251
|
Maffei A, Lembo G, Carnevale D. PI3Kinases in Diabetes Mellitus and Its Related Complications. Int J Mol Sci 2018; 19:ijms19124098. [PMID: 30567315 PMCID: PMC6321267 DOI: 10.3390/ijms19124098] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 02/07/2023] Open
Abstract
Recent studies have shown that phosphoinositide 3-kinases (PI3Ks) have become the target of many pharmacological treatments, both in clinical trials and in clinical practice. PI3Ks play an important role in glucose regulation, and this suggests their possible involvement in the onset of diabetes mellitus. In this review, we gather our knowledge regarding the effects of PI3K isoforms on glucose regulation in several organs and on the most clinically-relevant complications of diabetes mellitus, such as cardiomyopathy, vasculopathy, nephropathy, and neurological disease. For instance, PI3K α has been proven to be protective against diabetes-induced heart failure, while PI3K γ inhibition is protective against the disease onset. In vessels, PI3K γ can generate oxidative stress, while PI3K β inhibition is anti-thrombotic. Finally, we describe the role of PI3Ks in Alzheimer’s disease and ADHD, discussing the relevance for diabetic patients. Given the high prevalence of diabetes mellitus, the multiple effects here described should be taken into account for the development and validation of drugs acting on PI3Ks.
Collapse
Affiliation(s)
- Angelo Maffei
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161 Rome, Italy.
| |
Collapse
|
252
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 718] [Impact Index Per Article: 102.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
253
|
Chodkowska KA, Ciecierska A, Majchrzak K, Ostaszewski P, Sadkowski T. Simultaneous miRNA and mRNA Transcriptome Profiling of Differentiating Equine Satellite Cells Treated with Gamma-Oryzanol and Exposed to Hydrogen Peroxide. Nutrients 2018; 10:nu10121871. [PMID: 30513813 PMCID: PMC6316332 DOI: 10.3390/nu10121871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/25/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022] Open
Abstract
Gamma-oryzanol (GO) is a popular supplement for performance horses, dogs, and humans. Previous studies indicated that GO supplementation decreases creatine kinase activity and lactate level after exercise and may affect oxidative stress in Thoroughbred horses. GO may change genes expression in equine satellite cells (ESC). The purpose of this study was to evaluate the effect of GO on miRNA, gene expression, oxidative stress, and cell damage and viability in differentiating ESC pretreated with hydrogen peroxide (H2O2). ESCs were obtained from a young horse’s skeletal muscle. ESCs were pre-incubated with GO (24 h) and then exposed to H2O2 for one hour. For the microRNA and gene expression assessment, the microarray technique was used. Identified miRNAs and genes were validated using real time-quantitative polymerase chain reaction. Several tests related to cell viability, cell damage, and oxidative stress were performed. The microarray analysis revealed differences in 17 miRNAs and 202 genes between GO-treated and control ESC. The tests related to apoptosis, cell viability, and oxidative stress showed that GO affects these processes to varying degrees. Our results suggest that GO can change miRNA and gene expression and may impact the processes involved in tissue repairing after an injury.
Collapse
Affiliation(s)
- Karolina A Chodkowska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | - Anna Ciecierska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | - Kinga Majchrzak
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | - Piotr Ostaszewski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| |
Collapse
|
254
|
Xu L, Chen W, Ma M, Chen A, Tang C, Zhang C, Cai L. Microarray profiling analysis identifies the mechanism of miR-200b-3p/mRNA-CD36 affecting diabetic cardiomyopathy via peroxisome proliferator activated receptor-γ signaling pathway. J Cell Biochem 2018; 120:5193-5206. [PMID: 30506990 DOI: 10.1002/jcb.27795] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/10/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Current study focused on the influence of miR-200b-3p on cardiocyte apoptosis of diabetic cardiomyopathy (DCM) by regulating CD36 and peroxisome proliferator-activated receptor γ (PPAR-γ) signaling pathway. METHODS Bioinformatic analysis was used to analyze differentially expressed microRNA (miRNAs), messenger RNAs (mRNAs) and activated pathways in DCM. And then quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to verify expression of miR-200b-3p and CD36 in DCM model rats and glucose treated H9c2 cell line. Luciferase reporter assay was used to verify the transcriptional regulation of agomiR-200b-3p and investigate the relationship between miR-200b-3p and CD36. Flow cytometry was performed to assess cardiocyte apoptosis in different interference conditions. Echocardiography was used to illustrate the ejection fraction rate and fraction shortening rate of DCM model rats. Next, hematoxylin-eosin (H&E) staining assay was carried out to reveal structures of cardiocyte tissues with transfection in different conditions. Masson trichrome staining was used to evaluate myocardial fibrosis. Western blot analysis was used to detect the expression levels of PPAR-γ signaling-related protein PPAR-γ and Bcl-2. RESULTS miR-200b-3p was low-expressed while CD36 was overexpressed in DCM. AgomiR-200b-3p could inhibit the expression of CD36 to regulate cardiocyte apoptosis in DCM. CD36 activated PPAR-γ signaling pathway in DCM. Silencing CD36 or GW9662 treatment protect rat against DCM. CONCLUSION miR-200b-3p targeted CD36 to regulate cardiocyte apoptosis of DCM by activating PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Liqiong Xu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Cardiology, The First People's Hospital of Shuangliu District, Chengdu, China
| | - Wei Chen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Cardiology, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, China
| | - Min Ma
- Department of Cardiology, The Sixth People's Hospital of Chengdu, Chengdu, China
| | - Anfang Chen
- Department of Cardiology, The First People's Hospital of Shuangliu District, Chengdu, China
| | - Chengyue Tang
- Department of Cardiology, The First People's Hospital of Shuangliu District, Chengdu, China
| | - Chengwei Zhang
- Department of Cardiology, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, China
| | - Lin Cai
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Cardiology, The Third People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
255
|
Utility of curcumin for the treatment of diabetes mellitus: Evidence from preclinical and clinical studies. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
256
|
Zhang J, Qiu H, Huang J, Ding S, Huang B, Wu Q, Jiang Q. Establishment of a diabetic myocardial hypertrophy model in Mus musculus castaneus mouse. Int J Exp Pathol 2018; 99:295-303. [PMID: 30614094 PMCID: PMC6384502 DOI: 10.1111/iep.12296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to establish a robust model of diabetic myocardial hypertrophy in Mus musculus castaneus mice. Mice were fed a high-fat diet for four weeks and then given streptozotocin (STZ, 40 mg kg-1 d-1 for 5 days, intraperitoneally) and fasting blood glucose (FBG) levels were tested after seven days. Mice with FBG levels above 11.1 mmol/L were considered diabetic. Diabetic mice continued to have access to the high-fat diet until cardiac hypertrophy developed. FBG and body weight (BW) were measured weekly. Myocardial hypertrophy was confirmed by left ventricle (LV) hypertrophy index (LVHI), LV/BW, LV histopathological observation and atrial natriuretic factor (ANF) mRNA expression. Serum insulin and plasma haemoglobin A1c (HbA1c) levels, total cholesterol (TCH) and triglyceride (TG) were measured, and then an insulin resistance index (HOMA.IR) was calculated. The level of FBG in the model group remained above 11.1 mmol/L, and the BW showed significant weight loss, compared with the control group (P < 0.01). The high levels of HbA1c, HOME.IR, TCH and TG, and the low level of insulin suggested that glucose metabolism was not balanced with insulin resistance; meanwhile, higher TCH and TG showed that dyslipidaemia had also developed. After the diabetic mice were kept on the high-energy diet for another four weeks, histopathological observation showed myocardial injuries, much more surface area and collagen fibres, higher LVHI and LV/BW, and elevated expression of ANF mRNA (P < 0.01), suggesting that myocardial hypertrophy had appeared in Mus musculus castaneus mice under the current experimental conditions. Thus a robust model of diabetic myocardial hypertrophy was established four weeks after confirmation of diabetes, which was induced by feeding a high-fat diet for four weeks combined with a repeated low-dose STZ exposure, in Mus musculus castaneus mice.
Collapse
Affiliation(s)
- Jie Zhang
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Hongmei Qiu
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Jiajun Huang
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Shumei Ding
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Bo Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityGuizhouChina
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityGuizhouChina
| | - Qingsong Jiang
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| |
Collapse
|
257
|
Ye P, Gu Y, Zhu YR, Chao YL, Kong XQ, Luo J, Ren XM, Zuo GF, Zhang DM, Chen SL. Exogenous hydrogen sulfide attenuates the development of diabetic cardiomyopathy via the FoxO1 pathway. J Cell Physiol 2018; 233:9786-9798. [PMID: 30078216 DOI: 10.1002/jcp.26946] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/12/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Previous studies have suggested that exogenous hydrogen sulfide can alleviate the development of diabetic cardiomyopathy (DCM) by inhibiting oxidative stress, inflammation, and apoptosis. However, the underlying mechanism is not fully understood. Nuclear expression and function of the transcription factor Forkhead box protein O (FoxO1) have been associated with cardiovascular diseases, and thus, the importance of FoxO1 in DCM has gained increasing attention. This study was designed to investigate the interactions between hydrogen sulfide (H2 S) and nuclear FoxO1 in DCM. METHODS Diabetes was induced in adult male C57BL/6J mice by intraperitoneal injection of streptozotocin and was treated with H2 S donor sodium hydrosulfide for 12 weeks. The H9C2 cardiomyoblast cell line and neonatal rat cardiomyocytes (NRCMs) were treated with the slow-releasing H2 S donor GYY4137 before high-glucose (HG) exposure with or without pretreatment with the Akt inhibitor MK-2206 2HCl. Changes in FoxO1 protein phosphorylation and subcellular localization were determined in H9C2 cells, NRCMs, and cardiac tissues from normal and diabetic mice. Cardiac structure and function in the diabetic mice were evaluated by echocardiography and histological analysis and compared with those in control animals. RESULTS The echocardiographic and histopathological data indicated that exogenous H2 S improved cardiac function and attenuated cardiac hypertrophy and myocardial fibrosis in diabetic mice. H2 S also improved HG-induced oxidative stress and apoptosis in cardiac tissue and NRCMs. In addition, H2 S induced FoxO1 phosphorylation and nuclear exclusion in vitro and in vivo, and this function was not inhibited by MK-2206 2HCl. Alanine substitution mutation of three sites in FoxO1-enhanced FoxO1 transcriptional activity, and subsequent treatment with exogenous H2 S could not prevent HG-induced nuclear retention. CONCLUSIONS Our data indicate that H2 S is a novel regulator of FoxO1 in cardiac cells and provide evidence supporting the potential of H2 S in inhibiting the progression of DCM.
Collapse
Affiliation(s)
- Peng Ye
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yan-Rong Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue-Lin Chao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang-Quan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Min Ren
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guang-Feng Zuo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dai-Min Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
258
|
Fan L, Xiao Q, Zhang L, Wang X, Huang Q, Li S, Zhao X, Li Z. CAPE-pNO2 attenuates diabetic cardiomyopathy through the NOX4/NF-κB pathway in STZ-induced diabetic mice. Biomed Pharmacother 2018; 108:1640-1650. [DOI: 10.1016/j.biopha.2018.10.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/29/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022] Open
|
259
|
El-Mohtadi F, d'Arcy R, Tirelli N. Oxidation-Responsive Materials: Biological Rationale, State of the Art, Multiple Responsiveness, and Open Issues. Macromol Rapid Commun 2018; 40:e1800699. [DOI: 10.1002/marc.201800699] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/13/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Farah El-Mohtadi
- Division of Pharmacy and Optometry; School of Health Sciences; Faculty of Biology; Medicine, and Health; The University of Manchester; Manchester M13 9PT UK
| | - Richard d'Arcy
- Laboratory of Polymers and Biomaterials; Fondazione Istituto Italiano di Tecnologia; 16163 Genova Italy
| | - Nicola Tirelli
- Division of Pharmacy and Optometry; School of Health Sciences; Faculty of Biology; Medicine, and Health; The University of Manchester; Manchester M13 9PT UK
- Laboratory of Polymers and Biomaterials; Fondazione Istituto Italiano di Tecnologia; 16163 Genova Italy
| |
Collapse
|
260
|
Wang Q, Sun Y, Li T, Liu L, Zhao Y, Li L, Zhang L, Meng Y. Function of BRD4 in the pathogenesis of high glucose‑induced cardiac hypertrophy. Mol Med Rep 2018; 19:499-507. [PMID: 30483785 PMCID: PMC6297744 DOI: 10.3892/mmr.2018.9681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Diabetic cardiomyopathy is one of the major complications of diabetes, and due to the increasing number of patients with diabetes it is a growing concern. Diabetes-induced cardiomyopathy has a complex pathogenesis and histone deacetylase-mediated epigenetic processes are of prominent importance. The olfactory bromodomain-containing protein 4 (BRD4) is a protein that recognizes and binds acetylated lysine. It has been reported that the high expression of BRD4 is involved in the process of cardiac hypertrophy. The aim of the present study was to investigate the function of BRD4 in the process of high glucose (HG)-induced cardiac hypertrophy, and to clarify whether epigenetic regulation involving BRD4 is an important mechanism. It was revealed that BRD4 expression levels were increased in H9C2 cells following 48 h of HG stimulation. This result was also observed in a diabetic rat model. Furthermore, HG stimulation resulted in the upregulation of the myocardial hypertrophy marker, atrial natriuretic peptide, the cytoskeletal protein α-actin and fibrosis-associated genes including transforming growth factor-β, SMAD family member 3, connective tissue growth factor and collagen, type 1, α1. However, administration of the specific BRD4 inhibitor JQ1 (250 nM) for 48 h reversed this phenomenon. Furthermore, protein kinase B (AKT) phosphorylation was activated by HG stimulation and suppressed by JQ1. In conclusion, BRD4 serves an important role in the pathogenesis of HG-induced cardiomyocyte hypertrophy through the AKT pathway.
Collapse
Affiliation(s)
- Qian Wang
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuxin Sun
- Department of Otorhinolaryngology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tianshu Li
- Department of Functional Science Experiment Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lianqin Liu
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yunxia Zhao
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liyuan Li
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ling Zhang
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Meng
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
261
|
Jiang J, Liang G, Wu Z, Mo H, You Q, Wang Z, Wu K, Guo R. Naringenin alleviates high D-glucose-induced injuries through upregulation of microRNA-30d-5p level in human AC16 cardiac cells. J Appl Biomed 2018. [DOI: 10.1016/j.jab.2018.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
262
|
Borghetti G, von Lewinski D, Eaton DM, Sourij H, Houser SR, Wallner M. Diabetic Cardiomyopathy: Current and Future Therapies. Beyond Glycemic Control. Front Physiol 2018; 9:1514. [PMID: 30425649 PMCID: PMC6218509 DOI: 10.3389/fphys.2018.01514] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus and the associated complications represent a global burden on human health and economics. Cardiovascular diseases are the leading cause of death in diabetic patients, who have a 2–5 times higher risk of developing heart failure than age-matched non-diabetic patients, independent of other comorbidities. Diabetic cardiomyopathy is defined as the presence of abnormal cardiac structure and performance in the absence of other cardiac risk factors, such coronary artery disease, hypertension, and significant valvular disease. Hyperglycemia, hyperinsulinemia, and insulin resistance mediate the pathological remodeling of the heart, characterized by left ventricle concentric hypertrophy and perivascular and interstitial fibrosis leading to diastolic dysfunction. A change in the metabolic status, impaired calcium homeostasis and energy production, increased inflammation and oxidative stress, as well as an accumulation of advanced glycation end products are among the mechanisms implicated in the pathogenesis of diabetic cardiomyopathy. Despite a growing interest in the pathophysiology of diabetic cardiomyopathy, there are no specific guidelines for diagnosing patients or structuring a treatment strategy in clinical practice. Anti-hyperglycemic drugs are crucial in the management of diabetes by effectively reducing microvascular complications, preventing renal failure, retinopathy, and nerve damage. Interestingly, several drugs currently in use can improve cardiac health beyond their ability to control glycemia. GLP-1 receptor agonists and sodium-glucose co-transporter 2 inhibitors have been shown to have a beneficial effect on the cardiovascular system through a direct effect on myocardium, beyond their ability to lower blood glucose levels. In recent years, great improvements have been made toward the possibility of modulating the expression of specific cardiac genes or non-coding RNAs in vivo for therapeutic purpose, opening up the possibility to regulate the expression of key players in the development/progression of diabetic cardiomyopathy. This review summarizes the pathogenesis of diabetic cardiomyopathy, with particular focus on structural and molecular abnormalities occurring during its progression, as well as both current and potential future therapies.
Collapse
Affiliation(s)
- Giulia Borghetti
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Dirk von Lewinski
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Deborah M Eaton
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Markus Wallner
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
263
|
Leukocytic toll-like receptor 2 knockout protects against diabetes-induced cardiac dysfunction. Biochem Biophys Res Commun 2018; 506:668-673. [PMID: 30454704 DOI: 10.1016/j.bbrc.2018.10.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/13/2018] [Indexed: 12/26/2022]
Abstract
Diabetic cardiomyopathy is characterized by the deterioration of the myocardial function. Emerging evidences have indicated that leukocytic toll-like receptor 2 (TLR2) played an important role in the development of diabetic cardiomyopathy. Our study aimed to investigate whether TLR2 knockout (KO) exerted a cardioprotective effect in vivo. The establishment of diabetes model was set up in mice via intraperitoneal injection of streptozotocin (STZ). Results demonstrated that blocking of TLR2 significantly suppressed the enhanced left ventricular end-diastolic dimension (LVEDD), left ventricular end systolic diameter (LVESD) and the reduced the heart rate in diabetic cardiomyopathy mice. The decreased resting cell length, PS, TPS and + dL/dt while increased TR90 and - dL/dt caused by diabetic cardiomyopathy were remarkably inhibited by TLR2 KO. Besides that, the alleviated ΔFFI (360/380), decreased SERCA2a and p-NFATc3 expressions, extended Ca2+ decay time and elevated Calcineurin A induced by diabetic cardiomyopathy were vastly repressed by TLR2 KO in cardiocytes. Moreover, TLR2 gene silence could ameliorate oxidative stress-induced apoptosis, evidences were the up-regulated superoxide generation and Bax/Bcl-2 expression while restrained GSH/GSSG ratio caused by diabetic cardiomyopathy were tremendously repressed in TLR2 KO mice. Furthermore, blocking of TLR2 remarkably attenuated the augmented fibrosis areas of heart tissues in mice with diabetic cardiomyopathy. The result of the enhanced α-SMA and collagenⅠ caused by diabetic cardiomyopathy were suppressed in heart tissues of TLR2 KO mice further validate it. All in all, our study demonstrated that diabetes-induced cardiac dysfunction could be attenuated by TLR2 KO.
Collapse
|
264
|
Wanrooy BJ, Kumar KP, Wen SW, Qin CX, Ritchie RH, Wong CHY. Distinct contributions of hyperglycemia and high-fat feeding in metabolic syndrome-induced neuroinflammation. J Neuroinflammation 2018; 15:293. [PMID: 30348168 PMCID: PMC6198529 DOI: 10.1186/s12974-018-1329-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
Background High-fat feeding and hyperglycemia, key risk factors for the development of metabolic syndrome (MetS), are emerging to associate with increased risk of developing dementia and cognitive decline. Despite this, clinical and experimental studies have yet to elucidate the specific contributions of either high-fat feeding or hyperglycemia to potential neuroinflammatory components. In this study, we delineate these individual components of MetS in the development of neuroinflammation. Methods Male C57Bl/6 J adult mice were treated with either citrate vehicle (CIT) or streptozotocin (STZ; 55 mg/kg) 3, 5 and 7 days before commencement of either a normal or high-fat diet for 9 or 18 weeks. By creating separate models of high-fat feeding, STZ-induced hyperglycemia, as well as in combination, we were able to delineate the specific effects of a high-fat diet and hyperglycemia on the brain. Throughout the feeding regime, we measured the animals’ body weight and fasting blood glucose levels. At the experimental endpoint, we assessed plasma levels of insulin, glycated haemoglobin and performed glucose tolerance testing. In addition, we examined the effect of high fat-feeding and hyperglycemia on the levels of systemic inflammatory cytokines, gliosis in the hippocampus and immune infiltration in cerebral hemispheric tissue. Furthermore, we used intravital multiphoton microscopy to assess leukocyte-endothelial cell interactions in the cerebral vasculature of mice in vivo. Results We showed that acute hyperglycemia induces regional-specific effects on the brain by elevating microglial numbers and promotes astrocytosis in the hippocampus. In addition, we demonstrated that chronic hyperglycemia supported the recruitment of peripheral GR1+ granulocytes to the cerebral microvasculature in vivo. Moreover, we provided evidence that these changes were independent of the systemic inflammation associated with high-fat feeding. Conclusions Hyperglycemia alone preferentially induces microglial numbers and astrocytosis in the hippocampus and is associated with the peripheral recruitment of leukocytes to the cerebrovasculature, but not systemic inflammation. High-fat feeding alone, and in combination with hyperglycemia, increases the systemic pro-inflammatory cytokine milieu but does not result in brain-specific immune gliosis. These results shed light on the specific contributions of high-fat feeding and hyperglycemia as key factors of MetS in the development of neuroinflammation. Electronic supplementary material The online version of this article (10.1186/s12974-018-1329-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brooke J Wanrooy
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia
| | - Kathryn Prame Kumar
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia
| | - Shu Wen Wen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia
| | - Cheng Xue Qin
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Diabetes, Monash University, Melbourne, Australia
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
265
|
Shah S, Akhtar MS, Hassan M, Akhtar M, Paudel YN, Najmi AK. EGFR tyrosine kinase inhibition decreases cardiac remodeling and SERCA2a/NCX1 depletion in streptozotocin induced cardiomyopathy in C57/BL6 mice. Life Sci 2018; 210:29-39. [DOI: 10.1016/j.lfs.2018.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/28/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022]
|
266
|
Qi K, Zhong J. LncRNA HOTAIR improves diabetic cardiomyopathy by increasing viability of cardiomyocytes through activation of the PI3K/Akt pathway. Exp Ther Med 2018; 16:4817-4823. [PMID: 30542437 DOI: 10.3892/etm.2018.6755] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/26/2018] [Indexed: 12/19/2022] Open
Abstract
The current study aimed to investigate the role of long non-coding RNA (lncRNA) homeobox transcript antisense RNA (HOTAIR) in the pathogenesis of diabetic cardiomyopathy. Patients with diabetic cardiomyopathy, patients with diabetes but without cardiomyopathy and healthy controls were included in the current study. All participants underwent myocardial biopsy to collect myocardial tissues. Blood samples were also collected from each participant to prepare serum. Expression of HOTAIR in myocardial tissues was detected by reverse transcription-quantitative polymerase chain reaction. Receiver operating characteristic curve analysis was performed to evaluate the diagnostic value of serum HOTAIR for diabetic cardiomyopathy. AC16 human cardiomyocyte cells were treated with high glucose to observe the changes in expression of HOTAIR and phosphorylation of Akt. HOTAIR expression vector was transfected into cells of AC16 cell line and the effects of HOTAIR overexpression on cell viability and Akt phosphorylation were detected by MTT assay and western blot analysis, respectively. HOTAIR expression was significantly downregulated in myocardial tissues and serum of patients with diabetic cardiomyopathy compared with patients with diabetes and healthy controls. Serum HOTAIR could be used to effectively distinguish patients with diabetic cardiomyopathy from healthy controls. High glucose treatment inhibited HOTAIR expression and Akt phosphorylation. HOTAIR overexpression promoted Akt phosphorylation. HOTAIR overexpression improved AC16 cell viability, while PI3K/Akt inhibitor treatment reduced this effect. LncRNA HOTAIR may improve diabetic cardiomyopathy by increasing the viability of cardiomyocytes through activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Kun Qi
- Department of Gerontology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jianke Zhong
- Department of Gerontology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
267
|
Vichit P, Rungsipipat A, Surachetpong SD. Changes of cardiac function in diabetic dogs. J Vet Cardiol 2018; 20:438-450. [PMID: 30217497 DOI: 10.1016/j.jvc.2018.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 07/24/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE This study aimed to evaluate cardiac function and compare the concentration of cardiac biomarkers including cardiac troponin I (cTnI), galectin-3 (Gal-3), and N-terminal pro B-type natriuretic peptides (NT-proBNP) in diabetic and control dogs. ANIMALS Thirty-nine dogs were included. The diabetic and control groups consisted of 19 and 20 dogs, respectively. METHODS Plasma cTnI, Gal-3, and NT-proBNP concentrations were measured in the diabetic and control groups. Echocardiography was performed in all dogs to evaluate cardiac structure and function. Echocardiographic values and cardiac biomarker concentrations between the two groups were compared with the Mann-Whitney U test. The p-value < 0.05 was considered statistical significance. RESULTS No evidence of cardiac structural changes was detected in diabetic dogs on two-dimensional echocardiography. The echocardiographic values of diabetic and control dogs were within reference intervals. Echocardiographic changes indicating diastolic dysfunction assessed by spectral flow Doppler echocardiography and tissue Doppler imaging were found in diabetic dogs (42.10%) compared with control dogs (10.00%; p = 0.022). Diabetic dogs with durations of diabetes mellitus > 1 year had an increased left ventricular wall thickness and echocardiographic changes suggesting diastolic dysfunction compared with those with duration of diabetes mellitus < 1 year. No evidence of systolic dysfunction was detected in diabetic dogs. No significant difference in plasma cTnI, Gal-3, and NT-proBNP concentrations was found between the two groups. CONCLUSIONS Echocardiographic changes suggested that left ventricular diastolic dysfunction was detected in diabetic dogs without changes in the concentration of cardiac biomarkers including cTnI, Gal-3, and NT-proBNP compared with the age- and breed-matched control dogs.
Collapse
Affiliation(s)
- P Vichit
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, 10330, Thailand
| | - A Rungsipipat
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, 10330, Thailand
| | - S D Surachetpong
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, 10330, Thailand.
| |
Collapse
|
268
|
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
|
269
|
Dai B, Li H, Fan J, Zhao Y, Yin Z, Nie X, Wang DW, Chen C. MiR-21 protected against diabetic cardiomyopathy induced diastolic dysfunction by targeting gelsolin. Cardiovasc Diabetol 2018; 17:123. [PMID: 30180843 PMCID: PMC6122727 DOI: 10.1186/s12933-018-0767-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/25/2018] [Indexed: 12/17/2022] Open
Abstract
Background Diabetes is a leading cause of mortality and morbidity across the world. Over 50% of deaths among diabetic patients are caused by cardiovascular diseases. Cardiac diastolic dysfunction is one of the key early signs of diabetic cardiomyopathy, which often occurs before systolic dysfunction. However, no drug is currently licensed for its treatment. Methods Type 9 adeno-associated virus combined with cardiac Troponin T promoter were employed to manipulate miR-21 expression in the leptin receptor-deficient (db/db) mice. Cardiac structure and functions were measured by echocardiography and hemodynamic examinations. Primary cardiomyocytes and cardiomyocyte cell lines were used to perform gain/loss-of-function assays in vitro. Results We observed a significant reduction of miR-21 in the diastolic dysfunctional heart of db/db mice. Remarkably, delivery of miR-21 efficiently protected against the early impairment in cardiac diastolic dysfunction, represented by decreased ROS production, increased bioavailable NO and relieved diabetes-induced cardiomyocyte hypertrophy in db/db mice. Through bioinformatic analysis and Ago2 co-immunoprecipitation, we identified that miR-21 directly targeted gelsolin, a member of the actin-binding proteins, which acted as a transcriptional cofactor in signal transduction. Moreover, down-regulation of gelsolin by siRNA also attenuated the early phase of diabetic cardiomyopathy. Conclusion Our findings reveal a new role of miR-21 in attenuating diabetic cardiomyopathy by targeting gelsolin, and provide a molecular basis for developing a miRNA-based therapy against diabetic cardiomyopathy. Electronic supplementary material The online version of this article (10.1186/s12933-018-0767-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Beibei Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Jiahui Fan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Yanru Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Zhongwei Yin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Xiang Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China. .,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan, 430030, China. .,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
270
|
Curcumin-mediated effects on anti-diabetic drug-induced cardiotoxicity. 3 Biotech 2018; 8:399. [PMID: 30221112 DOI: 10.1007/s13205-018-1425-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/01/2018] [Indexed: 01/02/2023] Open
Abstract
The present study was designed to compare the cardiotoxicity of two very commonly used anti-diabetic drugs namely pioglitazone (Pio) and metformin (Met); and to study the effects of curcumin (Curc) against these drug-induced cardiotoxicity. Curc, being an anti-oxidant molecule and having cardio-protective potential, can have promising synergistic effects in reducing the cardiac stress induced by anti-diabetic therapies. Various dose and time-dependent cell viability and oxidative stress assays were conducted to study cardiotoxic side-effects and Curc-mediated effects in cardiomyoblasts. Effects of Curc were also studied in hyperglycaemia induced cardiac stress in the presence of drugs. Quantitative assays for cell growth, reactive oxygen species (ROS) generation, lipid peroxidation and mitochondrial permeability followed by anti-oxidant enzymes and caspases activity assays were done to study the mechanism of action of the induced cardiotoxicity. Significant dose and time mediated deleterious effects of Pio and Met were witnessed. Oxidative stress studies showed a remarkable increase in ROS with increasing dose of anti-diabetic drugs. Increased caspase activity and altered mitochondrial integrity were also witnessed in presence of Met and Pio in cardiomyoblasts. These alterations were found to be significantly reduced when treated with Curc simultaneously. The study confirms that Met and Pio exert toxic effects on cardiac cells by generating oxidative stress. Curc, being an anti-oxidative molecule, can suppress this effect and, therefore, can be used as a supplement with anti-diabetic drugs to suppress the induced cardiac stress.
Collapse
|
271
|
Glycophagy: An emerging target in pathology. Clin Chim Acta 2018; 484:298-303. [DOI: 10.1016/j.cca.2018.06.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/08/2018] [Accepted: 06/08/2018] [Indexed: 12/14/2022]
|
272
|
Duan P, Wang J, Li Y, Wei S, Su F, Zhang S, Duan Y, Wang L, Zhu Q. Opening of mitoKATP improves cardiac function and inhibits apoptosis via the AKT-Foxo1 signaling pathway in diabetic cardiomyopathy. Int J Mol Med 2018; 42:2709-2719. [PMID: 30132505 PMCID: PMC6192784 DOI: 10.3892/ijmm.2018.3832] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Decreasing phosphorylation of AKT-Foxo1 is closely associated with the onset of insulin resistance and apoptosis during diabetic cardiomyopathy (DCM). Opening of mitochondrial ATP-sensitive potassium channels (mitoKATP) increases the expression of p-AKT in the process of reperfusion injury. It was therefore hypothesized that opening of mitoKATP may regulate the AKT-Foxo1 signaling pathway and improve cardiac function in DCM. In the present study, opening of mitoKATP by diazoxide (DZX) was found to improve cardiac function and attenuate cardiomyocyte apoptosis in db/db mice. DZX also significantly increased the expression of p-AKT and p-Foxo1. Similarly, DZX decreased the expression of the heart failure marker NT-proBNP, increased mitochondrial membrane potential, inhibited apoptosis, and increased the expression of p-AKT and p-Foxo1 when mimicking insulin resistance in cultured cardiomyocytes. Moreover, the protective effects of DZX were completely blocked by the specific AKT inhibitor MK-2206. These data suggest that the regulation of the AKT-Foxo1 signaling pathway by mitoKATP plays an important role in improving cardiac function and inhibiting apoptosis in DCM, and may therefore be a new potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Peng Duan
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jinxin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Shiqiang Wei
- Department of Cardiology, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Feng Su
- Department of Medical Administration, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Sanlin Zhang
- Department of Cardiology, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Yuhui Duan
- Department of Cardiology, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Lin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Qinglei Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
273
|
Liu Z, Zheng S, Wang X, Qiu C, Guo Y. Novel ASK1 inhibitor AGI-1067 improves AGE-induced cardiac dysfunction by inhibiting MKKs/p38 MAPK and NF-κB apoptotic signaling. FEBS Open Bio 2018; 8:1445-1456. [PMID: 30186746 PMCID: PMC6120242 DOI: 10.1002/2211-5463.12499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/14/2018] [Accepted: 06/25/2018] [Indexed: 12/28/2022] Open
Abstract
Heart failure has been identified as one of the clinical manifestations of diabetic cardiovascular complications. Excessive myocardium apoptosis characterizes cardiac dysfunctions, which are correlated with an increased level of advanced glycation end products (AGEs). In this study, we investigated the participation of reactive oxygen species (ROS) and the involvements of apoptosis signal-regulating kinase 1 (ASK1)/mitogen-activated protein kinase (MAPK) kinases (MKKs)/p38 MAPK and nuclear factor κB (NF-κB) pathways in AGE-induced apoptosis-mediated cardiac dysfunctions. The antioxidant and therapeutic effects of a novel ASK1 inhibitor, AGI-1067, were also studied. Myocardium and isolated primary myocytes were exposed to AGEs and treated with AGI-1067. Invasive hemodynamic and echocardiographic assessments were used to evaluate the cardiac functions. ROS formation was evaluated by dihydroethidium fluorescence staining. A terminal deoxynucleotidyl transferase dUTP nick end labelling assay was used to detect the apoptotic cells. ASK1 and NADPH activities were determined by kinase assays. The association between ASK1 and thioredoxin 1 (Trx1) was assessed by immunoprecipitation. Western blotting was used to evaluate the phosphorylation and expression levels of proteins. Our results showed that AGE exposure significantly activated ASK1/MKKs/p38 MAPK, which led to increased cardiac apoptosis and cardiac impairments. AGI-1067 administration inhibited the activation of MKKs/p38 MAPK by inhibiting the disassociation of ASK1 and Trx1, which suppressed the AGE-induced myocyte apoptosis. Moreover, the NF-κB activation as well as the ROS generation was inhibited. As a result, cardiac functions were improved. Our findings suggested that AGI-1067 recovered AGE-induced cardiac dysfunction by blocking both ASK1/MKKs/p38 and NF-κB apoptotic signaling pathways.
Collapse
Affiliation(s)
- Zhongwei Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology Xi'an Jiaotong University China.,Department of Cardiology Shaanxi Provincial People's Hospital Xi'an China.,Department of Vascular Surgery Brigham and Women's Hospital Boston MA USA
| | - Shixiang Zheng
- Department of Vascular Surgery Brigham and Women's Hospital Boston MA USA.,Department of Critical Care Medicine Union Hospital of Fujian Medical University Fuzhou China
| | - Xi Wang
- Department of Vascular Surgery Brigham and Women's Hospital Boston MA USA.,Department of Obstetrics and Gynecology The Second Xiangya Hospital Central South University Changsha China
| | - Chuan Qiu
- Department of Biostatistics & Bioinformatics School of Public Health & Tropical Medicine Tulane University New Orleans LA USA
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology Xi'an Jiaotong University China
| |
Collapse
|
274
|
Chen H, Zhou W, Ruan Y, Yang L, Xu N, Chen R, Yang R, Sun J, Zhang Z. Reversal of angiotensin ll-induced β-cell dedifferentiation via inhibition of NF-κb signaling. Mol Med 2018; 24:43. [PMID: 30134927 PMCID: PMC6092859 DOI: 10.1186/s10020-018-0044-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is characterized by pancreatic β-cell failure, which arises from metabolic stress and results in β cell dedifferentiation, leading to β-cell death. Pathological activation of the renin–angiotensin system (RAS) contributes to increase cell stress, while RAS intervention reduces the onset of T2DM in high-risk populations and promotes insulin secretion in rodents. In this study, we investigated whether and how RAS induces β-cell dedifferentiation and the mechanism underlying this process. Methods In vitro, with the methods of quantitative real-time reverse transcriptase-PCR (qRT-PCR) and western blotting, we examined the change of cell identity-related gene expression, progenitor like gene expression, cellular function, and nuclear factor kappa b (NF-κb) signaling activity in β cell lines after exposure to angiotensin II (AngII) and disruption of RAS. In vivo, parallel studies were performed using db/db mice. Related protein expression was detected by Immunofluorescence analysis. Result Activation of RAS induced dedifferentiation and impaired insulin secretion, eventually leading to β-cell failure. Mechanistically, Angll induced β-cell dedifferentiation via NF-κb signaling, while treatment with lrbesartan and sc-514 reversed the progenitor state of β cells. Conclusion The present study found that RAS might induce β-cell dedifferentiation via angiotensin II receptor type 1 activation, which was promoted by NF-κb signaling. Therefore, blocking RAS or NF-kb signaling efficiently reversed the dedifferentiated status of β cells, suggesting a potential therapy for patients with type 2 diabetes. Electronic supplementary material The online version of this article (10.1186/s10020-018-0044-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Chen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Wenjun Zhou
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Yuting Ruan
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Lei Yang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Ningning Xu
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Rongping Chen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Rui Yang
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Jia Sun
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China.
| | - Zhen Zhang
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, 253, Gongyedadao Middle, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
275
|
Dong S, Zhang S, Chen Z, Zhang R, Tian L, Cheng L, Shang F, Sun J. Berberine Could Ameliorate Cardiac Dysfunction via Interfering Myocardial Lipidomic Profiles in the Rat Model of Diabetic Cardiomyopathy. Front Physiol 2018; 9:1042. [PMID: 30131709 PMCID: PMC6090155 DOI: 10.3389/fphys.2018.01042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Diabetic cardiomyopathy (DCM) is considered to be a distinct clinical entity independent of concomitant macro- and microvascular disorders, which is initiated partly by disturbances in energy substrates. This study was to observe the dynamic modulations of berberine in DCM rats and explore the changes of lipidomic profiles of myocardial tissue. Methods: Sprague-Dawley (SD) rats were fed high-sucrose and high-fat diet (HSHFD) for totally 22 weeks and intraperitoneally (i.p.) injected with 30 mg/kg of streptozotocin (STZ) at the fifth week to induce DCM. Seventy-two hours after STZ injection, the rats were orally given with berberine at 10, 30 mg/kg and metformin at 200 mg/kg, respectively. Dynamic changes of cardiac function, heart mass ratios and blood lipids were observed at f 4, 10, 16, and 22, respectively. Furthermore, lipid metabolites in myocardial tissue at week 16 were profiled by the ultra-high-performance liquid chromatography coupled to a quadruple time of flight mass spectrometer (UPLC/Q-TOF/MS) approach. Results: Berberine could protect against cardiac diastolic and systolic dysfunctions, as well as cardiac hypertrophy, and the most effective duration is with 16-week of administration. Meanwhile, 17 potential biomarkers of phosphatidylcholines (PCs), phosphatidylethanolamines (PEs) and sphingolipids (SMs) of DCM induced by HSFD/STZ were identified. The perturbations of lipidomic profiles could be partly reversed with berberine intervention, i.e., PC (16:0/20:4), PC (18:2/0:0), PC (18:0/18:2), PC (18:0/22:5), PC (20:4/0:0), PC (20:4/18:0), PC (20:4/18:1), PC (20:4/20:2), PE (18:2/0:0), and SM (d18:0/16:0). Conclusions: These results indicated a close relationship between PCs, PEs and SMs and cardiac damage mechanisms during development of DCM. The therapeutic effects of berberine on DCM are partly caused by interferences with PCs, PEs, and SMs metabolisms.
Collapse
Affiliation(s)
- Shifen Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shuofeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhirong Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Rong Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linyue Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Long Cheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fei Shang
- Department of Pharmacology, Analysis and Testing Center, Beijing University of Chemical Technology, Beijing, China
| | - Jianning Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
276
|
Ma L, Li XP, Ji HS, Liu YF, Li EZ. Baicalein Protects Rats with Diabetic Cardiomyopathy Against Oxidative Stress and Inflammation Injury via Phosphatidylinositol 3-Kinase (PI3K)/AKT Pathway. Med Sci Monit 2018; 24:5368-5375. [PMID: 30070262 PMCID: PMC6085984 DOI: 10.12659/msm.911455] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background The aim of this study was to explore the effect of baicalein on diabetic cardiomyopathy (DCM) rats and the mechanisms involved, and to determine the theoretical basis for clinical anti-tumor therapy. Material/Methods DCM rat model was induced with a single injection of streptozotocin. Then, DCM rats were treated with baicalein alone or co-treated with baicalein and PI3K/Akt inhibitor. Myocardial pathological changes were detected by HE and Masson staining. The activities of SOD, GSH-Px, and MDA in myocardial tissue were measured by biochemical tests. The levels of TNF-α, IL-1β, and cTn-I were examined by ELISA. NADP+/NADPH ratio was measured with the NADP+/NADPH assay kit. RT-PCR was used to detect the levels of PI3K and Akt. The levels of Bax, Bcl-2, Caspase-3, GSK-3β, PI3K, and Akt were detected by Western blot. Results Baicalein could improve pathological injury. SOD and GSH-Px activity decreased while the level of MDA increased in myocardial tissue. Baicalein treatment enhanced SOD activity in a dose-dependent manner but markedly reduced MDA. Similar changes were observed in both serum inflammatory factors and the NADP+/NADPH ratio. After adding PI3K-Akt inhibitor, the levels of PI3K and Akt mRNA expression were significantly decreased, but were not significantly different from the DCM group. Levels of Bcl-2, PI3K, p-GSK-3β/GSK-3β, and p-Akt were decreased in the DCM group, while the levels of Bax and Caspase-3 were obviously increased. Conclusions Baicalein can protect DCM rats against damage from oxidative stress and inflammation in myocardial tissue, and PI3K/Akt signaling pathway may be involved to mediating these effects.
Collapse
Affiliation(s)
- Lin Ma
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Xiang Peng Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Heng Sheng Ji
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Yue Fen Liu
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - En Ze Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
277
|
Dludla PV, Dias SC, Obonye N, Johnson R, Louw J, Nkambule BB. A Systematic Review on the Protective Effect of N-Acetyl Cysteine Against Diabetes-Associated Cardiovascular Complications. Am J Cardiovasc Drugs 2018; 18:283-298. [PMID: 29623672 DOI: 10.1007/s40256-018-0275-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Heart failure is the leading cause of death in patients with diabetes. No treatment currently exists to specifically protect these patients at risk of developing cardiovascular complications. Accelerated oxidative stress-induced tissue damage due to persistent hyperglycemia is one of the major factors implicated in deteriorated cardiac function within a diabetic state. N-acetyl cysteine (NAC), through its enhanced capacity to endogenously synthesize glutathione, a potent antioxidant, has displayed abundant health-promoting properties and has a favorable safety profile. OBJECTIVE An increasing number of experimental studies have reported on the strong ameliorative properties of NAC. We systematically reviewed the data on the cardioprotective potential of this compound to provide an informative summary. METHODS Two independent reviewers systematically searched major databases, including PubMed, Cochrane Library, Google scholar, and Embase for available studies reporting on the ameliorative effects of NAC as a monotherapy or in combination with other therapies against diabetes-associated cardiovascular complications. We used the ARRIVE and JBI appraisal guidelines to assess the quality of individual studies included in the review. A meta-analysis could not be performed because the included studies were heterogeneous and data from randomized clinical trials were unavailable. RESULTS Most studies support the ameliorative potential of NAC against a number of diabetes-associated complications, including oxidative stress. We discuss future prospects, such as identification of additional molecular mechanisms implicated in diabetes-induced cardiac damage, and highlight limitations, such as insufficient studies reporting on the comparative effect of NAC with common glucose-lowering therapies. Information on the comparative analysis of NAC, in terms of dose selection, administration mode, and its effect on different cardiovascular-related markers is important for translation into clinical studies. CONCLUSIONS NAC exhibits strong potential for the protection of the diabetic heart at risk of myocardial infarction through inhibition of oxidative stress. The effect of NAC in preventing both ischemia and non-ischemic-associated cardiac damage is also of interest. Consistency in dose selection in most studies reported remains important in dose translation for clinical relevance.
Collapse
|
278
|
Wang S, Zhao Z, Feng X, Cheng Z, Xiong Z, Wang T, Lin J, Zhang M, Hu J, Fan Y, Reiter RJ, Wang H, Sun D. Melatonin activates Parkin translocation and rescues the impaired mitophagy activity of diabetic cardiomyopathy through Mst1 inhibition. J Cell Mol Med 2018; 22:5132-5144. [PMID: 30063115 PMCID: PMC6156356 DOI: 10.1111/jcmm.13802] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022] Open
Abstract
Mitophagy eliminates dysfunctional mitochondria and thus plays a cardinal role in diabetic cardiomyopathy (DCM). We observed the favourable effects of melatonin on cardiomyocyte mitophagy in mice with DCM and elucidated their underlying mechanisms. Electron microscopy and flow cytometric analysis revealed that melatonin reduced the number of impaired mitochondria in the diabetic heart. Other than decreasing mitochondrial biogenesis, melatonin increased the clearance of dysfunctional mitochondria in mice with DCM. Melatonin increased LC3 II expression as well as the colocalization of mitochondria and lysosomes in HG‐treated cardiomyocytes and the number of typical autophagosomes engulfing mitochondria in the DCM heart. These results indicated that melatonin promoted mitophagy. When probing the mechanism, increased Parkin translocation to the mitochondria may be responsible for the up‐regulated mitophagy exerted by melatonin. Parkin knockout counteracted the beneficial effects of melatonin on the cardiac mitochondrial morphology and bioenergetic disorders, thus abolishing the substantial effects of melatonin on cardiac remodelling with DCM. Furthermore, melatonin inhibited Mammalian sterile 20‐like kinase 1 (Mst1) phosphorylation, thus enhancing Parkin‐mediated mitophagy, which contributed to mitochondrial quality control. In summary, this study confirms that melatonin rescues the impaired mitophagy activity of DCM. The underlying mechanism may be attributed to activation of Parkin translocation via inhibition of Mst1.
Collapse
Affiliation(s)
- Shanjie Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhijing Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyu Feng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zheng Cheng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Tingting Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mingming Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianqiang Hu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanhong Fan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, Texas
| | - Haichang Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
279
|
Zhang H, Chen X, Zong B, Yuan H, Wang Z, Wei Y, Wang X, Liu G, Zhang J, Li S, Cheng G, Wang Y, Ma Y. Gypenosides improve diabetic cardiomyopathy by inhibiting ROS-mediated NLRP3 inflammasome activation. J Cell Mol Med 2018; 22:4437-4448. [PMID: 29993180 PMCID: PMC6111804 DOI: 10.1111/jcmm.13743] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/26/2018] [Indexed: 12/17/2022] Open
Abstract
NLRP3 inflammasome activation plays an important role in diabetic cardiomyopathy (DCM), which may relate to excessive production of reactive oxygen species (ROS). Gypenosides (Gps), the major ingredients of Gynostemma pentaphylla (Thunb.) Makino, have exerted the properties of anti-hyperglycaemia and anti-inflammation, but whether Gps improve myocardial damage and the mechanism remains unclear. Here, we found that high glucose (HG) induced myocardial damage by activating the NLRP3 inflammasome and then promoting IL-1β and IL-18 secretion in H9C2 cells and NRVMs. Meanwhile, HG elevated the production of ROS, which was vital to NLRP3 inflammasome activation. Moreover, the ROS activated the NLRP3 inflammasome mainly by cytochrome c influx into the cytoplasm and binding to NLRP3. Inhibition of ROS and cytochrome c dramatically down-regulated NLRP3 inflammasome activation and improved the cardiomyocyte damage induced by HG, which was also detected in cells treated by Gps. Furthermore, Gps also reduced the levels of the C-reactive proteins (CRPs), IL-1β and IL-18, inhibited NLRP3 inflammasome activation and consequently improved myocardial damage in vivo. These findings provide a mechanism that ROS induced by HG activates the NLRP3 inflammasome by cytochrome c binding to NLRP3 and that Gps may be potential and effective drugs for DCM via the inhibition of ROS-mediated NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Hailong Zhang
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Xi Chen
- Department of General Pathology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Beibei Zong
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Hongmin Yuan
- Department of Thyroid Breast Surgery, Huaihe Hospital, Henan University, Kaifeng, China
| | - Zhizeng Wang
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Yinxiang Wei
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Xuance Wang
- Centre for Translational Medicine, Huaihe Hospital, Henan University, Kaifeng, China
| | - Guangchao Liu
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Jun Zhang
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Shulian Li
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Guanchang Cheng
- Department of Cardiac Surgery, Huaihe Hospital, Henan University, Kaifeng, China
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| | - Yuanfang Ma
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, School of Basic Medicine, Henan University, Kaifeng, China
| |
Collapse
|
280
|
Lahnwong C, Chattipakorn SC, Chattipakorn N. Potential mechanisms responsible for cardioprotective effects of sodium-glucose co-transporter 2 inhibitors. Cardiovasc Diabetol 2018; 17:101. [PMID: 29991346 PMCID: PMC6038192 DOI: 10.1186/s12933-018-0745-5] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus currently affects over 350 million patients worldwide and is associated with many deaths from cardiovascular complications. Sodium-glucose co-transporter 2 (SGLT-2) inhibitors are a novel class of antidiabetic drugs with cardiovascular benefits beyond other antidiabetic drugs. In the EMPA-REG OUTCOME trial, empagliflozin significantly decreases the mortality rate from cardiovascular causes [38% relative risk reduction (RRR)], the mortality rate from all-causes (32% RRR) and the rate of heart failure hospitalization (35% RRR) in diabetic patients with established cardiovascular diseases. The possible mechanisms of SGLT-2 inhibitors are proposed to be systemic effects by hemodynamic and metabolic actions. However, the direct mechanisms are not fully understood. In this review, reports concerning the effects of SGLT-2 inhibitors in models of diabetic cardiomyopathy, heart failure and myocardial ischemia from in vitro, in vivo as well as clinical reports are comprehensively summarized and discussed. By current evidences, it may be concluded that the direct effects of SGLT-2 inhibitors are potentially mediated through their ability to reduce cardiac inflammation, oxidative stress, apoptosis, mitochondrial dysfunction and ionic dyshomeostasis.
Collapse
Affiliation(s)
- Charshawn Lahnwong
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
281
|
The aging heart. Clin Sci (Lond) 2018; 132:1367-1382. [PMID: 29986877 DOI: 10.1042/cs20171156] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/10/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Abstract
As the elderly segment of the world population increases, it is critical to understand the changes in cardiac structure and function during the normal aging process. In this review, we outline the key molecular pathways and cellular processes that underlie the phenotypic changes in the heart and vasculature that accompany aging. Reduced autophagy, increased mitochondrial oxidative stress, telomere attrition, altered signaling in insulin-like growth factor, growth differentiation factor 11, and 5'- AMP-activated protein kinase pathways are among the key molecular mechanisms underlying cardiac aging. Aging promotes structural and functional changes in the atria, ventricles, valves, myocardium, pericardium, the cardiac conduction system, and the vasculature. We highlight the factors known to accelerate and attenuate the intrinsic aging of the heart and vessels in addition to potential preventive and therapeutic avenues. A greater understanding of the processes involved in cardiac aging may facilitate our ability to mitigate the escalating burden of CVD in older individuals and promote healthy cardiac aging.
Collapse
|
282
|
Tang SG, Liu XY, Ye JM, Hu TT, Yang YY, Han T, Tan W. Isosteviol ameliorates diabetic cardiomyopathy in rats by inhibiting ERK and NF-κB signaling pathways. J Endocrinol 2018; 238:47-60. [PMID: 29720537 DOI: 10.1530/joe-17-0681] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/16/2018] [Indexed: 12/23/2022]
Abstract
Diabetes-induced injury of myocardium, defined as diabetic cardiomyopathy (DCM), accounts for significant mortality and morbidity in diabetic population. Alleviation of DCM by a potent drug remains considerable interests in experimental and clinical researches because hypoglycemic drugs cannot effectively control this condition. Here, we explored the beneficial effects of isosteviol sodium (STVNa) on type 1 diabetes-induced DCM and the potential mechanisms involved. Male Wistar rats were induced to diabetes by injection of streptozotocin (STZ). One week later, diabetic rats were randomly grouped to receive STVNa (STZ/STVNa) or its vehicle (STZ). After 11 weeks of treatment or 11 weeks treatment following 4 weeks of removal of the treatment, the cardiac function and structure were evaluated and related mechanisms were investigated. In diabetic rats, oxidative stress, inflammation, blood glucose and plasma advanced glycation end products (AGEs) were significantly increased, whereas superoxide dismutase 2 (SOD-2) expression and activity were decreased. STVNa treatment inhibited cardiac hypertrophy, fibrosis and inflammation, showed similar ratio of heart to body weight and antioxidant capacities almost similar to the normal controls, which can be sustained at least 4 weeks. Moreover, STVNa inhibited diabetes-inducted stimulation of both extracellular signal-regulated kinase (ERK) and nuclear factor κB (NF-κB) signal pathways. However, blood glucose, plasma AGE and insulin levels were not altered by STVNa treatment. These results indicate that STVNa may be developed into a potent therapy for DCM. The mechanism underlying this therapeutic effect involves the suppression of oxidative stress and inflammation by inhibiting ERK and NF-κB without changing blood glucose or AGEs.
Collapse
Affiliation(s)
- Sheng-Gao Tang
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Xiao-Yu Liu
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ji-Ming Ye
- Molecular Pharmacology for DiabetesSchool of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Ting-Ting Hu
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ying-Ying Yang
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ting Han
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Wen Tan
- Institute of Biomedical & Pharmaceutical ScienceGuangdong University of Technology, Guangzhou, China
| |
Collapse
|
283
|
Louis XL, Raj P, McClinton KJ, Yu L, Suh M, Netticadan T. Supplementation of Type 1 Diabetic Rats with Carrot Powder Lowers Blood Glucose without Improving Cardiac Structure and Function. Prev Nutr Food Sci 2018; 23:115-121. [PMID: 30018889 PMCID: PMC6047871 DOI: 10.3746/pnf.2018.23.2.115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/05/2018] [Indexed: 01/17/2023] Open
Abstract
Foods and food bioactives have shown to be effective in preventing some human disease conditions. In this study, we examined the effects of carrot powder, rich in carotenoids, as a dietary supplement for the prevention of cardiac anomalies in streptozotocin (STZ) induced type 1 diabetic rats. Male Wistar rats were fed either control or carrot powder containing diet for 3 weeks. Type 1 diabetes was induced with STZ injection (65 mg/kg body weight) in half of the rats in each group. All rats were continued on their respective diet for a further 9 weeks. Cardiac structural and functional parameters were measured using echocardiography at 8 weeks post STZ administration. In comparison to non-diabetic rats, diabetic rats showed significant increase in isovolumetric relaxation time and a significant decrease in systolic function parameter, cardiac output. Left ventricular internal dimension and left ventricular posterior wall thickness were significantly higher in diabetic animals. Blood glucose levels were significantly lower in carrot supplemented diabetic rats when compared with non-treated diabetic rats. Diabetic rats treated and untreated had elevated level of lipid peroxidation. Catalase levels were significantly elevated in the carrot powder supplemented diabetic rats when compared to the control rats. Carrot supplementation lowered blood glucose levels significantly but did not normalize it to control levels. It had no effect on cardiac abnormalities and anti-oxidant status in rats with type 1 diabetes.
Collapse
Affiliation(s)
- Xavier Lieben Louis
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg R3T2N2, Canada
| | - Pema Raj
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg R2H 2A6, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg R3E0J, Canada
| | - Kathleen J McClinton
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg R3T2N2, Canada
| | - Liping Yu
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg R2H 2A6, Canada.,Agriculture and Agri-Food Canada, Winnipeg R2H 2A6, Canada
| | - Miyoung Suh
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg R3T2N2, Canada
| | - Thomas Netticadan
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg R2H 2A6, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg R3E0J, Canada.,Agriculture and Agri-Food Canada, Winnipeg R2H 2A6, Canada
| |
Collapse
|
284
|
Bayrami G, Alihemmati A, Karimi P, Javadi A, Keyhanmanesh R, Mohammadi M, Zadi-Heydarabad M, Badalzadeh R. Combination of Vildagliptin and Ischemic Postconditioning in Diabetic Hearts as a Working Strategy to Reduce Myocardial Reperfusion Injury by Restoring Mitochondrial Function and Autophagic Activity. Adv Pharm Bull 2018; 8:319-329. [PMID: 30023334 PMCID: PMC6046419 DOI: 10.15171/apb.2018.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/12/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023] Open
Abstract
Purpose: Diabetic hearts are resistant to cardioprotection by ischemic-postconditioning (IPostC). Protection of diabetic hearts and finding related interfering mechanisms would have clinical benefits. This study investigated the combination effects of vildagliptin (Vilda) and IPostC on cardioprotection and the levels of autophagy and mitochondrial function following myocardial ischemia/reperfusion (I/R) injury in type-II diabetic rats. Methods: Diabetes was established by high fat diet/low dose of streptozotocin and lasted for 12 weeks. The diabetic rats received Vilda (6 mg/kg/day, orally) for one month before I/R. Myocardial regional ischemia was induced through the ligation of left coronary artery, and IPostC was applied immediately at the onset of reperfusion. The infarct size was assessed by a computerised planimetry and left ventricles samples were harvested for cardiac mitochondrial function studies (ROS production, membrane potential and staining) and western blotting was used for determination of autophagy markers. Results: None of Vilda or IPostC but combination of them could significantly reduce the infarct size of diabetic hearts, comparing to control (P<0.001). IPostC could not significantly affect p62 expression level in diabetic hearts, but pre-treatment with Vilda alone (p<0.05) and in combination with IPostC (p<0.01) more significantly decreased p62 expression in comparison with corresponding control group. The expression of LC3B-II and LC3BII/LC3BI as well as mitochondrial ROS production were decreased significantly in treatment groups (p<0.001). Mitochondrial membrane depolarization was significantly higher and mitochondrial density was lower in untreated diabetic I/R hearts than treated groups (p<0.001). IPostC in combination with vildagliptin prevented the mitochondrial membrane depolarization and increased the mitochondrial content more potent than IPostC alone in diabetic hearts. Conclusion: Combination of vildagliptin and IPostC in diabetic hearts was a well-working strategy to reduce myocardial I/R damages by restoring mitochondrial membrane potential and ROS production and modulating the autophagic activity in I/R hearts.
Collapse
Affiliation(s)
- Goltaj Bayrami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aniseh Javadi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mustafa Mohammadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Badalzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
285
|
Zhao Q, Jia TZ, Cao QC, Tian F, Ying WT. A Crude 1-DNJ Extract from Home Made Bombyx Batryticatus Inhibits Diabetic Cardiomyopathy-Associated Fibrosis in db/db Mice and Reduces Protein N-Glycosylation Levels. Int J Mol Sci 2018; 19:ijms19061699. [PMID: 29880742 PMCID: PMC6032278 DOI: 10.3390/ijms19061699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/26/2018] [Accepted: 05/29/2018] [Indexed: 01/01/2023] Open
Abstract
The traditional Chinese drug Bombyx Batryticatus (BB), which is also named the white stiff silkworm, has been widely used in Chinese clinics for thousands of years. It is famous for its antispasmodic and blood circulation-promoting effects. Cardiomyocyte hypertrophy, interstitial cell hyperplasia, and myocardial fibrosis are closely related to the N-glycosylation of key proteins. To examine the alterations of N-glycosylation that occur in diabetic myocardium during the early stage of the disease, and to clarify the therapeutic effect of 1-Deoxynojirimycin (1-DNJ) extracted from BB, we used the db/db (diabetic) mouse model and an approach based on hydrophilic chromatography solid-phase extraction integrated with an liquid Chromatograph Mass Spectrometer (LC-MS) identification strategy to perform a site-specific N-glycosylation analysis of left ventricular cardiomyocyte proteins. Advanced glycation end products (AGEs), hydroxyproline, connective tissue growth factor (CTGF), and other serum biochemical indicators were measured with enzyme-linked immunosorbent assays (ELISA). In addition, the α-1,6-fucosylation of N-glycans was profiled with lens culinaris agglutinin (LCA) lectin blots and fluorescein isothiocyanate (FITC)-labelled lectin affinity histochemistry. The results indicated that 1-DNJ administration obviously downregulated myocardium protein N-glycosylation in db/db mice. The expression levels of serum indicators and fibrosis-related cytokines were reduced significantly by 1-DNJ in a dose-dependent manner. The glycan α-1,6-fucosylation level of the db/db mouse myocardium was elevated, and the intervention effect of 1-DNJ administration on N-glycan α-1,6-fucosylation was significant. To verify this result, the well-known transforming growth factor-β (TGF-β)/Smad2/3 pathway was selected, and core α-1,6-fucosylated TGF-β receptor II (TGFR-βII) was analysed semi-quantitatively with western blotting. The result supported the conclusions obtained from LCA lectin affinity histochemistry and lectin blot analysis. The expression level of α-1,6-fucosyltransferase (FUT8) mRNA was also detected, and the results showed that 1-DNJ administration did not cause obvious inhibitory effects on FUT8 expression. Therefore, the mechanism of 1-DNJ for relieving diabetic cardiomyopathy (DCM)-associated fibrosis can be concluded as the inhibition of N-acetylglucosamine (N-GlcNAc) formation and the reduction of substrate concentration.
Collapse
Affiliation(s)
- Qing Zhao
- The Key Laboratory of Chinese Materia Medica Processing Principle Analysis of the State Administration of Traditional Chinese Medicine, Pharmaceutical College of Liaoning Traditional Chinese Medicine University, Chinese Materia Medica Processing Engineering Technology Research Center of Liaoning Province, Dalian 110060, China.
- Chinese Materia Medica Department, Traditional Chinese Medicine College of Hebei University, Baoding 071000, China.
- Beijing Institute of Lifeomics, Beijing Proteome Research Center, Beijing 102206, China.
| | - Tian Zhu Jia
- The Key Laboratory of Chinese Materia Medica Processing Principle Analysis of the State Administration of Traditional Chinese Medicine, Pharmaceutical College of Liaoning Traditional Chinese Medicine University, Chinese Materia Medica Processing Engineering Technology Research Center of Liaoning Province, Dalian 110060, China.
| | - Qi Chen Cao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
| | - Fang Tian
- Beijing Institute of Lifeomics, Beijing Proteome Research Center, Beijing 102206, China.
| | - Wan Tao Ying
- Beijing Institute of Lifeomics, Beijing Proteome Research Center, Beijing 102206, China.
| |
Collapse
|
286
|
Burgeiro A, Fonseca A, Espinoza D, Carvalho L, Lourenço N, Antunes M, Carvalho E. Proteostasis in epicardial versus subcutaneous adipose tissue in heart failure subjects with and without diabetes. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2183-2198. [PMID: 29625179 PMCID: PMC6375688 DOI: 10.1016/j.bbadis.2018.03.025] [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: 11/30/2017] [Revised: 03/14/2018] [Accepted: 03/29/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are leading cause of death and primary cause of morbidity and mortality in diabetic population. Epicardial adipose tissue (EAT) covers the heart's surface and is a source of biomolecules regulating heart and blood vessel physiology. The protective activation of the unfolded protein response (UPR) and autophagy allows the cardiomyocyte reticular network to restore energy and/or nutrient homeostasis and to avoid cell death. However, an excessive or prolonged UPR activation can trigger cell death. UPR activation is an early event of diabetic cardiomyopathies and deregulated autophagy is associated with CVDs. RESULTS An upregulation of UPR markers (glucose-regulated protein 78 KDa, glucose-regulated protein 94 KDa, inositol-requiring enzyme 1α, protein kinase RNA-like ER kinase and CCAAT/-enhancer-binding protein homologous protein (CHOP) gene) in EAT compared to subcutaneous adipose tissue (SAT), was observed as well as the UPR-related apoptosis marker caspase-4/procaspase-4 ratio but not in CHOP protein levels. Additionally, levels of ubiquitin and ubiquitinated proteins were decreased in EAT. Moreover, upregulation of autophagy markers (5' adenosine monophosphate-activated protein kinase, mechanistic target of rapamycin, Beclin 1, microtubule-associated protein light chain 3-II, lysosome-associated membrane protein 2, and PTEN-induced putative kinase 1) was observed, as well as an increase in the apoptotic Bim but not the ratio between Bim and the anti-apoptotic Bcl-2 in EAT. Diabetic patients show alterations in UPR activation markers but not in autophagy or apoptosis markers. CONCLUSION UPR and autophagy are increased in EAT compared to SAT, opening doors to the identification of early biomarkers for cardiomyopathies and novel therapeutic targets.
Collapse
Affiliation(s)
- A. Burgeiro
- Center of Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - A.C. Fonseca
- Center of Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - D. Espinoza
- Center of Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - L. Carvalho
- Institute of Pathology, Faculty of Medicine, University of Coimbra, 3004-517 Coimbra, Portugal
| | - N. Lourenço
- Centre for Informatics and Systems of the University of Coimbra (CISUC), Department of Informatics Engineering, University of Coimbra, Portugal
| | - M. Antunes
- Cardiothoracic Surgery Unit at the Coimbra University Hospital Centre, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - E. Carvalho
- Center of Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal,The Portuguese Diabetes Association (APDP), 1250-203 Lisbon, Portugal,Arkansas Children's Research Institute, Little Rock, Arkansas 72202, United States,Corresponding author: Eugénia Carvalho, , Address for correspondence: Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Rua Larga, Faculdade de Medicina, Pólo I, 1° andar,3004-504 Coimbra, Phone number: 00351 239820190, Fax number: 00351 239822776
| |
Collapse
|
287
|
[Exendin-4 alleviates diabetic cardiomyopathy in mice by regulating Sirt1/PGC1α]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38. [PMID: 29891446 PMCID: PMC6743905 DOI: 10.3969/j.issn.1673-4254.2018.05.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To investigate the protective effect of exendin-4 against diabetic cardiomyopathy in mice and explore the underlying mechanism. METHODS C57BL/6J mice were randomly divided into normal control group with normal diet and diabetic group with high-fat diet for 4 weeks before streptozotocin injection. The successfully established diabetic mouse models were divided into diabetic group with exendin-4 treatment and diabetic control group for daily treatment with intraperitoneal injection of 1 nmol/kg exendin-4 and saline of equivalent volume for 8 weeks, respectively. The physiological parameters such as blood glucose and body weight were recorded. RT-PCR was used to examine the transcription levels of genes related with myocardial hypertrophy and fibrosis and the genes related with mitochondrial functions including PGC1α, NRF and CytoC. The expressions of oxidative stress markers and Sirt1/PGC1 proteins were measured using Western blotting. and HE staining was used to observe the myocardial structural changes in the mice. RESULTS Compared with the normal control mice, the mice in diabetic control group showed significantly increased blood glucose and blood lipid levels (P<0.001), which were obviously improved by Exendin-4 treatment. The expressions of ANP, BNP, TGFβ1, CytoC1 and NOX1 were significantly increased (P<0.05) while Sirt1, PGC1α, NRF and SOD1 expression were markedly decreased in the myocardial tissue of the diabetic mice (P<0.05). Exendin-4 treatment resulted in obviously reduced expressions of ANP, BNP, TGFβ1, CytoC1 and NOX1 (P<0.05) and increased expressions of Sirt1, PGC1α, NRF and SOD1 (P<0.05) in the diabetic mice. CONCLUSIONS Exendin-4 protects against myocardial injury in diabetic mice by improving mitochondrial function and inhibiting oxidative stress through the Sirt1/PGC1α signaling pathway.
Collapse
|
288
|
Resveratrol Ameliorates Cardiac Dysfunction by Inhibiting Apoptosis via the PI3K/Akt/FoxO3a Pathway in a Rat Model of Diabetic Cardiomyopathy. J Cardiovasc Pharmacol 2018; 70:184-193. [PMID: 28678055 DOI: 10.1097/fjc.0000000000000504] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this study was to explore the effect and mechanism of action of resveratrol (RSV) on cardiac function in diabetic cardiomyopathy (DCM). Hyperglycemia-induced apoptosis contributes to the pathogenic changes in DCM. RSV treatment inhibited high glucose-induced apoptosis of neonatal rat ventricular myocytes. Additionally, high glucose decreased cell viability, prevented serine-threonine kinase (Akt) and FoxO3a phosphorylation, and suppressed cytoplasmic translocation of FoxO3a. However, these effects of apoptosis were reversed by 10 μM of RSV. The PI3K inhibitor LY294002 abolished the RSV protective effect in vitro. RSV (5 or 50 mg·kg·d orally for 8 weeks) prevented the deterioration of cardiac function and structural cardiomyopathy in a streptozotocin-induced rat model of diabetes and reduced apoptosis in diabetic myocardium. Furthermore, it restored streptozotocin-impaired phosphorylation of Akt and FoxO3a (p-Akt and p-FoxO3a) and suppressed nuclear translocation of FoxO3a in vivo. Together, these data indicate that RSV has therapeutic potential against DCM by inhibiting apoptosis via the PI3K/Akt/FoxO3a pathway.
Collapse
|
289
|
Ng HH, Leo CH, Parry LJ, Ritchie RH. Relaxin as a Therapeutic Target for the Cardiovascular Complications of Diabetes. Front Pharmacol 2018; 9:501. [PMID: 29867503 PMCID: PMC5962677 DOI: 10.3389/fphar.2018.00501] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular complications are the major cause of mortality in patients with diabetes. This is closely associated with both macrovascular and microvascular complications of diabetes, which lead to organ injuries in diabetic patients. Previous studies have consistently demonstrated the beneficial effects of relaxin treatment for protection of the vasculature, with evidence of antioxidant and anti-remodeling actions. Relaxin enhances nitric oxide, prostacyclin and endothelium-derived hyperpolarization (EDH)-type-mediated relaxation in various vascular beds. These effects of relaxin on the systemic vasculature, coupled with its cardiac actions, reduce pulmonary capillary wedge pressure and pulmonary artery pressure. This results in an overall decrease in systemic and pulmonary vascular resistance in heart failure patients. The anti-fibrotic actions of relaxin are well established, a desirable property in the context of diabetes. Further, relaxin ameliorates diabetic wound healing, with accelerated angiogenesis and vasculogenesis. Relaxin-mediated stimulation of vascular endothelial growth factor (VEGF) and stromal cell-derived factor 1-α, as well as regulation of metalloproteinase expression, ameliorates cardiovascular fibrosis in diabetic mice. In the heart, relaxin is a cardioprotective molecule in several experimental animal models, exerting anti-fibrotic, anti-hypertrophy and anti-apoptotic effects in diabetic pathologies. Collectively, these studies provide a foundation to propose the therapeutic potential for relaxin as an adjunctive agent in the prevention or treatment of diabetes-induced cardiovascular complications. This review provides a comprehensive overview of the beneficial effects of relaxin, and identifies its therapeutic possibilities for alleviating diabetes-related cardiovascular injury.
Collapse
Affiliation(s)
- Hooi Hooi Ng
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Chen Huei Leo
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
- Science and Math Cluster, Singapore University of Technology and Design, Singapore, Singapore
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H. Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology & Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
290
|
Mohammed Yusof NL, Zainalabidin S, Mohd Fauzi N, Budin SB. Hibiscus sabdariffa (roselle) polyphenol-rich extract averts cardiac functional and structural abnormalities in type 1 diabetic rats. Appl Physiol Nutr Metab 2018; 43:1224-1232. [PMID: 29726706 DOI: 10.1139/apnm-2018-0084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is often associated with cardiac functional and structural alteration, an initial event leading to cardiovascular complications. Roselle (Hibiscus sabdariffa) has been widely proven as an antioxidant and recently has incited research interest for its potential in treating cardiovascular disease. Therefore, this study aimed to determine the cardioprotective effects of H. sabdariffa (roselle) polyphenol-rich extract (HPE) in type-1-induced diabetic rats. Twenty-four male Sprague-Dawley rats were randomized into 4 groups (n = 6/group): nondiabetic, diabetic alone (DM), diabetic supplemented with HPE (DM+HPE), and diabetic supplemented with metformin. Type-1 diabetes was induced with streptozotocin (55 mg/kg intraperitoneally). Rats were forced-fed with HPE (100 mg/kg) and metformin (150 mg/kg) daily for 8 weeks. Results showed that HPE supplementation improved hyperglycemia and dyslipidemia significantly (p < 0.05) in the DM+HPE compared with the DM group. HPE supplementation attenuated cardiac oxidative damage in the DM group, indicated by low malondialdehyde and advanced oxidation protein product. As for the antioxidant status, HPE significantly (p < 0.05) increased glutathione level, as well as catalase and superoxide dismutase 1 and 2 activities. These findings correlate with cardiac function, whereby left ventricle developed pressure in DM+HPE (79.13 ± 3.08 mm Hg) was higher significantly compared with DM (45.84 ± 1.65 mm Hg). Coronary flow of DM+HPE (17.43 ± 0.62 mL/min) was also greater compared with DM (13.02 ± 0.6 mL/min), showing that HPE supplementation improved cardiac contractility and relaxation rate significantly (p < 0.05). Histological analysis showed a marked decrease in cardiomyocyte hypertrophy and fibrosis in DM+HPE compared with the DM group. Ultrastructural changes and impairment of mitochondria induced by diabetes were minimized by HPE supplementation. Collectively, these findings suggest that HPE is a potential cardioprotective agent in a diabetic setting through its hypoglycemic, anti-hyperlipidemia, and antioxidant properties.
Collapse
Affiliation(s)
- Nur Liyana Mohammed Yusof
- a Programme of Biomedical Science, School of Applied Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- a Programme of Biomedical Science, School of Applied Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Norsyahida Mohd Fauzi
- b Faculty of Pharmacy, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Siti Balkis Budin
- a Programme of Biomedical Science, School of Applied Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| |
Collapse
|
291
|
Toedebusch R, Belenchia A, Pulakat L. Diabetic Cardiomyopathy: Impact of Biological Sex on Disease Development and Molecular Signatures. Front Physiol 2018; 9:453. [PMID: 29773993 PMCID: PMC5943496 DOI: 10.3389/fphys.2018.00453] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Diabetic cardiomyopathy refers to a unique set of heart-specific pathological variables induced by hyperglycemia and insulin resistance. Given that cardiovascular disease (CVD) is the leading cause of death in the world, and type 2 diabetes incidence continues to rise, understanding the complex interplay between these two morbidities and developing novel therapeutic strategies is vital. Two hallmark characteristics specific to diabetic cardiomyopathy are diastolic dysfunction and cardiac structural mal-adaptations, arising from cardiac cellular responses to the complex toxicity induced by hyperglycemia with or without hyperinsulinemia. While type 2 diabetes is more prevalent in men compared to women, cardiovascular risk is higher in diabetic women than in diabetic men, suggesting that diabetic women take a steeper path to cardiomyopathy and heart failure. Accumulating evidence from randomized clinical trials indicate that although pre-menopausal women have lower risk of CVDs, compared to age-matched men, this advantage is lost in diabetic pre-menopausal women, which suggests estrogen availability does not protect from increased cardiovascular risk. Notably, few human studies have assessed molecular and cellular mechanisms regarding similarities and differences in the progression of diabetic cardiomyopathy in men versus women. Additionally, most pre-clinical rodent studies fail to include female animals, leaving a void in available data to truly understand the impact of biological sex differences in diabetes-induced dysfunction of cardiovascular cells. Elegant reviews in the past have discussed in detail the roles of estrogen-mediated signaling in cardiovascular protection, sex differences associated with telomerase activity in the heart, and cardiac responses to exercise. In this review, we focus on the emerging cellular and molecular markers that define sex differences in diabetic cardiomyopathy based on the recent clinical and pre-clinical evidence. We also discuss miR-208a, MED13, and AT2R, which may provide new therapeutic targets with hopes to develop novel treatment paradigms to treat diabetic cardiomyopathy uniquely between men and women.
Collapse
Affiliation(s)
- Ryan Toedebusch
- Cardiovascular Medicine Division, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Anthony Belenchia
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Lakshmi Pulakat
- Cardiovascular Medicine Division, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
292
|
Shin YK, Hsieh YS, Kwon S, Lee HS, Seol GH. Linalyl acetate restores endothelial dysfunction and hemodynamic alterations in diabetic rats exposed to chronic immobilization stress. J Appl Physiol (1985) 2018; 124:1274-1283. [DOI: 10.1152/japplphysiol.01018.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although stress is one of the risk factors of diabetes, few studies have assessed the effects of stress on diabetic rats. This study, therefore, analyzed differences in cardiovascular-related factors among control, nonstressed diabetic, and stressed diabetic rats as well as assessed the effects of linalyl acetate (LA) on stressed diabetic rats. Male Sprague-Dawley rats were subjected to immobilization stress throughout the experimental period, and diabetes was induced on day 15 by a single injection of streptozotocin. After confirming the induction of diabetes, stressed diabetic rats were administered LA (10 or 100 mg/kg) or metformin (500 mg/kg) for the last 7 days. Compared with nonstressed diabetic rats, stressed diabetic rats had significantly lower body weight, body fat percentage, ACh-induced vasorelaxation, systolic blood pressure (SBP), diastolic blood pressure (DBP), heart rate (HR), and NF-κB expression as well as increased serum nitrite concentration. Although metformin increased serum insulin concentration significantly, 100 mg/kg LA showed only an increasing tendency. However, treatment with 100 mg/kg LA not only reduced serum glucose and NF-κB expression, but also restored ACh-induced vasorelaxation, SBP, DBP, HR, AMP-activated protein kinase expression, and serum nitrite almost to control levels. Importantly, 100 mg/kg LA was more effective than metformin in ameliorating serum glucose, endothelial nitric oxide synthase expression, HR, and serum nitrite. These findings suggest that chronic stress can aggravate endothelial dysfunction and hemodynamic alterations in diabetes and that LA may have potent therapeutic efficacy in diabetic patients with cardiovascular disease complications or chronic stress. NEW & NOTEWORTHY To our knowledge, this is the first study to assess the effects of linalyl acetate (LA) on cardiovascular-related factors in diabetic rats exposed to chronic stress. Treatment with LA restored acetylcholine-induced vasorelaxation, blood pressure, heart rate, and AMP-activated protein kinase and serum nitrite levels. The present results suggest that LA may have potent therapeutic efficacy in diabetic patients with complications of cardiovascular disease or chronic stress.
Collapse
Affiliation(s)
- You Kyoung Shin
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| | - Yu Shan Hsieh
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| | - Soonho Kwon
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| | - Hui Su Lee
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| |
Collapse
|
293
|
Pickering RJ, Rosado CJ, Sharma A, Buksh S, Tate M, de Haan JB. Recent novel approaches to limit oxidative stress and inflammation in diabetic complications. Clin Transl Immunology 2018; 7:e1016. [PMID: 29713471 PMCID: PMC5905388 DOI: 10.1002/cti2.1016] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 12/25/2022] Open
Abstract
Diabetes is considered a major burden on the healthcare system of Western and non‐Western societies with the disease reaching epidemic proportions globally. Diabetic patients are highly susceptible to developing micro‐ and macrovascular complications, which contribute significantly to morbidity and mortality rates. Over the past decade, a plethora of research has demonstrated that oxidative stress and inflammation are intricately linked and significant drivers of these diabetic complications. Thus, the focus now has been towards specific mechanism‐based strategies that can target both oxidative stress and inflammatory pathways to improve the outcome of disease burden. This review will focus on the mechanisms that drive these diabetic complications and the feasibility of emerging new therapies to combat oxidative stress and inflammation in the diabetic milieu.
Collapse
Affiliation(s)
- Raelene J Pickering
- Department of Diabetes Central Clinical School Monash University Melbourne VIC Australia
| | - Carlos J Rosado
- Department of Diabetes Central Clinical School Monash University Melbourne VIC Australia
| | - Arpeeta Sharma
- Oxidative Stress Laboratory Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| | - Shareefa Buksh
- Oxidative Stress Laboratory Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| | - Mitchel Tate
- Heart Failure Pharmacology Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| | - Judy B de Haan
- Oxidative Stress Laboratory Basic Science Domain Baker Heart and Diabetes Institute Melbourne VIC Australia
| |
Collapse
|
294
|
Shen Y, Tang G, Gao P, Zhang B, Xiao H, Si LY. Activation of adenosine A2b receptor attenuates high glucose-induced apoptosis in H9C2 cells via PI3K/Akt signaling. In Vitro Cell Dev Biol Anim 2018; 54:384-391. [PMID: 29626279 DOI: 10.1007/s11626-018-0241-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/04/2018] [Indexed: 12/13/2022]
Abstract
High glucose plays a vital role in apoptosis in H9C2 cells. However, the exact molecular mechanism remains unclear. In this study, we aimed to evaluate the cardio-protective role of A2b receptor in high glucose-induced cardiomyocyte apoptosis via PI3K/Akt pathway. Adenosine A2b receptor agonist (Bay506583), antagonist (MRS1754), and Akt inhibitor (LY294002) were applied respectively to H9C2 cells before exposed to high glucose for 12 h. Apoptosis of H9C2 cells was determined by TUNEL assay and the apoptosis rate by flow cytometry. The protein level of adenosine A2b receptor, p-Akt, total Akt, cleaved capase-3, cleaved capase-9, bax, and bcl-2 was measured by western blotting. The results demonstrated that apoptosis of H9C2 cardiomyocytes triggered by high-glucose treatment was time-dependent. The protein level of A2b receptor and activated Akt was both decreased in cardiomyocyte with high-glucose treatment. Moreover, we found that high glucose-induced apoptosis in H9C2 cells could be attenuated by administration of adenosine A2b receptor agonist Bay606583. This effect could be reversed by Akt inhibitor LY294002. In conclusion, activation of A2b receptor could prevent high glucose-induced apoptosis of H9C2 cells in vitro to a certain extent by activating PI3K/Akt signaling. In conclusion, these results suggested that activation of A2b receptor could be a novel therapeutic approach to high glucose-induced cardiomyocyte injury.
Collapse
Affiliation(s)
- Yi Shen
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Gang Tang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pan Gao
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bin Zhang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hang Xiao
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Liang-Yi Si
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
295
|
An Intervention Target for Myocardial Fibrosis: Autophagy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6215916. [PMID: 29850542 PMCID: PMC5911341 DOI: 10.1155/2018/6215916] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 02/07/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
Myocardial fibrosis (MF) is the result of metabolic imbalance of collagen synthesis and metabolism, which is widespread in various cardiovascular diseases. Autophagy is a lysosomal degradation pathway which is highly conserved. In recent years, research on autophagy has been increasing and the researchers have also become cumulatively aware of the specified association between autophagy and MF. This review highlights the role of autophagy in MF and the potential effects through the administration of medicine.
Collapse
|
296
|
Chang X, Lu K, Wang L, Lv M, Fu W. Astraglaus polysaccharide protects diabetic cardiomyopathy by activating NRG1/ErbB pathway. Biosci Trends 2018; 12:149-156. [PMID: 29607874 DOI: 10.5582/bst.2018.01027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Diabetic cardiomyopathy (DCM) is one of the main cardiac complications among diabetic patients. According to previous studies, the pathogenesis of DCM is associated with oxidative stress, apoptosis and proliferation of local cardiac cells. It showed, NRG1 can improve the function of mitochondria, and thereby, increasing proliferation and decreasing apoptosis of cardiac muscle cell via ErbB/AKT signaling, also, exert antioxidative function. Besides, NRG1/ErbB pathway was impaired in the DCM model which suggested this signaling played key role in DCM. Astraglaus polysaccharide (APS), one of the active components of Astragalus mongholicus, showed striking antioxidative effect. Here, in this study, our data showed that APS can promote proliferation and decrease apoptosis in AGE-induced DCM cell model, besides, APS can decrease intracellular ROS level, increase activity of SOD, GSH-Px and lower level of MDA and NO in DCM cell model, indicating APS exerted antioxidative function in DCM model cells. Besides, western blot results revealed APS induced NRG1 expressing and the phosphorylation level of ErbB2/4. In addition, the elevated NRG1 promoted AKT and PI3k phosphorylation which indicated APS may exert its function by NRG1/ErbB and the downstream AKT/PI3K signaling. Canertinib is ErbB inhibitor. The effect of APS on proliferation, apoptosis, antioxidation and NRG1/ErbB pathway was partly abolished after the cells were co-treated with APS and canertinib. Taken together, these results suggested APS may display its protective function in DCM cells by activating NGR1/ErbB signaling pathway. And our study increased potential for prevention and therapy to DCM.
Collapse
Affiliation(s)
- Xiao Chang
- Department of intensive care unit, Shenzhen Traditional Chinese Medicine Hospital
| | - Kang Lu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine
| | - Ling Wang
- Department of intensive care unit, Shenzhen Traditional Chinese Medicine Hospital
| | - Min Lv
- Department of intensive care unit, Shenzhen Traditional Chinese Medicine Hospital
| | - Wenjun Fu
- South China Research Center for Acupuncture and Moxibustion, School of Basic Medical Science, Guangzhou University of Chinese Medicine
| |
Collapse
|
297
|
Gong TA, Hall SA. Targeting Other Modifiable Risk Factors for the Prevention of Heart Failure: Diabetes, Smoking, Obesity, and Inactivity. CURRENT CARDIOVASCULAR RISK REPORTS 2018. [DOI: 10.1007/s12170-018-0574-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
298
|
Black Seed Thymoquinone Improved Insulin Secretion, Hepatic Glycogen Storage, and Oxidative Stress in Streptozotocin-Induced Diabetic Male Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8104165. [PMID: 29686746 PMCID: PMC5857299 DOI: 10.1155/2018/8104165] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/19/2017] [Indexed: 01/24/2023]
Abstract
Diabetes mellitus is one of the metabolic diseases having several complications. Nigella sativa oil (NSO) might have beneficial effects in the treatment of diabetic complications. Thirty-two mature male Wistar rats were equally divided into four experimental groups: control, control NSO 2 mL/kg, streptozotocin- (STZ-) induced diabetic, and diabetic (STZ-induced) treated with oral NSO 2 mg/kg for 30 days. Fasting blood glucose (FBG), insulin, and lipid profile levels were determined. Pancreatic and hepatic tissues were used for catalase and GSH. Histopathology, hepatic glycogen contents, insulin immunohistochemistry, and pancreatic islet morphometry were performed. NSO 2 mL/kg was noticed to decrease (P < 0.05) FBG and increase (P < 0.05) insulin levels in diabetic rats than in diabetic nontreated animals. Lipid profile showed significant (P < 0.5) improvement in diabetic rats that received NSO 2 mL/kg than in the diabetic group. Both pancreatic and hepatic catalase and GSH activities revealed a significant (P < 0.05) increment in the diabetic group treated with NSO than in the diabetic animals. NSO improved the histopathological picture and hepatic glycogen contents of the diabetic group as well as increased (P < 0.05) insulin immunoreactive parts % and mean pancreatic islet diameter. NSO exerts ameliorative and therapeutic effects on the STZ-induced diabetic male Wistar rats.
Collapse
|
299
|
Glezeva N, Chisale M, McDonald K, Ledwidge M, Gallagher J, Watson CJ. Diabetes and complications of the heart in Sub-Saharan Africa: An urgent need for improved awareness, diagnostics and management. Diabetes Res Clin Pract 2018; 137:10-19. [PMID: 29287838 DOI: 10.1016/j.diabres.2017.12.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/21/2017] [Accepted: 12/21/2017] [Indexed: 12/24/2022]
Abstract
Type 2 diabetes mellitus is no longer a disease of high income countries but a global health pandemic. With the continued and rapid increase in its prevalence worldwide it is forecasted that diabetes will be a leading cause of morbidity and mortality. A major concern stems from its role in development and progression of cardiovascular disease, including cardiac dysfunction and heart failure. Within low- and middle-income areas such as Sub-Saharan Africa the burden of diabetes is already significant driven by many factors, including, socioeconomic (urbanisation), nutritional (high-calorie "western-diet", obesity) and lifestyle (physical inactivity) changes. Insufficient economic and community resources, poor health care system development and chronic disease management, poor education, and a lack of preventative and diagnostic measures further aggravate the severity of the diabetes problem. This review outlines the burden of type 2 diabetes mellitus in Sub-Saharan Africa and highlights the need for improved community health care and regulations to reduce its epidemiological spread and devastating impact on health.
Collapse
Affiliation(s)
- Nadezhda Glezeva
- gHealth Research Group, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | | | - Kenneth McDonald
- gHealth Research Group, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Mark Ledwidge
- gHealth Research Group, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Joe Gallagher
- gHealth Research Group, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Chris J Watson
- Centre for Experimental Medicine, Queen's University Belfast, Northern Ireland, United Kingdom.
| |
Collapse
|
300
|
Min JJ, Shin BS, Lee JH, Jeon Y, Ryu DK, Kim S, Shin YH. Effects of Pravastatin on Type 1 Diabetic Rat Heart with or without Blood Glycemic Control. J Diabetes Res 2018; 2018:1067853. [PMID: 29682576 PMCID: PMC5850894 DOI: 10.1155/2018/1067853] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/29/2017] [Indexed: 01/11/2023] Open
Abstract
Although statins have been suggested to attenuate the progression of diabetic cardiomyopathy, its effect without glycemic control remains unclear. Therefore, we evaluated the effect of pravastatin on diabetic rat hearts according to glycemic control. Rats were randomly divided into five groups: control (C), diabetes (D), diabetes with insulin (I), diabetes with pravastatin (P), and diabetes with insulin and pravastatin (IP). Eight weeks after allocated treatments, the heart was extracted and analyzed following echocardiography. Cardiac fibrosis was measured using Masson's trichrome stain. Cardiac expression of collagen I/III, matrix metalloproteinase (MMP)-2, MMP-9, and angiotensin-converting enzyme (ACE)/ACE2 was evaluated by immunohistochemistry and/or Western blot. Enzyme-linked immunosorbent assay was used for measuring reactive oxygen species (ROS). Diabetic groups without glycemic control (D and P) showed significantly impaired diastolic function and increased levels of cardiac fibrosis, collagen I/III, MMP-2, MMP-9, and ROS production. However, there were little significant differences in the outcomes among the control and two glucose-controlled diabetic groups (I and IP). Groups C and IP showed more preserved ACE2 and lower ACE expressions than the other groups did (D, I, and P). Our study suggested glycemic control would be more important to attenuate the progression of diabetic cardiomyopathy than pravastatin medication.
Collapse
Affiliation(s)
- Jeong Jin Min
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byung-Seop Shin
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Hwan Lee
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yunseok Jeon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dae Kyun Ryu
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sojin Kim
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Hee Shin
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|