201
|
Li W, Yang J, Lyu Q, Wu G, Lin S, Yang Q, Hu J. Taurine attenuates isoproterenol-induced H9c2 cardiomyocytes hypertrophy by improving antioxidative ability and inhibiting calpain-1-mediated apoptosis. Mol Cell Biochem 2020; 469:119-132. [PMID: 32304004 DOI: 10.1007/s11010-020-03733-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
Pathological cardiac hypertrophy is ultimately accompanied by cardiomyocyte apoptosis. Apoptosis mainly related to calpain-1-mediated apoptotic pathways. Studies had proved that taurine can maintain heart health through antioxidation and antiapoptotic functions, but the effect of taurine on cardiac hypertrophy is still unclear. This study aimed to determine whether taurine could inhibit calpain-1-mediated mitochondria-dependent apoptotic pathways in isoproterenol (ISO)-induced hypertrophic cardiomyocytes. We found that taurine could inhibit the increase in cell surface area and reduce the protein expression levels of the hypertrophic markers atrial natriuretic peptide, brain natriuretic polypeptide, and β-myosin heavy chain. Taurine also reduced ROS, intracellular Ca2+ overload and mitochondrial membrane potential. Moreover, taurine inhibited cardiomyocyte apoptosis by decreasing the protein expression of calpain-1, Bax, t-Bid, cytosolic cytochrome c, Apaf-1, cleaved caspase-9 and cleaved caspase-3 and by enhancing calpastatin and Bcl-2 protein expression. Calpain-1 small interfering RNA transfection results showed similar antiapoptotic effects as the taurine prevention group. However, compared with the two treatments, taurine inhibited the expression of cleaved caspase-9 more significantly. Therefore, we believe that taurine prevents ISO-induced H9c2 cardiomyocyte hypertrophy by inhibiting oxidative stress, intracellular Ca2+ overload, the calpain-1-mediated mitochondria-dependent apoptotic pathway and cleaved caspase-9 levels.
Collapse
Affiliation(s)
- Weiwei Li
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Qiufeng Lyu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Shumei Lin
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Qunhui Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China.
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China.
| |
Collapse
|
202
|
Yang Y, Li T, Li Z, Liu N, Yan Y, Liu B. Role of Mitophagy in Cardiovascular Disease. Aging Dis 2020; 11:419-437. [PMID: 32257551 PMCID: PMC7069452 DOI: 10.14336/ad.2019.0518] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/18/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and mitochondrial dysfunction is the primary contributor to these disorders. Recent studies have elaborated on selective autophagy-mitophagy, which eliminates damaged and dysfunctional mitochondria, stabilizes mitochondrial structure and function, and maintains cell survival and growth. Numerous recent studies have reported that mitophagy plays an important role in the pathogenesis of various cardiovascular diseases. This review summarizes the mechanisms underlying mitophagy and advancements in studies on the role of mitophagy in cardiovascular disease.
Collapse
Affiliation(s)
- Yibo Yang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Youyou Yan
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
203
|
Zhang M, Wang X, Liu M, Liu D, Pan J, Tian J, Jin T, Xu Y, An F. Inhibition of PHLPP1 ameliorates cardiac dysfunction via activation of the PI3K/Akt/mTOR signalling pathway in diabetic cardiomyopathy. J Cell Mol Med 2020; 24:4612-4623. [PMID: 32150791 PMCID: PMC7176843 DOI: 10.1111/jcmm.15123] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/18/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Background Pleckstrin homology (PH) domain leucine‐rich repeat protein phosphatase 1 (PHLPP1) is a kind of serine/threonine phosphatase, whose dysregulation is accompanied with numerous human diseases. However, its role in diabetic cardiomyopathy remains unclear. We explored the underlying function and mechanism of PHLPP1 in diabetic cardiomyopathy (DCM). Method In vivo, Type 1 diabetic rats were induced by intraperitoneal injection of 60 mg/kg streptozotocin (STZ). Lentivirus‐mediated short hairpin RNA (shRNA) was used to knock down the expression of PHLPP1. In vitro, primary neonatal rat cardiomyocytes and H9C2 cells were incubated in 5.5 mmol/L glucose (normal glucose, NG) or 33.3 mmol/L glucose (high glucose, HG). PHLPP1 expression was inhibited by PHLPP1‐siRNA to probe into the function of PHLPP1 in high glucose‐induced apoptosis in H9c2 cells. Results Diabetic rats showed up‐regulated PHLPP1 expression, left ventricular dysfunction, increased myocardial apoptosis and fibrosis. PHLPP1 inhibition alleviated cardiac dysfunction. Additionally, PHLPP1 inhibition significantly reduced HG‐induced apoptosis and restored PI3K/AKT/mTOR pathway activity in H9c2 cells. Furthermore, pretreatment with LY294002, an inhibitor of PI3K/Akt/mTOR pathway, abolished the anti‐apoptotic effect of PHLPP1 inhibition. Conclusion Our study indicated that PHLPP1 inhibition alleviated cardiac dysfunction via activating the PI3K/Akt/mTOR signalling pathway in DCM. Therefore, PHLPP1 may be a novel therapeutic target for human DCM.
Collapse
Affiliation(s)
- Mingjun Zhang
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xuyang Wang
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Ming Liu
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Dian Liu
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jinyu Pan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital of Shandong First Medical University, Jinan, China
| | - Jingjing Tian
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Jin
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yunfan Xu
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Fengshuang An
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
204
|
Prakoso D, De Blasio MJ, Tate M, Kiriazis H, Donner DG, Qian H, Nash D, Deo M, Weeks KL, Parry LJ, Gregorevic P, McMullen JR, Ritchie RH. Gene therapy targeting cardiac phosphoinositide 3-kinase (p110α) attenuates cardiac remodeling in type 2 diabetes. Am J Physiol Heart Circ Physiol 2020; 318:H840-H852. [PMID: 32142359 DOI: 10.1152/ajpheart.00632.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Diabetic cardiomyopathy is a distinct form of heart disease that represents a major cause of death and disability in diabetic patients, particularly, the more prevalent type 2 diabetes patient population. In the current study, we investigated whether administration of recombinant adeno-associated viral vectors carrying a constitutively active phosphoinositide 3-kinase (PI3K)(p110α) construct (rAAV6-caPI3K) at a clinically relevant time point attenuates diabetic cardiomyopathy in a preclinical type 2 diabetes (T2D) model. T2D was induced by a combination of a high-fat diet (42% energy intake from lipid) and low-dose streptozotocin (three consecutive intraperitoneal injections of 55 mg/kg body wt), and confirmed by increased body weight, mild hyperglycemia, and impaired glucose tolerance (all P < 0.05 vs. nondiabetic mice). After 18 wk of untreated diabetes, impaired left ventricular (LV) systolic dysfunction was evident, as confirmed by reduced fractional shortening and velocity of circumferential fiber shortening (Vcfc, all P < 0.01 vs. baseline measurement). A single tail vein injection of rAAV6-caPI3K gene therapy (2×1011vector genomes) was then administered. Mice were followed for an additional 8 wk before end point. A single injection of cardiac targeted rAAV6-caPI3K attenuated diabetes-induced cardiac remodeling by limiting cardiac fibrosis (reduced interstitial and perivascular collagen deposition, P < 0.01 vs. T2D mice) and cardiomyocyte hypertrophy (reduced cardiomyocyte size and Nppa gene expression, P < 0.001 and P < 0.05 vs. T2D mice, respectively). The diabetes-induced LV systolic dysfunction was reversed with rAAV6-caPI3K, as demonstrated by improved fractional shortening and velocity of circumferential fiber shortening (all P < 0.05 vs pre-AAV measurement). This cardioprotection occurred in combination with reduced LV reactive oxygen species (P < 0.05 vs. T2D mice) and an associated decrease in markers of endoplasmic reticulum stress (reduced Grp94 and Chop, all P < 0.05 vs. T2D mice). Together, our findings demonstrate that a cardiac-selective increase in PI3K(p110α), via rAAV6-caPI3K, attenuates T2D-induced diabetic cardiomyopathy, providing proof of concept for potential translation to the clinic.NEW & NOTEWORTHY Diabetes remains a major cause of death and disability worldwide (and its resultant heart failure burden), despite current care. The lack of existing management of heart failure in the context of the poorer prognosis of concomitant diabetes represents an unmet clinical need. In the present study, we now demonstrate that delayed intervention with PI3K gene therapy (rAAV6-caPI3K), administered as a single dose in mice with preexisting type 2 diabetes, attenuates several characteristics of diabetic cardiomyopathy, including diabetes-induced impairments in cardiac remodeling, oxidative stress, and function. Our discovery here contributes to the previous body of work, suggesting the cardioprotective effects of PI3K(p110α) could be a novel therapeutic approach to reduce the progression to heart failure and death in diabetes-affected patients.
Collapse
Affiliation(s)
- Darnel Prakoso
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,School of Biosciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Miles J De Blasio
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,School of Biosciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Mitchel Tate
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Hongwei Qian
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - David Nash
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Minh Deo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| | - Laura J Parry
- School of Biosciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia.,Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Neurology, The University of Washington, Seattle, Washington
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Physiology, Monash University, Clayton, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| | - Rebecca Helen Ritchie
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Diabetes, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
205
|
Xia X, Liang Y, Zheng W, Lin D, Sun S. miR-410-5p promotes the development of diabetic cardiomyopathy by suppressing PIM1-induced anti-apoptosis. Mol Cell Probes 2020; 52:101558. [PMID: 32145290 DOI: 10.1016/j.mcp.2020.101558] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a common complication of diabetes mellitus that can cause many severe symptoms, such as heart failure, arrhythmia, and sudden death. However, the molecular mechanisms underlying cardiac dysfunction in DCM remain elusive. In this study, we found that miR-410-5p was increased in the myocardial tissue of a diabetes mellitus (DM) rat model. Further studies confirmed that inhibition of miR-410-5p reduced cell apoptosis by regulating the Bcl-2/Bax axis. Through bioinformatics analysis and luciferase reporter assays, we observed that miR-410-5p directly targets PIM1. Moreover, knockdown of miR-410-5p by antagomir-410-5p improved diabetes-induced cardiac function and myocardial tissue structure. In conclusion, our study demonstrated that miR-410-5p might be involved in the progression of DCM by targeting PIM1 and might be a promising therapeutic target for DCM in the future.
Collapse
Affiliation(s)
- Xiaofen Xia
- Department of Cardiovascular Medicine, Hangzhou Lin'an District People's Hospital, 548 Yijin Road, Lin'an, Hangzhou, Zhejiang, 311300, China
| | - Yan Liang
- Department of Endocrinology, Shanxian Dongda Hospital, 001 Shunshi East Road, Shanxian, Shandong, China
| | - Weihong Zheng
- School of Life Science, Huzhou University, 759 Erhuan East Road, Huzhou, Zhejiang, China
| | - Dongming Lin
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, 318 Chaowang Road, Hangzhou, Zhejiang, China
| | - Shuping Sun
- Department of Cardiovascular Medicine, Hangzhou Lin'an District People's Hospital, 548 Yijin Road, Lin'an, Hangzhou, Zhejiang, 311300, China.
| |
Collapse
|
206
|
Induction of caveolin-3/eNOS complex by nitroxyl (HNO) ameliorates diabetic cardiomyopathy. Redox Biol 2020; 32:101493. [PMID: 32182574 PMCID: PMC7078438 DOI: 10.1016/j.redox.2020.101493] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Nitroxyl (HNO), one-electron reduced and protonated sibling of nitric oxide (NO), is a potential regulator of cardiovascular functions. It produces positive inotropic, lusitropic, myocardial anti-hypertrophic and vasodilator properties. Despite of these favorable actions, the significance and the possible mechanisms of HNO in diabetic hearts have yet to be fully elucidated. H9c2 cells or primary neonatal mouse cardiomyocytes were incubated with normal glucose (NG) or high glucose (HG). Male C57BL/6 mice received intraperitoneal injection of streptozotocin (STZ) to induce diabetes. Here, we demonstrated that the baseline fluorescence signals of HNO in H9c2 cells were reinforced by both HNO donor Angeli's salt (AS), and the mixture of hydrogen sulfide (H2S) donor sodium hydrogen sulfide (NaHS) and NO donor sodium nitroprusside (SNP), but decreased by HG. Pretreatment with AS significantly reduced HG-induced cell vitality injury, apoptosis, reactive oxygen species (ROS) generation, and hypertrophy in H9c2 cells. This effect was mediated by induction of caveolin-3 (Cav-3)/endothelial nitric oxide (NO) synthase (eNOS) complex. Disruption of Cav-3/eNOS by pharmacological manipulation or small interfering RNA (siRNA) abolished the protective effects of AS in HG-incubated H9c2 cells. In STZ-induced diabetic mice, administration of AS ameliorated the development of diabetic cardiomyopathy, as evidenced by improved cardiac function and reduced cardiac hypertrophy, apoptosis, oxidative stress and myocardial fibrosis without affecting hyperglycemia. This study shed light on how interaction of NO and H2S regulates cardiac pathology and provide new route to treat diabetic cardiomyopathy with HNO.
Collapse
|
207
|
Krog S, Ludvigsen TP, Nielsen OL, Kirk RK, Lykkegaard K, Wulff EM, Møller JE, Pedersen HD, Olsen LH. Myocardial Changes in Diabetic and Nondiabetic Nonhuman Primates. Vet Pathol 2020; 57:332-343. [PMID: 32096447 DOI: 10.1177/0300985820901332] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Diabetic human patients have increased risk of heart failure compared to healthy subjects. The underlying mechanisms for this are not fully understood, and to help develop improved treatment strategies, well-characterized animal models are essential. To investigate cardiac dysfunction in diabetes, this study evaluated myocardial changes in 10 aging rhesus monkeys with and without diabetes. Based on evaluation of plasma glycosylated hemoglobin and glucose, 7 of 10 rhesus macaques had diabetes for a minimum of 11 months, while 3 of 10 were categorized as nondiabetic. A detailed histological examination of formalin-fixed left ventricular myocardial samples was followed by a semiquantitative evaluation of myocardial fibrosis and fat infiltration; digital quantifications of myocardial collagen, lipofuscin, and nuclear area fractions; and measurements of cardiomyocyte diameter. Histological myocardial evaluation revealed the presence of lipofuscin; large nuclei; interstitial, replacement, and vascular fibrosis; adipocyte infiltration; and vacuolar degeneration with atrophy of cardiomyocytes and fibrosis. However, there were no differences between groups for semiquantitative fat infiltration, fibrosis, cardiomyocyte size, collagen, or nuclear and lipofuscin area fraction. Lipofuscin area fraction correlated with plasma insulin, triglyceride, total cholesterol, and high-density lipoprotein cholesterol concentrations. In conclusion, myocardial pathological changes were found in left ventricular myocardium in aged rhesus macaques, independent of the stage of diabetes. The duration of diabetes might have been too short to cause differences between groups.
Collapse
Affiliation(s)
- Simone Krog
- Faculty of Health and Medical Sciences, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Ole L Nielsen
- Faculty of Health and Medical Sciences, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Rikke K Kirk
- Novo Nordisk A/S, Global Drug Discovery, Måløv, Denmark
| | | | - Erik M Wulff
- Novo Nordisk A/S, Global Drug Discovery, Måløv, Denmark
| | - Jacob E Møller
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Henrik D Pedersen
- Faculty of Health and Medical Sciences, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.,Ellegaard Göttingen Minipigs A/S, Dalmose, Denmark
| | - Lisbeth H Olsen
- Faculty of Health and Medical Sciences, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
208
|
De Blasio MJ, Huynh N, Deo M, Dubrana LE, Walsh J, Willis A, Prakoso D, Kiriazis H, Donner DG, Chatham JC, Ritchie RH. Defining the Progression of Diabetic Cardiomyopathy in a Mouse Model of Type 1 Diabetes. Front Physiol 2020; 11:124. [PMID: 32153425 PMCID: PMC7045054 DOI: 10.3389/fphys.2020.00124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence of diabetes and its association with increased cardiovascular disease risk represents a major health issue worldwide. Diabetes-induced hyperglycemia is implicated as a central driver of responses in the diabetic heart such as cardiomyocyte hypertrophy, fibrosis, and oxidative stress, termed diabetic cardiomyopathy. The onset of these responses in the setting of diabetes has not been studied to date. This study aimed to determine the time course of development of diabetic cardiomyopathy in a model of type 1 diabetes (T1D) in vivo. Diabetes was induced in 6-week-old male FVB/N mice via streptozotocin (55 mg/kg i.p. for 5 days; controls received citrate vehicle). At 2, 4, 8, 12, and 16 weeks of untreated diabetes, left ventricular (LV) function was assessed by echocardiography before post-mortem quantification of markers of LV cardiomyocyte hypertrophy, collagen deposition, DNA fragmentation, and changes in components of the hexosamine biosynthesis pathway (HBP) were assessed. Blood glucose and HbA1c levels were elevated by 2 weeks of diabetes. LV and muscle (gastrocnemius) weights were reduced from 8 weeks, whereas liver and kidney weights were increased from 2 and 4 weeks of diabetes, respectively. LV diastolic function declined with diabetes progression, demonstrated by a reduction in E/A ratio from 4 weeks of diabetes, and an increase in peak A-wave amplitude, deceleration time, and isovolumic relaxation time (IVRT) from 4–8 weeks of diabetes. Systemic and local inflammation (TNFα, IL-1β, CD68) were increased with diabetes. The cardiomyocyte hypertrophic marker Nppa was increased from 8 weeks of diabetes while β-myosin heavy chain was increased earlier, from 2 weeks of diabetes. LV fibrosis (picrosirius red; Ctgf and Tgf-β gene expression) and DNA fragmentation (a marker of cardiomyocyte apoptosis) increased with diabetes progression. LV Nox2 and Cd36 expression were elevated after 16 weeks of diabetes. Markers of the LV HBP (Ogt, Oga, Gfat1/2 gene expression), and protein abundance of OGT and total O-GlcNAcylation, were increased by 16 weeks of diabetes. This is the first study to define the progression of cardiac markers contributing to the development of diabetic cardiomyopathy in a mouse model of T1D, confirming multiple pathways contribute to disease progression at various time points.
Collapse
Affiliation(s)
- Miles J De Blasio
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Nguyen Huynh
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Minh Deo
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Leslie E Dubrana
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jesse Walsh
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Andrew Willis
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Darnel Prakoso
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Helen Kiriazis
- Experimental Cardiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Daniel G Donner
- Experimental Cardiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - John C Chatham
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
209
|
Jelinic M, Kahlberg N, Leo CH, Ng HH, Rosli S, Deo M, Li M, Finlayson S, Walsh J, Parry LJ, Ritchie RH, Qin CX. Annexin-A1 deficiency exacerbates pathological remodelling of the mesenteric vasculature in insulin-resistant, but not insulin-deficient, mice. Br J Pharmacol 2020; 177:1677-1691. [PMID: 31724161 DOI: 10.1111/bph.14927] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/04/2019] [Accepted: 10/27/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Arterial stiffness, a characteristic feature of diabetes, increases the risk of cardiovascular complications. Potential mechanisms that promote arterial stiffness in diabetes include oxidative stress, glycation and inflammation. The anti-inflammatory protein annexin-A1 has cardioprotective properties, particularly in the context of ischaemia. However, the role of endogenous annexin-A1 in the vasculature in both normal physiology and pathophysiology remains largely unknown. Hence, this study investigated the role of endogenous annexin-A1 in diabetes-induced remodelling of mouse mesenteric vasculature. EXPERIMENTAL APPROACH Insulin-resistance was induced in male mice (AnxA1+/+ and AnxA1-/- ) with the combination of streptozotocin (55mg/kg i.p. x 3 days) with high fat diet (42% energy from fat) or citrate vehicle with normal chow diet (20-weeks). Insulin-deficiency was induced in a separate cohort of mice using a higher total streptozocin dose (55mg/kg i.p. x 5 days) on chow diet (16-weeks). At study endpoint, mesenteric artery passive mechanics were assessed by pressure myography. KEY RESULTS Insulin-resistance induced significant outward remodelling but had no impact on passive stiffness. Interestingly, vascular stiffness was significantly increased in AnxA1-/- mice when subjected to insulin-resistance. In contrast, insulin-deficiency induced outward remodelling and increased volume compliance in mesenteric arteries, regardless of genotype. In addition, the annexin-A1 / formyl peptide receptor axis is upregulated in both insulin-resistant and insulin-deficient mice. CONCLUSION AND IMPLICATIONS Our study provided the first evidence that endogenous AnxA1 may play an important vasoprotective role in the context of insulin-resistance. AnxA1-based therapies may provide additional benefits over traditional anti-inflammatory strategies for reducing vascular injury in diabetes.
Collapse
Affiliation(s)
- Maria Jelinic
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Nicola Kahlberg
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia
| | - Chen Huei Leo
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia.,Science, Math and Technology, Singapore University of Technology and Design, Singapore
| | - Hooi Hooi Ng
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia.,Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Sarah Rosli
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Minh Deo
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mandy Li
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Siobhan Finlayson
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jesse Walsh
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Laura J Parry
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
210
|
Erukainure OL, Chukwuma CI, Matsabisa MG, Salau VF, Koorbanally NA, Islam MS. Buddleja saligna Willd (Loganiaceae) inhibits angiotensin-converting enzyme activity in oxidative cardiopathy with concomitant modulation of nucleotide hydrolyzing enzymatic activities and dysregulated lipid metabolic pathways. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112358. [PMID: 31676404 DOI: 10.1016/j.jep.2019.112358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Buddleja saligna Willd (Loganiaceae), mostly indigenous to South Africa is traditionally used in the treatment cardio-dysfunctional related ailments amongst other diseases. AIMS The cardio-protective effect of B. saligna was investigated in ferric-induced oxidative cardiopathy. METHODS Hearts harvested from healthy male SD rats were incubated with 0.1 mM FeSO4 to induce oxidative damage and co-incubated with B. saligna extract. Reaction mixtures without the extract served as negative control, while tissues without the extract or standard antioxidant (gallic acid) and pro-oxidant served as the normal control. The tissues were analyzed for levels of glutathione, malondialdehyde, and nitric oxide as well as cholinergic, angiotensin-converting enzyme (ACE), lipase, and purinergic enzymes activities, lipid profiles, fatty acid metabolic pathways and metabolites. RESULTS Induction of oxidative damage significantly (p < 0.05) depleted the levels of GSH, SOD, catalase, and ENTPDase activities, while concomitantly elevating the levels of MDA, NO, ACE, acetylcholinesterase, lipase and ATPase activities. These levels and activities were significantly reversed on treatment with B. saligna. Treatment with B. saligna also led to depletion of cardiac cholesterol and LDL-c levels, while elevating triglyceride and HDL-c level. It also depleted oxidative-induced lipid metabolites with concomitant generation of thirteen other metabolites. B. saligna also inactivated oxidative-induced pathways for beta oxidation of very long chain fatty acids, glycerolipid metabolism, and fatty acid elongation in mitochondria. CONCLUSION These results suggest that B. saligna protects against ferric-induced oxidative cardiopathy by mitigating oxidative stress, while concomitantly inhibiting ACE, acetylcholinesterase and lipase activities, and modulating lipid spectrum and dysregulated metabolic pathways.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Chika I Chukwuma
- Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, 9300, South Africa
| | - Motlalepula G Matsabisa
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa.
| | - Veronica F Salau
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban, 4000, South Africa
| | - Neil A Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal, (Westville Campus), Durban, 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban, 4000, South Africa
| |
Collapse
|
211
|
Effects of Anchomanes difformis on Inflammation, Apoptosis, and Organ Toxicity in STZ-Induced Diabetic Cardiomyopathy. Biomedicines 2020; 8:biomedicines8020029. [PMID: 32046294 PMCID: PMC7168158 DOI: 10.3390/biomedicines8020029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/29/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022] Open
Abstract
Persistent hyperglycemia is known to cause enhanced generation of reactive oxygen species in diabetes. Several inflammatory cytokines are induced by oxidative stress, and their release also leads to increased oxidative stress; this makes oxidative stress one of the important factors in the development of chronic inflammation and other immune responses. These have been implicated in the development of diabetic complications such as nephropathy and cardiomyopathy. Anchomanes difformis has been shown to possess antioxidant and anti-inflammatory potentials. The present study investigated the immunomodulatory potential and the antiapoptotic ability of Anchomanes difformis to ameliorate heart toxicity and injury in type II diabetes. Two weeks of fructose (10%) administration followed by single intraperitoneal injection of streptozotocin (40 mg/kg) were used to induce type II diabetes in male Wistar rats. Leaf extract (aqueous) of Anchomanes difformis (200 and 400 mg/kg) was administered orally for six weeks. Blood glucose concentrations and body weights before and after interventions were determined. Interleukin (IL)-1β, IL-6, IL-10, IL-18, monocyte chemoattractant protein 1 (MCP-1), and tumor necrosis factor alpha (TNFα) were measured in the heart homogenates. Catalase (CAT), superoxide dismutase (SOD), total protein, oxygen radical absorbance capacity (ORAC), ferric reducing antioxidant power (FRAP), thiobarbituric acid reactive substances (TBARS), and heart-type fatty acid-binding protein (H-FABP) levels were determined. Expressions of transcription factors (Nrf 2 and NFkB/p65) and apoptotic markers were also investigated in the heart. Anchomanesdifformis administration reduced pro-inflammatory cytokines, increased anti-inflammatory markers, and enhanced antioxidant defense in the heart of diabetic treated animals. Anchomanesdifformis is a new, promising therapeutic agent that can be explored for the treatment of pathological conditions associated with immune responses and will be a useful tool in the management of associated diabetic complications.
Collapse
|
212
|
Zhang SY, Yang KL, Long ZY, Li WQ, Huang HY. Use of a Systematic Pharmacological Methodology to Explore the Mechanism of Shengmai Powder in Treating Diabetic Cardiomyopathy. Med Sci Monit 2020; 26:e919029. [PMID: 32023237 PMCID: PMC7020766 DOI: 10.12659/msm.919029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/08/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cardiovascular complications, such as diabetic cardiomyopathy (DCM), are the leading cause of death in diabetic patients. Shengmai Powder (SMP) was found to have cardioprotective effects. MATERIAL AND METHODS Based on the systematic pharmacological methodology, this research determined the genes of DCM and the known targets of SMP, predicted potential compounds and targets of SMP, constructed networks for DCM and SMP, and performed network analysis. RESULTS Five network were constructed: (1) the DCM gene PPI network; (2) the Compound-compound target network of SMP; (3) the SMP-DCM PPI network; (4) the Compound-known target network of SMP; (5) and the SMP known target-DCM PPI network. Several DCM and treatment related targets, clusters, signaling pathways, and biological processes were found. CONCLUSIONS SMP is able to regulate glycometabolism-related, lipid metabolism-related, inflammatory response-related, oxidative stress-related signaling pathways, and biological processes and targets, which suggests that SMP may have a therapeutic effect on DCM.
Collapse
Affiliation(s)
- Shi-ying Zhang
- Department of Traditional Chinese Medicine, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
- Hunan University of Chinese Medicine, Changsha, Hunan, P.R. China
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
- Department of Traditional Chinese Medicine, Shenzhen Luohu Hospital Group Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Kai-lin Yang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, P.R. China
- Capital Medical University, Beijing, P.R. China
| | - Zhi-yong Long
- Shantou University Medical College, Shantou University, Shantou, Guangdong, P.R. China
- Department of Rehabilitation Medicine, Guangdong Geriatric Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Wei-qing Li
- Department of Traditional Chinese Medicine, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
- Department of Traditional Chinese Medicine, Shenzhen Luohu Hospital Group Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Hui-yong Huang
- Hunan University of Chinese Medicine, Changsha, Hunan, P.R. China
| |
Collapse
|
213
|
Taurine protects against cardiac dysfunction induced by pressure overload through SIRT1-p53 activation. Chem Biol Interact 2020; 317:108972. [PMID: 32017914 DOI: 10.1016/j.cbi.2020.108972] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/21/2019] [Accepted: 01/31/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND Heart failure (HF) is an epidemic disease with increased incidence annually. It has been reported that taurine can improve cardiac function. This study investigated the cardioprotective effects of taurine in pressure-loaded HF mice and elucidated the possible mechanism. METHODS HF models were established by transverse aortic constriction (TAC). Animals were treated with either taurine for 9 weeks and/or the SIRT1 inhibitor EX527 (5 mg/kg/day, every 2days) after TAC operation. Cardiac function and geometry were revealed by echocardiography. Myocardial hypertrophy and fibrosis were assessed using Fluorescent wheat germ agglutinin (WGA) staining and Masson's trichrome staining. Western blot and RT-PCR were performed to elucidate the expression of target proteins and genes respectively. Apoptosis in cardiomyocytes was detected by TUNEL staining. Myocardial oxidative stress was assessed by detecting the concentration of myocardial super oxidative dismutase (SOD) and malonyldialdehyde (MDA) and reactive oxygen species (ROS). Taurine concentrations and NAD+/NADH ratio were determined by taurine and NAD+/NADH assay kit. RESULTS Taurine notably relieved cardiac dysfunction after TAC. The mechanisms were attributed to reduced myocyte hypertrophy and fibrosis, and alleviated apoptosis and oxidative stress. Meanwhile, taurine increased NAD+/NADH ratio,promoted the expression of SIRT1 and suppressed p53 acetylation. However, EX-527(inhibitor of SIRT1) decreased NAD+/NADH ratio and increased acetyl-p53 levels, and abolished the cardioprotective effects of taurine on mice subjected to TAC and increased apoptosis and oxidative stress. CONCLUSION The mechanism responsible for cardiac-protective effects of taurine in HF induced by pressure overload is associated with the activation of the SIRT1-p53 pathway.
Collapse
|
214
|
Fu T, Mohan M, Brennan EP, Woodman OL, Godson C, Kantharidis P, Ritchie RH, Qin CX. Therapeutic Potential of Lipoxin A 4 in Chronic Inflammation: Focus on Cardiometabolic Disease. ACS Pharmacol Transl Sci 2020; 3:43-55. [PMID: 32259087 DOI: 10.1021/acsptsci.9b00097] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Several studies have shown that failure to resolve inflammation may contribute to the progression of many chronic inflammatory disorders. It has been suggested targeting the resolution of inflammation might be a novel therapeutic approach for chronic inflammatory diseases, including inflammatory bowel disease, diabetic complications, and cardiometabolic disease. Lipoxins [LXs] are a class of endogenously generated mediators that promote the resolution of inflammation. Biological actions of LXs include inhibition of neutrophil infiltration, promotion of macrophage polarization, increase of macrophage efferocytosis, and restoration of tissue homeostasis. Recently, several studies have demonstrated that LXs and synthetic analogues protect tissues from acute and chronic inflammation. The mechanism includes down-regulation of pro-inflammatory cytokines and chemokines (e.g., interleukin-1β and tumor necrosis factor-α), inhibition of the activation of the master pro-inflammatory pathway (e.g., nuclear factor κ-light-chain-enhancer of activated B cells pathway) and increased release of the pro-resolving cytokines (e.g., interleukin-10). Three generations of LXs analogues are well described in the literature, and more recently a fourth generation has been generated that appears to show enhanced potency. In this review, we will briefly discuss the potential therapeutic opportunity provided by lipoxin A4 as a novel approach to treat chronic inflammatory disorders, focusing on cardiometabolic disease and the current drug development in this area.
Collapse
Affiliation(s)
- Ting Fu
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Muthukumar Mohan
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia
| | - Eoin P Brennan
- UCD Diabetes Complications Research Centre, UCD Conway Institute, UCD School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Owen L Woodman
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD Conway Institute, UCD School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Phillip Kantharidis
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia
| | - Rebecca H Ritchie
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia.,Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
215
|
Wang SY, Zhu S, Wu J, Zhang M, Xu Y, Xu W, Cui J, Yu B, Cao W, Liu J. Exercise enhances cardiac function by improving mitochondrial dysfunction and maintaining energy homoeostasis in the development of diabetic cardiomyopathy. J Mol Med (Berl) 2020; 98:245-261. [PMID: 31897508 DOI: 10.1007/s00109-019-01861-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a major cause of morbidity and mortality in diabetic patients. Reactive oxygen species (ROS) produced by oxidative stress play an important role in the development of DCM. DCM involves abnormal energy metabolism, thereby reducing energy production. Exercise has been reported to be effective in protecting the heart against ROS accumulation during the development of DCM. We hypothesize that the AMPK/PGC-1α axis may play a crucial role in exercise-induced bioenergetic metabolism and aerobic respiration on oxidative stress parameters in the development of diabetic cardiomyopathy. Using a streptozotocin/high-fat diet mouse to generate a diabetic model, our aim was to evaluate the effects of exercise on the cardiac function, mitochondrial oxidative capacity, mitochondrial function, and cardiac expression of PGC-1α. Mice fed a high-fat diet were given MO-siPGC-1α or treated with AMPK inhibitor. Mitochondrial structure and effects of switching between the Warburg effect and aerobic respiration were analysed. Exercise improved blood pressure and systolic dysfunction in diabetic mouse hearts. The beneficial effects of exercise were also observed in a mitochondrial function study, as reflected by an enhanced oxidative phosphorylation level, increased membrane potential, and decreased ROS level and oxygen consumption. On the other hand, depletion of PGC-1α attenuated the effects of exercise on the enhancement of mitochondrial function. In addition, PGC-1α may be responsible for reversing the Warburg effect to aerobic respiration, thus enhancing mitochondrial metabolism and energy homoeostasis. In this study, we demonstrate the protective effects of exercise on shifting energy metabolism from fatty acid oxidation to glucose oxidation in an established diabetic stage. These data suggest that exercise is effective at ameliorating diabetic cardiomyopathy by improving mitochondrial function and reducing metabolic disturbances.
Collapse
Affiliation(s)
- Shawn Yongshun Wang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China.,Department of Biomedical Science, University of Hong Kong, Pokfulam, Hong Kong
| | - Siyu Zhu
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Jian Wu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Maomao Zhang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Yousheng Xu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Wei Xu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Jinjin Cui
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Bo Yu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Wei Cao
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China. .,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China.
| | - Jingjin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China. .,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China. .,Department of Anesthesiology, University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
216
|
Qin CX, Anthonisz J, Leo CH, Kahlberg N, Velagic A, Li M, Jap E, Woodman OL, Parry LJ, Horowitz JD, Kemp-Harper BK, Ritchie RH. Nitric Oxide Resistance, Induced in the Myocardium by Diabetes, Is Circumvented by the Nitric Oxide Redox Sibling, Nitroxyl. Antioxid Redox Signal 2020; 32:60-77. [PMID: 31680536 DOI: 10.1089/ars.2018.7706] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aim: Impairment of tissue responsiveness to exogenous and endogenous nitric oxide (NO•), known as NO• resistance, occurs in many cardiovascular disease states, prominently in diabetes and especially in the presence of marked hyperglycemia. In this study, we sought to determine in moderate and severe diabetes (i) whether NO• resistance also occurs in the myocardium, and (ii) whether the NO• redox sibling nitroxyl (HNO) circumvents this. Results: The spectrum of acute NO• effects (induced by diethylamine-NONOate), including vasodilation, and enhanced myocardial contraction and relaxation were impaired by moderately diabetic rats ([blood glucose] ∼20 mM). In contrast, acute HNO effects (induced by isopropylamine-NONOate) were preserved even in more severe diabetes ([blood glucose] >28 mM). Intriguingly, the positive inotropic effects of HNO were significantly enhanced in diabetic rat hearts. Further, progressive attenuation of soluble guanylyl cyclase (sGC) contribution to myocardial NO• responses occurred with increasing severity of diabetes. Nevertheless, activation of sGC by HNO remained intact in the myocardium. Innovation: Diabetes is associated with marked attenuation of vascular and myocardial effects of NO and NO donors, and this NO• resistance is circumvented by HNO, suggesting potential therapeutic utility for HNO donors in cardiovascular emergencies in diabetics. Conclusion: These results provide the first evidence that NO• resistance occurs in diabetic hearts, and that HNO largely circumvents this problem. Further, the positive inotropic and lusitropic effects of HNO are enhanced in a severely diabetic myocardium, a finding that warrants further mechanistic interrogation. The results support a potential role for therapeutic HNO administration in acute treatment of ischemia and/or heart failure in diabetics.
Collapse
Affiliation(s)
- Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| | - Jarryd Anthonisz
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| | - Chen Huei Leo
- School of Biosciences, University of Melbourne, Parkville, Australia.,Science and Maths Cluster, Singapore University of Technology & Design, Singapore Singapore
| | - Nicola Kahlberg
- School of Biosciences, University of Melbourne, Parkville, Australia
| | - Anida Velagic
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| | - Mandy Li
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Edwina Jap
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Owen L Woodman
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Laura J Parry
- School of Biosciences, University of Melbourne, Parkville, Australia
| | - John D Horowitz
- Cardiology Unit, The Queen Elizabeth Hospital, Basil Hetzel Institute, The University of Adelaide, Woodville SA, Australia
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia.,Department of Medicine (Central Clinical School), Monash University, Melbourne, Australia
| |
Collapse
|
217
|
Sefidgari-Abrasi S, Karimi P, Roshangar L, Morshedi M, Bavafa-Valenlia K, Saghafi-Asl M, Mohiti S, Rahimiyan-Heravan M. Lactobacillus plantarum And Inulin: Therapeutic Agents to Enhance Cardiac Ob Receptor Expression and Suppress Cardiac Apoptosis in Type 2 Diabetic Rats. J Diabetes Res 2020; 2020:4745389. [PMID: 32509880 PMCID: PMC7246403 DOI: 10.1155/2020/4745389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/23/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND T2DM may cause increased levels of oxidative stress and cardiac apoptosis through elevated blood glucose. The present study investigated the effects of Lactobacillus plantarum (L. plantarum) as a probiotic strain and inulin as a prebiotic supplement on cardiac oxidative stress and apoptotic markers in type 2 diabetes mellitus (T2DM) rats. METHODS A high-fat diet and a low dose of streptozotocin were used to induce type 2 diabetes. The rats were divided into six groups which were supplemented with L. plantarum, inulin, or their combination for 8 weeks. RESULTS The results showed improved activity of cardiac antioxidant parameters including total antioxidant capacity (TAC), superoxide dismutase (SOD), and glutathione peroxidase (GPx) (P < 0.001, P < 0.01, and P < 0.01, respectively) and decreased level of cardiac malondialdehyde (MDA) concentration (P < 0.05). These changes were accompanied with increased protein expression of cardiac obesity receptor (Ob-R) (P = 0.05) and reduced apoptotic markers such as tumor necrosis factor-alpha (TNF-α), Fas ligand (FasL), and caspase proteins (P < 0.001, P = 0.003, and P < 0.01, respectively) in T2DM rats after concurrent L. plantarum and inulin supplementation. Moreover, a remarkable correlation of cardiac Ob-R and oxidative stress parameters with cardiac apoptotic markers was observed (P < 0.01). CONCLUSION The concurrent use of L. plantarum and inulin seems to be beneficial, as they can lead to decreased heart complications of T2DM via reducing cardiac apoptotic markers.
Collapse
Affiliation(s)
- Safa Sefidgari-Abrasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Morshedi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Bavafa-Valenlia
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Saghafi-Asl
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Mohiti
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Rahimiyan-Heravan
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
218
|
Luo J, Yan D, Li S, Liu S, Zeng F, Cheung CW, Liu H, Irwin MG, Huang H, Xia Z. Allopurinol reduces oxidative stress and activates Nrf2/p62 to attenuate diabetic cardiomyopathy in rats. J Cell Mol Med 2019; 24:1760-1773. [PMID: 31856386 PMCID: PMC6991641 DOI: 10.1111/jcmm.14870] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/15/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022] Open
Abstract
Allopurinol (ALP) attenuates oxidative stress and diabetic cardiomyopathy (DCM), but the mechanism is unclear. Activation of nuclear factor erythroid 2‐related factor 2 (Nrf2) following the disassociation with its repressor Keap1 under oxidative stress can maintain inner redox homeostasis and attenuate DCM with concomitant attenuation of autophagy. We postulated that ALP treatment may activate Nrf2 to mitigate autophagy over‐activation and consequently attenuate DCM. Streptozotocin‐induced type 1 diabetic rats were untreated or treated with ALP (100 mg/kg/d) for 4 weeks and terminated after heart function measurements by echocardiography and pressure‐volume conductance system. Cardiomyocyte H9C2 cells infected with Nrf2 siRNA or not were incubated with high glucose (HG, 25 mmol/L) concomitantly with ALP treatment. Cell viability, lactate dehydrogenase, 15‐F2t‐Isoprostane and superoxide dismutase (SOD) were measured with colorimetric enzyme‐linked immunosorbent assays. ROS, apoptosis, was assessed by dihydroethidium staining and TUNEL, respectively. The Western blot and qRT‐PCR were used to assess protein and mRNA variations. Diabetic rats showed significant reductions in heart rate (HR), left ventricular eject fraction (LVEF), stroke work (SW) and cardiac output (CO), left ventricular end‐systolic volume (LVVs) as compared to non‐diabetic control and ALP improved or normalized HR, LVEF, SW, CO and LVVs in diabetic rats (all P < .05). Hearts of diabetic rats displayed excessive oxidative stress manifested as increased levels of 15‐F2t‐Isoprostane and superoxide anion production, increased apoptotic cell death and cardiomyocytes autophagy that were concomitant with reduced expressions of Nrf2, heme oxygenase‐1 (HO‐1) and Keap1. ALP reverted all the above‐mentioned diabetes‐induced biochemical changes except that it did not affect the levels of Keap1. In vitro, ALP increased Nrf2 and reduced the hyperglycaemia‐induced increases of H9C2 cardiomyocyte hypertrophy, oxidative stress, apoptosis and autophagy, and enhanced cellular viability. Nrf2 gene silence cancelled these protective effects of ALP in H9C2 cells. Activation of Nrf2 subsequent to the suppression of Keap1 and the mitigation of autophagy over‐activation may represent major mechanisms whereby ALP attenuates DCM.
Collapse
Affiliation(s)
- Jierong Luo
- Department of Anesthesiology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Dan Yan
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Sisi Li
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Shiming Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fei Zeng
- Department of Anesthesiology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chi Wai Cheung
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA, USA
| | - Michael G Irwin
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Huansen Huang
- Department of Anesthesiology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
219
|
Karan A, Bhakkiyalakshmi E, Jayasuriya R, Sarada DVL, Ramkumar KM. The pivotal role of nuclear factor erythroid 2-related factor 2 in diabetes-induced endothelial dysfunction. Pharmacol Res 2019; 153:104601. [PMID: 31838079 DOI: 10.1016/j.phrs.2019.104601] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/23/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Endothelial dysfunction (ED) is a key event in the onset and progression of vascular complications associated with diabetes. Regulation of endothelial function and the underlying signaling mechanisms in the progression of diabetes-induced vascular complications have been well established. Recent studies indicate that increased oxidative stress is an important determinant of endothelial injury and patients with hypertension display ED mediated by impaired Nitric Oxide (NO) availability. Further, oxidative stress is known to be associated with inflammation and ED in vascular remodeling and diabetes-associated hypertension. Numerous strategies have been developed to improve the function of endothelial cells and increasing number of evidences highlight the indispensable role of antioxidants in modulation of endothelium-dependent vasodilation responses. Nuclear factor Erythroid 2-related factor 2 (Nrf2), is the principal transcriptional regulator, that is central in mediating oxidative stress signal response. Having unequivocally established the relationship between type 2 diabetes mellitus (T2DM) and oxidative stress, the pivotal role of Nrf2/Keap1/ARE network, has taken the center stage as target for developing therapies towards maintaining the cellular redox environment. Several activators of Nrf2 are known to combat diabetes-induced ED and few are currently in clinical trials. Focusing on their therapeutic value in diabetes-induced ED, this review highlights some natural and synthetic molecules that are involved in the modulation of the Nrf2/Keap1/ARE network and its underlying molecular mechanisms in the regulation of ED. Further emphasis is also laid on the therapeutic benefits of directly up-regulating Nrf2-mediated antioxidant defences in regulating endothelial redox homeostasis for countering diabetes-induced ED.
Collapse
Affiliation(s)
- Amin Karan
- Life Science Division, SRM Research Institute, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamilnadu, India; Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Elango Bhakkiyalakshmi
- Life Science Division, SRM Research Institute, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamilnadu, India; Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Ravichandran Jayasuriya
- Life Science Division, SRM Research Institute, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamilnadu, India; Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - D V L Sarada
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Life Science Division, SRM Research Institute, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamilnadu, India; Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
220
|
Ramalingam A, Budin SB, Mohd Fauzi N, Ritchie RH, Zainalabidin S. Angiotensin II Type I Receptor Antagonism Attenuates Nicotine-Induced Cardiac Remodeling, Dysfunction, and Aggravation of Myocardial Ischemia-Reperfusion Injury in Rats. Front Pharmacol 2019; 10:1493. [PMID: 31920673 PMCID: PMC6920178 DOI: 10.3389/fphar.2019.01493] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
Increased exposure to nicotine contributes to the development of cardiac dysfunction by promoting oxidative stress, fibrosis, and inflammation. These deleterious events altogether render cardiac myocytes more susceptible to acute cardiac insults such as ischemia-reperfusion (I/R) injury. This study sought to elucidate the role of angiotensin II type I (AT1) receptors in cardiac injury resulting from prolonged nicotine administration in a rat model. Male Sprague-Dawley rats were given nicotine (0.6 mg/kg ip) for 28 days to induce cardiac dysfunction, alone or in combination with the AT1 receptor antagonist, irbesartan (10 mg/kg, po). Vehicle-treated rats were used as controls. Rat hearts isolated from each experimental group at study endpoint were examined for changes in function, histology, gene expression, and susceptibility against acute I/R injury determined ex vivo. Rats administered nicotine alone exhibited significantly increased cardiac expression of angiotensin II and angiotensin-converting enzyme (ACE) in addition to elevated systolic blood pressure (SBP) and heart rate. Furthermore, nicotine administration markedly reduced left ventricular (LV) performance with concomitant increases in myocardial oxidative stress, fibrosis, and inflammation. Concomitant treatment with irbesartan attenuated these effects, lowering blood pressure, heart rate, oxidative stress, and expression of fibrotic and inflammatory genes. Importantly, the irbesartan-treated group also manifested reduced susceptibility to I/R injury ex vivo. These findings suggest that AT1 receptors play an important role in nicotine-induced cardiac dysfunction, and pharmacological approaches targeting cardiac AT1 receptors may thus benefit patients with sustained exposure to nicotine.
Collapse
Affiliation(s)
- Anand Ramalingam
- Programme of Biomedical Science, Centre for Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Balkis Budin
- Programme of Biomedical Science, Centre for Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Norsyahida Mohd Fauzi
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Satirah Zainalabidin
- Programme of Biomedical Science, Centre for Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
221
|
Zhang ZY, Wang N, Qian LL, Miao LF, Dang SP, Wu Y, Wang RX. Glucose Fluctuations Promote Aortic Fibrosis through the ROS/p38 MAPK/Runx2 Signaling Pathway. J Vasc Res 2019; 57:24-33. [PMID: 31715615 DOI: 10.1159/000503608] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/23/2019] [Indexed: 11/19/2022] Open
Abstract
AIM Glucose fluctuations may be responsible for, or further the onset of arterial hypertension, but the exact mechanisms remain unclear. The purpose of this study was to investigate the mechanisms behind and related to aortic fibrosis and aortic stiffening induced by glucose fluctuations. METHODS Sprague-Dawley rats were injected with streptozotocin (STZ) and randomly divided into three treatment groups: controlled STZ-induced diabetes (C-STZ); uncontrolled STZ-induced diabetes (U-STZ); and STZ-induced diabetes with glucose fluctuations (STZ-GF). After 3 weeks, rat blood pressure (BP) was tested, and aortic fibrosis was detected by using the Masson trichrome staining technique. Levels of p38 mitogen-activated protein kinase (p38 MAPK), runt-related transcription factor 2 (Runx2), collagen type 1 (collagen I), and NADPH oxidases were determined by Western blot.Rat vascular smooth muscle cells in vitro were used to explore underlying mechanisms. RESULTS The systolic BP of diabetic rats in the C-STZ, U-STZ, and STZ-GF groups was 127.67 ± 6.53, 150.03 ± 5.24, and 171.63 ± 3.53 mm Hg, respectively (p< 0.05). The mean BP of diabetic rats in the three groups was 91.20 ± 10.07, 117.29 ± 4.28, and 140.58 ± 2.14 mm Hg, respectively (p< 0.05). The diastolic BP of diabetic rats in the three groups was 73.20 ± 12.63, 101.93 ± 5.79, and 125.37 ± 4.62 mm Hg, respectively (p< 0.05). The ratios of fibrosis areas in the aortas of the three groups were 11.85 ± 1.23, 29.00 ± 0.87, and 48.36 ± 0.55, respectively (p< 0.05). The expressions of p38 MAPK, Runx2, and collagen I were significantly increased in the STZ-GF group. In vitro, applications of inhibitors of reactive oxygen species (ROS) and p38 MAPK successfully reversed glucose fluctuations that would have possibly induced aortic fibrosis. CONCLUSIONS Blood glucose fluctuations aggravate aortic fibrosis via affecting the ROS/p38 MAPK /Runx2 signaling pathway.
Collapse
Affiliation(s)
- Zhen-Ye Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ning Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ling-Ling Qian
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ling-Feng Miao
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Shi-Peng Dang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ying Wu
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ru-Xing Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China,
| |
Collapse
|
222
|
D'Amario D, Migliaro S, Borovac JA, Restivo A, Vergallo R, Galli M, Leone AM, Montone RA, Niccoli G, Aspromonte N, Crea F. Microvascular Dysfunction in Heart Failure With Preserved Ejection Fraction. Front Physiol 2019; 10:1347. [PMID: 31749710 PMCID: PMC6848263 DOI: 10.3389/fphys.2019.01347] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an increasingly studied entity accounting for 50% of all diagnosed heart failure and that has claimed its own dignity being markedly different from heart failure with reduced EF in terms of etiology and natural history (Graziani et al., 2018). Recently, a growing body of evidence points the finger toward microvascular dysfunction as the major determinant of the pathological cascade that justifies clinical manifestations (Crea et al., 2017). The high burden of comorbidities such as metabolic syndrome, hypertension, atrial fibrillation, chronic kidney disease, obstructive sleep apnea, and similar, could lead to a systemic inflammatory state that impacts the physiology of the endothelium and the perivascular environment, engaging complex molecular pathways that ultimately converge to myocardial fibrosis, stiffening, and dysfunction (Paulus and Tschope, 2013). These changes could even self-perpetrate with a positive feedback where hypoxia and locally released inflammatory cytokines trigger interstitial fibrosis and hypertrophy (Ohanyan et al., 2018). Identifying microvascular dysfunction both as the cause and the maintenance mechanism of this condition has opened the field to explore specific pharmacological targets like nitric oxide (NO) pathway, sarcomeric titin, transforming growth factor beta (TGF-β) pathway, immunomodulators or adenosine receptors, trying to tackle the endothelial impairment that lies in the background of this syndrome (Graziani et al., 2018;Lam et al., 2018). Yet, many questions remain, and the new data collected still lack a translation to improved treatment strategies. To further elaborate on this tangled and exponentially growing topic, we will review the evidence favoring a microvasculature-driven etiology of this condition, its clinical correlations, the proposed diagnostic workup, and the available/hypothesized therapeutic options to address microvascular dysfunction in the failing heart.
Collapse
Affiliation(s)
- Domenico D'Amario
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Stefano Migliaro
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Josip A Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split, Croatia
| | - Attilio Restivo
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Rocco Vergallo
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mattia Galli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Maria Leone
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Rocco A Montone
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giampaolo Niccoli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Nadia Aspromonte
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
223
|
Dludla PV, Orlando P, Silvestri S, Mazibuko-Mbeje SE, Johnson R, Marcheggiani F, Cirilli I, Muller CJF, Louw J, Obonye N, Nyawo T, Nkambule BB, Tiano L. N-Acetyl cysteine ameliorates hyperglycemia-induced cardiomyocyte toxicity by improving mitochondrial energetics and enhancing endogenous Coenzyme Q 9/10 levels. Toxicol Rep 2019; 6:1240-1245. [PMID: 31799124 PMCID: PMC6883304 DOI: 10.1016/j.toxrep.2019.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/23/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Hyperglycemia is known to accelerate oxidative stress-induced myocardial injury. Mitochondrial energetics is an important mechanism to explore in the diabetic heart. NAC protects against hyperglycemia-induced cardiomyocyte toxicity. NAC improves mitochondrial energetics and enhances endogenous CoQ levels. CoQ supports the process of bioenergetics in addition to its antioxidant activities.
The diabetic heart has been linked with reduced endogenous levels of coenzyme Q9/10 (CoQ), an important antioxidant and component of the electron transport chain. Although CoQ has displayed cardioprotective potential in experimental models of diabetes, the impact of N-acetyl cysteine (NAC) on mitochondrial energetics and endogenous levels of CoQ remains to be clarified. To explore these effects, high glucose-exposed H9c2 cardiomyocytes were used as an experimental model of hyperglycemia-induced cardiac injury. The results showed that high glucose exposure caused an increased production of reactive oxygen species (ROS), which was associated with impaired mitochondrial energetics as confirmed by a reduction of maximal respiration rate and depleted ATP levels. These detrimental effects were consistent with significantly reduced endogenous CoQ levels and accelerated cell toxicity. Although metformin demonstrated similar effects on mitochondrial energetics and cell viability, NAC demonstrated a more pronounced effect in ameliorating cytosolic and mitochondrial ROS production. Interestingly, the ameliorative effects of NAC against hyperglycemia-induced injury were linked with its capability to enhance endogenous CoQ levels. Although such data are to be confirmed in other models, especially in vivo studies, the overall findings provide additional evidence on the therapeutic mechanisms by which NAC protects against diabetes-induced cardiac injury.
Collapse
Key Words
- ATP, adenosine triphosphate
- CoQ9/10, Coenzyme Q9/10
- Coenzyme Q
- DCFH-DA, dichlorofluorescein diacetate
- DMEM, Dulbecco’s Modified Eagle’s Medium
- Diabetes
- ECAR, extracellular acidification rates
- FBS, fetal bovine serum
- HPLC, high-performance liquid chromatograph
- Hyperglycemia
- MET, metformin
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Mitochondrial energetics
- N-Acetyl cysteine
- NAC, N-acetyl cysteine
- PBS, Phosphate buffered saline
- ROS, reactive oxygen species
- Reactive oxygen species
Collapse
Affiliation(s)
- Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Sithandiwe E Mazibuko-Mbeje
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Ilenia Cirilli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Nnini Obonye
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Thembeka Nyawo
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| |
Collapse
|
224
|
MiR-207 inhibits autophagy and promotes apoptosis of cardiomyocytes by directly targeting LAMP2 in type 2 diabetic cardiomyopathy. Biochem Biophys Res Commun 2019; 520:27-34. [DOI: 10.1016/j.bbrc.2019.09.092] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023]
|
225
|
Abstract
Patients with diabetes mellitus have >2× the risk for developing heart failure (HF; HF with reduced ejection fraction and HF with preserved ejection fraction). Cardiovascular outcomes, hospitalization, and prognosis are worse for patients with diabetes mellitus relative to those without. Beyond the structural and functional changes that characterize diabetic cardiomyopathy, a complex underlying, and interrelated pathophysiology exists. Despite the success of many commonly used antihyperglycemic therapies to lower hyperglycemia in type 2 diabetes mellitus the high prevalence of HF persists. This, therefore, raises the possibility that additional factors beyond glycemia might contribute to the increased HF risk in diabetes mellitus. This review summarizes the state of knowledge about the impact of existing antihyperglycemic therapies on HF and discusses potential mechanisms for beneficial or deleterious effects. Second, we review currently approved pharmacological therapies for HF and review evidence that addresses their efficacy in the context of diabetes mellitus. Dysregulation of many cellular mechanisms in multiple models of diabetic cardiomyopathy and in human hearts have been described. These include oxidative stress, inflammation, endoplasmic reticulum stress, aberrant insulin signaling, accumulation of advanced glycated end-products, altered autophagy, changes in myocardial substrate metabolism and mitochondrial bioenergetics, lipotoxicity, and altered signal transduction such as GRK (g-protein receptor kinase) signaling, renin angiotensin aldosterone signaling and β-2 adrenergic receptor signaling. These pathophysiological pathways might be amenable to pharmacological therapy to reduce the risk of HF in the context of type 2 diabetes mellitus. Successful targeting of these pathways could alter the prognosis and risk of HF beyond what is currently achieved using existing antihyperglycemic and HF therapeutics.
Collapse
Affiliation(s)
- Helena C Kenny
- From the Fraternal Order of Eagles Diabetes Research Center, and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| | - E Dale Abel
- From the Fraternal Order of Eagles Diabetes Research Center, and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| |
Collapse
|
226
|
Quantitative proteomic analysis reveals high interference on protein expression of H9c2 cells activated with glucose and cardiotonic steroids. J Proteomics 2019; 211:103536. [PMID: 31629057 DOI: 10.1016/j.jprot.2019.103536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/09/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023]
Abstract
In recent decades, the incidence of death and morbidity due to diabetes has increased worldwide, causing a high social and economic impact. Diabetes is a major cause of blindness, kidney failure, heart attack, stroke and lower limb amputation. However, the molecular mechanisms that make the heart and kidneys the main targets of diabetes are not completely understood. To better understand the complex biochemical mechanism of diabetic cardiomyopathy, we investigated the effects of hyperglycemia with concomitant digoxin and ouabain stimulation in H9c2 cells. Total extracted proteins were analyzed by label-free LC-MS/MS, quantified by Scaffold software and validated by parallel reaction monitoring (PRM) methodology. Here, we show that the eukaryotic initiation factors (Eifs) and elongation factors (Eefs) Eif3f, Eef2 and Eif4a1 are overexpressed following cardiotonic steroid (CTS) stimulation. Similarly, the expression of four 14-3-3 proteins that play a key role in cardiac ventricular compaction was altered after CTS stimulation. In total, the expression of nine protein groups was altered in response to the stimulation of H9c2 cells. Here, the biological consequences of these changes are discussed in depth. SIGNIFICANCE: Hyperglycemia is the main physiological condition that provokes tissue and vascular injuries in heart of diabetic patients. However, the changings at large scale in the expression of proteins of cardiomyocytes generated by this condition was not yet studied. Here we report for the first time the altered biosynthesis of nine groups of proteins of H9c2 cells activated by high glucose concentrations and by cardiotonic steroids (CTS). Furthermore, the increased biosynthesis of Eifs, Eefs and 14-3-3 protein groups by CTS, which play a crucial role in cardiomyopathies are original data reported in this work. These findings not only enhance our knowledge concerning to the effects of hyperglycemia and CTS on H9c2 cells but also indicate potential molecular targets to interfere in diabetes cardiomyopathy progression.
Collapse
|
227
|
Zhang ZY, Miao LF, Qian LL, Wang N, Qi MM, Zhang YM, Dang SP, Wu Y, Wang RX. Molecular Mechanisms of Glucose Fluctuations on Diabetic Complications. Front Endocrinol (Lausanne) 2019; 10:640. [PMID: 31620092 PMCID: PMC6759481 DOI: 10.3389/fendo.2019.00640] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence indicates the occurrence and development of diabetic complications relates to not only constant high plasma glucose, but also glucose fluctuations which affect various kinds of molecular mechanisms in various target cells and tissues. In this review, we detail reactive oxygen species and their potentially damaging effects upon glucose fluctuations and resultant downstream regulation of protein signaling pathways, including protein kinase C, protein kinase B, nuclear factor-κB, and the mitogen-activated protein kinase signaling pathway. A deeper understanding of glucose-fluctuation-related molecular mechanisms in the development of diabetic complications may enable more potential target therapies in future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ru-Xing Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
228
|
Xu X, Kobayashi S, Timm D, Huang Y, Zhao F, Shou W, Liang Q. Enhanced mTOR complex 1 signaling attenuates diabetic cardiac injury in OVE26 mice. FASEB J 2019; 33:12800-12811. [PMID: 31469601 DOI: 10.1096/fj.201901206r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The protein kinase mechanistic target of rapamycin (mTOR) performs diverse cellular functions through 2 distinct multiprotein complexes, mTOR complex (mTORC)1 and 2. Numerous studies using rapamycin, an mTORC1 inhibitor, have implicated a role for mTORC1 in several types of heart disease. People with diabetes are more susceptible to heart failure. mTORC1 activity is increased in the diabetic heart, but its functional significance remains controversial. To investigate the role of mTORC1 in the diabetic heart, we crossed OVE26 type 1 diabetic mice with transgenic mice expressing a constitutively active mTOR (mTORca) or kinase-dead mTOR (mTORkd) in the heart. The expression of mTORca or mTORkd affected only mTORC1 but not mTORC2 activities, with corresponding changes in the activities of autophagy, a cellular degradation pathway negatively regulated by mTORC1. Diabetic cardiac damage in OVE26 mice was dramatically reduced by mTORca but exacerbated by mTORkd expression as assessed by changes in cardiac function, oxidative stress, and myocyte apoptosis. These findings demonstrated that the enhanced mTORC1 signaling in the OVE26 diabetic heart was an adaptive response that limited cardiac dysfunction, suggesting that manipulations that enhance mTORC1 activity may reduce diabetic cardiac injury, in sharp contrast to the results previously obtained with rapamycin.-Xu, X., Kobayashi, S., Timm, D., Huang, Y., Zhao, F., Shou, W., Liang, Q. Enhanced mTOR complex 1 signaling attenuates diabetic cardiac injury in OVE26 mice.
Collapse
Affiliation(s)
- Xianmin Xu
- Sanford Research, Sioux Falls, South Dakota, USA
| | - Satoru Kobayashi
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| | - Derek Timm
- Sanford Research, Sioux Falls, South Dakota, USA
| | - Yuan Huang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| | - Fengyi Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Weinian Shou
- Department of Pediatrics, Riley Heart Center, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Qiangrong Liang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, USA
| |
Collapse
|
229
|
Abstract
Heart failure and related morbidity and mortality are increasing at an alarming rate, in large part, because of increases in aging, obesity, and diabetes mellitus. The clinical outcomes associated with heart failure are considerably worse for patients with diabetes mellitus than for those without diabetes mellitus. In people with diabetes mellitus, the presence of myocardial dysfunction in the absence of overt clinical coronary artery disease, valvular disease, and other conventional cardiovascular risk factors, such as hypertension and dyslipidemia, has led to the descriptive terminology, diabetic cardiomyopathy. The prevalence of diabetic cardiomyopathy is increasing in parallel with the increase in diabetes mellitus. Diabetic cardiomyopathy is initially characterized by myocardial fibrosis, dysfunctional remodeling, and associated diastolic dysfunction, later by systolic dysfunction, and eventually by clinical heart failure. Impaired cardiac insulin metabolic signaling, mitochondrial dysfunction, increases in oxidative stress, reduced nitric oxide bioavailability, elevations in advanced glycation end products and collagen-based cardiomyocyte and extracellular matrix stiffness, impaired mitochondrial and cardiomyocyte calcium handling, inflammation, renin-angiotensin-aldosterone system activation, cardiac autonomic neuropathy, endoplasmic reticulum stress, microvascular dysfunction, and a myriad of cardiac metabolic abnormalities have all been implicated in the development and progression of diabetic cardiomyopathy. Molecular mechanisms linked to the underlying pathophysiological changes include abnormalities in AMP-activated protein kinase, peroxisome proliferator-activated receptors, O-linked N-acetylglucosamine, protein kinase C, microRNA, and exosome pathways. The aim of this review is to provide a contemporary view of these instigators of diabetic cardiomyopathy, as well as mechanistically based strategies for the prevention and treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Guanghong Jia
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.)
| | - Michael A Hill
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.)
| | - James R Sowers
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.).
| |
Collapse
|
230
|
Zhang X, Zhong Z, Li W. Downregulation of TRAP1 aggravates injury of H9c2 cardiomyocytes in a hyperglycemic state. Exp Ther Med 2019; 18:2681-2686. [PMID: 31572516 DOI: 10.3892/etm.2019.7847] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/31/2019] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy increases the risk of heart failure and is one of the major causes of death in patients with diabetes. The present study investigated the expression and function of tumor necrosis factor receptor-associated protein 1 (TRAP1) in cardiomyocytes in a hyperglycemic state. For the in vitro study, H9c2 cells (rat cardiomyoblasts) were treated with normal glucose, high glucose. TRAP1 expression was determined by reverse transcription-quantitative PCR and western blot analysis. Viability of cardiomyocytes was detected using the CellTiter 96® AQueous One Solution assay. The intracellular reactive oxygen species (ROS) content was detected using a fluorescent 2',7'-dichlorodihydrofluorescein diacetate probe, and the change in mitochondrial membrane potential was detected by JC-1 fluorescent staining. Changes in cell viability, ROS content and mitochondrial membrane potential were determined following small interfering (si) RNA-mediated knockdown of TRAP1. Results demonstrated that compared with the normal control group, the expression of TRAP1 in H9c2 cells decreased in the high glucose group which was accompanied by a reduction in mitochondrial membrane potential and cell viability, and increased intracellular ROS production. TRAP1 expression was significantly decreased following TRAP1-siRNA transfection which was accompanied by enhanced ROS production, lower mitochondrial membrane potential and impaired cell viability. In conclusion, the present findings suggested that the decrease in cardiomyocyte TRAP1 expression under high glucose conditions was associated with myocardial injury. It was hypothesized that TRAP1 may have a protective role on cardiomyocytes under high glucose surroundings.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Zhen Zhong
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Wangen Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| |
Collapse
|
231
|
Jia T, Wang YN, Zhang J, Hao X, Zhang D, Xu X. Cinaciguat in combination with insulin induces a favorable effect on implant osseointegration in type 2 diabetic rats. Biomed Pharmacother 2019; 118:109216. [PMID: 31319371 DOI: 10.1016/j.biopha.2019.109216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 01/17/2023] Open
Abstract
The osseointegration process of implant is seriously impaired in type 2 diabetes mellitus (T2DM) that causes high failure rate, and insufficiency exists in current insulin therapy, creating a demand for new bone-synergistic agent. Cinaciguat, a novel type of soluble guanylate cyclase (sGC) activator, plays a vital role in glucose metabolism, inflammation control and bone regeneration. We hypothesized that the combined application of cinaciguat and insulin could reverse poor implant osseointegration in diabetes. To test this hypothesis, streptozotocin-induced diabetic rats were placed implants in the femur, and divided into five groups: control, T2DM, cinaciguat-treated T2DM (7 μg/kg), insulin-treated T2DM (12 IU/kg), cinaciguat plus insulin combination-treated T2DM (7 μg/kg and 12 IU/kg respectively), according to different treatment received. The weight and glucose levels of rats were evaluated at fixed times, and plasma level of cyclic guanosine monophosphate (cGMP) was determined before euthanasia. Three months after therapy, the femurs were isolated for pull-out test, environmental scanning electron microscope observation, microscopic computerized tomography evaluation and various histology analysis. Results revealed that diabetic rats showed the highest blood glucose level and lowest cGMP content, which led to the worst structural damage and least osseointegration. Combined treatment could attenuate the diabetes induced hyperglycemia to be normal, restore the cGMP content, protein kinase G II (PKG II) expression, phosphodiesterase-5 (PDE5) activity and ameliorate the mechanical strength, the impaired bone microarchitecture and osseointegration to the highest level. Meanwhile, monotreatment (insulin or cinaciguat) also showed restorative effect, but less. Our findings demonstrated that the cGMP/PKG II signaling pathway activated by cinaciguat mediated the favorable effects of the combined application on improving implant fixation under T2DM condition.
Collapse
Affiliation(s)
- Tingting Jia
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China; Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Ya-Nan Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China; Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Jiajia Zhang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China; Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Xinyu Hao
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China; Department of Pediatric Dentistry, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Dongjiao Zhang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China; Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong Province, China.
| | - Xin Xu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China; Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
232
|
Bubb KJ, Drummond GR, Figtree GA. New opportunities for targeting redox dysregulation in cardiovascular disease. Cardiovasc Res 2019; 116:532-544. [DOI: 10.1093/cvr/cvz183] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/02/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Abstract
Despite substantial promise, the use of antioxidant therapy to improve cardiovascular outcomes has been disappointing. Whilst the fundamental biology supporting their use continues to build, the challenge now is to differentially target dysregulated redox signalling domains and to identify new ways to deliver antioxidant substances. Looking further afield to other disciplines, there is an emerging ‘tool-kit’ containing sophisticated molecular and drug delivery applications. Applying these to the cardiovascular redox field could prove a successful strategy to combat the increasing disease burden. Excessive reactive oxygen species production and protein modifications in the mitochondria has been the target of successful drug development with several positive outcomes emerging in the cardiovascular space, harnessing both improved delivery mechanisms and enhanced understanding of the biological abnormalities. Using this as a blueprint, similar strategies could be applied and expanded upon in other redox-hot-spots, such as the caveolae sub-cellular region, which houses many of the key cardiovascular redox proteins such as NADPH oxidase, endothelial nitric oxide synthase, angiotensin II receptors, and beta adrenoceptors. The expanded tool kit of drug development, including gene and miRNA therapies, nanoparticle technology and micropeptide targeting, can be applied to target dysregulated redox signalling in subcellular compartments of cardiovascular cells. In this review, we consider the opportunities for improving cardiovascular outcomes by utilizing new technology platforms to target subcellular ‘bonfires’ generated by dysregulated redox pathways, to improve clinical outcomes.
Collapse
Affiliation(s)
- Kristen J Bubb
- Cardiothoracic and Vascular Health, Kolling Institute and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Gemma A Figtree
- Cardiothoracic and Vascular Health, Kolling Institute and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
233
|
Filardi T, Ghinassi B, Di Baldassarre A, Tanzilli G, Morano S, Lenzi A, Basili S, Crescioli C. Cardiomyopathy Associated with Diabetes: The Central Role of the Cardiomyocyte. Int J Mol Sci 2019; 20:ijms20133299. [PMID: 31284374 PMCID: PMC6651183 DOI: 10.3390/ijms20133299] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
Abstract
The term diabetic cardiomyopathy (DCM) labels an abnormal cardiac structure and performance due to intrinsic heart muscle malfunction, independently of other vascular co-morbidity. DCM, accounting for 50%–80% of deaths in diabetic patients, represents a worldwide problem for human health and related economics. Optimal glycemic control is not sufficient to prevent DCM, which derives from heart remodeling and geometrical changes, with both consequences of critical events initially occurring at the cardiomyocyte level. Cardiac cells, under hyperglycemia, very early undergo metabolic abnormalities and contribute to T helper (Th)-driven inflammatory perturbation, behaving as immunoactive units capable of releasing critical biomediators, such as cytokines and chemokines. This paper aims to focus onto the role of cardiomyocytes, no longer considered as “passive” targets but as “active” units participating in the inflammatory dialogue between local and systemic counterparts underlying DCM development and maintenance. Some of the main biomolecular/metabolic/inflammatory processes triggered within cardiac cells by high glucose are overviewed; particular attention is addressed to early inflammatory cytokines and chemokines, representing potential therapeutic targets for a prompt early intervention when no signs or symptoms of DCM are manifesting yet. DCM clinical management still represents a challenge and further translational investigations, including studies at female/male cell level, are warranted.
Collapse
Affiliation(s)
- Tiziana Filardi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Susanna Morano
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", Piazza L. de Bosis 6, 00135 Rome, Italy.
| |
Collapse
|
234
|
Wang J, Wang S, Wang W, Chen J, Zhang Z, Zheng Q, Liu Q, Cai L. Protection against diabetic cardiomyopathy is achieved using a combination of sulforaphane and zinc in type 1 diabetic OVE26 mice. J Cell Mol Med 2019; 23:6319-6330. [PMID: 31270951 PMCID: PMC6714218 DOI: 10.1111/jcmm.14520] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/20/2019] [Accepted: 06/15/2019] [Indexed: 12/17/2022] Open
Abstract
Sulforaphane (SFN) can effectively induce nuclear factor E2–related factor 2 (Nrf2), and zinc (Zn) can effectively induce metallothionein (MT), both of which have been shown to protect against diabetic cardiomyopathy (DCM). However, it is unclear whether combined treatment with SFN and Zn offers better cardiac protection than either one alone. Here, we treated 5‐week‐old OVE mice that spontaneously develop type 1 diabetes with SFN and/or Zn for 18 weeks. Cardiac dysfunction, by echocardiography, and pathological alterations and remodelling, shown by cardiac hypertrophy, fibrosis, inflammation and oxidative damage, examined by histopathology, Western blotting and real‐time PCR, were observed in OVE mice. All these dysfunction and pathological abnormalities seen in OVE mice were attenuated in OVE mice with treatment of either SFN, Zn or SFN/Zn, and the combined treatment with SFN/Zn was better than single treatments at ameliorating DCM. In addition, combined SFN and Zn treatment increased Nrf2 function and MT expression in the heart of OVE mice to a greater extent than SFN or Zn alone. This indicates that the dual activation of Nrf2 and MT by combined treatment with SFN and Zn may be more effective than monotherapy at preventing the development of DCM via complementary, additive mechanisms.
Collapse
Affiliation(s)
- Jiqun Wang
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky, USA
| | - Shudong Wang
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, China
| | - Wanning Wang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky, USA.,Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Jing Chen
- Department of Otolaryngology, Stanford University, Palo Alto, California, USA
| | - Zhiguo Zhang
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, China
| | - Qi Zheng
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, Kentucky, USA
| | - Quan Liu
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky, USA.,Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
235
|
Li Q, Li P, Su J, Liu S, Yang X, Yang Y, Niu S. LncRNA NKILA was upregulated in diabetic cardiomyopathy with early prediction values. Exp Ther Med 2019; 18:1221-1225. [PMID: 31316617 PMCID: PMC6601402 DOI: 10.3892/etm.2019.7671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor-κB interacting long non-coding RNA (LncRNA NKILA) is a well-studied tumor suppressor lncRNA in several types of malignancies. The present study reports the involvement of this lncRNA in diabetic cardiomyopathy (DC). A 8-year-follow-up study on 312 diabetic patients without exhibiting obvious complications demonstrated that plasma lncRNA NKILA levels were upregulated specifically in diabetic patients who developed DC but not in patients with other complications. Plasma levels of lncRNA NKILA at 6 months prior to diagnosis is sufficient to distinguish patients with DC from other diabetic patients without significant complications. Although in vitro experiments demonstrated that lncRNA NKILA expression in cardiomyocyte cells was not affected by high-glucose treatment, ectopic lncRNA NKILA expression and lncRNA NKILA knockdown potentiated, and inhibited cardiomyocyte apoptosis, respectively. Therefore, the data from the present study suggests that overexpression of lncRNA NKILA is involved in DC, and overexpression of lncRNA NKILA may serve as a therapeutic target for treating DC.
Collapse
Affiliation(s)
- Qingyong Li
- The Second Department of Cardiology, The People's Hospital of Puyang, Puyang, Henan 457000, P.R. China
| | - Panrong Li
- The Second Department of Cardiology, The People's Hospital of Puyang, Puyang, Henan 457000, P.R. China
| | - Jinling Su
- The Second Department of Cardiology, The People's Hospital of Puyang, Puyang, Henan 457000, P.R. China
| | - Suping Liu
- The Second Department of Cardiology, The People's Hospital of Puyang, Puyang, Henan 457000, P.R. China
| | - Xiaoman Yang
- The Second Department of Cardiology, The People's Hospital of Puyang, Puyang, Henan 457000, P.R. China
| | - Yuejin Yang
- Fuwai Cardiovascular Hospital, Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Suocheng Niu
- The Second Department of Cardiology, The People's Hospital of Puyang, Puyang, Henan 457000, P.R. China
| |
Collapse
|
236
|
Astragaloside IV Exerts a Myocardial Protective Effect against Cardiac Hypertrophy in Rats, Partially via Activating the Nrf2/HO-1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4625912. [PMID: 31285785 PMCID: PMC6594267 DOI: 10.1155/2019/4625912] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/13/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Previous evidence suggested that astragaloside IV (ASIV) had a cardioprotective effect, but the potential mechanisms were undetermined. This study is aimed at validating the prevention of cardiac hypertrophy in chronic heart failure (CHF) rats and hypertrophy in H9c2 cardiomyocytes by ASIV and at exploring the potential mechanism involved. CHF rat models of abdominal aortic constriction (AAC) were used with the aim of determining the protective effect of ASIV in cardiac hypertrophy in the rats. We proved that ASIV could attenuate cardiac hypertrophy by improving left ventricular function and structure and showed that the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) and its downstream gene heme oxygenase-1 (HO-1) increased in the high-dose ASIV intervention group. To further investigate the specific mechanism of ASIV, we hypothesized that ASIV might prevent cardiac hypertrophy via activating the Nrf2/HO-1 signaling pathway. We established a cardiomyocyte hypertrophy model induced by angiotensin II (Ang II), which was then transfected with Nrf2 shRNA, to knock down the expression of the Nrf2 gene. We found that the protective effect of ASIV against Ang II-induced cardiomyocyte hypertrophy was abolished in the Nrf2 shRNA transfection group, ultimately aggravating cardiomyocyte hypertrophy induced by Ang II, and it is possible that oxidative stress may be involved in this process. Our results demonstrated that ASIV improved cardiac function and inhibited cardiac hypertrophy by upregulating Nrf2, and this effect was partially achieved by stimulating the Nrf2/HO-1 signaling pathway, suggesting that ASIV could have therapeutic potential for the treatment of cardiac hypertrophy and CHF.
Collapse
|
237
|
Long non-coding RNA MEG3 serves as a ceRNA for microRNA-145 to induce apoptosis of AC16 cardiomyocytes under high glucose condition. Biosci Rep 2019; 39:BSR20190444. [PMID: 31085717 PMCID: PMC6554216 DOI: 10.1042/bsr20190444] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the most serious complications of diabetes, but its pathogenesis remains largely unclear. In the present study, we aimed to explore the potential role of long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3) and to investigate the underlying mechanisms in human AC16 cardiomyocytes under high glucose (HG) condition. The results demonstrated that MEG3 was overexpressed in HG-treated AC16 cells, and MEG3 knockdown suppressed the HG-induced apoptosis in AC16 cells. Mechanistically, MEG3 directly binds to miR-145 in AC16 cells, thereby up-regulating the expression of PDCD4. Rescue experiments showed that the role of MEG3 in HG-treated AC16 cells was partly dependent on its suppression on miR-145. In summary, our findings suggested that the role of MEG3 in HG-treated human cardiomyocytes is to serve as a competing endogenous RNA (ceRNA), which negatively regulates miR-145. These findings may provide a valuable and promising therapeutic target for the treatment of DCM in the future.
Collapse
|
238
|
Guan Y, Zhou L, Zhang Y, Tian H, Li A, Han X. Effects of PP2A/Nrf2 on experimental diabetes mellitus-related cardiomyopathy by regulation of autophagy and apoptosis through ROS dependent pathway. Cell Signal 2019; 62:109339. [PMID: 31173878 DOI: 10.1016/j.cellsig.2019.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 01/08/2023]
Abstract
Diabetes mellitus-related cardiomyopathy (DMCMP) has been defined as ventricular dysfunction that occurs in diabetic patients independent of a recognized cause such as coronary artery disease or hypertension. Mechanisms underlying DMCMP have not been fully elucidated. In this study, the roles of protein phosphatase 2A/nuclear factor NF-E2-related factor 2 (PP2A/Nrf2) in experimental DMCMP induced by high glucose were studied in vitro and in vivo. The results showed that high glucose could induce experimental DMCMP and increase ROS generation, increase the expression and nuclear translocation of Nrf2, down-regulate the expression of PI3K/Akt/mTOR and up-regulate the expression of ERK, and activate the autophagy of cardiomyocytes. The activity or expression of PP2A in DMCMP increased. PP2A could up-regulate the expression of Nrf2 and promote cardiomyocytes autophagy and apoptosis. Inhibition of PP2A could reduce the expression of Nrf2 and inhibit the autophagy and apoptosis of cardiomyocytes. The results suggested that hyperglycemic-induced experimental DMCMP may be related to up-regulating the expression of Nrf2 through PP2A/Nrf2 pathway. These results will be helpful to elucidate the pathogenesis and mechanism of DMCMP and find targets for the development of new drugs to prevent or treat DMCMP.
Collapse
Affiliation(s)
- Yanhui Guan
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Lichun Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Huiqin Tian
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Department of Pharmacology, Shandong college of Traditional Chinese Medicine, 508 East Binhai road, Yantai 264199, China
| | - Anqi Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Key Laboratory of Chemical Biology of Natural Products, Ministry of Education, Shandong University, Jinan, China.
| |
Collapse
|
239
|
Li L, Luo W, Qian Y, Zhu W, Qian J, Li J, Jin Y, Xu X, Liang G. Luteolin protects against diabetic cardiomyopathy by inhibiting NF-κB-mediated inflammation and activating the Nrf2-mediated antioxidant responses. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152774. [PMID: 31009852 DOI: 10.1016/j.phymed.2018.11.034] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 05/27/2023]
Abstract
BACKGROUND Diabetes mellitus is a well-known risk factor for the development of heart failure. Inflammation and oxidative stress play a key role in the development of diabetic cardiomyopathy (DCM), and this nexus represents an attractive target to combat this disease. Naturally occurring flavonoid luteolin exhibits both anti-inflammatory and antioxidant activities in various systems. HYPOTHESIS/PURPOSE In this study, we aimed to investigate potential cardioprotective effects of luteolin in cultured cardiomyocytes and in mice with type 1 diabetes. METHODS C57BL/6 mice were intraperitoneal injection of streptozotocin (STZ) to induce DCM. High glucose (HG) was used to induce H9C2 cells injury in vitro. Cardiac fibrosis, hypertrophy, inflammation and oxidative stress were studied both in vitro and in vivo. RESULTS Our studies show that luteolin significantly reduces HG-induced inflammatory phenotype and oxidative stress in H9C2 cardiomyocytes. We found that the mechanisms involved inhibition of nuclear factor-kappa B (NF-κB) pathway and the activation of antioxidant nuclear factor-erythroid 2 related factor 2 (Nrf2) signaling pathway. Modulation of these pathways resulted in reduced expression of matrix proteins and cellular hypertrophy. Luteolin also prevented cardiac fibrosis, hypertrophy, and dysfunction in STZ-induced diabetic mice. These readouts were also associated with reduced levels of inflammatory cytokines and oxidative stress biomarkers. CONCLUSION Our results indicate that luteolin protects heart tissues in STZ-induced diabetic mice through modulating Nrf2-mediated oxidative stress and NF-κB-mediated inflammatory responses. These findings suggest that luteolin may be a potential therapeutic agent for DCM.
Collapse
Affiliation(s)
- Li Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Anesthesiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yuanyuan Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weiwei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jieli Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yiyi Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuzhong Xu
- Department of Anesthesiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
240
|
Yin MS, Zhang YC, Xu SH, Liu JJ, Sun XH, Liang C, Wang Y, Li J, Wang FW, Wang QL, Mu YL. Puerarin prevents diabetic cardiomyopathy in vivo and in vitro by inhibition of inflammation. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2019; 21:476-493. [PMID: 29322879 DOI: 10.1080/10286020.2017.1405941] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/13/2017] [Indexed: 06/07/2023]
Abstract
Diabetic cardiomyopathy (DCM) is one of the chief diabetes mellitus complications. Inflammation factors may be one reason for the damage from DM. The purpose of this research is to study the potential protective effects of puerarin on DM and the possible mechanisms of action related to NF-κB signal pathway. Following administration of puerarin to the disease model rat, several changes were observed including the changes of serum biochemical index, improved diastolic dysfunction, and enhanced endogenous antioxidant enzymes activities, further NF-κB signaling activation. Puerarin showed cardio-protective effects on DCM by inhibiting inflammation, and it might be a potential candidate for the treatment of DCM.
Collapse
Affiliation(s)
- Mao-Shan Yin
- a Center for Drug Evaluation , Food and Drug Administration , Beijing 100038 , China
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Yi-Chi Zhang
- c Department of Pharmacy , Jinan Children's Hospital , Jinan 250022 , China
| | - Shu-Hong Xu
- d State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica , Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100050 , China
| | - Jing-Jing Liu
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Xiao-Hui Sun
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Chen Liang
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Yan Wang
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Jie Li
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Fu-Wen Wang
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Qing-Li Wang
- a Center for Drug Evaluation , Food and Drug Administration , Beijing 100038 , China
| | - Yan-Ling Mu
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| |
Collapse
|
241
|
Li X, Ke X, Li Z, Li B. Vaspin prevents myocardial injury in rats model of diabetic cardiomyopathy by enhancing autophagy and inhibiting inflammation. Biochem Biophys Res Commun 2019; 514:1-8. [PMID: 31014675 DOI: 10.1016/j.bbrc.2019.04.110] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/14/2019] [Indexed: 12/21/2022]
Abstract
NLRP3 inflammasome activation plays an important role in diabetic cardiomyopathy (DCM). It is known that autophagy is related to the activation of inflammasomes during oxidative stress. Visceral adipose tissue-derived serine protease inhibitor (Vaspin), is an adipocytokine that has been shown to exert a protective effect on autophagic activity, but whether and how Vaspin improves myocardial damage in DCM remain unclear. In this study, we explored the role of Vaspin in DCM using a streptozotocin (STZ)-induced diabetes model. Cardiac function, cardiomyocyte apoptosis, myocardial tissue morphology, and mitochondrial morphology in diabetic rats were improved after eight weeks of Vaspin treatment. Vaspin treatment augmented autophagy activation in diabetic rat hearts. Moreover, the activation of NLRP3 inflammasome was inhibited by Vaspin, followed by a decrease in the cleavage of caspase-1 and maturation of IL-1β and TNF-ɑ. In vitro studies found that the mitochondrial reactive oxygen species (ROS) generation as well as the depolarization of the mitochondrial membrane in H9C2cells induced by high glucose were attenuated by Vaspin. This inhibitory effect of Vaspin on NLRP3 inflammasome activation was due to the protection of autophagy activity and was abolished after the treatment of autophagy inhibitor (3-MA). These results demonstrate that Vaspin alleviates STZ-induced myocardial injury and renders a cardioprotective effect by suppressing NLRP3 inflammasome activation and promoting autophagy.
Collapse
Affiliation(s)
- Xuelian Li
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, China.
| | - Xuan Ke
- Department of Cardiology, Wuhan Third Hospital, Wuhan, Hubei, 430000, China.
| | - Zhiyang Li
- Grade 2016 Class 2, College of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Bingong Li
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China; Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, China.
| |
Collapse
|
242
|
Relationship of High-Density Lipoprotein-Associated Arylesterase Activity to Systolic Heart Failure in Patients with and without Type 2 Diabetes. Sci Rep 2019; 9:5979. [PMID: 30979955 PMCID: PMC6461629 DOI: 10.1038/s41598-019-42518-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/02/2019] [Indexed: 02/02/2023] Open
Abstract
High-density lipoprotein (HDL) confers protection against cardiovascular disease partly attributable to its robust anti-oxidant activities, which is largely impaired in diabetic conditions. In this study, we analyzed the anti-oxidant activity of HDL, as represented by the arylesterase activity of paraoxonase 1 (PON1) in HDL particles, in 216 consecutive HF patients with (n = 79) or without (n = 137) type 2 diabetes, and age- and gender-matched 112 diabetic and 189 non-diabetic non-HF controls. We found arylesterase activity was significantly decreased in patients with than without HF, and was further decreased when comorbid with diabetes. After adjusting for conventional risk factors and apolipoprotein A-I levels, arylesterase activity remained correlated positively with left ventricular ejection fraction in diabetic (r = 0.325, P = 0.020) but not non-diabetic patients (r = 0.089, P = 0.415), and negatively with NT-proBNP and NYHA functional class in both subgroups. In regression analyses, a higher risk of HF was observed in diabetic than non-diabetic patients when having low arylesterase activities. In conclusion, our data demonstrate that impaired serum arylesterase activity in patients with HF is further reduced when comorbid with diabetes. The relationship of impaired arylesterase activity to HF is especially enhanced in diabetic patients.
Collapse
|
243
|
Farkhondeh T, Samarghandian S, Roshanravan B. Impact of chrysin on the molecular mechanisms underlying diabetic complications. J Cell Physiol 2019; 234:17144-17158. [DOI: 10.1002/jcp.28488] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences Birjand Iran
| | - Saeed Samarghandian
- Noncommunicable Disease Research Center, Neyshabur University of Medical Sciences Neyshabur Iran
- Department of Basic Medical Sciences Neyshabur University of Medical Sciences Neyshabur Iran
| | - Babak Roshanravan
- Student Research Committee, Birjand University of Medical Sciences Birjand Iran
| |
Collapse
|
244
|
Zhao T, Chen H, Xu F, Wang J, Liu Y, Xing X, Guo L, Zhang M, Lu Q. Liraglutide alleviates cardiac fibrosis through inhibiting P4hα-1 expression in STZ-induced diabetic cardiomyopathy. Acta Biochim Biophys Sin (Shanghai) 2019; 51:293-300. [PMID: 30883649 DOI: 10.1093/abbs/gmy177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/25/2018] [Indexed: 01/09/2023] Open
Abstract
Diabetic cardiomyopathy is an important contributor to morbidity and mortality of diabetic patients by causing heart failure. Interstitial and perivascular fibrosis plays a crucial role in diabetic cardiomyopathy. However, there is a lack of effective specific treatments available for diabetic cardiomyopathy. In the present study, we aim to explore the effects of Liraglutide, a GLP-1 analogue, on diabetic cardiomyopathy in STZ-induced diabetic rats fed with high-fat diet. A total of 60 male Wistar rats were randomly assigned to three groups, i.e. normal group, model group, and Liraglutide group, with 20 rats in each group. Serum levels of TC, TG, LDL-C, NEFA, and hydroxyproline were measured using commercial kits. Cardiac function was evaluated by QRS waves, LVEDd, LVESd, and LVEF. Myocardial fibrosis was measured by immunohistochemistry. Our results demonstrated that chronic administration of Liraglutide decreased the level of blood glucose and significantly alleviated lipid metabolic disturbance compared with the model group. Furthermore, Liraglutide was found to improve the damaged cardiac function. In line with this, we also found that the alleviation of cardiac dysfunction was associated with the decreased fibrosis in diabetic myocardial tissues, which was reflected by the decreased expressions of P4hα-1, COL-1, COL-3, MMP-1, and MMP-9. Our results thus suggest that Liraglutide might have a myocardial protective effect by inhibiting P4hα-1-mediated myocardial fibrosis.
Collapse
Affiliation(s)
- Tong Zhao
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Huiqiang Chen
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Fei Xu
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Juan Wang
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Yusheng Liu
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Xiaowei Xing
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Linlin Guo
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research of the Chinese Ministry of Education and Public Health, Shandong University Qilu Hospital, Jinan, China
| | - Qinghua Lu
- Department of Cardiology, the Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
245
|
Nakos I, Kadoglou NPE, Gkeka P, Tzallas AT, Giannakeas N, Tsalikakis DG, Katsimpoulas M, Mantziaras G, Kostomitsopoulos N, Liapis CD, Kakisis J. Exercise Training Attenuates the Development of Cardiac Autonomic Dysfunction in Diabetic Rats. In Vivo 2019; 32:1433-1441. [PMID: 30348698 DOI: 10.21873/invivo.11396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIM Exercise training usually complements pharmacological therapy of type 1 diabetes mellitus, however, little is known about its impact on cardiac autonomic neuropathy. Our aim was to evaluate the impact of exercise on electrocardiographic parameters and heart rate variability in diabetic rats. MATERIALS AND METHODS Wistar rats were randomly assigned to four groups (n=12): Sedentary control (SC), sedentary diabetic (SD), exercise control (EC), and exercise diabetic (ED). Diabetes was induced by a single intraperitoneal injection of streptozotocin (45 mg/kg). Exercise groups underwent 8 weeks of training on a treadmill. At the end of the study, echocardiography was performed and continuous electrocardiographic recording was obtained by intra-abdominally implanted telemetric devices. Diabetes induction significantly reduced the heart rate and increased the blood glucose level (p<0.001) and R-wave amplitude (p<0.05). Frequency-domain spectral variables were also analyzed. The SD group had a significantly lower absolute high-frequency component (p<0.05) and higher normalized low-frequency component, as well as low-frequency power divided by the high-frequency power ratio when compared to the SC and EC groups (p<0.05). All these diabetes-related adverse changes in heart rate variability parameters were significantly reversed by exercise training (p<0.05). Overall, our study shows that early initiation of systemic exercise training prevents the development of cardiac autonomic neuropathy in rats with type 1 diabetes mellitus, by favorable change in the balance between parasympathetic and sympathetic activity.
Collapse
Affiliation(s)
- Ioannis Nakos
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece
| | - Nikolaos P E Kadoglou
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece.,The Heart Hospital, University College London, London, U.K
| | - Paraskevi Gkeka
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece
| | - Alexandros T Tzallas
- Department of Computer Engineering, School of Applied Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Nikolaos Giannakeas
- Department of Computer Engineering, School of Applied Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Dimitrios G Tsalikakis
- Research and Analysis Laboratory, Department of Informatics and Telecommunication Engineering, University of Western Macedonia, Kozani, Greece
| | - Michalis Katsimpoulas
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece
| | - Georgios Mantziaras
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece
| | - Christos D Liapis
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece.,Athens Medical Center, Vascular and Endovascular Surgery Clinic, Athens, Greece
| | - John Kakisis
- Biomedical Research Foundation, Center of Clinical, Experimental Surgery, and Translational Research, Academy of Athens, Athens, Greece.,Athens Medical Center, Vascular and Endovascular Surgery Clinic, Athens, Greece
| |
Collapse
|
246
|
Chen Y, Tan S, Liu M, Li J. LncRNA TINCR is downregulated in diabetic cardiomyopathy and relates to cardiomyocyte apoptosis. SCAND CARDIOVASC J 2019; 52:335-339. [PMID: 30453794 DOI: 10.1080/14017431.2018.1546896] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Yongheng Chen
- Department of Cardiovascular Internal Medicine, the First Affiliated Hospital of Changsha Medical University, Changsha City, P.R. China
| | - Shan Tan
- Department of Cardiovascular Internal Medicine, the First Affiliated Hospital of Changsha Medical University, Changsha City, P.R. China
| | - Mei Liu
- Department of Cardiovascular Internal Medicine, the First Affiliated Hospital of Changsha Medical University, Changsha City, P.R. China
| | - Jianming Li
- Department of Anatomy, Changsha Medical University, Changsha City, P.R. China
| |
Collapse
|
247
|
Rodgers JL, Iyer D, Rodgers LE, Vanthenapalli S, Panguluri SK. Impact of hyperoxia on cardiac pathophysiology. J Cell Physiol 2019; 234:12595-12603. [PMID: 30652312 DOI: 10.1002/jcp.28136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/20/2018] [Indexed: 01/09/2023]
Abstract
Mechanical ventilation with high oxygen therapy (hyperoxia) is widely implemented in critical care and ICU settings. Although supplemental oxygen is beneficial to treat hypoxia, its use is also associated with poor outcomes and high mortality in patients. Lung injury due to hyperoxia exposure has been well-documented in patients, including in adults and neonates. Thus, lung injury due to hyperoxia has been extensively researched in both preclinical and clinical studies. However, hyperoxia has also been shown to be associated with hemodynamic changes in patients in ICU, including reductions in heart rate, stroke volume, and cardiac output. In addition, certain experimental studies report that hyperoxia exposure in neonates results in cardiac dysfunction in later adult life. Despite this, until recently, the impact of hyperoxia within the heart has not been well studied, or reported, specifically in adult experimental models. To close this significant gap, our lab has sought to clarify hyperoxia-induced cardiac pathophysiology in adult murine models. This review discusses the current findings regarding the cardiovascular impact of hyperoxia exposure.
Collapse
Affiliation(s)
- Jennifer L Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Drishya Iyer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Lydia E Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Sahit Vanthenapalli
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Siva K Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| |
Collapse
|
248
|
Wang J, Tang Z, Zhang Y, Qiu C, Zhu L, Zhao N, Liu Z. Matrine alleviates AGEs- induced cardiac dysfunctions by attenuating calcium overload via reducing ryanodine receptor 2 activity. Eur J Pharmacol 2019; 842:118-124. [DOI: 10.1016/j.ejphar.2018.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/01/2018] [Accepted: 10/10/2018] [Indexed: 12/29/2022]
|
249
|
Chang GJ, Yeh YH, Chen WJ, Ko YS, Pang JHS, Lee HY. Inhibition of Advanced Glycation End Products Formation Attenuates Cardiac Electrical and Mechanical Remodeling and Vulnerability to Tachyarrhythmias in Diabetic Rats. J Pharmacol Exp Ther 2019; 368:66-78. [PMID: 30381326 DOI: 10.1124/jpet.118.252080] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/23/2018] [Indexed: 03/08/2025] Open
Abstract
Diabetic patients with cardiomyopathy show a higher incidence of arrhythmias and sudden death. Chronic hyperglycemia induces the formation of advanced glycation end products (AGEs), which contribute to the pathogenesis of diabetic cardiomyopathy. This study investigated whether inhibition of AGEs formation by aminoguanidine (AG) could prevent cardiac electromechanical and arrhythmogenic remodeling in diabetes mellitus. Streptozotocin-induced diabetic rats received AG (100 mg/kg daily, i.p.) or vehicle (normal saline, i.p.) for 5 weeks. The rats underwent hemodynamic recording to evaluate cardiac function, and heart preparations were used to determine the electrical, mechanical, and biochemical functions. In vitro high glucose-induced AGEs formation, reactive oxygen species (ROS) generation, and action potential changes were examined in HL-1 atrial cells. AG treatment improved the diabetes-induced depression in left ventricular pressure and the relaxation rate, and normalized the prolongation of QTc intervals in anesthetized rats. AG reduced the vulnerabilities to atrial and ventricular tachyarrhythmias in perfused diabetic hearts. AG normalized the prolonged action potential duration in diabetic atrial and ventricular muscles, which was correlated with the restoration of both transient outward (I to) and steady-state outward (I SS) K+ current densities in cardiomyocytes. The abnormal kinetics of Ca2+ transients and contraction were reversed in cardiomyocytes from AG-treated diabetic rats, along with parallel preservation of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA2a) expression. Furthermore, ex vivo and in vitro studies showed AG attenuated AGEs and ROS formation. Thus, long-term administration of AG ameliorated cardiac electromechanical remodeling and arrhythmogenicity in diabetic rats and may present an effective strategy for the prevention of diabetes-associated arrhythmias.
Collapse
Affiliation(s)
- Gwo-Jyh Chang
- Graduate Institute of Clinical Medicinal Sciences, College of Medicine, Chang Gung University (G.-J.C., J.-H.S.P., H.-Y.L.), and Cardiovascular Medicine, Chang Gung Memorial Hospital (G.-J.C., Y.-H.Y., W.-J.C., Y.-S.K.), Tao-Yuan, Taiwan
| | - Yung-Hsin Yeh
- Graduate Institute of Clinical Medicinal Sciences, College of Medicine, Chang Gung University (G.-J.C., J.-H.S.P., H.-Y.L.), and Cardiovascular Medicine, Chang Gung Memorial Hospital (G.-J.C., Y.-H.Y., W.-J.C., Y.-S.K.), Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Graduate Institute of Clinical Medicinal Sciences, College of Medicine, Chang Gung University (G.-J.C., J.-H.S.P., H.-Y.L.), and Cardiovascular Medicine, Chang Gung Memorial Hospital (G.-J.C., Y.-H.Y., W.-J.C., Y.-S.K.), Tao-Yuan, Taiwan
| | - Yu-Shien Ko
- Graduate Institute of Clinical Medicinal Sciences, College of Medicine, Chang Gung University (G.-J.C., J.-H.S.P., H.-Y.L.), and Cardiovascular Medicine, Chang Gung Memorial Hospital (G.-J.C., Y.-H.Y., W.-J.C., Y.-S.K.), Tao-Yuan, Taiwan
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medicinal Sciences, College of Medicine, Chang Gung University (G.-J.C., J.-H.S.P., H.-Y.L.), and Cardiovascular Medicine, Chang Gung Memorial Hospital (G.-J.C., Y.-H.Y., W.-J.C., Y.-S.K.), Tao-Yuan, Taiwan
| | - Hsiao-Yu Lee
- Graduate Institute of Clinical Medicinal Sciences, College of Medicine, Chang Gung University (G.-J.C., J.-H.S.P., H.-Y.L.), and Cardiovascular Medicine, Chang Gung Memorial Hospital (G.-J.C., Y.-H.Y., W.-J.C., Y.-S.K.), Tao-Yuan, Taiwan
| |
Collapse
|
250
|
Li J, Zhao Y, Zhou N, Li L, Li K. Dexmedetomidine Attenuates Myocardial Ischemia-Reperfusion Injury in Diabetes Mellitus by Inhibiting Endoplasmic Reticulum Stress. J Diabetes Res 2019; 2019:7869318. [PMID: 31886285 PMCID: PMC6914963 DOI: 10.1155/2019/7869318] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/24/2019] [Accepted: 10/25/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE With the increasing incidence of diabetes mellitus (DM) combined with myocardial ischemia, how to reduce myocardial ischemia-reperfusion injury in DM patients has become a major problem faced by clinicians. We investigated the therapeutic effects of dexmedetomidine (DEX) on myocardial ischemia-reperfusion injury in DM rats and its effect on endoplasmic reticulum stress. METHODS SD rats with SPF grade were randomly divided into 6 groups: non-DM rats were divided into the sham operation group (NDM-S group), ischemia-reperfusion group (NDM-IR group), and dexmedetomidine group (NDM-DEX group); DM rats were divided into the diabetic sham operation group (DM-S group), diabetes-reperfusion group (DM-IR group), and diabetes-dexmedetomidine (DM-DEX) group, with 10 rats in each group. Then the effects of DEX on the changes of CK-MB and cTnT levels were examined. The effects of myocardial pathological damage and myocardial infarct size were detected. The apoptosis of cardiomyocytes was detected. The apoptosis of heart tissue cells was also tested through the expressions of cleaved caspase-3, Bcl-2, and Bax proteins. The expression of endoplasmic reticulum stress-related proteins GRP78, CHOP, ERO1α, ERO1β, and PDI was examined. The hypoxia/reoxygenation (H/R) injury cell model was established, the effects of DEX, DEX+ ERS agonist on cell apoptosis was also detected. RESULTS The myocardial damage of DM-IR was more severe than that of NDM-IR rats. DEX could reduce the expression of CK-MB and cTnT, reduce pathological damage, and reduce scar formation and improve fibrosis. DEX can reduce the expression of GRP78, CHOP, ERO1α, ERO1β, and PDI proteins in vivo and in vitro. And the effect of DEX on cell apoptosis could be blocked by ERS agonist. CONCLUSION DEX attenuates myocardial ischemia-reperfusion injury in DM rats and H/R injury cell, which is associated with the reduction of ERS-induced cardiomyocyte apoptosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Line
- Creatine Kinase, MB Form/blood
- Dexmedetomidine/pharmacology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Endoplasmic Reticulum Stress/drug effects
- Fibrosis
- Heat-Shock Proteins/metabolism
- Male
- Membrane Glycoproteins/metabolism
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/complications
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Protein Disulfide-Isomerases/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- Transcription Factor CHOP/metabolism
- Troponin T/blood
Collapse
Affiliation(s)
- Jinjie Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin 130033, China
| | - Ying Zhao
- Department of Nephrology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun Jilin 130021, China
| | - Nan Zhou
- Department of Anesthesia, The General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Longyun Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin 130033, China
| | - Kai Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin 130033, China
| |
Collapse
|