1
|
Lv F, Xie L, Li L, Lin J. LMK235 ameliorates inflammation and fibrosis after myocardial infarction by inhibiting LSD1-related pathway. Sci Rep 2024; 14:23450. [PMID: 39379699 PMCID: PMC11461967 DOI: 10.1038/s41598-024-74887-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Histone deacetylase 4 (HDAC4) and histone deacetylase 5 (HDAC5) are two isoforms of class IIa HDACs, and LMK235 is an HDAC inhibitor with higher selectivity for HDAC4/5. This study aimed to explore the expression and subcellular localization of HDAC4/5 and determine the mechanisms underlying the impact of LMK235 on ventricular remodelling post-MI. METHODS The MI model was established by left anterior descending branch (LAD) ligation, and LMK235 or vehicle was intraperitoneally injected daily for 21 days. Cardiac function was determined by echocardiography. Inflammation was evaluated by HE staining and measuring inflammatory cytokine expression, and fibrosis was evaluated by Masson staining and measuring fibrotic biomarker expression. RESULTS We found that LMK235 ameliorated cardiac dysfunction post-MI by suppressing inflammation and fibrosis, and LMK235 inhibited upregulation of lysine-specific demethylase 1 (LSD1) expression post-MI. In macrophages, LMK235 attenuated lipopolysaccharide (LPS) - induced inflammatory cytokine expression and inhibited LSD1 expression, while overexpression of LSD1 abrogated the anti-inflammatory effect of LMK235. In cardiac fibroblasts, LMK235 attenuated transforming growth factor-β1 (TGF-β1) - induced fibrotic biomarker expression and inhibited LSD1 expression, while overexpression of LSD1 abrogated the antifibrotic effect of LMK235. CONCLUSION LMK235 attenuates chronic inflammation and fibrosis post-MI, leading to improved cardiac function. The anti-inflammatory effect of LMK235 may result from inhibition of the LSD1-NF-κB pathway in macrophages. The antifibrotic effect of LMK235 may result from inhibition of the LSD1-Smad2/3 pathway in cardiac fibroblasts.
Collapse
Affiliation(s)
- Fangzhou Lv
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Laidi Xie
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jiafeng Lin
- Department of Cardiology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Liu YB, Wang Q, Song YL, Song XM, Fan YC, Kong L, Zhang JS, Li S, Lv YJ, Li ZY, Dai JY, Qiu ZK. Abnormal phosphorylation / dephosphorylation and Ca 2+ dysfunction in heart failure. Heart Fail Rev 2024; 29:751-768. [PMID: 38498262 DOI: 10.1007/s10741-024-10395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Heart failure (HF) can be caused by a variety of causes characterized by abnormal myocardial systole and diastole. Ca2+ current through the L-type calcium channel (LTCC) on the membrane is the initial trigger signal for a cardiac cycle. Declined systole and diastole in HF are associated with dysfunction of myocardial Ca2+ function. This disorder can be correlated with unbalanced levels of phosphorylation / dephosphorylation of LTCC, endoplasmic reticulum (ER), and myofilament. Kinase and phosphatase activity changes along with HF progress, resulting in phased changes in the degree of phosphorylation / dephosphorylation. It is important to realize the phosphorylation / dephosphorylation differences between a normal and a failing heart. This review focuses on phosphorylation / dephosphorylation changes in the progression of HF and summarizes the effects of phosphorylation / dephosphorylation of LTCC, ER function, and myofilament function in normal conditions and HF based on previous experiments and clinical research. Also, we summarize current therapeutic methods based on abnormal phosphorylation / dephosphorylation and clarify potential therapeutic directions.
Collapse
Affiliation(s)
- Yan-Bing Liu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
- Medical College, Qingdao University, Qingdao, China
| | - Qian Wang
- Medical College, Qingdao University, Qingdao, China
| | - Yu-Ling Song
- Department of Pediatrics, Huantai County Hospital of Traditional Chinese Medicine, Zibo, China
| | | | - Yu-Chen Fan
- Medical College, Qingdao University, Qingdao, China
| | - Lin Kong
- Medical College, Qingdao University, Qingdao, China
| | | | - Sheng Li
- Medical College, Qingdao University, Qingdao, China
| | - Yi-Ju Lv
- Medical College, Qingdao University, Qingdao, China
| | - Ze-Yang Li
- Medical College, Qingdao University, Qingdao, China
| | - Jing-Yu Dai
- Department of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| | - Zhen-Kang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
3
|
Li Z, Chen J, Huang H, Zhan Q, Wang F, Chen Z, Lu X, Sun G. Post-translational modifications in diabetic cardiomyopathy. J Cell Mol Med 2024; 28:e18158. [PMID: 38494853 PMCID: PMC10945092 DOI: 10.1111/jcmm.18158] [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: 07/30/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 03/19/2024] Open
Abstract
The increasing attention towards diabetic cardiomyopathy as a distinctive complication of diabetes mellitus has highlighted the need for standardized diagnostic criteria and targeted treatment approaches in clinical practice. Ongoing research is gradually unravelling the pathogenesis of diabetic cardiomyopathy, with a particular emphasis on investigating various post-translational modifications. These modifications dynamically regulate protein function in response to changes in the internal and external environment, and their disturbance of homeostasis holds significant relevance for the development of chronic ailments. This review provides a comprehensive overview of the common post-translational modifications involved in the initiation and progression of diabetic cardiomyopathy, including O-GlcNAcylation, phosphorylation, methylation, acetylation and ubiquitination. Additionally, the review discusses drug development strategies for targeting key post-translational modification targets, such as agonists, inhibitors and PROTAC (proteolysis targeting chimaera) technology that targets E3 ubiquitin ligases.
Collapse
Affiliation(s)
- Zhi Li
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Jie Chen
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Hailong Huang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Qianru Zhan
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Fengzhi Wang
- Department of Neurology, People's Hospital of Liaoning ProvincePeople's Hospital of China Medical UniversityShenyangChina
| | - Zihan Chen
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Xinwei Lu
- Department of CardiologySiping Central People's HospitalSipingChina
| | - Guozhe Sun
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
4
|
Herwig M, Begovic M, Budde H, Delalat S, Zhazykbayeva S, Sieme M, Schneider L, Jaquet K, Mügge A, Akin I, El-Battrawy I, Fielitz J, Hamdani N. Protein Kinase D Plays a Crucial Role in Maintaining Cardiac Homeostasis by Regulating Post-Translational Modifications of Myofilament Proteins. Int J Mol Sci 2024; 25:2790. [PMID: 38474037 DOI: 10.3390/ijms25052790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Protein kinase D (PKD) enzymes play important roles in regulating myocardial contraction, hypertrophy, and remodeling. One of the proteins phosphorylated by PKD is titin, which is involved in myofilament function. In this study, we aimed to investigate the role of PKD in cardiomyocyte function under conditions of oxidative stress. To do this, we used mice with a cardiomyocyte-specific knock-out of Prkd1, which encodes PKD1 (Prkd1loxP/loxP; αMHC-Cre; PKD1 cKO), as well as wild type littermate controls (Prkd1loxP/loxP; WT). We isolated permeabilized cardiomyocytes from PKD1 cKO mice and found that they exhibited increased passive stiffness (Fpassive), which was associated with increased oxidation of titin, but showed no change in titin ubiquitination. Additionally, the PKD1 cKO mice showed increased myofilament calcium (Ca2+) sensitivity (pCa50) and reduced maximum Ca2+-activated tension. These changes were accompanied by increased oxidation and reduced phosphorylation of the small myofilament protein cardiac myosin binding protein C (cMyBPC), as well as altered phosphorylation levels at different phosphosites in troponin I (TnI). The increased Fpassive and pCa50, and the reduced maximum Ca2+-activated tension were reversed when we treated the isolated permeabilized cardiomyocytes with reduced glutathione (GSH). This indicated that myofilament protein oxidation contributes to cardiomyocyte dysfunction. Furthermore, the PKD1 cKO mice exhibited increased oxidative stress and increased expression of pro-inflammatory markers interleukin (IL)-6, IL-18, and tumor necrosis factor alpha (TNF-α). Both oxidative stress and inflammation contributed to an increase in microtubule-associated protein 1 light chain 3 (LC3)-II levels and heat shock response by inhibiting the mammalian target of rapamycin (mTOR) in the PKD1 cKO mouse myocytes. These findings revealed a previously unknown role for PKD1 in regulating diastolic passive properties, myofilament Ca2+ sensitivity, and maximum Ca2+-activated tension under conditions of oxidative stress. Finally, we emphasized the importance of PKD1 in maintaining the balance of oxidative stress and inflammation in the context of autophagy, as well as cardiomyocyte function.
Collapse
Affiliation(s)
- Melissa Herwig
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Merima Begovic
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Heidi Budde
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Simin Delalat
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Saltanat Zhazykbayeva
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Marcel Sieme
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Luca Schneider
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Kornelia Jaquet
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, UK RUB, Ruhr University Bochum, 44789 Bochum, Germany
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ibrahim El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, UK RUB, Ruhr University Bochum, 44789 Bochum, Germany
| | - Jens Fielitz
- Department of Molecular Cardiology, DZHK (German Center for Cardiovascular Research), Partner Site, 17475 Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, 44801 Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Physiology, University Maastricht, 6211 LK Maastricht, The Netherlands
- HCEMM-SU Cardiovascular Comorbidities Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| |
Collapse
|
5
|
Tang J, Tam E, Song E, Xu A, Sweeney G. Crosstalk between myocardial autophagy and sterile inflammation in the development of heart failure. AUTOPHAGY REPORTS 2024; 3:2320605. [PMID: 40395524 PMCID: PMC11864620 DOI: 10.1080/27694127.2024.2320605] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 05/22/2025]
Abstract
Heart failure, a leading driver of global mortality, remains a topic of intense contemporary research interest due to the prevailing unmet need in cardiometabolic therapeutics. Numerous mechanisms with the potential to influence the onset and development of heart failure remain incompletely understood. Firstly, myocardial autophagy, which involves lysosomal degradation of damaged cellular components, confers context-dependent beneficial and detrimental effects. Secondly, sterile inflammation may arise following cardiac stress and exacerbate the progression of heart failure. Inflammation changes in a temporal manner and its onset must be adequately resolved to limit progression of heart failure. Mitochondria are an important factor in contributing to sterile inflammation by releasing damage associated molecular patterns (DAMPs) including mitochondrial DNA (mtDNA). Accordingly, this is one reason why the selective autophagy of mitochondria to maintain optimal function is important in determining cardiac function. In this review, we examine the increasing evidence suggesting crosstalk between autophagy and sterile inflammation together with their role in the development of heart failure. In particular, this is exemplified in the preclinical models of ischaemia/reperfusion injury and pressure overload induced heart failure. We also highlight potential therapeutic approaches focusing on autophagy and addressing sterile inflammation, aiming to enhance outcomes in heart failure.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, Toronto, ON, Canada
| | - Eddie Tam
- Department of Biology, York University, Toronto, ON, Canada
| | - Erfei Song
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Metabolic and Bariatric Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
6
|
Zhu C, Piao Z, Jin L. HDAC5 inhibition attenuates ventricular remodeling and cardiac dysfunction. Orphanet J Rare Dis 2023; 18:266. [PMID: 37667300 PMCID: PMC10476361 DOI: 10.1186/s13023-023-02896-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND This study aimed to investigate the role of histone deacetylase 5 (HDAC5) in ventricular remodeling and explore the therapeutic potential of the HDAC5 inhibitor LMK235. METHODS A transverse aortic constriction (TAC) mouse model and angiotensin II (Ang II)-treated H9C2 cells were used to evaluate the effects of HDAC5 inhibition with LMK235 on ventricular remodeling and cardiac dysfunction. Additionally, the involvement of the extracellular signal-regulated kinase (ERK)/early growth response protein 1 (EGR1) signaling pathway in regulating myocyte enhancer factor 2 A (MEF2A) expression was assessed. RESULTS HDAC5 was upregulated in TAC mice and Ang II-treated H9C2 cells, suggesting its involvement in ventricular remodeling and cardiac dysfunction. LMK235 treatment significantly improved cardiac function in TAC mice and attenuated TAC-induced ventricular remodeling and Ang II-induced H9C2 cell hypertrophy. Mechanically, HDAC5 inhibition activated the ERK/EGR1 signaling pathway. CONCLUSIONS Our findings demonstrate that HDAC5 may suppress the activation of ERK/EGR1 signaling to regulate MEF2A expression and therefore participate in cardiac pathophysiology.
Collapse
Affiliation(s)
- Chenxi Zhu
- Department of Cardiology, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, No. 306 Hualongqiao Road, Wenzhou, Zhejiang, 325000, China
| | - Zhehao Piao
- Department of Cardiology, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, No. 306 Hualongqiao Road, Wenzhou, Zhejiang, 325000, China
| | - Li Jin
- Department of Cardiology, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, No. 306 Hualongqiao Road, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
7
|
Mira Hernandez J, Ko CY, Mandel AR, Shen EY, Baidar S, Christensen AR, Hellgren K, Morotti S, Martin JL, Hegyi B, Bossuyt J, Bers DM. Cardiac Protein Kinase D1 ablation alters the myocytes β-adrenergic response. J Mol Cell Cardiol 2023; 180:33-43. [PMID: 37149124 PMCID: PMC11186477 DOI: 10.1016/j.yjmcc.2023.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
β-adrenergic (β-AR) signaling is essential for the adaptation of the heart to exercise and stress. Chronic stress leads to the activation of Ca2+/calmodulin-dependent kinase II (CaMKII) and protein kinase D (PKD). Unlike CaMKII, the effects of PKD on excitation-contraction coupling (ECC) remain unclear. To elucidate the mechanisms of PKD-dependent ECC regulation, we used hearts from cardiac-specific PKD1 knockout (PKD1 cKO) mice and wild-type (WT) littermates. We measured calcium transients (CaT), Ca2+ sparks, contraction and L-type Ca2+ current in paced cardiomyocytes under acute β-AR stimulation with isoproterenol (ISO; 100 nM). Sarcoplasmic reticulum (SR) Ca2+ load was assessed by rapid caffeine (10 mM) induced Ca2+ release. Expression and phosphorylation of ECC proteins phospholambam (PLB), troponin I (TnI), ryanodine receptor (RyR), sarcoendoplasmic reticulum Ca2+ ATPase (SERCA) were evaluated by western blotting. At baseline, CaT amplitude and decay tau, Ca2+ spark frequency, SR Ca2+ load, L-type Ca2+ current, contractility, and expression and phosphorylation of ECC protein were all similar in PKD1 cKO vs. WT. However, PKD1 cKO cardiomyocytes presented a diminished ISO response vs. WT with less increase in CaT amplitude, slower [Ca2+]i decline, lower Ca2+ spark rate and lower RyR phosphorylation, but with similar SR Ca2+ load, L-type Ca2+ current, contraction and phosphorylation of PLB and TnI. We infer that the presence of PKD1 allows full cardiomyocyte β-adrenergic responsiveness by allowing optimal enhancement in SR Ca2+ uptake and RyR sensitivity, but not altering L-type Ca2+ current, TnI phosphorylation or contractile response. Further studies are necessary to elucidate the specific mechanisms by which PKD1 is regulating RyR sensitivity. We conclude that the presence of basal PKD1 activity in cardiac ventricular myocytes contributes to normal β-adrenergic responses in Ca2+ handling.
Collapse
Affiliation(s)
- Juliana Mira Hernandez
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America; Research Group in Veterinary Medicine-GIVET, School of Veterinary Medicine, University Corporation Lasallista, Caldas, Antioquia, Colombia
| | - Christopher Y Ko
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Avery R Mandel
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Erin Y Shen
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Sonya Baidar
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Ashley R Christensen
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Kim Hellgren
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Jody L Martin
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America; Cardiovascular Research Institute, University of California, Davis, Davis, CA 95616, United States of America
| | - Bence Hegyi
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America.
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, United States of America.
| |
Collapse
|
8
|
Bossuyt J, Borst JM, Verberckmoes M, Bailey LRJ, Bers DM, Hegyi B. Protein Kinase D1 Regulates Cardiac Hypertrophy, Potassium Channel Remodeling, and Arrhythmias in Heart Failure. J Am Heart Assoc 2022; 11:e027573. [PMID: 36172952 DOI: 10.1161/jaha.122.027573] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Structural and electrophysiological remodeling characterize heart failure (HF) enhancing arrhythmias. PKD1 (protein kinase D1) is upregulated in HF and mediates pathological hypertrophic signaling, but its role in K+ channel remodeling and arrhythmogenesis in HF is unknown. Methods and Results We performed echocardiography, electrophysiology, and expression analysis in wild-type and PKD1 cardiomyocyte-specific knockout (cKO) mice following transverse aortic constriction (TAC). PKD1-cKO mice exhibited significantly less cardiac hypertrophy post-TAC and were protected from early decline in cardiac contractile function (3 weeks post-TAC) but not the progression to HF at 7 weeks post-TAC. Wild-type mice exhibited ventricular action potential duration prolongation at 8 weeks post-TAC, which was attenuated in PKD1-cKO, consistent with larger K+ currents via the transient outward current, sustained current, inward rectifier K+ current, and rapid delayed rectifier K+ current and increased expression of corresponding K+ channels. Conversely, reduction of slowly inactivating K+ current was independent of PKD1 in HF. Acute PKD inhibition slightly increased transient outward current in TAC and sham wild-type myocytes but did not alter other K+ currents. Sham PKD1-cKO versus wild-type also exhibited larger transient outward current and faster early action potential repolarization. Tachypacing-induced action potential duration alternans in TAC animals was increased and independent of PKD1, but diastolic arrhythmogenic activities were reduced in PKD1-cKO. Conclusions Our data indicate an important role for PKD1 in the HF-related hypertrophic response and K+ channel downregulation. Therefore, PKD1 inhibition may represent a therapeutic strategy to reduce hypertrophy and arrhythmias; however, PKD1 inhibition may not prevent disease progression and reduced contractility in HF.
Collapse
Affiliation(s)
- Julie Bossuyt
- Department of Pharmacology University of California Davis CA
| | - Johanna M Borst
- Department of Pharmacology University of California Davis CA
| | | | | | - Donald M Bers
- Department of Pharmacology University of California Davis CA
| | - Bence Hegyi
- Department of Pharmacology University of California Davis CA
| |
Collapse
|
9
|
Guttzeit S, Backs J. Post-translational modifications talk and crosstalk to class IIa histone deacetylases. J Mol Cell Cardiol 2021; 162:53-61. [PMID: 34416247 DOI: 10.1016/j.yjmcc.2021.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/08/2021] [Accepted: 08/12/2021] [Indexed: 12/30/2022]
Abstract
Epigenetic modifications, such as histone or DNA modifications are key regulators of gene transcription and changes are often associated with maladaptive processes underlying cardiovascular disease. Epigenetic regulators therefore likely play a crucial role in cardiomyocyte homeostasis and facilitate the cellular adaption to various internal and external stimuli, responding to different intercellular and extracellular cues. Class IIa histone deacetylases are a class of epigenetic regulators that possess a myriad of post-transcriptional modification sites that modulate their activity in response to oxidative stress, altered catecholamine signalling or changes in the cellular metabolism. This review summaries the known reversible, post-translational modifications (PTMs) of class IIa histone deacetylases (HDACs) that ultimately drive transcriptional changes in homeostasis and disease. We also highlight the idea of a crosstalk of various PTMs on class IIa HDACs potentially leading to compensatory or synergistic effects on the class IIa HDAC-regulated cell behavior.
Collapse
Affiliation(s)
- Sebastian Guttzeit
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
10
|
McCarty MF. Nutraceutical, Dietary, and Lifestyle Options for Prevention and Treatment of Ventricular Hypertrophy and Heart Failure. Int J Mol Sci 2021; 22:ijms22073321. [PMID: 33805039 PMCID: PMC8037104 DOI: 10.3390/ijms22073321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Although well documented drug therapies are available for the management of ventricular hypertrophy (VH) and heart failure (HF), most patients nonetheless experience a downhill course, and further therapeutic measures are needed. Nutraceutical, dietary, and lifestyle measures may have particular merit in this regard, as they are currently available, relatively safe and inexpensive, and can lend themselves to primary prevention as well. A consideration of the pathogenic mechanisms underlying the VH/HF syndrome suggests that measures which control oxidative and endoplasmic reticulum (ER) stress, that support effective nitric oxide and hydrogen sulfide bioactivity, that prevent a reduction in cardiomyocyte pH, and that boost the production of protective hormones, such as fibroblast growth factor 21 (FGF21), while suppressing fibroblast growth factor 23 (FGF23) and marinobufagenin, may have utility for preventing and controlling this syndrome. Agents considered in this essay include phycocyanobilin, N-acetylcysteine, lipoic acid, ferulic acid, zinc, selenium, ubiquinol, astaxanthin, melatonin, tauroursodeoxycholic acid, berberine, citrulline, high-dose folate, cocoa flavanols, hawthorn extract, dietary nitrate, high-dose biotin, soy isoflavones, taurine, carnitine, magnesium orotate, EPA-rich fish oil, glycine, and copper. The potential advantages of whole-food plant-based diets, moderation in salt intake, avoidance of phosphate additives, and regular exercise training and sauna sessions are also discussed. There should be considerable scope for the development of functional foods and supplements which make it more convenient and affordable for patients to consume complementary combinations of the agents discussed here. Research Strategy: Key word searching of PubMed was employed to locate the research papers whose findings are cited in this essay.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA
| |
Collapse
|
11
|
Yuan H, Xiang Q, Yang L, Geng J. Protein kinase D participates in cardiomyocyte hypertrophy by regulating extracellular signal-regulated and myocyte enhancer factor 2D. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.repce.2020.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
12
|
CaMKII and PKA-dependent phosphorylation co-regulate nuclear localization of HDAC4 in adult cardiomyocytes. Basic Res Cardiol 2021; 116:11. [PMID: 33590335 PMCID: PMC7884572 DOI: 10.1007/s00395-021-00850-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Nuclear histone deacetylase 4 (HDAC4) represses MEF2-mediated transcription, implicated in the development of heart failure. CaMKII-dependent phosphorylation drives nucleus-to-cytoplasm HDAC4 shuttling, but protein kinase A (PKA) is also linked to HDAC4 translocation. However, the interplay of CaMKII and PKA in regulating adult cardiomyocyte HDAC4 translocation is unclear. Here we sought to determine the interplay of PKA- and CaMKII-dependent HDAC4 phosphorylation and translocation in adult mouse, rabbit and human ventricular myocytes. Confocal imaging and protein analyses revealed that inhibition of CaMKII-but not PKA, PKC or PKD-raised nucleo-to-cytoplasmic HDAC4 fluorescence ratio (FNuc/FCyto) by ~ 50%, indicating baseline CaMKII activity that limits HDAC4 nuclear localization. Further CaMKII activation (via increased extracellular [Ca2+], high pacing frequencies, angiotensin II or overexpression of CaM or CaMKIIδC) led to significant HDAC4 nuclear export. In contrast, PKA activation by isoproterenol or forskolin drove HDAC4 into the nucleus (raising FNuc/FCyto by > 60%). These PKA-mediated effects were abolished in cells pretreated with PKA inhibitors and in cells expressing mutant HDAC4 in S265/266A mutant. In physiological conditions where both kinases are active, PKA-dependent nuclear accumulation of HDAC4 was predominant in the very early response, while CaMKII-dependent HDAC4 export prevailed upon prolonged stimuli. This orchestrated co-regulation was shifted in failing cardiomyocytes, where CaMKII-dependent effects predominated over PKA-dependent response. Importantly, human cardiomyocytes showed similar CaMKII- and PKA-dependent HDAC4 shifts. Collectively, CaMKII limits nuclear localization of HDAC4, while PKA favors HDAC4 nuclear retention and S265/266 is essential for PKA-mediated regulation. These pathways thus compete in HDAC4 nuclear localization and transcriptional regulation in cardiac signaling.
Collapse
|
13
|
Hegyi B, Pölönen RP, Hellgren KT, Ko CY, Ginsburg KS, Bossuyt J, Mercola M, Bers DM. Cardiomyocyte Na + and Ca 2+ mishandling drives vicious cycle involving CaMKII, ROS, and ryanodine receptors. Basic Res Cardiol 2021; 116:58. [PMID: 34648073 PMCID: PMC8516771 DOI: 10.1007/s00395-021-00900-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022]
Abstract
Cardiomyocyte Na+ and Ca2+ mishandling, upregulated Ca2+/calmodulin-dependent kinase II (CaMKII), and increased reactive oxygen species (ROS) are characteristics of various heart diseases, including heart failure (HF), long QT (LQT) syndrome, and catecholaminergic polymorphic ventricular tachycardia (CPVT). These changes may form a vicious cycle of positive feedback to promote cardiac dysfunction and arrhythmias. In HF rabbit cardiomyocytes investigated in this study, the inhibition of CaMKII, late Na+ current (INaL), and leaky ryanodine receptors (RyRs) all attenuated the prolongation and increased short-term variability (STV) of action potential duration (APD), but in age-matched controls these inhibitors had no or minimal effects. In control cardiomyocytes, we enhanced RyR leak (by low [caffeine] plus isoproterenol mimicking CPVT) which markedly increased STV and delayed afterdepolarizations (DADs). These proarrhythmic changes were significantly attenuated by both CaMKII inhibition and mitochondrial ROS scavenging, with a slight synergy with INaL inhibition. Inducing LQT by elevating INaL (by Anemone toxin II, ATX-II) caused markedly prolonged APD, increased STV, and early afterdepolarizations (EADs). Those proarrhythmic ATX-II effects were largely attenuated by mitochondrial ROS scavenging, and partially reduced by inhibition of CaMKII and pathological leaky RyRs using dantrolene. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) bearing LQT3 mutation SCN5A N406K, dantrolene significantly attenuated cell arrhythmias and APD prolongation. Targeting critical components of the Na+-Ca2+-CaMKII-ROS-INaL arrhythmogenic vicious cycle may exhibit important on-target and also trans-target effects (e.g., INaL and RyR inhibition can alter INaL-mediated LQT3 effects). Incorporating this vicious cycle into therapeutic strategies provides novel integrated insight for treating cardiac arrhythmias and diseases.
Collapse
Affiliation(s)
- Bence Hegyi
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Risto-Pekka Pölönen
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA ,grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Kim T. Hellgren
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Christopher Y. Ko
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Kenneth S. Ginsburg
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Julie Bossuyt
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Mark Mercola
- grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Donald M. Bers
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| |
Collapse
|
14
|
Protein kinase D participates in cardiomyocyte hypertrophy by regulating extracellular signal-regulated and myocyte enhancer factor 2D. Rev Port Cardiol 2020; 40:191-200. [PMID: 33334620 DOI: 10.1016/j.repc.2020.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/13/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Cardiomyocyte hypertrophy is an important feature of hypertension. However, its molecular underpinnings, especially the signaling cascades, remain unclear. Here we hypothesized that a protein kinase D (PKD)-dependent extracellular signal-regulated kinase 5 (ERK5) pathway was able to regulate downstream myocyte enhancer factor 2D (MEF2D), affecting prohypertrophic responses to angiotensin II (Ang II). METHODS Neonatal rat cardiomyocytes from 2- to 3-day-old Sprague-Dawley rats were prepared and Western blot, real-time quantitative PCR and immunofluorescence staining were used to assess the activation and translocation of pathway signaling molecules. Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) expression and [3H]-leucine (Leu) incorporation were measured to determine cell hypertrophy. RESULTS Elevated levels of phosphorylated PKD (p-PKD) and ERK5 (p-ERK5) were observed in cardiomyocytes stimulated with Ang II, while silencing protein kinase C epsilon (PKCɛ) resulted in significantly lower levels of p-PKD. Furthermore, Ang II-induced ERK5 activated translocation was mediated by the PKD pathway. Consequently, inhibiting PKCɛ, PKD and ERK5 by siRNA significantly attenuated Ang II-induced MEF2D activation, ANP and BNP mRNA expression, and [3H]-Leu incorporation. CONCLUSIONS Our studies are the first to show that the PKCɛ/PKD/ERK5/MEF2D pathway plays an important role in the cardiomyocyte hypertrophy response to Ang II.
Collapse
|
15
|
Takano APC, Senger N, Barreto-Chaves MLM. The endocrinological component and signaling pathways associated to cardiac hypertrophy. Mol Cell Endocrinol 2020; 518:110972. [PMID: 32777452 DOI: 10.1016/j.mce.2020.110972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Although myocardial growth corresponds to an adaptive response to maintain cardiac contractile function, the cardiac hypertrophy is a condition that occurs in many cardiovascular diseases and typically precedes the onset of heart failure. Different endocrine factors such as thyroid hormones, insulin, insulin-like growth factor 1 (IGF-1), angiotensin II (Ang II), endothelin (ET-1), catecholamines, estrogen, among others represent important stimuli to cardiomyocyte hypertrophy. Thus, numerous endocrine disorders manifested as changes in the local environment or multiple organ systems are especially important in the context of progression from cardiac hypertrophy to heart failure. Based on that information, this review summarizes experimental findings regarding the influence of such hormones upon signalling pathways associated with cardiac hypertrophy. Understanding mechanisms through which hormones differentially regulate cardiac hypertrophy could open ways to obtain therapeutic approaches that contribute to prevent or delay the onset of heart failure related to endocrine diseases.
Collapse
Affiliation(s)
| | - Nathalia Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | |
Collapse
|
16
|
Cardiac CaMKII δ and Wenxin Keli Prevents Ang II-Induced Cardiomyocyte Hypertrophy by Modulating CnA-NFATc4 and Inflammatory Signaling Pathways in H9c2 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9502651. [PMID: 33149757 PMCID: PMC7603598 DOI: 10.1155/2020/9502651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/18/2020] [Accepted: 09/20/2020] [Indexed: 01/23/2023]
Abstract
Previous studies have demonstrated that calcium-/calmodulin-dependent protein kinase II (CaMKII) and calcineurin A-nuclear factor of activated T-cell (CnA-NFAT) signaling pathways play key roles in cardiac hypertrophy (CH). However, the interaction between CaMKII and CnA-NFAT signaling remains unclear. H9c2 cells were cultured and treated with angiotensin II (Ang II) with or without silenced CaMKIIδ (siCaMKII) and cyclosporine A (CsA, a calcineurin inhibitor) and subsequently treated with Wenxin Keli (WXKL). Patch clamp recording was conducted to assess L-type Ca2+ current (ICa-L), and the expression of proteins involved in signaling pathways was measured by western blotting. Myocardial cytoskeletal protein and nuclear translocation of target proteins were assessed by immunofluorescence. The results indicated that siCaMKII suppressed Ang II-induced CH, as evidenced by reduced cell surface area and ICa-L. Notably, siCaMKII inhibited Ang II-induced activation of CnA and NFATc4 nuclear transfer. Inflammatory signaling was inhibited by siCaMKII and WXKL. Interestingly, CsA inhibited CnA-NFAT pathway expression but activated CaMKII signaling. In conclusion, siCaMKII may improve CH, possibly by blocking CnA-NFAT and MyD88 signaling, and WXKL has a similar effect. These data suggest that inhibiting CaMKII, but not CnA, may be a promising approach to attenuate CH and arrhythmia progression.
Collapse
|
17
|
Li J, Gao Q, Wang S, Kang Z, Li Z, Lei S, Sun X, Zhao M, Chen X, Jiao G, Hu H, Hao L. Sustained increased CaMKII phosphorylation is involved in the impaired regression of isoproterenol-induced cardiac hypertrophy in rats. J Pharmacol Sci 2020; 144:30-42. [DOI: 10.1016/j.jphs.2020.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 12/28/2022] Open
|
18
|
Ljubojevic-Holzer S, Herren AW, Djalinac N, Voglhuber J, Morotti S, Holzer M, Wood BM, Abdellatif M, Matzer I, Sacherer M, Radulovic S, Wallner M, Ivanov M, Wagner S, Sossalla S, von Lewinski D, Pieske B, Brown JH, Sedej S, Bossuyt J, Bers DM. CaMKIIδC Drives Early Adaptive Ca 2+ Change and Late Eccentric Cardiac Hypertrophy. Circ Res 2020; 127:1159-1178. [PMID: 32821022 PMCID: PMC7547876 DOI: 10.1161/circresaha.120.316947] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Supplemental Digital Content is available in the text. CaMKII (Ca2+-Calmodulin dependent protein kinase) δC activation is implicated in pathological progression of heart failure (HF) and CaMKIIδC transgenic mice rapidly develop HF and arrhythmias. However, little is known about early spatio-temporal Ca2+ handling and CaMKII activation in hypertrophy and HF.
Collapse
Affiliation(s)
- Senka Ljubojevic-Holzer
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria.,Department of Pharmacology, University of California, Davis, CA (S.L.-H., A.W.H., S.M., B.M.W., J.B., D.M.B.).,BioTechMed Graz, Austria (S.L.-H., J.V., S. Sedej)
| | - Anthony W Herren
- Department of Pharmacology, University of California, Davis, CA (S.L.-H., A.W.H., S.M., B.M.W., J.B., D.M.B.)
| | - Natasa Djalinac
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Julia Voglhuber
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria.,BioTechMed Graz, Austria (S.L.-H., J.V., S. Sedej)
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, CA (S.L.-H., A.W.H., S.M., B.M.W., J.B., D.M.B.)
| | - Michael Holzer
- Otto-Loewi Research Centre, Division of Pharmacology (M.H.), Medical University of Graz, Austria
| | - Brent M Wood
- Department of Pharmacology, University of California, Davis, CA (S.L.-H., A.W.H., S.M., B.M.W., J.B., D.M.B.)
| | - Mahmoud Abdellatif
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Ingrid Matzer
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Michael Sacherer
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Snjezana Radulovic
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Markus Wallner
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Milan Ivanov
- Institute for Medical Research, University of Belgrade, Serbia (M.I.)
| | - Stefan Wagner
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Germany (S.W., S. Sossalla)
| | - Samuel Sossalla
- Klinik für Kardiologie und Pneumologie, Georg-August-Universität Göttingen, Germany (S. Sossalla).,Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Germany (S.W., S. Sossalla)
| | - Dirk von Lewinski
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine Berlin, Germany (B.P.)
| | - Joan Heller Brown
- Department of Pharmacology, University of California San Diego, La Jolla (J.H.B.)
| | - Simon Sedej
- Department of Cardiology (S.L.-H., N.D., J.V., M.A., I.M., M.S., S.R., M.W., D.v.L., S. Sedej), Medical University of Graz, Austria.,BioTechMed Graz, Austria (S.L.-H., J.V., S. Sedej).,Faculty of Medicine, Institute of Physiology, University of Maribor, Slovenia (S. Sedej)
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, CA (S.L.-H., A.W.H., S.M., B.M.W., J.B., D.M.B.)
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA (S.L.-H., A.W.H., S.M., B.M.W., J.B., D.M.B.)
| |
Collapse
|
19
|
Herwig M, Kolijn D, Lódi M, Hölper S, Kovács Á, Papp Z, Jaquet K, Haldenwang P, Dos Remedios C, Reusch PH, Mügge A, Krüger M, Fielitz J, Linke WA, Hamdani N. Modulation of Titin-Based Stiffness in Hypertrophic Cardiomyopathy via Protein Kinase D. Front Physiol 2020; 11:240. [PMID: 32351396 PMCID: PMC7174613 DOI: 10.3389/fphys.2020.00240] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
The giant protein titin performs structure-preserving functions in the sarcomere and is important for the passive stiffness (Fpassive) of cardiomyocytes. Protein kinase D (PKD) enzymes play crucial roles in regulating myocardial contraction, hypertrophy, and remodeling. PKD phosphorylates myofilament proteins, but it is not known whether the giant protein titin is also a PKD substrate. Here, we aimed to determine whether PKD phosphorylates titin and thereby modulates cardiomyocyte Fpassive in normal and failing myocardium. The phosphorylation of titin was assessed in cardiomyocyte-specific PKD knock-out mice (cKO) and human hearts using immunoblotting with a phosphoserine/threonine and a phosphosite-specific titin antibody. PKD-dependent site-specific titin phosphorylation in vivo was quantified by mass spectrometry using stable isotope labeling by amino acids in cell culture (SILAC) of SILAC-labeled mouse heart protein lysates that were mixed with lysates isolated from hearts of either wild-type control (WT) or cKO mice. Fpassive of single permeabilized cardiomyocytes was recorded before and after PKD and HSP27 administration. All-titin phosphorylation was reduced in cKO compared to WT hearts. Multiple conserved PKD-dependent phosphosites were identified within the Z-disk, A-band and M-band regions of titin by quantitative mass spectrometry, and many PKD-dependent phosphosites detected in the elastic titin I-band region were significantly decreased in cKO. Analysis of titin site-specific phosphorylation showed unaltered or upregulated phosphorylation in cKO compared to matched WT hearts. Fpassive was elevated in cKO compared to WT cardiomyocytes and PKD administration lowered Fpassive of WT and cKO cardiomyocytes. Cardiomyocytes from hypertrophic cardiomyopathy (HCM) patients showed higher Fpassive compared to control hearts and significantly lower Fpassive after PKD treatment. In addition, we found higher phosphorylation at CaMKII-dependent titin sites in HCM compared to control hearts. Expression and phosphorylation of HSP27, a substrate of PKD, were elevated in HCM hearts, which was associated with increased PKD expression and phosphorylation. The relocalization of HSP27 in HCM away from the sarcomeric Z-disk and I-band suggested that HSP27 failed to exert its protective action on titin extensibility. This protection could, however, be restored by administration of HSP27, which significantly reduced Fpassive in HCM cardiomyocytes. These findings establish a previously unknown role for PKDin regulating diastolic passive properties of healthy and diseased hearts.
Collapse
Affiliation(s)
- Melissa Herwig
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Detmar Kolijn
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Mária Lódi
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Soraya Hölper
- Sanofi-Aventis Deutschland GmbH Industriepark Höchst, Frankfurt, Germany
| | - Árpád Kovács
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kornelia Jaquet
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Peter Haldenwang
- Department of Cardiothoracic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Cris Dos Remedios
- School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, NSW, Australia
| | - Peter H Reusch
- Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Mügge
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Jens Fielitz
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, University of Münster, Münster, Germany
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
20
|
Chang PC, Wo HT, Lee HL, Lin SF, Chu Y, Wen MS, Chou CC. Sacubitril/Valsartan Therapy Ameliorates Ventricular Tachyarrhythmia Inducibility in a Rabbit Myocardial Infarction Model. J Card Fail 2020; 26:527-537. [PMID: 32209390 DOI: 10.1016/j.cardfail.2020.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Coronary artery disease is the most common cause of heart failure (HF) in developed countries. The aim of this study was to elucidate the mechanisms of reduction of arrhythmias after sacubitril/valsartan (LCZ696) therapy in a myocardial infarction (MI)-HF rabbit model. METHODS AND RESULTS Chronic MI in rabbits with HF were divided into 3 groups: placebo control, valsartan 30 mg/day and LCZ696 60 mg/day. After 4 weeks of therapy, an electrophysiologic study and a dual voltage-calcium optical mapping study were performed. The LCZ696 group had significantly better left ventricular ejection fraction and lower ventricular tachyarrhythmia inducibility than the valsartan and placebo groups. The most common ventricular tachyarrhythmia pattern was 1 or 2 ectopic beats originating from the peri-infarct areas, followed by re-entrant beats surrounding phase singularity points. Compared to the valsartan and placebo groups, the LCZ696 group had significantly shorter action-potential duration, shorter intracellular calcium tau constant, faster conduction velocity, and shorter pacing cycle length to induce arrhythmogenic alternans. LCZ696 therapy reduced the phosphorylated calmodulin-dependent protein kinase II (CaMKII-p) expression. CONCLUSIONS In a rabbit model with chronic MI and HF, LCZ696 therapy ameliorated postinfarct heart function impairment and electrophysiologic remodeling and altered CaMKII-p expression, leading to reduced ventricular tachyarrhythmia inducibility.
Collapse
Affiliation(s)
- Po-Cheng Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou; Chang Gung University College of Medicine, Taoyuan
| | - Hung-Ta Wo
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou
| | - Hui-Ling Lee
- Department of Anesthesia, Chang Gung Memorial Hospital, Taipei
| | - Shien-Fong Lin
- Institute of Biomedical Engineering, National Chiao Tung University, Hsin Chu, Taiwan
| | - Yen Chu
- Division of Thoracic Surgery, Chang Gung Memorial Hospital, Linkou; Chang Gung University College of Medicine, Taoyuan
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou; Chang Gung University College of Medicine, Taoyuan
| | - Chung-Chuan Chou
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou; Chang Gung University College of Medicine, Taoyuan.
| |
Collapse
|
21
|
Wang Y, Liu X, Shi H, Yu Y, Yu Y, Li M, Chen R. NLRP3 inflammasome, an immune-inflammatory target in pathogenesis and treatment of cardiovascular diseases. Clin Transl Med 2020; 10:91-106. [PMID: 32508013 PMCID: PMC7240865 DOI: 10.1002/ctm2.13] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammation is an important process involved in several cardiovascular diseases (CVDs), and nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is a vital player in innate immunity and inflammation. In this review, we aim to provide a comprehensive summary of the current knowledge on the role and involvement of NLRP3 inflammasome in the pathogenesis and treatment of CVDs. NLRP3 inflammasome functions as a molecular platform, and triggers the activation of caspase-1 and cleavage of pro-IL-1β, pro-IL-18, and gasdermin D (GSDMD). Cleaved NT-GSDMD forms pores in the cell membrane and initiates pyroptosis, inducing cell death and release of many intracellular pro-inflammatory molecules. NLRP3 inflammasome activation is triggered via inter-related pathways downstream of K+ efflux, lysosomal disruption, and mitochondrial dysfunction. In addition, the Golgi apparatus and noncoding RNAs are gradually being recognized to play important roles in NLRP3 inflammasome activation. Many investigations have revealed the association between NLRP3 inflammasome and CVDs, including atherosclerosis, ischemia/reperfusion (I/R) injury and heart failure induced by pressure overload or cardiomyopathy. Some existing medications, including orthodox and natural medicines, used for CVD treatment have been newly discovered to act via NLRP3 inflammasome. In addition, NLRP3 inflammasome pathway components such as NLRP3, caspase-1, and IL-1β may be considered as novel therapeutic targets for CVDs. Thus, NLRP3 inflammasome is a key molecule involved in the pathogenesis of CVDs, and further research focused on development of NLRP3 inflammasome-based targeted therapies for CVDs and the clinical evaluation of these therapies is essential.
Collapse
Affiliation(s)
- Yucheng Wang
- Department of CardiologyZhongshan HospitalShanghai Institute of Cardiovascular DiseasesShanghai Medical College of Fudan UniversityShanghaiChina
| | - Xiaoxiao Liu
- Department of CardiologyZhongshan HospitalShanghai Institute of Cardiovascular DiseasesShanghai Medical College of Fudan UniversityShanghaiChina
| | - Hui Shi
- Department of CardiologyZhongshan HospitalShanghai Institute of Cardiovascular DiseasesShanghai Medical College of Fudan UniversityShanghaiChina
| | - Yong Yu
- Department of CardiologyZhongshan HospitalShanghai Institute of Cardiovascular DiseasesShanghai Medical College of Fudan UniversityShanghaiChina
| | - Ying Yu
- Department of General PracticeZhongshan HospitalShanghai Medical College of Fudan UniversityShanghaiChina
| | - Minghui Li
- Department of CardiologyZhongshan HospitalShanghai Institute of Cardiovascular DiseasesShanghai Medical College of Fudan UniversityShanghaiChina
| | - Ruizhen Chen
- Department of CardiologyZhongshan HospitalShanghai Institute of Cardiovascular DiseasesShanghai Medical College of Fudan UniversityShanghaiChina
| |
Collapse
|
22
|
Shemarova IV, Nesterov VP. Molecular Basis of Cardioprotection in Ischemic Heart Disease. J EVOL BIOCHEM PHYS+ 2019. [DOI: 10.1134/s0022093019030013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Bagchi RA, Weeks KL. Histone deacetylases in cardiovascular and metabolic diseases. J Mol Cell Cardiol 2019; 130:151-159. [PMID: 30978343 DOI: 10.1016/j.yjmcc.2019.04.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) regulate gene transcription by catalyzing the removal of acetyl groups from key lysine residues in nucleosomal histones and via the recruitment of other epigenetic regulators to DNA promoter/enhancer regions. Over the past two decades, HDACs have been implicated in multiple processes pertinent to cardiovascular and metabolic diseases, including cardiac hypertrophy and remodeling, fibrosis, calcium handling, inflammation and energy metabolism. The development of small molecule HDAC inhibitors and genetically modified loss- and gain-of-function mouse models has allowed interrogation of the roles of specific HDAC isoforms in these processes. Isoform-selective HDAC inhibitors may prove to be powerful therapeutic agents for the treatment of cardiovascular diseases, obesity and diabetes.
Collapse
Affiliation(s)
- Rushita A Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
24
|
Karppinen S, Hänninen SL, Rapila R, Tavi P. Sarcoplasmic reticulum Ca 2+ -induced Ca 2+ release regulates class IIa HDAC localization in mouse embryonic cardiomyocytes. Physiol Rep 2019; 6. [PMID: 29380950 PMCID: PMC5789715 DOI: 10.14814/phy2.13522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
In embryonic cardiomyocytes, sarcoplasmic reticulum (SR)‐derived Ca2+ release is required to induce Ca2+ oscillations for contraction and to control cardiac development through Ca2+‐activated pathways. Here, our aim was to study how SR Ca2+ release regulates cytosolic and nuclear Ca2+ distribution and the subsequent effects on the Ca2+‐dependent localization of class IIa histone deacetylases (HDAC) and cardiac‐specific gene expression in embryonic cardiomyocytes. Confocal microscopy was used to study changes in Ca2+‐distribution and localization of immunolabeled HDAC4 and HDAC5 upon changes in SR Ca2+ release in mouse embryonic cardiomyocytes. Dynamics of translocation were also observed with a confocal microscope, using HDAC5‐green fluorescent protein transfected myocytes. Expression of class IIa HDACs in differentiating myocytes and changes in cardiac‐specific gene expression were studied using real‐time quantitative PCR. Inhibition of SR Ca2+ release caused a significant decrease in intranuclear Ca2+ concentration, a rapid nuclear import of HDAC5 and subnuclear redistribution of HDAC4. Endogenous localization of HDAC5 and HDAC4 was mostly cytosolic and at the nuclear periphery, respectively. Downregulated expression of cardiac‐specific genes was also observed upon SR Ca2+ release inhibition. Electrical stimulation of sarcolemmal Ca2+ influx was not sufficient to rescue either the HDAC localization or the gene expression changes. SR Ca2+ release controls subcellular Ca2+ distribution and regulates localization of HDAC4 and HDAC5 in embryonic cardiomyocytes. Changes in SR Ca2+ release also caused changes in expression of the developmental phase‐specific genes, which may be due to the changes in HDAC‐localization.
Collapse
Affiliation(s)
- Sari Karppinen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sandra L Hänninen
- Institute of Biomedicine, Department of Physiology and Biocenter Oulu, University of Oulu, Finland
| | - Risto Rapila
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
25
|
Hegyi B, Bers DM, Bossuyt J. CaMKII signaling in heart diseases: Emerging role in diabetic cardiomyopathy. J Mol Cell Cardiol 2019; 127:246-259. [PMID: 30633874 DOI: 10.1016/j.yjmcc.2019.01.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) is upregulated in diabetes and significantly contributes to cardiac remodeling with increased risk of cardiac arrhythmias. Diabetes is frequently associated with atrial fibrillation, coronary artery disease, and heart failure, which may further enhance CaMKII. Activation of CaMKII occurs downstream of neurohormonal stimulation (e.g. via G-protein coupled receptors) and involve various posttranslational modifications including autophosphorylation, oxidation, S-nitrosylation and O-GlcNAcylation. CaMKII signaling regulates diverse cellular processes in a spatiotemporal manner including excitation-contraction and excitation-transcription coupling, mechanics and energetics in cardiac myocytes. Chronic activation of CaMKII results in cellular remodeling and ultimately arrhythmogenic alterations in Ca2+ handling, ion channels, cell-to-cell coupling and metabolism. This review addresses the detrimental effects of the upregulated CaMKII signaling to enhance the arrhythmogenic substrate and trigger mechanisms in the heart. We also briefly summarize preclinical studies using kinase inhibitors and genetically modified mice targeting CaMKII in diabetes. The mechanistic understanding of CaMKII signaling, cardiac remodeling and arrhythmia mechanisms may reveal new therapeutic targets and ultimately better treatment in diabetes and heart disease in general.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
26
|
Hendrickx JO, van Gastel J, Leysen H, Santos-Otte P, Premont RT, Martin B, Maudsley S. GRK5 - A Functional Bridge Between Cardiovascular and Neurodegenerative Disorders. Front Pharmacol 2018; 9:1484. [PMID: 30618771 PMCID: PMC6304357 DOI: 10.3389/fphar.2018.01484] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/03/2018] [Indexed: 12/15/2022] Open
Abstract
Complex aging-triggered disorders are multifactorial programs that comprise a myriad of alterations in interconnected protein networks over a broad range of tissues. It is evident that rather than being randomly organized events, pathophysiologies that possess a strong aging component such as cardiovascular diseases (hypertensions, atherosclerosis, and vascular stiffening) and neurodegenerative conditions (dementia, Alzheimer's disease, mild cognitive impairment, Parkinson's disease), in essence represent a subtly modified version of the intricate molecular programs already in place for normal aging. To control such multidimensional activities there are layers of trophic protein control across these networks mediated by so-called "keystone" proteins. We propose that these "keystones" coordinate and interconnect multiple signaling pathways to control whole somatic activities such as aging-related disease etiology. Given its ability to control multiple receptor sensitivities and its broad protein-protein interactomic nature, we propose that G protein coupled receptor kinase 5 (GRK5) represents one of these key network controllers. Considerable data has emerged, suggesting that GRK5 acts as a bridging factor, allowing signaling regulation in pathophysiological settings to control the connectivity between both the cardiovascular and neurophysiological complications of aging.
Collapse
Affiliation(s)
- Jhana O. Hendrickx
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| | - Jaana van Gastel
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| | - Hanne Leysen
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| | - Paula Santos-Otte
- Institute of Biophysics, Humboldt-Universitat zu Berlin, Berlin, Germany
| | - Richard T. Premont
- Harrington Discovery Institute, Case Western Reserve University, Cleveland, GA, United States
| | - Bronwen Martin
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| |
Collapse
|
27
|
Wood BM, Simon M, Galice S, Alim CC, Ferrero M, Pinna NN, Bers DM, Bossuyt J. Cardiac CaMKII activation promotes rapid translocation to its extra-dyadic targets. J Mol Cell Cardiol 2018; 125:18-28. [PMID: 30321537 PMCID: PMC6279589 DOI: 10.1016/j.yjmcc.2018.10.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 10/03/2018] [Accepted: 10/11/2018] [Indexed: 10/28/2022]
Abstract
Calcium-calmodulin dependent protein kinase IIδ (CaMKIIδ) is an important regulator of cardiac electrophysiology, calcium (Ca) balance, contraction, transcription, arrhythmias and progression to heart failure. CaMKII is readily activated at mouths of dyadic cleft Ca channels, but because of its low Ca-calmodulin affinity and presumed immobility it is less clear how CaMKII gets activated near other known, extra-dyad targets. CaMKII is typically considered to be anchored in cardiomyocytes, but while untested, mobility of active CaMKII could provide a mechanism for broader target phosphorylation in cardiomyocytes. We therefore tested CaMKII mobility and how this is affected by kinase activation in adult rabbit cardiomyocytes. We measured translocation of both endogenous and fluorescence-tagged CaMKII using immunocytochemistry, fluorescence recovery after photobleach (FRAP) and photoactivation of fluorescence. In contrast to the prevailing view that CaMKII is anchored near its myocyte targets, we found CaMKII to be highly mobile in resting myocytes, which was slowed by Ca chelation and accelerated by pacing. At low [Ca], CaMKII was concentrated at Z-lines near the dyad but spread throughout the sarcomere upon pacing. Nuclear exchange of CaMKII was also enhanced upon pacing- and heart failure-induced chronic activation. This mobilization of active CaMKII and its intrinsic memory may allow CaMKII to be activated in high [Ca] regions and then move towards more distant myocyte target sites.
Collapse
Affiliation(s)
- Brent M Wood
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Mitchell Simon
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Samuel Galice
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Chidera C Alim
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Maura Ferrero
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Natalie N Pinna
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| |
Collapse
|
28
|
Cardiac specific PRMT1 ablation causes heart failure through CaMKII dysregulation. Nat Commun 2018; 9:5107. [PMID: 30504773 PMCID: PMC6269446 DOI: 10.1038/s41467-018-07606-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of Ca2+/calmodulin-dependent protein kinase (CaMK)II is closely linked with myocardial hypertrophy and heart failure. However, the mechanisms that regulate CaMKII activity are incompletely understood. Here we show that protein arginine methyltransferase 1 (PRMT1) is essential for preventing cardiac CaMKII hyperactivation. Mice null for cardiac PRMT1 exhibit a rapid progression to dilated cardiomyopathy and heart failure within 2 months, accompanied by cardiomyocyte hypertrophy and fibrosis. Consistently, PRMT1 is downregulated in heart failure patients. PRMT1 depletion in isolated cardiomyocytes evokes hypertrophic responses with elevated remodeling gene expression, while PRMT1 overexpression protects against pathological responses to neurohormones. The level of active CaMKII is significantly elevated in PRMT1-deficient hearts or cardiomyocytes. PRMT1 interacts with and methylates CaMKII at arginine residues 9 and 275, leading to its inhibition. Accordingly, pharmacological inhibition of CaMKII restores contractile function in PRMT1-deficient mice. Thus, our data suggest that PRMT1 is a critical regulator of CaMKII to maintain cardiac function. The mechanisms that regulate the activity of Ca2 +/calmodulin-dependent protein kinase II (CaMKII) in the context of heart failure are incompletely understood. Here the authors show that protein arginine methyltransferase 1 (PRMT1) prevents cardiac hyperactivation of CaMKII and heart failure development by methylating CaMKII at arginine residues 9 and 275.
Collapse
|
29
|
Assessment of PKA and PKC inhibitors on force and kinetics of non-failing and failing human myocardium. Life Sci 2018; 215:119-127. [PMID: 30399377 DOI: 10.1016/j.lfs.2018.10.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023]
Abstract
AIMS Heart failure (HF) is a prevalent disease that is considered the foremost reason for hospitalization in the United States. Most protein kinases (PK) are activated in heart disease and their inhibition has been shown to improve cardiac function in both animal and human studies. However, little is known about the direct impact of PKA and PKC inhibitors on human cardiac contractile function. MATERIAL AND METHODS We investigated the ex vivo effect of such inhibitors on force as well as on kinetics of left ventricular (LV) trabeculae dissected from non-failing and failing human hearts. In these experiments, we applied 0.5 μM of H-89 and GF109203X, which are PKA and PKC inhibitors, respectively, in comparison to their vehicle DMSO (0.05%). KEY FINDINGS AND CONCLUSION Statistical analyses revealed no significant effect for H-89 and GF109203X on either contractile force or kinetics parameters of both non-failing and failing muscles even though they were used at a concentration higher than the reported IC50s and Kis. Therefore, several factors such as selectivity, concentration, and treatment time, which are related to these PK inhibitors according to previous studies require further exploration.
Collapse
|
30
|
Converse role of class I and class IIa HDACs in the progression of atrial fibrillation. J Mol Cell Cardiol 2018; 125:39-49. [PMID: 30321539 DOI: 10.1016/j.yjmcc.2018.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/25/2022]
Abstract
Atrial fibrillation (AF), the most common persistent clinical tachyarrhythmia, is associated with altered gene transcription which underlies cardiomyocyte dysfunction, AF susceptibility and progression. Recent research showed class I and class IIa histone deacetylases (HDACs) to regulate pathological and fetal gene expression, and thereby induce hypertrophy and cardiac contractile dysfunction. Whether class I and class IIa HDACs are involved in AF promotion is unknown. We aim to elucidate the role of class I and class IIa HDACs in tachypacing-induced contractile dysfunction in experimental model systems for AF and clinical AF. METHODS AND RESULTS: Class I and IIa HDACs were overexpressed in HL-1 cardiomyocytes followed by calcium transient (CaT) measurements. Overexpression of class I HDACs, HDAC1 or HDAC3, significantly reduced CaT amplitude in control normal-paced (1 Hz) cardiomyocytes, which was further reduced by tachypacing (5 Hz) in HDAC3 overexpressing cardiomyocytes. HDAC3 inhibition by shRNA or by the specific inhibitor, RGFP966, prevented contractile dysfunction in both tachypaced HL-1 cardiomyocytes and Drosophila prepupae. Conversely, overexpression of class IIa HDACs (HDAC4, HDAC5, HDAC7 or HDAC9) did not affect CaT in controls, with HDAC5 and HDAC7 overexpression even protecting against tachypacing-induced CaT loss. Notably, the protective effect of HDAC5 and HDAC7 was abolished in cardiomyocytes overexpressing a dominant negative HDAC5 or HDAC7 mutant, bearing a mutation in the binding domain for myosin enhancer factor 2 (MEF2). Furthermore, tachypacing induced phosphorylation of HDAC5 and promoted its translocation from the nucleus to cytoplasm, leading to up-regulation of MEF2-related fetal gene expression (β-MHC, BNP). In accord, boosting nuclear localization of HDAC5 by MC1568 or Go6983 attenuated CaT loss in tachypaced HL-1 cardiomyocytes and preserved contractile function in Drosophila prepupae. Findings were expanded to clinical AF. Here, patients with AF showed a significant increase in expression levels and activity of HDAC3, phosphorylated HDAC5 and fetal genes (β-MHC, BNP) in atrial tissue compared to controls in sinus rhythm. CONCLUSION: Class I and class IIa HDACs display converse roles in AF progression. Whereas overexpression of Class I HDAC3 induces cardiomyocyte dysfunction, class IIa HDAC5 overexpression reveals protective properties. Accordingly, HDAC3 inhibitors and HDAC5 nuclear boosters show protection from tachypacing-induced changes and therefore may represent interesting therapeutic options in clinical AF.
Collapse
|
31
|
Activation of CaMKIIδA promotes Ca 2+ leak from the sarcoplasmic reticulum in cardiomyocytes of chronic heart failure rats. Acta Pharmacol Sin 2018; 39:1604-1612. [PMID: 29900930 DOI: 10.1038/aps.2018.20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/19/2018] [Indexed: 12/14/2022]
Abstract
Activation of the Ca2+/calmodulin-dependent protein kinase II isoform δA (CaMKIIδA) disturbs intracellular Ca2+ homeostasis in cardiomyocytes during chronic heart failure (CHF). We hypothesized that upregulation of CaMKIIδA in cardiomyocytes might enhance Ca2+ leak from the sarcoplasmic reticulum (SR) via activation of phosphorylated ryanodine receptor type 2 (P-RyR2) and decrease Ca2+ uptake by inhibition of SR calcium ATPase 2a (SERCA2a). In this study, CHF was induced in rats by ligation of the left anterior descending coronary artery. We found that CHF caused an increase in the expression of CaMKIIδA and P-RyR2 in the left ventricle (LV). The role of CaMKIIδA in regulation of P-RyR2 was elucidated in cardiomyocytes isolated from neonatal rats in vitro. Hypoxia induced upregulation of CaMKIIδA and activation of P-RyR2 in the cardiomyocytes, which both were attenuated by knockdown of CaMKIIδA. Furthermore, we showed that knockdown of CaMKIIδA significantly decreased the Ca2+ leak from the SR elicited by hypoxia in the cardiomyocytes. In addition, CHF also induced a downregulation of SERCA2a in the LV of CHF rats. Knockdown of CaMKIIδA normalized hypoxia-induced downregulation of SERCA2a in cardiomyocytes in vitro. The results demonstrate that the inhibition of CaMKIIδA may improve cardiac function by preventing SR Ca2+ leak through downregulation of P-RyR2 and upregulation of SERCA2a expression in cardiomyocytes in CHF.
Collapse
|
32
|
Yeh ST, Zambrano CM, Koch WJ, Purcell NH. PH domain leucine-rich repeat protein phosphatase 2 (PHLPP2) regulates G-protein-coupled receptor kinase 5 (GRK5)-induced cardiac hypertrophy in vitro. J Biol Chem 2018; 293:8056-8064. [PMID: 29628444 DOI: 10.1074/jbc.m117.809913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/30/2018] [Indexed: 12/20/2022] Open
Abstract
PH domain leucine-rich repeat protein phosphatase (PHLPP) is a serine/threonine phosphatase that has been shown to regulate cell growth and survival through dephosphorylation of several members of the AGC family of kinases. G-protein-coupled receptor kinase 5 (GRK5) is an AGC kinase that regulates phenylephrine (PE)-induced cardiac hypertrophy through its noncanonical function of directly targeting proteins to the nucleus to regulate transcription. Here we investigated the possibility that the PHLPP2 isoform can regulate GRK5-induced cardiomyocyte hypertrophy in neonatal rat ventricular myocytes (NRVMs). We show that removal of PHLPP2 by siRNA induces hypertrophic growth of NRVMs as measured by cell size changes at baseline, potentiated PE-induced cell size changes, and re-expression of fetal genes atrial natriuretic factor and brain natriuretic peptide. Endogenous GRK5 and PHLPP2 were found to interact in NRVMs, and PE-induced nuclear accumulation of GRK5 was enhanced upon down-regulation of PHLPP2. Conversely, overexpression of PHLPP2 blocked PE-induced hypertrophic growth, re-expression of fetal genes, and nuclear accumulation of GRK5, which depended on its phosphatase activity. Finally, using siRNA against GRK5, we found that GRK5 was necessary for the hypertrophic response induced by PHLPP2 knockdown. Our findings demonstrate for the first time a novel regulation of GRK5 by the phosphatase PHLPP2, which modulates hypertrophic growth. Understanding the signaling pathways affected by PHLPP2 has potential for new therapeutic targets in the treatment of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Szu-Tsen Yeh
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Cristina M Zambrano
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California 92093
| | - Walter J Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Nicole H Purcell
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California 92093.
| |
Collapse
|
33
|
Bussey CT, Erickson JR. Physiology and pathology of cardiac CaMKII. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Duran J, Lagos D, Pavez M, Troncoso MF, Ramos S, Barrientos G, Ibarra C, Lavandero S, Estrada M. Ca 2+/Calmodulin-Dependent Protein Kinase II and Androgen Signaling Pathways Modulate MEF2 Activity in Testosterone-Induced Cardiac Myocyte Hypertrophy. Front Pharmacol 2017; 8:604. [PMID: 28955223 PMCID: PMC5601904 DOI: 10.3389/fphar.2017.00604] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/21/2017] [Indexed: 11/25/2022] Open
Abstract
Testosterone is known to induce cardiac hypertrophy through androgen receptor (AR)-dependent and -independent pathways, but the molecular underpinnings of the androgen action remain poorly understood. Previous work has shown that Ca2+/calmodulin-dependent protein kinase II (CaMKII) and myocyte-enhancer factor 2 (MEF2) play key roles in promoting cardiac myocyte growth. In order to gain mechanistic insights into the action of androgens on the heart, we investigated how testosterone affects CaMKII and MEF2 in cardiac myocyte hypertrophy by performing studies on cultured rat cardiac myocytes and hearts obtained from adult male orchiectomized (ORX) rats. In cardiac myocytes, MEF2 activity was monitored using a luciferase reporter plasmid, and the effects of CaMKII and AR signaling pathways on MEF2C were examined by using siRNAs and pharmacological inhibitors targeting these two pathways. In the in vivo studies, ORX rats were randomly assigned to groups that were administered vehicle or testosterone (125 mg⋅kg-1⋅week-1) for 5 weeks, and plasma testosterone concentrations were determined using ELISA. Cardiac hypertrophy was evaluated by measuring well-characterized hypertrophy markers. Moreover, western blotting was used to assess CaMKII and phospholamban (PLN) phosphorylation, and MEF2C and AR protein levels in extracts of left-ventricle tissue from control and testosterone-treated ORX rats. Whereas testosterone treatment increased the phosphorylation levels of CaMKII (Thr286) and phospholambam (PLN) (Thr17) in cardiac myocytes in a time- and concentration-dependent manner, testosterone-induced MEF2 activity and cardiac myocyte hypertrophy were prevented upon inhibition of CaMKII, MEF2C, and AR signaling pathways. Notably, in the hypertrophied hearts obtained from testosterone-administered ORX rats, both CaMKII and PLN phosphorylation levels and AR and MEF2 protein levels were increased. Thus, this study presents the first evidence indicating that testosterone activates MEF2 through CaMKII and AR signaling. Our findings suggest that an orchestrated mechanism of action involving signal transduction and transcription pathways underlies testosterone-induced cardiac myocyte hypertrophy.
Collapse
Affiliation(s)
- Javier Duran
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Daniel Lagos
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Mario Pavez
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Mayarling F Troncoso
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Sebastián Ramos
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Genaro Barrientos
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Cristian Ibarra
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas and Facultad Medicina, Universidad de ChileSantiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, DallasTX, United States
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| |
Collapse
|
35
|
The reduced myofilament responsiveness to calcium contributes to the negative force-frequency relationship in rat cardiomyocytes: role of reactive oxygen species and p-38 map kinase. Pflugers Arch 2017; 469:1663-1673. [DOI: 10.1007/s00424-017-2058-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/11/2017] [Indexed: 01/01/2023]
|
36
|
Cai L, Fan G, Wang F, Liu S, Li T, Cong X, Chun J, Chen X. Protective Role for LPA 3 in Cardiac Hypertrophy Induced by Myocardial Infarction but Not by Isoproterenol. Front Physiol 2017; 8:356. [PMID: 28611684 PMCID: PMC5447740 DOI: 10.3389/fphys.2017.00356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/15/2017] [Indexed: 01/04/2023] Open
Abstract
Background: We previously reported that lysophosphatidic acid (LPA) promoted cardiomyocyte hypertrophy in vitro via one of its G protein-coupled receptor subtypes, LPA3. In this study, we examined the role of LPA3 in cardiac hypertrophy induced by isoproterenol (ISO) and myocardial infarction. Methods:In vitro, neonatal rat cardiomyocytes (NRCMs) were subjected to LPA3 knocked-down, or pretreated with a β-adrenergic receptor (β-AR) antagonist (propranolol) before LPA/ISO treatment. Cardiomyocyte size and hypertrophic gene (ANP, BNP) mRNA levels were determined. In vivo, [Formula: see text] and wild-type mice were implanted subcutaneously with an osmotic mini-pump containing ISO or vehicle for 2 weeks; echocardiography was performed to determine the heart weight/body weight ratio, cardiomyocyte cross-sectional area, and level of ANP mRNA expression. [Formula: see text] and wild-type mice were subjected to permanent coronary artery ligation or sham surgery for 4 weeks; cardiac function, including the degree of hypertrophy and infarction size, was determined. Results:In vitro, we found that knocked-down LPA3 in NRCMs did not attenuate ISO-induced hypertrophy, and propranolol was unable to abolish LPA-induced hypertrophy. In vivo, chronic ISO infusion caused cardiac hypertrophy in wild-type mice, while hypertrophic responses to ISO infusion were not attenuated in [Formula: see text] mice. However, in a myocardial infarction (MI) model, [Formula: see text] mice exhibited reduced cardiac hypertrophy compared to wild-type mice at 4 weeks post-MI, which was associated with reduced cardiac function and increased infarct size. Conclusions: Our data show that LPA3 appears to play a protective role in myocardial hypertrophy post-MI, but does not appear to be involved in the hypertrophy that occurs in response to β-AR stimulation in vivo and in vitro. These results implicate LPA-LPA3 lipid signaling in cardiac hypertrophy occurring after pathological insults like MI, which presents a new variable in β-AR-independent hypertrophy. Thus, modulation of LPA3 signaling might represent a new strategy for preventing the stressed myocardium from ischemia injury.
Collapse
Affiliation(s)
- Lin Cai
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Guangpu Fan
- Cardiovascular Surgery Department, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Fang Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Si Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Tiewei Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Xiangfeng Cong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery InstituteLa Jolla, CA, United States
| | - Xi Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
37
|
Gentzel M, Schambony A. Dishevelled Paralogs in Vertebrate Development: Redundant or Distinct? Front Cell Dev Biol 2017; 5:59. [PMID: 28603713 PMCID: PMC5445114 DOI: 10.3389/fcell.2017.00059] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/12/2017] [Indexed: 01/21/2023] Open
Abstract
Dishevelled (DVL) proteins are highly conserved in the animal kingdom and are important key players in β-Catenin-dependent and -independent Wnt signaling pathways. Vertebrate genomes typically comprise three DVL genes, DVL1, DVL2, and DVL3. Expression patterns and developmental functions of the three vertebrate DVL proteins however, are only partially redundant in any given species. Moreover, expression and function of DVL isoforms have diverged between different vertebrate species. All DVL proteins share basic functionality in Wnt signal transduction. Additional, paralog-specific interactions and functions combined with context-dependent availability of DVL isoforms may play a central role in defining Wnt signaling specificity and add selectivity toward distinct downstream pathways. In this review, we recapitulate briefly cellular functions of DVL paralogs, their role in vertebrate embryonic development and congenital disease.
Collapse
Affiliation(s)
- Marc Gentzel
- Molecular Analysis-Mass Spectrometry, Center for Molecular and Cellular Bioengineering (CMCB), TU DresdenDresden, Germany
| | - Alexandra Schambony
- Developmental Biology, Biology Department, Friedrich-Alexander University Erlangen-NurembergErlangen, Germany
| |
Collapse
|
38
|
Chen J, Wang D, Wang F, Shi S, Chen Y, Yang B, Tang Y, Huang C. Exendin-4 inhibits structural remodeling and improves Ca 2+ homeostasis in rats with heart failure via the GLP-1 receptor through the eNOS/cGMP/PKG pathway. Peptides 2017; 90:69-77. [PMID: 28242257 DOI: 10.1016/j.peptides.2017.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) agonist exendin-4 is a long-acting analog of GLP-1, which stimulates insulin secretion and is clinically used in the treatment of type 2 diabetes. Previous studies have demonstrated that GLP-1 agonists and analogs serve as cardioprotective factors in various conditions. Disturbances in calcium cycling are characteristic of heart failure (HF); therefore, the aim of this study was to investigate the effect of exendin-4 (a GLP-1 mimetic) on the regulation of calcium handling and to identify the underlying mechanisms in an HF rat model after myocardial infarction (MI). Rats underwent surgical ligation of the left anterior descending coronary artery or sham surgery prior to infusion with vehicle, exendin-4, or exendin-4 and exendin9-39 for 4 weeks. Exendin-4 treatment decreased MI size, suppressed chamber dilation, myocyte hypertrophy, and fibrosis and improved in vivo heart function in the rats subjected to MI. Exendin-4 resulted in an increase in circulating GLP-1 and GLP-1R in ventricular tissues. Additionally, exendin-4 activated the eNOS/cGMP/PKG signaling pathway and inhibited the Ca2+/calmodulin-dependent kinase II (CaMKII) pathways. Myocytes isolated from exendin-4-treated hearts displayed higher Ca2+ transients, higher sarcoplasmic reticulum Ca2+ content, and higher l-type Ca2+ current densities than MI hearts. Exendin-4 treatment restored the protein expression of sarcoplasmic reticulum Ca2+ uptake ATPase (SERCA2a), phosphorylated phospholamban (PLB) and Cav1.2 and decreased the levels of phosphorylated ryanodine receptor (RyR). Moreover, the favorable effects of exendin-4 were significantly inhibited by exendin9-39 (a GLP-1 receptor antagonist). Exendin-4 treatment of an HF rat model after MI inhibited cardiac and cardiomyocytes progressive remodeling. In addition, Ca2+ handling and its molecular modulation were also improved by exendin-4 treatment. The beneficial effects of exendin-4 on cardiac remodeling may be mediated through activation of the eNOS/cGMP/PKG pathway.
Collapse
Affiliation(s)
- Jingjing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Dandan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Fangai Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Yuting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
39
|
Wood BM, Bossuyt J. Emergency Spatiotemporal Shift: The Response of Protein Kinase D to Stress Signals in the Cardiovascular System. Front Pharmacol 2017; 8:9. [PMID: 28174535 PMCID: PMC5258689 DOI: 10.3389/fphar.2017.00009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022] Open
Abstract
Protein Kinase D isoforms (PKD 1-3) are key mediators of neurohormonal, oxidative, and metabolic stress signals. PKDs impact a wide variety of signaling pathways and cellular functions including actin dynamics, vesicle trafficking, cell motility, survival, contractility, energy substrate utilization, and gene transcription. PKD activity is also increasingly linked to cancer, immune regulation, pain modulation, memory, angiogenesis, and cardiovascular disease. This increasing complexity and diversity of PKD function, highlights the importance of tight spatiotemporal control of the kinase via protein–protein interactions, post-translational modifications or targeting via scaffolding proteins. In this review, we focus on the spatiotemporal regulation and effects of PKD signaling in response to neurohormonal, oxidant and metabolic signals that have implications for myocardial disease. Precise targeting of these mechanisms will be crucial in the design of PKD-based therapeutic strategies.
Collapse
Affiliation(s)
- Brent M Wood
- Department of Pharmacology, University of California, Davis, Davis CA, USA
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, Davis CA, USA
| |
Collapse
|
40
|
Mitochondria and Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:203-226. [DOI: 10.1007/978-3-319-55330-6_11] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
41
|
Enhanced nucleoplasmic Ca2+ signaling in ventricular myocytes from young hypertensive rats. J Mol Cell Cardiol 2016; 101:58-68. [DOI: 10.1016/j.yjmcc.2016.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 10/31/2016] [Accepted: 11/01/2016] [Indexed: 11/20/2022]
|
42
|
Shimizu I, Minamino T. Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol 2016; 97:245-62. [PMID: 27262674 DOI: 10.1016/j.yjmcc.2016.06.001] [Citation(s) in RCA: 661] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The heart must continuously pump blood to supply the body with oxygen and nutrients. To maintain the high energy consumption required by this role, the heart is equipped with multiple complex biological systems that allow adaptation to changes of systemic demand. The processes of growth (hypertrophy), angiogenesis, and metabolic plasticity are critically involved in maintenance of cardiac homeostasis. Cardiac hypertrophy is classified as physiological when it is associated with normal cardiac function or as pathological when associated with cardiac dysfunction. Physiological hypertrophy of the heart occurs in response to normal growth of children or during pregnancy, as well as in athletes. In contrast, pathological hypertrophy is induced by factors such as prolonged and abnormal hemodynamic stress, due to hypertension, myocardial infarction etc. Pathological hypertrophy is associated with fibrosis, capillary rarefaction, increased production of pro-inflammatory cytokines, and cellular dysfunction (impairment of signaling, suppression of autophagy, and abnormal cardiomyocyte/non-cardiomyocyte interactions), as well as undesirable epigenetic changes, with these complex responses leading to maladaptive cardiac remodeling and heart failure. This review describes the key molecules and cellular responses involved in physiological/pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
43
|
Lkhagva B, Kao YH, Chen YC, Chao TF, Chen SA, Chen YJ. Targeting histone deacetylases: A novel therapeutic strategy for atrial fibrillation. Eur J Pharmacol 2016; 781:250-7. [PMID: 27089819 DOI: 10.1016/j.ejphar.2016.04.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/18/2016] [Accepted: 04/15/2016] [Indexed: 12/28/2022]
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia associated with high mortality and morbidity. Current treatments of AF have limited efficacy and considerable side effects. Histone deacetylases (HDACs) play critical roles in the pathophysiology of cardiovascular diseases and contribute to the genesis of AF. Therefore, HDAC inhibition may prove a novel therapeutic strategy for AF through upstream therapy and modifications of AF electrical and structural remodeling. In this review, we provide an update of the knowledge of the effects of HDACs and HDAC inhibitors on AF, and dissect potential underlying mechanisms.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Tze-Fan Chao
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Ann Chen
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
44
|
Abstract
Calcium (Ca) is a universal second messenger involved in the regulation of various cellular processes, including electrical signaling, contraction, secretion, memory, gene transcription, and cell death. In heart, Ca governs cardiomyocyte contraction, is central in electrophysiological properties, and controls major signaling pathway implicated in gene transcription. How cardiomyocytes decode Ca signal to regulate gene expression without interfering with, or being controlled by, "contractile" Ca that floods the entire cytosol during each heartbeat is still elusive. In this review, we summarize recent findings on nuclear Ca regulation and its downstream signaling in cardiomyocytes. We will address difficulties in reliable quantification of nuclear Ca fluxes and discuss its role in the development and progression of cardiac hypertrophy and heart failure. We also point out key open questions to stimulate future work.
Collapse
|
45
|
Coughlan KA, Valentine RJ, Sudit BS, Allen K, Dagon Y, Kahn BB, Ruderman NB, Saha AK. PKD1 Inhibits AMPKα2 through Phosphorylation of Serine 491 and Impairs Insulin Signaling in Skeletal Muscle Cells. J Biol Chem 2016; 291:5664-5675. [PMID: 26797128 DOI: 10.1074/jbc.m115.696849] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 01/27/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is an energy-sensing enzyme whose activity is inhibited in settings of insulin resistance. Exposure to a high glucose concentration has recently been shown to increase phosphorylation of AMPK at Ser(485/491) of its α1/α2 subunit; however, the mechanism by which it does so is not known. Diacylglycerol (DAG), which is also increased in muscle exposed to high glucose, activates a number of signaling molecules including protein kinase (PK)C and PKD1. We sought to determine whether PKC or PKD1 is involved in inhibition of AMPK by causing Ser(485/491) phosphorylation in skeletal muscle cells. C2C12 myotubes were treated with the PKC/D1 activator phorbol 12-myristate 13-acetate (PMA), which acts as a DAG mimetic. This caused dose- and time-dependent increases in AMPK Ser(485/491) phosphorylation, which was associated with a ∼60% decrease in AMPKα2 activity. Expression of a phosphodefective AMPKα2 mutant (S491A) prevented the PMA-induced reduction in AMPK activity. Serine phosphorylation and inhibition of AMPK activity were partially prevented by the broad PKC inhibitor Gö6983 and fully prevented by the specific PKD1 inhibitor CRT0066101. Genetic knockdown of PKD1 also prevented Ser(485/491) phosphorylation of AMPK. Inhibition of previously identified kinases that phosphorylate AMPK at this site (Akt, S6K, and ERK) did not prevent these events. PMA treatment also caused impairments in insulin-signaling through Akt, which were prevented by PKD1 inhibition. Finally, recombinant PKD1 phosphorylated AMPKα2 at Ser(491) in cell-free conditions. These results identify PKD1 as a novel upstream kinase of AMPKα2 Ser(491) that plays a negative role in insulin signaling in muscle cells.
Collapse
Affiliation(s)
- Kimberly A Coughlan
- From the Endocrinology, Diabetes, and Nutrition Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Rudy J Valentine
- Department of Kinesiology, Iowa State University, Ames, Iowa 50011, and
| | - Bella S Sudit
- From the Endocrinology, Diabetes, and Nutrition Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Katherine Allen
- From the Endocrinology, Diabetes, and Nutrition Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Yossi Dagon
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Neil B Ruderman
- From the Endocrinology, Diabetes, and Nutrition Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Asish K Saha
- From the Endocrinology, Diabetes, and Nutrition Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118,.
| |
Collapse
|
46
|
Promiscuous actions of small molecule inhibitors of the protein kinase D-class IIa HDAC axis in striated muscle. FEBS Lett 2015; 589:1080-8. [PMID: 25816750 DOI: 10.1016/j.febslet.2015.03.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/15/2015] [Accepted: 03/16/2015] [Indexed: 11/23/2022]
Abstract
PKD-mediated phosphorylation of class IIa HDACs frees the MEF2 transcription factor to activate genes that govern muscle differentiation and growth. Studies of the regulation and function of this signaling axis have involved MC1568 and Gö-6976, which are small molecule inhibitors of class IIa HDAC and PKD catalytic activity, respectively. We describe unanticipated effects of these compounds. MC1568 failed to inhibit class IIa HDAC catalytic activity in vitro, and exerted divergent effects on skeletal muscle differentiation compared to a bona fide inhibitor of these HDACs. In cardiomyocytes, Gö-6976 triggered calcium signaling and activated stress-inducible kinases. Based on these findings, caution is warranted when employing MC1568 and Gö-6976 as pharmacological tool compounds to assess functions of class IIa HDACs and PKD.
Collapse
|
47
|
Mattiazzi A, Bassani RA, Escobar AL, Palomeque J, Valverde CA, Vila Petroff M, Bers DM. Chasing cardiac physiology and pathology down the CaMKII cascade. Am J Physiol Heart Circ Physiol 2015; 308:H1177-91. [PMID: 25747749 DOI: 10.1152/ajpheart.00007.2015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/16/2015] [Indexed: 11/22/2022]
Abstract
Calcium dynamics is central in cardiac physiology, as the key event leading to the excitation-contraction coupling (ECC) and relaxation processes. The primary function of Ca(2+) in the heart is the control of mechanical activity developed by the myofibril contractile apparatus. This key role of Ca(2+) signaling explains the subtle and critical control of important events of ECC and relaxation, such as Ca(2+) influx and SR Ca(2+) release and uptake. The multifunctional Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) is a signaling molecule that regulates a diverse array of proteins involved not only in ECC and relaxation but also in cell death, transcriptional activation of hypertrophy, inflammation, and arrhythmias. CaMKII activity is triggered by an increase in intracellular Ca(2+) levels. This activity can be sustained, creating molecular memory after the decline in Ca(2+) concentration, by autophosphorylation of the enzyme, as well as by oxidation, glycosylation, and nitrosylation at different sites of the regulatory domain of the kinase. CaMKII activity is enhanced in several cardiac diseases, altering the signaling pathways by which CaMKII regulates the different fundamental proteins involved in functional and transcriptional cardiac processes. Dysregulation of these pathways constitutes a central mechanism of various cardiac disease phenomena, like apoptosis and necrosis during ischemia/reperfusion injury, digitalis exposure, post-acidosis and heart failure arrhythmias, or cardiac hypertrophy. Here we summarize significant aspects of the molecular physiology of CaMKII and provide a conceptual framework for understanding the role of the CaMKII cascade on Ca(2+) regulation and dysregulation in cardiac health and disease.
Collapse
Affiliation(s)
- Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina;
| | - Rosana A Bassani
- Centro de Engenharia Biomédica, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Ariel L Escobar
- Biological Engineering and Small Scale Technologies, School of Engineering, University of California, Merced, California; and
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Carlos A Valverde
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Martín Vila Petroff
- Centro de Investigaciones Cardiovasculares, The National Scientific and Technical Research Council-La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, California
| |
Collapse
|
48
|
Louch WE, Koivumäki JT, Tavi P. Calcium signalling in developing cardiomyocytes: implications for model systems and disease. J Physiol 2015; 593:1047-63. [PMID: 25641733 PMCID: PMC4358669 DOI: 10.1113/jphysiol.2014.274712] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 12/28/2014] [Indexed: 12/15/2022] Open
Abstract
Adult cardiomyocytes exhibit complex Ca(2+) homeostasis, enabling tight control of contraction and relaxation. This intricate regulatory system develops gradually, with progressive maturation of specialized structures and increasing capacity of Ca(2+) sources and sinks. In this review, we outline current understanding of these developmental processes, and draw parallels to pathophysiological conditions where cardiomyocytes exhibit a striking regression to an immature state of Ca(2+) homeostasis. We further highlight the importance of understanding developmental physiology when employing immature cardiomyocyte models such as cultured neonatal cells and stem cells.
Collapse
Affiliation(s)
- William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo0424, Oslo, Norway
- K. G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo0316, Oslo, Norway
| | - Jussi T Koivumäki
- Simula Research Laboratory, Center for Cardiological Innovation and Center for Biomedical ComputingOslo, Norway
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern FinlandKuopio, Finland
| |
Collapse
|
49
|
Bossuyt J, Bers DM. Assessing GPCR and G protein signaling to the nucleus in live cells using fluorescent biosensors. Methods Mol Biol 2015; 1234:149-159. [PMID: 25304355 DOI: 10.1007/978-1-4939-1755-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
G protein-coupled receptor (GPCR) signaling cascades regulate a wide variety of cellular processes and feature prominently in many cardiovascular pathologies. As such they represent major drug targets and discovering novel aspects of GPCR signaling provide important opportunities to identify additional potential therapeutic approaches to reverse or prevent cardiac remodeling and failure. Monitoring cellular trafficking of signaling components and specific protein kinase activities using fluorescent biosensors has provided key insight into stress/GPCR-induced kinase signaling networks and their effect on cardiac gene expression. Herein we describe the protocols for the expression, visualization (by confocal microscopy), and interpretation of data obtained with such biosensors expressed in adult cardiomyocytes. Our focus is on the cellular trafficking of class II histone deacetylases (i.e., HDAC5) and on the FRET sensor (Camui) for calmodulin-dependent protein kinase II (CaMKII).
Collapse
Affiliation(s)
- Julie Bossuyt
- Department of Pharmacology, University of California, Davis, CA, 95616-8636, USA
| | | |
Collapse
|
50
|
Aguiar CJ, Rocha-Franco JA, Sousa PA, Santos AK, Ladeira M, Rocha-Resende C, Ladeira LO, Resende RR, Botoni FA, Barrouin Melo M, Lima CX, Carballido JM, Cunha TM, Menezes GB, Guatimosim S, Leite MF. Succinate causes pathological cardiomyocyte hypertrophy through GPR91 activation. Cell Commun Signal 2014; 12:78. [PMID: 25539979 PMCID: PMC4296677 DOI: 10.1186/s12964-014-0078-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 11/28/2014] [Indexed: 12/28/2022] Open
Abstract
Background Succinate is an intermediate of the citric acid cycle as well as an extracellular circulating molecule, whose receptor, G protein-coupled receptor-91 (GPR91), was recently identified and characterized in several tissues, including heart. Because some pathological conditions such as ischemia increase succinate blood levels, we investigated the role of this metabolite during a heart ischemic event, using human and rodent models. Results We found that succinate causes cardiac hypertrophy in a GPR91 dependent manner. GPR91 activation triggers the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), the expression of calcium/calmodulin dependent protein kinase IIδ (CaMKIIδ) and the translocation of histone deacetylase 5 (HDAC5) into the cytoplasm, which are hypertrophic-signaling events. Furthermore, we found that serum levels of succinate are increased in patients with cardiac hypertrophy associated with acute and chronic ischemic diseases. Conclusions These results show for the first time that succinate plays an important role in cardiomyocyte hypertrophy through GPR91 activation, and extend our understanding of how ischemia can induce hypertrophic cardiomyopathy. Electronic supplementary material The online version of this article (doi:10.1186/s12964-014-0078-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carla J Aguiar
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - João A Rocha-Franco
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Pedro A Sousa
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Anderson K Santos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Marina Ladeira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Cibele Rocha-Resende
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Luiz O Ladeira
- Department of Physics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Fernando A Botoni
- Department of Medicine, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Marcos Barrouin Melo
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Cristiano X Lima
- Department of Medicine, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - José M Carballido
- Novartis Institutes for Biomedical Research, Basel, CH-4002, Switzerland.
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto, Medical School, University of São Paulo, São Paulo, Brazil.
| | - Gustavo B Menezes
- Department of Morphology, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| | - M Fatima Leite
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG - CEP: 31270-901, Brazil.
| |
Collapse
|