1
|
Kang S, Noh Y, Oh SJ, Yoon HJ, Im S, Kwon HT, Pak YK. Neuroprotective Effects of Aldehyde-Reducing Composition in an LPS-Induced Neuroinflammation Model of Parkinson's Disease. Molecules 2023; 28:7988. [PMID: 38138478 PMCID: PMC10745824 DOI: 10.3390/molecules28247988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease in which neuroinflammation and oxidative stress interact to contribute to pathogenesis. This study investigates the in vivo neuroprotective effects of a patented yeast extract lysate in a lipopolysaccharide (LPS)-induced neuroinflammation model. The yeast extract lysate, named aldehyde-reducing composition (ARC), exhibited potent antioxidant and anti-aldehyde activities in vitro. Oral administration of ARC at 10 or 20 units/kg/day for 3 days prior to intraperitoneal injection of LPS (10 mg/kg) effectively preserved dopaminergic neurons in the substantia nigra (SN) and striatum by preventing LPS-induced cell death. ARC also normalized the activation of microglia and astrocytes in the SN, providing further evidence for its neuroprotective properties. In the liver, ARC downregulated the LPS-induced increase in inflammatory cytokines and reversed the LPS-induced decrease in antioxidant-related genes. These findings indicate that ARC exerts potent antioxidant, anti-aldehyde, and anti-inflammatory effects in vivo, suggesting its potential as a disease-modifying agent for the prevention and treatment of neuroinflammation-related diseases, including Parkinson's disease.
Collapse
Affiliation(s)
- Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Youngjin Noh
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Seung Jun Oh
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
| | - Hye Ji Yoon
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Suyeol Im
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
| | - Hung Taeck Kwon
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
2
|
Zhong L, Han J, Fan X, Huang Z, Su L, Cai X, Lin S, Chen X, Huang W, Dai S, Ye B. Novel GSDMD inhibitor GI-Y1 protects heart against pyroptosis and ischemia/reperfusion injury by blocking pyroptotic pore formation. Basic Res Cardiol 2023; 118:40. [PMID: 37782407 DOI: 10.1007/s00395-023-01010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
Activation of gasdermin D (GSDMD) and its concomitant cardiomyocyte pyroptosis are critically involved in multiple cardiac pathological conditions. Pharmacological inhibition or gene knockout of GSDMD could protect cardiomyocyte from pyroptosis and dysfunction. Thus, seeking and developing highly potent GSDMD inhibitors probably provide an attractive strategy for treating diseases targeting GSDMD. Through structure-based virtual screening, pharmacological screening and subsequent pharmacological validations, we preliminarily identified GSDMD inhibitor Y1 (GI-Y1) as a selective GSDMD inhibitor with cardioprotective effects. Mechanistically, GI-Y1 binds to GSDMD and inhibits lipid- binding and pyroptotic pore formation of GSDMD-N by targeting the Arg7 residue. Importantly, we confirmed the cardioprotective effect of GI-Y1 on myocardial I/R injury and cardiac remodeling by targeting GSDMD. More extensively, GI-Y1 also inhibited the mitochondrial binding of GSDMD-N and its concomitant mitochondrial dysfunction. The findings of this study identified a new drug (GI-Y1) for the treatment of cardiac disorders by targeting GSDMD, and provide a new tool compound for pyroptosis research.
Collapse
Affiliation(s)
- Lingfeng Zhong
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lan Su
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuang Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Ding WJ, Chen GH, Deng SH, Zeng KF, Lin KL, Deng B, Zhang SW, Tan ZB, Xu YC, Chen S, Chen JB, Chen TF, Tan YZ, Zhou YC, Zhang JZ, Liu B. Calycosin protects against oxidative stress-induced cardiomyocyte apoptosis by activating aldehyde dehydrogenase 2. Phytother Res 2023; 37:35-49. [PMID: 36059198 DOI: 10.1002/ptr.7591] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/08/2022] [Accepted: 08/02/2022] [Indexed: 01/19/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide, and oxidative stress is part of the process that causes MI. Calycosin, a naturally occurring substance with cardioprotective properties, is one of the major active constituents in Radix Astragali. In this study, effect of Calycosin was investigated in vivo and in vitro to determine whether it could alleviate oxidative stress and oxidative stress-induced cardiac apoptosis in neonatal cardiomyocytes (NCMs) via activation of aldehyde dehydrogenase 2 (ALDH2). Calycosin protected against oxidative stress and oxidative stress-induced apoptosis in NCMs. Molecular docking revealed that the ALDH2-Calycosin complex had a binding energy of -9.885 kcal/mol. In addition, molecular docking simulations demonstrated that the ALDH2-Calycosin complex was stable. Using BLI assays, we confirmed that Calycosin could interact with ALDH2 (KD = 1.9 × 10-4 M). Furthermore, an ALDH2 kinase activity test revealed that Calycosin increased ALDH2 activity, exhibiting an EC50 of 91.79 μM. Pre-incubation with ALDH2 inhibitor (CVT-10216 or disulfiram) reduced the cardio-protective properties Calycosin. In mice with MI, Calycosin therapy substantially reduced myocardial apoptosis, oxidative stress, and activated ALDH2. Collectively, our findings clearly suggest that Calycosin reduces oxidative stress and oxidative stress-induced apoptosis via the regulation of ALDH2 signaling, which supports potential therapeutic use in MI.
Collapse
Affiliation(s)
- Wen-Jun Ding
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guang-Hong Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, China
| | - Sui-Hui Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ke-Feng Zeng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai-Li Lin
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Bo Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang-Wei Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhang-Bin Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - You-Cai Xu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Si Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun-Bang Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting-Fang Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong-Zhen Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, China
| | - Jing-Zhi Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Pan G, Roy B, Giri S, Lanfear DE, Thandavarayan RA, Guha A, Ortiz PA, Palaniyandi SS. Aldehyde Dehydrogenase 2 Activator Augments the Beneficial Effects of Empagliflozin in Mice with Diabetes-Associated HFpEF. Int J Mol Sci 2022; 23:10439. [PMID: 36142350 PMCID: PMC9499333 DOI: 10.3390/ijms231810439] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 01/24/2023] Open
Abstract
To ameliorate diabetes mellitus-associated heart failure with preserved ejection fraction (HFpEF), we plan to lower diabetes-mediated oxidative stress-induced 4-hydroxy-2-nonenal (4HNE) accumulation by pharmacological agents that either decrease 4HNE generation or increase its detoxification.A cellular reactive carbonyl species (RCS), 4HNE, was significantly increased in diabetic hearts due to a diabetes-induced decrease in 4HNE detoxification by aldehyde dehydrogenase (ALDH) 2, a cardiac mitochondrial enzyme that metabolizes 4HNE. Therefore, hyperglycemia-induced 4HNE is critical for diabetes-mediated cardiotoxicity and we hypothesize that lowering 4HNE ameliorates diabetes-associated HFpEF. We fed a high-fat diet to ALDH2*2 mice, which have intrinsically low ALDH2 activity, to induce type-2 diabetes. After 4 months of diabetes, the mice exhibited features of HFpEF along with increased 4HNE adducts, and we treated them with vehicle, empagliflozin (EMP) (3 mg/kg/d) to reduce 4HNE and Alda-1 (10 mg/kg/d), and ALDH2 activator to enhance ALDH2 activity as well as a combination of EMP + Alda-1 (E + A), via subcutaneous osmotic pumps. After 2 months of treatments, cardiac function was assessed by conscious echocardiography before and after exercise stress. EMP + Alda-1 improved exercise tolerance, diastolic and systolic function, 4HNE detoxification and cardiac liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) pathways in ALDH2*2 mice with diabetes-associated HFpEF. This combination was even more effective than EMP alone. Our data indicate that ALDH2 activation along with the treatment of hypoglycemic agents may be a salient strategy to alleviate diabetes-associated HFpEF.
Collapse
Affiliation(s)
- Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University, Detroit, MI 48202, USA
| | - Bipradas Roy
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University, Detroit, MI 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - David E. Lanfear
- Heart and Vascular Institute, Henry Ford Hospital, Detroit, MI 48202, USA
- Center for Health Policy and Health Services Research, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | - Ashrith Guha
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Pablo A. Ortiz
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University, Detroit, MI 48202, USA
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
5
|
Pan G, Roy B, Palaniyandi SS. Diabetic Aldehyde Dehydrogenase 2 Mutant (ALDH2*2) Mice Are More Susceptible to Cardiac Ischemic-Reperfusion Injury Due to 4-Hydroxy-2-Nonenal Induced Coronary Endothelial Cell Damage. J Am Heart Assoc 2021; 10:e021140. [PMID: 34482710 PMCID: PMC8649540 DOI: 10.1161/jaha.121.021140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background Aldehyde dehydrogenase‐2 (ALDH2), a mitochondrial enzyme, detoxifies reactive aldehydes such as 4‐hydroxy‐2‐nonenal (4HNE). A highly prevalent E487K mutation in ALDH2 (ALDH2*2) in East Asian people with intrinsic low ALDH2 activity is implicated in diabetic complications. 4HNE‐induced cardiomyocyte dysfunction was studied in diabetic cardiac damage; however, coronary endothelial cell (CEC) injury in myocardial ischemia‐reperfusion injury (IRI) in diabetic mice has not been studied. Therefore, we hypothesize that the lack of ALDH2 activity exacerbates 4HNE‐induced CEC dysfunction which leads to cardiac damage in ALDH2*2 mutant diabetic mice subjected to myocardial IRI. Methods and Results Three weeks after diabetes mellitus (DM) induction, hearts were subjected to IRI either in vivo via left anterior descending artery occlusion and release or ex vivo IRI by using the Langendorff system. The cardiac performance was assessed by conscious echocardiography in mice or by inserting a balloon catheter in the left ventricle in the ex vivo model. Just 3 weeks of DM led to an increase in cardiac 4HNE protein adducts and, cardiac dysfunction, and a decrease in the number of CECs along with reduced myocardial ALDH2 activity in ALDH2*2 mutant diabetic mice compared with their wild‐type counterparts. Systemic pretreatment with Alda‐1 (10 mg/kg per day), an activator of both ALDH2 and ALDH2*2, led to a reduction in myocardial infarct size and dysfunction, and coronary perfusion pressure upon cardiac IRI by increasing CEC population and coronary arteriole opening. Conclusions Low ALDH2 activity exacerbates 4HNE‐mediated CEC injury and thereby cardiac dysfunction in diabetic mouse hearts subjected to IRI, which can be reversed by ALDH2 activation.
Collapse
Affiliation(s)
- Guodong Pan
- Division of Hypertension and Vascular ResearchDepartment of Internal MedicineHenry Ford Health SystemDetroitMI
| | - Bipradas Roy
- Division of Hypertension and Vascular ResearchDepartment of Internal MedicineHenry Ford Health SystemDetroitMI
- Department of PhysiologyWayne State UniversityDetroitMI
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular ResearchDepartment of Internal MedicineHenry Ford Health SystemDetroitMI
- Department of PhysiologyWayne State UniversityDetroitMI
| |
Collapse
|
6
|
Cao C, Kwok D, Edie S, Li Q, Ding B, Kossinna P, Campbell S, Wu J, Greenberg M, Long Q. kTWAS: integrating kernel machine with transcriptome-wide association studies improves statistical power and reveals novel genes. Brief Bioinform 2021; 22:5985285. [PMID: 33200776 DOI: 10.1093/bib/bbaa270] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
The power of genotype-phenotype association mapping studies increases greatly when contributions from multiple variants in a focal region are meaningfully aggregated. Currently, there are two popular categories of variant aggregation methods. Transcriptome-wide association studies (TWAS) represent a set of emerging methods that select variants based on their effect on gene expressions, providing pretrained linear combinations of variants for downstream association mapping. In contrast to this, kernel methods such as sequence kernel association test (SKAT) model genotypic and phenotypic variance use various kernel functions that capture genetic similarity between subjects, allowing nonlinear effects to be included. From the perspective of machine learning, these two methods cover two complementary aspects of feature engineering: feature selection/pruning and feature aggregation. Thus far, no thorough comparison has been made between these categories, and no methods exist which incorporate the advantages of TWAS- and kernel-based methods. In this work, we developed a novel method called kernel-based TWAS (kTWAS) that applies TWAS-like feature selection to a SKAT-like kernel association test, combining the strengths of both approaches. Through extensive simulations, we demonstrate that kTWAS has higher power than TWAS and multiple SKAT-based protocols, and we identify novel disease-associated genes in Wellcome Trust Case Control Consortium genotyping array data and MSSNG (Autism) sequence data. The source code for kTWAS and our simulations are available in our GitHub repository (https://github.com/theLongLab/kTWAS).
Collapse
Affiliation(s)
- Chen Cao
- Department of Biochemistry & Molecular Biology, University of Calgary
| | - Devin Kwok
- Department of Mathematics & Statistics, University of Calgary
| | | | - Qing Li
- Department of Biochemistry & Molecular Biology, University of Calgary
| | - Bowei Ding
- Department of Mathematics & Statistics, University of Calgary
| | - Pathum Kossinna
- Department of Biochemistry & Molecular Biology, University of Calgary
| | | | - Jingjing Wu
- Department of Mathematics & Statistics, University of Calgary
| | | | - Quan Long
- Departments of Biochemistry & Molecular Biology, Medical Genetics and Mathematics & Statistics
| |
Collapse
|
7
|
A Novel ALDH2 Activator AD-9308 Improves Diastolic and Systolic Myocardial Functions in Streptozotocin-Induced Diabetic Mice. Antioxidants (Basel) 2021; 10:antiox10030450. [PMID: 33805825 PMCID: PMC7998151 DOI: 10.3390/antiox10030450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus has reached epidemic proportion worldwide. One of the diabetic complications is cardiomyopathy, characterized by early left ventricular (LV) diastolic dysfunction, followed by development of systolic dysfunction and ventricular dilation at a late stage. The pathogenesis is multifactorial, and there is no effective treatment yet. In recent years, 4-hydroxy-2-nonenal (4-HNE), a toxic aldehyde generated from lipid peroxidation, is implicated in the pathogenesis of cardiovascular diseases. Its high bioreactivity toward proteins results in cellular damage. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is the major enzyme that detoxifies 4-HNE. The development of small-molecule ALDH2 activator provides an opportunity for treating diabetic cardiomyopathy. This study found that AD-9308, a water-soluble andhighly selective ALDH2 activator, can improve LV diastolic and systolic functions, and wall remodeling in streptozotocin-induced diabetic mice. AD-9308 treatment dose-dependently lowered serum 4-HNE levels and 4-HNE protein adducts in cardiac tissue from diabetic mice, accompanied with ameliorated myocardial fibrosis, inflammation, and apoptosis. Improvements of mitochondrial functions, sarco/endoplasmic reticulumcalcium handling and autophagy regulation were also observed in diabetic mice with AD-9308 treatment. In conclusion, ADLH2 activation effectively ameliorated diabetic cardiomyopathy, which may be mediated through detoxification of 4-HNE. Our findings highlighted the therapeutic potential of ALDH2 activation for treating diabetic cardiomyopathy.
Collapse
|
8
|
Roy B, Palaniyandi SS. A role for aldehyde dehydrogenase (ALDH) 2 in angiotensin II-mediated decrease in angiogenesis of coronary endothelial cells. Microvasc Res 2021; 135:104133. [PMID: 33428883 DOI: 10.1016/j.mvr.2021.104133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 11/17/2022]
Abstract
Diabetes-induced coronary endothelial cell (CEC) dysfunction contributes to diabetic heart diseases. Angiotensin II (Ang II), a vasoactive hormone, is upregulated in diabetes, and is reported to increase oxidative stress in CECs. 4-hydroxy-2-nonenal (4HNE), a key lipid peroxidation product, causes cellular dysfunction by forming adducts with proteins. By detoxifying 4HNE, aldehyde dehydrogenase (ALDH) 2 reduces 4HNE mediated proteotoxicity and confers cytoprotection. Thus, we hypothesize that ALDH2 improves Ang II-mediated defective CEC angiogenesis by decreasing 4HNE-mediated cytotoxicity. To test our hypothesis, we treated the cultured mouse CECs (MCECs) with Ang II (0.1, 1 and 10 μM) for 2, 4 and 6 h. Next, we treated MCECs with Alda-1 (10 μM), an ALDH2 activator or disulfiram (2.5 μM)/ALDH2 siRNA (1.25 nM), the ALDH2 inhibitors, or blockers of angiotensin II type-1 and 2 receptors i.e. Losartan and PD0123319 respectively before challenging MCECs with 10 μM Ang II. We found that 10 μM Ang II decreased tube formation in MCECs with in vitro angiogenesis assay (P < .0005 vs control). 10 μM Ang II downregulated the levels of vascular endothelial growth factor receptor 1 (VEGFR1) (p < .005 for mRNA and P < .05 for protein) and VEGFR2 (p < .05 for mRNA and P < .005 for protein) as well as upregulated the levels of angiotensin II type-2 receptor (AT2R) (p < .05 for mRNA and P < .005 for protein) and 4HNE-adducts (P < .05 for protein) in cultured MCECs, compared to controls. ALDH2 inhibition with disulfiram/ALDH2 siRNA exacerbated 10 μM Ang II-induced decrease in coronary angiogenesis (P < .005) by decreasing the levels of VEGFR1 (P < .005 for mRNA and P < .05 for protein) and VEGFR2 (P < .05 for both mRNA and protein) and increasing the levels of AT2R (P < .05 for both mRNA and protein) and 4HNE-adducts (P < .05 for protein) relative to Ang II alone. AT2R inhibition per se improved angiogenesis in MCECs. Additionally, enhancing ALDH2 activity with Alda 1 rescued Ang II-induced decrease in angiogenesis by increasing the levels of VEGFR1, VEGFR2 and decreasing the levels of AT2R. In summary, ALDH2 can be an important target in reducing 4HNE-induced proteotoxicity and improving angiogenesis in MCECs. Finally, we conclude ALDH2 activation can be a therapeutic strategy to improve coronary angiogenesis to ameliorate cardiometabolic diseases.
Collapse
Affiliation(s)
- Bipradas Roy
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States of America; Department of Physiology, Wayne State University, Detroit, MI 48202, United States of America
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States of America; Department of Physiology, Wayne State University, Detroit, MI 48202, United States of America.
| |
Collapse
|
9
|
Pan G, Deshpande M, Pang H, Stemmer PM, Carruthers NJ, Shearn CT, Backos DS, Palaniyandi SS. 4-Hydroxy-2-nonenal attenuates 8-oxoguanine DNA glycosylase 1 activity. J Cell Biochem 2020; 121:4887-4897. [PMID: 32628320 PMCID: PMC7935017 DOI: 10.1002/jcb.29814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/18/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Elevated cellular oxidative stress and oxidative DNA damage are key contributors to impaired cardiac function in diabetes. During chronic inflammation, reactive oxygen species (ROS)-induced lipid peroxidation results in the formation of reactive aldehydes, foremost of which is 4-hydroxy-2-nonenal (4HNE). 4HNE forms covalent adducts with proteins, negatively impacting cellular protein function. During conditions of elevated oxidative stress, oxidative DNA damage such as modification by 8-hydroxydeoxyguanosine (8OHdG) is repaired by 8-oxoguanine glycosylase-1 (OGG-1). Based on these facts, we hypothesized that 4HNE forms adducts with OGG-1 inhibiting its activity, and thus, increases the levels of 8OHG in diabetic heart tissues. To test our hypothesis, we evaluated OGG-1 activity, 8OHG and 4HNE in the hearts of leptin receptor deficient db/db mice, a type-2 diabetic model. We also treated the recombinant OGG-1 with 4HNE to measure direct adduction. We found decreased OGG-1 activity (P > .05), increased 8OHG (P > .05) and increased 4HNE adducts (P > .05) along with low aldehyde dehydrogenase-2 activity (P > .05). The increased colocalization of OGG-1 and 4HNE in cardiomyocytes suggest 4HNE adduction on OGG-1. Furthermore, colocalization of 8OHG and OGG-1 with mitochondrial markers TOM 20 and aconitase, respectively, indicated significant levels of oxidatively-induced mtDNA damage and implicated a role for mitochondrial OGG-1 function. In vitro exposure of recombinant OGG-1 (rOGG-1) with increasing concentrations of 4HNE resulted in a concentration-dependent decrease in OGG-1 activity. Mass spectral analysis of trypsin digests of 4HNE-treated rOGG-1 identified 4HNE adducts on C28, C75, C163, H179, H237, C241, K249, H270, and H282. In silico molecular modeling of 4HNE-K249 OGG-1 and 4HNE-H270 OGG-1 mechanistically supported 4HNE-mediated enzymatic inhibition of OGG-1. In conclusion, these data support the hypothesis that inhibition of OGG-1 by direct modification by 4HNE contributes to decreased OGG-1 activity and increased 8OHG-modified DNA that are present in the diabetic heart.
Collapse
Affiliation(s)
- Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
| | - Mandar Deshpande
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
| | - Haiyan Pang
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences & Proteomics Facility Core, Wayne State University, Detroit, MI, USA, 48201
| | - Nicholas J Carruthers
- Institute of Environmental Health Sciences & Proteomics Facility Core, Wayne State University, Detroit, MI, USA, 48201
| | - Colin T. Shearn
- Department of Pediatrics Division of Pediatric Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80045
| | - Donald S. Backos
- School of Pharmacy, University of Colorado Anschutz Medical Center, Aurora, CO 80045
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
- Department of Physiology, Wayne State University, Detroit, MI, 48202
| |
Collapse
|
10
|
Goodnough CL, Gross ER. Precision Medicine Considerations for the Management of Heart Disease and Stroke in East Asians. CARDIOLOGY PLUS 2020; 5:101-108. [PMID: 33954271 PMCID: PMC8095722 DOI: 10.4103/cp.cp_17_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Heart disease is the leading cause of death in Asian Americans. Importantly, people of East Asian descent are more likely to carry a loss-of-function point mutation in aldehyde dehydrogenase 2 (ALDH2), ALDH2*2, which reduces ALDH2 enzymatic activity by at least 40% relative to wild type ALDH2. Given the role of ALDH2 in removing toxic aldehydes from the cell, ALDH2 is intimately involved in the cardioprotective mechanisms of ischemic preconditioning and the pathophysiology of ischemia reperfusion injury. The ALDH2*2 variant is associated with an increased incidence of coronary artery disease, myocardial infarction, and stroke. Furthermore, this variant is associated with insensitivity to nitroglycerin, which is commonly prescribed in patients with cardiovascular disease. In this review, we discuss the genetic susceptibility and pathophysiology associated with the ALDH2*2 variant in regards to cardiovascular disease. We also present the considerations for the management of heart disease and stroke specific to East Asians carrying the ALDH2*2 genetic variant.
Collapse
Affiliation(s)
- Candida L Goodnough
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
11
|
4-hydroxy-2-nonenal decreases coronary endothelial cell migration: Potentiation by aldehyde dehydrogenase 2 inhibition. Vascul Pharmacol 2020; 131:106762. [PMID: 32585188 DOI: 10.1016/j.vph.2020.106762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/07/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
4-hydroxynonenal (4HNE) is a reactive aldehyde, which is involved in oxidative stress associated pathogenesis. The cellular toxicity of 4HNE is mitigated by aldehyde dehydrogenase (ALDH) 2. Thus, we hypothesize that ALDH2 inhibition exacerbates 4HNE-induced decrease in coronary endothelial cell (EC) migration in vitro. To test our hypothesis, we pharmacologically inhibited ALDH2 in cultured mouse coronary ECs (MCECs) by disulfiram (DSF) (2.5 μM) before challenging the cells with different doses of 4HNE (25, 50 and 75 μM) for 4, 12, 16 and 24 h. We evaluated MCEC migration by scratch wound migration assay. 4HNE attenuated MCEC migration significantly relative to control (P < .05), which was exacerbated with DSF pretreatment (P < .05). DSF pretreatment exacerbated 4HNE-induced decrease in ALDH2 activity in MCECs. Next, we showed that 75 μM 4HNE significantly decreased the intracellular mRNA levels of vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), focal adhesion kinase (FAK) and other promigratory genes compared to control, which were further decreased by DSF pretreatment. 75 μM 4HNE also decreased the protein levels of VEGFR2, FAK, phospho-FAK, Src and paxillin in MCECs. Thus, we conclude that ALDH2 inhibition potentiates 4HNE-induced decrease in MCECs migration in vitro.
Collapse
|
12
|
唐 碧, 康 品, 郭 建, 朱 磊, 徐 庆, 高 琴, 张 恒, 王 洪. [Effects of mitochondrial aldehyde dehydrogenase 2 on autophagy-associated proteins in neonatal rat myocardial fibroblasts cultured in high glucose]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:523-527. [PMID: 31140414 PMCID: PMC6743934 DOI: 10.12122/j.issn.1673-4254.2019.05.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate whether autophagy mediates the effects of aldehyde dehydrogenase 2 (ALDH2) on the proliferation of neonatal rat cardiac fibroblasts cultured in high glucose. METHODS Cardiac fibroblasts were isolated from neonatal (within 3 days) SD rats and subcultured. The fibroblasts of the third passage, after identification with immunofluorescence staining for vimentin, were treated with 5.5 mmol/L glucose (control group), 30 mmol/L glucose (high glucose group), or 30 mmol/L glucose in the presence of Alda-1 (an ALDH2 agonist), daidzin (an ALDH2 2 inhibitor), or both. Western blotting was employed to detect ALDH2, microtubule-associated protein 1 light chain 3B subunit (LC3B) and Beclin-1 in the cells, and a hydroxyproline detection kit was used for determining hydroxyproline content in cell culture medium; CCK- 8 kit was used for assessing the proliferation ability of the cardiac fibroblasts after the treatments. RESULTS Compared with the control cells, the cells exposed to high glucose exhibited obviously decreased expressions of ALDH2, Beclin-1 and LC3B and increased cell number and hydroxyproline content in the culture medium. Treatment of the high glucose-exposed cells with Alda-1 significantly increased Beclin-1, LC3B, and ALDH2 protein expressions and lowered the cell number and intracellular hydroxyproline content, whereas the application of daidzin resulted in reverse changes in the expressions of ALDH2, Beclin-1 and LC3B, viable cell number and intracellular hydroxyproline content in high glucose-exposed cells. CONCLUSIONS Mitochondrial ALDH2 inhibits the proliferation of neonatal rat cardiac fibroblasts induced by high glucose, and the effect is possibly mediated by the up-regulation of autophagy-related proteins Beclin-1 and LC3B.
Collapse
Affiliation(s)
- 碧 唐
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 品方 康
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 建路 郭
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 磊 朱
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 庆梅 徐
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 琴 高
- 蚌埠医学院 生理学教研室,安徽 蚌埠 233030Department of Physiology Cardiovascular Research Center of BengBu Medical College, Bengbu 233030, China
- 蚌埠医学院 心血管病研究中心,安徽 蚌埠 233030Department of Physiology Bengbu Medical College, Bengbu 233030, China
| | - 恒 张
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 洪巨 王
- 蚌埠医学院第一附属医院心血管科,安徽 蚌埠 233004Department of Cardiovascular Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
13
|
Munukutla S, Pan G, Palaniyandi SS. Aldehyde Dehydrogenase (ALDH) 2 in Diabetic Heart Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1193:155-174. [PMID: 31368103 DOI: 10.1007/978-981-13-6260-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A major pathophysiological mechanism behind the development of diabetic heart diseases is oxidative stress mediated by toxic reactive aldehydes such as 4-hydroxynonenal (4HNE). Aldehyde dehydrogenase (ALDH) 2 is a mitochondrial enzyme that has been found to detoxify these deleterious aldehydes and thereby mitigate cardiac damage. Furthermore, its protective role in cellular signaling reverses aberrations caused by hyperglycemia, thereby protecting cardiac function. This chapter assesses the role of ALDH2 in diabetic heart diseases by examining preclinical studies where ALDH2 activity is perturbed in both decreased and increased directions. In doing so, issues in improving ALDH2 activity in select human populations are elucidated, and further research directions are discussed.
Collapse
Affiliation(s)
- Srikar Munukutla
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| | - Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| | - Suresh S Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA.
- Department of Physiology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
14
|
Cao R, Fang D, Wang J, Yu Y, Ye H, Kang P, Li Z, Wang H, Gao Q. ALDH2 Overexpression Alleviates High Glucose-Induced Cardiotoxicity by Inhibiting NLRP3 Inflammasome Activation. J Diabetes Res 2019; 2019:4857921. [PMID: 31871948 PMCID: PMC6906837 DOI: 10.1155/2019/4857921] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/02/2019] [Accepted: 10/25/2019] [Indexed: 12/19/2022] Open
Abstract
Although the underlying mechanisms of diabetes-induced myocardial injury have not been fully illuminated, the inflammation reaction has been reported intently linked with diabetes. The nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome, the key component of pyroptosis, is involved in inflammation reaction, which may be one of the important mechanisms in diabetes-induced myocardial injury. The purpose of this study was to investigate the changes of NLRP3 inflammasome and pyroptosis in high glucose-induced H9C2 cardiac cell injury and investigate whether overexpression of mitochondrial aldehyde dehydrogenase 2 (ALDH2) can reduce the occurrence of pyroptosis. The H9C2 cardiac cells were exposed to 35 mM glucose for 24 h to induce cytotoxicity. Mitochondrial ALDH2 overexpression cardiac cell line was constructed. The results showed in high glucose condition that ALDH2 overexpression significantly increased H9C2 cardiac cell viability, increased mitochondrial ALDH2 activity and protein expression, and reduced mitochondrial reactive oxygen species (ROS) production, 4-hydroxynonenal (4-HNE), and lactate dehydrogenase (LDH) levels; meanwhile, the pyroptosis key components-NLRP3 inflammasome-related proteins, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), cysteine-containing aspartate specific protease 1 (Caspase-1), and interleukin-18 (IL-18) protein expressions-were significantly decreased, and IL-18 and interleukin-1β (IL-1β) levels were also decreased. In high glucose-induced cardiac cell injury, ALDH2 overexpression may reduce ROS production, thereby inhibiting the activation of NLRP3 inflammation and cell pyroptosis. ALDH2 gene might play the potential role in the treatment of high glucose-induced H9C2 cardiac cell injury.
Collapse
Affiliation(s)
- Ruiping Cao
- Department of Physiology, Bengbu, Anhui 233030, China
| | - Dian Fang
- Department of Physiology, Bengbu, Anhui 233030, China
| | - Jiahui Wang
- Department of Physiology, Bengbu, Anhui 233030, China
| | - Ying Yu
- Department of Physiology, Bengbu, Anhui 233030, China
| | - Hongwei Ye
- Department of Physiology, Bengbu, Anhui 233030, China
| | - Pinfang Kang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Zhenghong Li
- Department of Physiology, Bengbu, Anhui 233030, China
| | - Hongju Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Qin Gao
- Department of Physiology, Bengbu, Anhui 233030, China
| |
Collapse
|
15
|
Guo J, Kang P, Zhu L, Sun S, Tao M, Zhang H, Tang B. [Mitochondrial aldehyde dehydrogenase 2 protects against high glucose-induced injury in neonatal rat cardiomyocytes by regulating CaN-NFAT3 signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1288-1293. [PMID: 30514674 DOI: 10.12122/j.issn.1673-4254.2018.11.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate whether CaN-NFAT3 pathway mediates the protective effects of aldehyde dehydrogenase (ALDH) 2 in high glucose-treated neonatal rat ventricular myocytes. METHODS The ventricular myocytes were isolated from the heart of neonatal (within 3 days) SD rats by enzyme digestion and cultured in the presence of 5-Brdu. After reaching confluence, the cultured ventricular myocytes were identified using immunofluorescence assay for α-SA protein. The cells were then cultured in either normal (5 mmol/L) or high glucose (30 mmol/L) medium in the presence of ALDH2 agonist Alda-1, ALDH 2 inhibitor Daidzin, or Alda-1 and NFAT3 inhibitor (11R-VIVIT). Fluorescent probe and ELISA were used to detect intracellular Ca2+ concentration and CaN content, respectively; ALDH2, CaN and NFAT3 protein expressions in the cells were detected using Western blotting. RESULTS Compared with cells cultured in normal glucose, the cells exposed to high glucose showed a significantly decreased expression of ALDH2 protein (P < 0.05) and increased expressions of CaN (P < 0.05) and NFAT3 proteins with also increased intracellular CaN and Ca2+ concentrations (P < 0.01). Alda-1 treatment significantly lowered Ca2+ concentration (P < 0.05), intracellular CaN content (P < 0.01), and CaN and NFAT3 protein expressions (P < 0.05), and increased ALDH2 protein expression (P < 0.05) in high glucose- exposed cells; Daidzin treatment significantly increased Ca2+ concentration (P < 0.01) and intracellular CaN content (P < 0.05) in the exposed cells. Compared with Alda-1 alone, treatment of the high glucose-exposed cells with both Alda-1 and 11R-VIVIT did not produce significant changes in the expression of ALDH2 protein (P>0.05) but significantly reduced the expression of NFAT3 protein (P < 0.05). CONCLUSIONS Mitochondrial ALDH2 protects neonatal rat cardiomyocytes against high glucose-induced injury possibly by negatively regulating Ca2+-CaN-NFAT3 signaling pathway.
Collapse
Affiliation(s)
- Jianlu Guo
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Pinfang Kang
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Lei Zhu
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Shuo Sun
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Min Tao
- Department of Cardiology, Huishan District People's Hospital, Wuxi 214100, China
| | - Heng Zhang
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Bi Tang
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
16
|
Pan G, Deshpande M, Pang H, Palaniyandi SS. Precision medicine approach: Empagliflozin for diabetic cardiomyopathy in mice with aldehyde dehydrogenase (ALDH) 2 * 2 mutation, a specific genetic mutation in millions of East Asians. Eur J Pharmacol 2018; 839:76-81. [PMID: 30240795 DOI: 10.1016/j.ejphar.2018.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 01/17/2023]
Abstract
A vast majority of type-2 diabetic patients (~65%) die of cardiovascular complications including heart failure (HF). In diabetic hearts, levels of 4-hydroxy-2-nonenal (4HNE), a reactive aldehyde that is produced upon lipid peroxidation, were increased. We also demonstrated that in diabetic hearts, there is a decrease in the activity of aldehyde dehydrogenase (ALDH) 2, a primary detoxifying enzyme present in cardiac mitochondria. A single point mutation at E487K of ALDH2 in East Asians known as ALDH2 * 2 intrinsically lowers ALDH2 activity. We hypothesize that Empagliflozin (EMP), a sodium-glucose cotransporter (SGLT) 2 inhibitor, can ameliorate diabetic cardiomyopathy by decreasing hyperglycemia-mediated 4HNE protein adducts in ALDH2 * 2 mutant mice which serve as a precision medicine tool as they mimic ALDH2 * 2 carriers. We induced type-2 diabetes in 11-14 month-old male and female ALDH2 * 2 mice through a high-fat diet. Chow-fed ALDH2 * 2 mice served as controls. At the end of 4 months, we treated the diabetic ALDH2 * 2 mice with EMP (3 mg/kg/d) or its vehicle (Veh). After 2 months of EMP treatment, cardiac function was assessed by conscious echocardiography after treadmill exercise stress. EMP improved the cardiac function and running distance and duration significantly compared to Veh-treated ALDH2 * 2 diabetic mice. These beneficial effects can be attributed to the EMP-mediated decrease in cardiac mitochondrial 4HNE adducts and increase in the levels of phospho AKT, AKT, phospho Akt substrate of 160 kDa (pAS160), AS160 and GLUT-4 in the skeletal muscle tissue of the ALDH2*2 mutant diabetic mice, respectively. Finally, our data implicate EMP can ameliorate diabetic cardiomyopathy in diabetic ALDH2 * 2 mutant patients.
Collapse
Affiliation(s)
- Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States
| | - Mandar Deshpande
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States
| | - Haiyan Pang
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States; Department of Physiology, Wayne State University, Detroit, MI 48202, United States.
| |
Collapse
|