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States of America
| | - Frank L. Conlon
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological & Genome Sciences, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biology, UNC-Chapel Hill, Chapel Hill, NC, United States of America
- * E-mail:
| |
Collapse
|
38
|
Yu LY, Shi WL, Guo XG. Cardio-Protective Role of Gingerol along with Prominent Anti-Diabetic Cardiomyopathy Action in A Streptozotocin-Induced Diabetes Mellitus Rat Model. CELL JOURNAL 2017; 19:469-475. [PMID: 28836409 PMCID: PMC5570412 DOI: 10.22074/cellj.2017.4509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/26/2016] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Diabetic cardiomyopathy (DCM) is characterized as a coronary heart disease which expands during diabetes due to alterations in the myocardial function and structure. The currentstudy intends to elucidate the protective effect of gingerol on DCM in a streptozotocin (STZ)-induced diabetes mellitus (DM) rat model. MATERIALS AND METHODS In this experimental study, the animals were divided into three groups: normal control, DM control, and DM+gingerol (10 mg/kg). The body weights of all rats were estimated at regular intervals. The myocardial profile, oxidative stress, and activities of metabolic enzymes were also scrutinized. The proinflammatory cytokine levels together with cellular protein expression connected with apoptosis were estimated via Western blot analysis. RESULTS The rats that suffered from DCM exhibited abnormal levels of myocardial markers, aberrant metabolic enzymatic activity, elevated concentrations of inflammatory factors, and enhanced oxidative stress parameters along with increased cell death apoptosis. Whereas gingerol showed protective effects on the treated rats by an improved antioxidant defense system. CONCLUSIONS The current findings suggested that gingerol is effective in the treatment of DCM by inhibition of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Li-Ya Yu
- Department of Cardiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Wen-Lei Shi
- Department of Cardiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Xin-GuiGuo Guo
- Department of Cardiology, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
39
|
Cardiac transcriptome profiling of diabetic Akita mice using microarray and next generation sequencing. PLoS One 2017; 12:e0182828. [PMID: 28837672 PMCID: PMC5570368 DOI: 10.1371/journal.pone.0182828] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/25/2017] [Indexed: 01/10/2023] Open
Abstract
Although diabetes mellitus (DM) causes cardiomyopathy and exacerbates heart failure, the underlying molecular mechanisms for diabetic cardiomyopathy/heart failure are poorly understood. Insulin2 mutant (Ins2+/-) Akita is a mouse model of T1DM, which manifests cardiac dysfunction. However, molecular changes at cardiac transcriptome level that lead to cardiomyopathy remain unclear. To understand the molecular changes in the heart of diabetic Akita mice, we profiled cardiac transcriptome of Ins2+/- Akita and Ins2+/+ control mice using next generation sequencing (NGS) and microarray, and determined the implications of differentially expressed genes on various heart failure signaling pathways using Ingenuity pathway (IPA) analysis. First, we validated hyperglycemia, increased cardiac fibrosis, and cardiac dysfunction in twelve-week male diabetic Akita. Then, we analyzed the transcriptome levels in the heart. NGS analyses on Akita heart revealed 137 differentially expressed transcripts, where Bone Morphogenic Protein-10 (BMP10) was the most upregulated and hairy and enhancer of split-related (HELT) was the most downregulated gene. Moreover, twelve long non-coding RNAs (lncRNAs) were upregulated. The microarray analyses on Akita heart showed 351 differentially expressed transcripts, where vomeronasal-1 receptor-180 (Vmn1r180) was the most upregulated and WD Repeat Domain 83 Opposite Strand (WDR83OS) was the most downregulated gene. Further, miR-101c and H19 lncRNA were upregulated but Neat1 lncRNA was downregulated in Akita heart. Eleven common genes were upregulated in Akita heart in both NGS and microarray analyses. IPA analyses revealed the role of these differentially expressed genes in key signaling pathways involved in diabetic cardiomyopathy. Our results provide a platform to initiate focused future studies by targeting these genes and/or non-coding RNAs, which are differentially expressed in Akita hearts and are involved in diabetic cardiomyopathy.
Collapse
|
40
|
Wang WK, Lu QH, Wang X, Wang B, Wang J, Gong HP, Wang L, Li H, Du YM. Ulinastatin attenuates diabetes-induced cardiac dysfunction by the inhibition of inflammation and apoptosis. Exp Ther Med 2017; 14:2497-2504. [PMID: 28962186 PMCID: PMC5609313 DOI: 10.3892/etm.2017.4824] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 05/19/2017] [Indexed: 12/20/2022] Open
Abstract
Ulinastatin exhibits anti-inflammatory activity and protects the heart from ischemia/reperfusion injury. However, whether ulinastatin has a protective effect in diabetic cardiomyopathy is yet to be elucidated. The aim of the present study was to investigate the protective effects of ulinastatin against diabetic cardiomyopathy and its underlying mechanisms. A C57/BL6J mice model of diabetic cardiomyopathy was used and mice were randomly assigned to three groups: Control group, diabetes mellitus (DM) group and DM + ulinastatin treatment group. Cardiac function was assessed using echocardiography and the level of inflammatory cytokine high mobility group box 1 (HMGB1) expression was measured using histopathological examination and reverse transcription-quantitative polymerase chain reaction. The levels of tumor necrosis factor (TNF)-α and interleukin (IL)-6 were measured using western blotting and ELISA. The apoptosis rate in the myocardium was assessed by TUNEL assay. Caspase-3 activation, expression of B-cell lymphoma 2 (Bcl-2) and Bcl-2 associated × (Bax) were measured using western blotting, as was the activity of the mitogen activated protein kinase (MAPK) signaling pathway. The results indicated that ulinastatin significantly improved cardiac function in mice with DM. Ulinastatin treatment significantly downregulated HMGB1, TNF-α and IL-6 expression (P<0.05) and significantly reduced the percentage of apoptotic cardiomyocytes (P<0.05) via reduction of caspase-3 activation and the ratio of Bax/Bcl-2 in diabetic hearts (P<0.05). In addition, ulinastatin attenuated the activation of the MAPK signaling pathway. In conclusion, ulinastatin had a protective effect against DM-induced cardiac dysfunction in a mouse model. This protective effect may be associated with the anti-inflammatory and anti-apoptotic abilities of ulinastatin via the MAPK signaling pathway.
Collapse
Affiliation(s)
- Wen-Ke Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Qing-Hua Lu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xin Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ben Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Juan Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Hui-Ping Gong
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lin Wang
- Department of Healthcare, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Hao Li
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yi-Meng Du
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
41
|
Lee WS, Kim J. Diabetic cardiomyopathy: where we are and where we are going. Korean J Intern Med 2017; 32:404-421. [PMID: 28415836 PMCID: PMC5432803 DOI: 10.3904/kjim.2016.208] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 01/08/2017] [Indexed: 12/15/2022] Open
Abstract
The global burden of diabetes mellitus and its related complications are currently increasing. Diabetes mellitus affects the heart through various mechanisms including microvascular impairment, metabolic disturbance, subcellular component abnormalities, cardiac autonomic dysfunction, and a maladaptive immune response. Eventually, diabetes mellitus can cause functional and structural changes in the myocardium without coronary artery disease, a disorder known as diabetic cardiomyopathy (DCM). There are many diagnostic tools and management options for DCM, although it is difficult to detect its development and effectively prevent its progression. In this review, we summarize the current research regarding the pathophysiology and pathogenesis of DCM. Moreover, we discuss emerging diagnostic evaluation methods and treatment strategies for DCM, which may help our understanding of its underlying mechanisms and facilitate the identification of possible new therapeutic targets.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Correspondence to Jaetaek Kim, M.D. Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea Tel: +82-2-6299-1397 Fax: +82-2-6299-1390 E-mail:
| |
Collapse
|
42
|
Qiao F, Longley KR, Feng S, Schnack S, Gao H, Li Y, Schlenker EH, Wang H. Reduced body weight gain in ubiquilin-1 transgenic mice is associated with increased expression of energy-sensing proteins. Physiol Rep 2017; 5:e13260. [PMID: 28420763 PMCID: PMC5408289 DOI: 10.14814/phy2.13260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 12/14/2022] Open
Abstract
Ubiquilin-1 (Ubqln1), a ubiquitin-like protein, is implicated in a variety of pathophysiological processes, but its role in mediating body weight gain or metabolism has not been determined. Here, we demonstrate that global overexpression of Ubqln1 in a transgenic (Tg) mouse reduces the animal's body weight gain. The decreased body weight gain in Tg mice is associated with lower visceral fat content and higher metabolic rate. The Ubqln1 Tg mice exhibited reduced leptin and insulin levels as well as increased insulin sensitivity manifested by homeostatic model assessment of insulin resistance. Additionally, the reduced body weight in Tg mice was associated with the upregulation of two energy-sensing proteins, sirtuin1 (SIRT1) in the hypothalamus and AMP-activated protein kinase (AMPK) in the skeletal muscle. Consistent with the in vivo results, overexpression of Ubqln1 significantly increased SIRT1 and AMPK levels in the mouse embryonic fibroblast cell culture. Thus, our results not only establish the link between Ubqln1 and body weight regulation but also indicate that the metabolic function of Ubqln1 on body weight may be through regulating energy-sensing proteins.
Collapse
Affiliation(s)
- Fangfang Qiao
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Kirsty R Longley
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Shelley Feng
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Sabrina Schnack
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Hongbo Gao
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Yifan Li
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Evelyn H Schlenker
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Hongmin Wang
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
43
|
Zhang B, Shen Q, Chen Y, Pan R, Kuang S, Liu G, Sun G, Sun X. Myricitrin Alleviates Oxidative Stress-induced Inflammation and Apoptosis and Protects Mice against Diabetic Cardiomyopathy. Sci Rep 2017; 7:44239. [PMID: 28287141 PMCID: PMC5347164 DOI: 10.1038/srep44239] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/06/2017] [Indexed: 12/22/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) has been increasingly considered as a main cause of heart failure and death in diabetic patients. At present, no effective treatment exists to prevent its development. In the present study, we describe the potential protective effects and mechanisms of myricitrin (Myr) on the cardiac function of streptozotosin-induced diabetic mice and on advanced glycation end products (AGEs)-induced H9c2 cardiomyocytes. In vitro experiments revealed that pretreatment with Myr significantly decreased AGEs-induced inflammatory cytokine expression, limited an increase in ROS levels, and reduced cell apoptosis, fibrosis, and hypertrophy in H9c2 cells. These effects are correlated with Nrf2 activation and NF-κB inhibition. In vivo investigation demonstrated that oral administration of Myr at 300 mg/kg/day for 8 weeks remarkably decreased the expression of enzymes associated with cardiomyopathy, as well as the expression of inflammatory cytokines and apoptotic proteins. Finally, Myr improved diastolic dysfunction and attenuated histological abnormalities. Mechanistically, Myr attenuated diabetes-induced Nrf2 inhibition via the regulation of Akt and ERK phosphorylation in the diabetic heart. Collectively, these results strongly indicate that Myr exerts cardioprotective effects against DCM through the blockage of inflammation, oxidative stress, and apoptosis. This suggests that Myr might be a potential therapeutic agent for the treatment of DCM.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Qiang Shen
- Center of Research and Development on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin 150076, China
| | - Yaping Chen
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Ruile Pan
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Guiyan Liu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| |
Collapse
|
44
|
Liu X, Liu S. Role of microRNAs in the pathogenesis of diabetic cardiomyopathy. Biomed Rep 2017; 6:140-145. [PMID: 28357065 DOI: 10.3892/br.2017.841] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/10/2017] [Indexed: 01/10/2023] Open
Abstract
The morbidity of diabetes mellitus has been increasing annually. As a progressive metabolic disorder, chronic complications occur in the late stage of diabetes. In addition, cardiovascular diseases account for the major cause of morbidity and mortality among the diabetic population worldwide. Diabetic cardiomyopathy (DCM) is a type of diabetic heart disease. Patients with DCM show symptoms and signs of heart failure while no specific cause, such as coronary disease, hypertension, alcohol consumption, or other structural heart diseases has been identified. The pathogenesis of DCM is complex and has not been well understood until recently. MicroRNAs (miRs) belong to a novel family of highly conserved, short, non-coding, single-stranded RNA molecules that regulate transcriptional and post-transcriptional gene expression. Furthermore, recent studies have demonstrated an association between miRs and DCM. In the current review, the role of miRs in the pathogenesis of DCM is summarized. It was concluded that miRs contribute to the regulation of cardiomyocyte hypertrophy, myocardial fibrosis, cardiomyocyte apoptosis, mitochondrial dysfunction, myocardial electrical remodeling, epigenetic modification and various other pathophysiological processes of DCM. These studies may provide novel insights into targets for prevention and treatment of the disease.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, P.R. China
| | - Shixue Liu
- Emergency Department, Rizhao Chinese Medicine Hospital, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
45
|
Salem ESB, Fan GC. Pathological Effects of Exosomes in Mediating Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 998:113-138. [PMID: 28936736 DOI: 10.1007/978-981-10-4397-0_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic subjects are at risk of developing cardiovascular disease, which accounts for 60-80% of diabetes-related mortality. Atherosclerosis is still considered as a leading cause of heart failure in diabetic patients, but it could also be an intrinsic and long-term effect of contractile cardiac cells malfunction, known as diabetic cardiomyopathy (DCM). Pathologically, this cardiac dysfunction is manifested by inflammation, apoptosis, fibrosis, hypertrophy and altered cardiomyocytes metabolism. However, the underlying molecular mechanisms of DCM pathophysiology are not clearly understood. Recent and several studies have suggested that exosomes are contributed to the regulation of cell-to-cell communication. Therefore, their in-depth investigation can interpret the complex pathophysiology of DCM. Structurally, exosomes are membrane-bounded vesicles (10-200 nm in diameter), which are actively released from all types of cells and detected in all biological fluids. They carry a wide array of bioactive molecules, including mRNAs, none-coding RNAs (e.g., microRNAs, lncRNAs, circRNAs, etc), proteins and lipids. Importantly, the abundance and nature of loaded molecules inside exosomes fluctuate with cell types and pathological conditions. This chapter summarizes currently available studies on the exosomes' role in the regulation of diabetic cardiomyopathy. Specifically, the advances on the pathological effects of exosomes in diabetic cardiomyopathy as well as the therapeutic potentials and perspectives are also discussed.
Collapse
Affiliation(s)
- Esam S B Salem
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 5872 Care Mail Loc-0575, Cincinnati, OH, 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 5872 Care Mail Loc-0575, Cincinnati, OH, 45267, USA.
| |
Collapse
|
46
|
Dong S, Zhang R, Liang Y, Shi J, Li J, Shang F, Mao X, Sun J. Changes of myocardial lipidomics profiling in a rat model of diabetic cardiomyopathy using UPLC/Q-TOF/MS analysis. Diabetol Metab Syndr 2017; 9:56. [PMID: 28736579 PMCID: PMC5520292 DOI: 10.1186/s13098-017-0249-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 06/28/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a serious cardiac dysfunction induced by changes in the structure and contractility of the myocardium that are initiated in part by alterations in energy substrates. The underlying mechanisms of DCM are still under controversial. The observation of lipids, especially lipidomics profiling, can provide an insight into the know the biomarkers of DCM. The aim of our research was to detect changes of myocardial lipidomics profiling in a rat model of diabetic cardiomyopathy. METHODS Diabetic cardiomyopathy was induced by feeding a high-sucrose/fat diet (HSFD) for 28 weeks and streptozotocin (30 mg/kg, intraperitoneally). The ultra-high-performance liquid chromatography (UPLC) coupled to quadruple time-of flight (QTOF) mass spectrometer was used to acquire and analyze the lipidomics profiling of myocardial tissue. Meanwhile, parameters of cardiac function were collected using cardiac catheterization, and the cardiac index was calculated, and fasting blood glucose and lipid levels were measured by an ultraviolet spectrophotometric method. RESULTS We detected 3023 positive ion peaks and 300 negative ion peaks. Levels of phosphatidylcholine (PC) (22:6/18:2), PC (22:6/18:1), PC (20:4/16:1), PC (16:1/18:3), phosphatidylethanolamine (PE) (20:4/18:2), and PE (20:4/16:0) were down-regulated, and PC (20:2/18:2), PC (18:0/16:0), and PC (20:4/18:0) were up-regulated in DCM model rats, when compared with control rats. Cardiac functions signed as values of left ventricular systolic pressure, maximal uprising velocity of left ventricular pressure and maximal decreasing velocity of left ventricular pressure were injured by 21-44%, and the cardiac index was increased by 25%, and fasting blood glucose and lipids were increased by 34-368%. Meanwhile, the cardiac lipid-related biomarkers have significant correlation with changes of cardiac function and cardiac index. CONCLUSIONS UPLC/Q-TOF/MS analysis data suggested changes of some potential lipid biomarkers in the development of cardiac dysfunction and hypertrophy of diabetic cardiomyopathy, which may serve as potential important targets for clinical diagnosis and therapeutic intervention of DCM in the future.
Collapse
Affiliation(s)
- Shifen Dong
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wang Jing Zhong Huan South Road, Chaoyang District, Beijing, 100102 China
| | - Rong Zhang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wang Jing Zhong Huan South Road, Chaoyang District, Beijing, 100102 China
| | - Yaoyue Liang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wang Jing Zhong Huan South Road, Chaoyang District, Beijing, 100102 China
| | - Jiachen Shi
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wang Jing Zhong Huan South Road, Chaoyang District, Beijing, 100102 China
| | - Jiajia Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wang Jing Zhong Huan South Road, Chaoyang District, Beijing, 100102 China
| | - Fei Shang
- Beijing University of Chemical Technology, Beijing, 100029 China
| | - Xuezhou Mao
- Biostatistics and Programming, Sanofi U.S., Bridgewater, NJ 08807 USA
| | - Jianning Sun
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wang Jing Zhong Huan South Road, Chaoyang District, Beijing, 100102 China
| |
Collapse
|
47
|
Nandi SS, Zheng H, Sharma NM, Shahshahan HR, Patel KP, Mishra PK. Lack of miR-133a Decreases Contractility of Diabetic Hearts: A Role for Novel Cross Talk Between Tyrosine Aminotransferase and Tyrosine Hydroxylase. Diabetes 2016; 65:3075-90. [PMID: 27411382 PMCID: PMC5033264 DOI: 10.2337/db16-0023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/01/2016] [Indexed: 01/21/2023]
Abstract
MicroRNAs (miRNAs) have a fundamental role in diabetic heart failure. The cardioprotective miRNA-133a (miR-133a) is downregulated, and contractility is decreased in diabetic hearts. Norepinephrine (NE) is a key catecholamine that stimulates contractility by activating β-adrenergic receptors (β-AR). NE is synthesized from tyrosine by the rate-limiting enzyme, tyrosine hydroxylase (TH), and tyrosine is catabolized by tyrosine aminotransferase (TAT). However, the cross talk/link between TAT and TH in the heart is unclear. To determine whether miR-133a plays a role in the cross talk between TH and TAT and regulates contractility by influencing NE biosynthesis and/or β-AR levels in diabetic hearts, Sprague-Dawley rats and miR-133a transgenic (miR-133aTg) mice were injected with streptozotocin to induce diabetes. The diabetic rats were then treated with miR-133a mimic or scrambled miRNA. Our results revealed that miR-133a mimic treatment improved the contractility of the diabetic rat's heart concomitant with upregulation of TH, cardiac NE, β-AR, and downregulation of TAT and plasma levels of NE. In miR-133aTg mice, cardiac-specific miR-133a overexpression prevented upregulation of TAT and suppression of TH in the heart after streptozotocin was administered. Moreover, miR-133a overexpression in CATH.a neuronal cells suppressed TAT with concomitant upregulation of TH, whereas knockdown and overexpression of TAT demonstrated that TAT inhibited TH. Luciferase reporter assay confirmed that miR-133a targets TAT. In conclusion, miR-133a controls the contractility of diabetic hearts by targeting TAT, regulating NE biosynthesis, and consequently, β-AR and cardiac function.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Diabetes Mellitus, Experimental/metabolism
- HEK293 Cells
- Hemodynamics/physiology
- Humans
- Immunohistochemistry
- Male
- Mice, Transgenic
- MicroRNAs/genetics
- MicroRNAs/metabolism
- MicroRNAs/physiology
- Myocardial Contraction/genetics
- Myocardial Contraction/physiology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Norepinephrine/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tyrosine 3-Monooxygenase/genetics
- Tyrosine 3-Monooxygenase/metabolism
- Tyrosine Transaminase/genetics
- Tyrosine Transaminase/metabolism
Collapse
Affiliation(s)
- Shyam Sundar Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| | - Hong Zheng
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| | - Neeru M Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| | - Hamid R Shahshahan
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
48
|
|
49
|
Zhang L, Ding WY, Wang ZH, Tang MX, Wang F, Li Y, Zhong M, Zhang Y, Zhang W. Early administration of trimetazidine attenuates diabetic cardiomyopathy in rats by alleviating fibrosis, reducing apoptosis and enhancing autophagy. J Transl Med 2016; 14:109. [PMID: 27121077 PMCID: PMC4848862 DOI: 10.1186/s12967-016-0849-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 03/31/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Trimetazidine, as an anti-ischemic and antioxidant agent, has been demonstrated to have many cardioprotective effects. However, whether early administration of trimetazidine has an effect on diabetic cardiomyopathy and the mechanisms underlying the effect have not yet been elucidated. METHODS We established a type 2 DCM rat model by high-fat diet and low-dose streptozotocin. Rats were separated into different groups: control, diabetes, and diabetes + trimetazidine (n = 6, each). Cardiac autophagy, cardiac functions, and cardiomyocyte apoptosis were monitored. RESULTS Rats with type 2 DCM showed severe insulin resistance, left ventricular dysfunction, increased cardiomyocyte apoptosis, and reduced cardiac autophagy. Collagen volume fraction (CVF) and perivascular collagen area/luminal area (PVCA/LA) ratio were significantly higher in the diabetic group than the control group. We found that trimetazidine treatment ameliorated metabolic disturbance and insulin resistance, reduced cardiomyocyte apoptosis, and restored cardiac autophagy. CVF and PVCA/LA ratio were also lower in the diabetes + trimetazidine group than the diabetic group (CVF, 4.75 ± 0.52 % vs. 11.04 ± 1.67 %, p < 0.05; PVCA/LA, 8.37 ± 0.51 vs. 17.97 ± 2.66, p < 0.05). Furthermore, trimetazidine inhibited phosphorylation of ERK and P38 MAPK to reduce myocardial fibrosis. Inhibited phosphorylation of AMPK was restored and the interaction between Bcl-2 and Beclin1 was enhanced in diabetes + trimetazidine group, resulting in the initiation of autophagy and alleviation of apoptosis. CONCLUSIONS Early administration of trimetazidine could ameliorate diabetic cardiomyopathy by inhibiting myocardial fibrosis and cardiomyocyte apoptosis and enhancing autophagy. Therefore, trimetazidine may be a good choice in the prevention of diabetic cardiomyopathy if applied at the early stage of diabetes.
Collapse
Affiliation(s)
- Lei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Jinan, People's Republic of China.,Department of Cardiology, Qilu Hospital of Shandong University, No.107 Wenhua West Road, Jinan, 250012, People's Republic of China
| | - Wen-Yuan Ding
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zhi-Hao Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Meng-Xiong Tang
- Department of Emergency, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Feng Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Jinan, People's Republic of China.,Department of Cardiology, Qilu Hospital of Shandong University, No.107 Wenhua West Road, Jinan, 250012, People's Republic of China
| | - Ya Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Jinan, People's Republic of China.,Department of Cardiology, Qilu Hospital of Shandong University, No.107 Wenhua West Road, Jinan, 250012, People's Republic of China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Jinan, People's Republic of China.,Department of Cardiology, Qilu Hospital of Shandong University, No.107 Wenhua West Road, Jinan, 250012, People's Republic of China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Jinan, People's Republic of China.,Department of Cardiology, Qilu Hospital of Shandong University, No.107 Wenhua West Road, Jinan, 250012, People's Republic of China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Jinan, People's Republic of China. .,Department of Cardiology, Qilu Hospital of Shandong University, No.107 Wenhua West Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
50
|
Rezk NA, Sabbah NA, Saad MSS. Role of MicroRNA 126 in screening, diagnosis, and prognosis of diabetic patients in Egypt. IUBMB Life 2016; 68:452-8. [PMID: 27118517 DOI: 10.1002/iub.1502] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 03/30/2016] [Indexed: 01/17/2023]
Abstract
MicroRNAs (miRNAs), family of non-coding small RNAs, play a vital role in the regulation of blood glucose level. We aimed to investigate the relation of serum miRNA-126 expression with impaired glucose tolerance as well as type 2 diabetes mellitus (T2DM) patients with and without complications. One hundred healthy controls, eighty-six patients with IGT, and one hundred patients with T2DM were recruited in this study. Serum miRNA-126 expression was assessed by quantitative real-time polymerase chain reaction. We found a significant decrease of serum miRNA-126 expression between IGT patients as well as diabetic patients when both compared with controls and between diabetic patients compared to IGT patients. A significant decrease of serum miRNA-126 expression was detected in diabetic patients with complications compared to those without evident complications especially those with diabetic macrovascular complications and diabetic retinopathy. Serum microRNA-126 expression could be a good marker for diagnosis of IGT and T2DM as well as for monitoring the outcomes of such disease. © 2016 IUBMB Life, 68(6):452-458, 2016.
Collapse
Affiliation(s)
- Noha A Rezk
- Faculty of Medicine, Medical Biochemistry Department, Zagazig University, Egypt
| | - Norhan A Sabbah
- Faculty of Medicine, Medical Biochemistry Department, Zagazig University, Egypt
| | - Mohamed S S Saad
- Faculty of Medicine, Internal Medicine Department, Zagazig University, Egypt
| |
Collapse
|