1
|
Ryu G, Ko M, Lee S, Park SI, Choi JW, Lee JY, Kim JY, Kwon HJ. Urolithin A Protects Hepatocytes from Palmitic Acid-Induced ER Stress by Regulating Calcium Homeostasis in the MAM. Biomolecules 2024; 14:1505. [PMID: 39766212 PMCID: PMC11673756 DOI: 10.3390/biom14121505] [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: 10/06/2024] [Revised: 11/01/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
An ellagitannin-derived metabolite, Urolithin A (UA), has emerged as a potential therapeutic agent for metabolic disorders due to its antioxidant, anti-inflammatory, and mitochondrial function-improving properties, but its efficacy in protecting against ER stress remains underexplored. The endoplasmic reticulum (ER) is a cellular organelle involved in protein folding, lipid synthesis, and calcium regulation. Perturbations in these functions can lead to ER stress, which contributes to the development and progression of metabolic disorders such as metabolic-associated fatty liver disease (MAFLD). In this study, we identified a novel target protein of UA and elucidated its mechanism for alleviating palmitic acid (PA)-induced ER stress. Cellular thermal shift assay (CETSA)-LC-MS/MS analysis revealed that UA binds directly to the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA), an important regulator of calcium homeostasis in mitochondria-associated ER membranes (MAMs). As an agonist of SERCA, UA attenuates abnormal calcium fluctuations and ER stress in PA-treated liver cells, thereby contributing to cell survival. The lack of UA activity in SERCA knockdown cells suggests that UA regulates cellular homeostasis through its interaction with SERCA. Collectively, our results demonstrate that UA protects against PA-induced ER stress and enhances cell survival by regulating calcium homeostasis in MAMs through SERCA. This study highlights the potential of UA as a therapeutic agent for metabolic disorders associated with ER stress.
Collapse
Affiliation(s)
- Gayoung Ryu
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Minjeong Ko
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Sooyeon Lee
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Se In Park
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| | - Jin-Woong Choi
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.-W.C.); (J.Y.L.); (J.Y.K.)
| | - Ju Yeon Lee
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.-W.C.); (J.Y.L.); (J.Y.K.)
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.-W.C.); (J.Y.L.); (J.Y.K.)
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Ho Jeong Kwon
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (G.R.); (M.K.); (S.L.); (S.I.P.)
| |
Collapse
|
2
|
Malikides O, Simantirakis E, Zacharis E, Fragkiadakis K, Kochiadakis G, Marketou M. Cardiac Remodeling and Ventricular Pacing: From Genes to Mechanics. Genes (Basel) 2024; 15:671. [PMID: 38927607 PMCID: PMC11203142 DOI: 10.3390/genes15060671] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiac remodeling and ventricular pacing represent intertwined phenomena with profound implications for cardiovascular health and therapeutic interventions. This review explores the intricate relationship between cardiac remodeling and ventricular pacing, spanning from the molecular underpinnings to biomechanical alterations. Beginning with an examination of genetic predispositions and cellular signaling pathways, we delve into the mechanisms driving myocardial structural changes and electrical remodeling in response to pacing stimuli. Insights into the dynamic interplay between pacing strategies and adaptive or maladaptive remodeling processes are synthesized, shedding light on the clinical implications for patients with various cardiovascular pathologies. By bridging the gap between basic science discoveries and clinical translation, this review aims to provide a comprehensive understanding of cardiac remodeling in the context of ventricular pacing, paving the way for future advancements in cardiovascular care.
Collapse
Affiliation(s)
- Onoufrios Malikides
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
| | - Emmanouel Simantirakis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - Evangelos Zacharis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - Konstantinos Fragkiadakis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - George Kochiadakis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - Maria Marketou
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
3
|
Wang W, Tayier B, Guan L, Yan F, Mu Y. Optimization of the cotransfection of SERCA2a and Cx43 genes for myocardial infarction complications. Life Sci 2023; 331:122067. [PMID: 37659592 DOI: 10.1016/j.lfs.2023.122067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
As our previous study showed, the therapeutic effect of two genes (SERCA2a and Cx43) on heart failure after myocardial infarction (MI) was greater than that of single gene (SERCA2a or Cx43) therapy for bone marrow stem cell (BMSC) transplantation. Based on previous research, the aim of this study was to investigate the optimal ratio of codelivery of SERCA2a and Cx43 genes for MI therapy after biotinylated microbubble (BMB) transplantation via ultrasonic-targeted microbubble destruction (UTMD). Forty rats underwent left anterior descending (LAD) ligation and BMSC injection into the infarct and border zones. Four weeks later, the genes SERCA2a and Cx43 were codelivered at different ratios (1:1, 1:2 and 2:1) into the infarcted heart via UTMD. Cardiac mechanoelectrical function was determined at 4 wks after gene delivery, and the hearts of the rats were harvested for measurement of MI size and detection of SERCA2a and Cx43 expression. Q-PCR analysis of the expression of Nkx2.5 and GATA4 in the myocardial infarct zone and measurement of neovascularization in infarcted hearts. After comparing the therapeutic effects of different cogene ratios, the SERCA2a/Cx43-1:2 group showed remarkable cardiac electrical stability and strengthened the role of anti-arrhythmia. In conclusion, the optimum ratio of the SERCA2a/Cx43 gene is 1:2, which is advantageous for maintaining cardiac electrophysiological stability.
Collapse
Affiliation(s)
- Wei Wang
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China; Department of Ultrasound, Urumqi Friendship Hospital, Urumqi, China
| | - Baihetiya Tayier
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Lina Guan
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Yuming Mu
- Department of Echocardiography, Xinjiang Medical University Affiliated First Hospital, Urumqi, China; Xinjiang Key Laboratory of Ultrasound Medicine, Urumqi, China.
| |
Collapse
|
4
|
Hock MT, Teitgen AE, McCabe KJ, Hirakis SP, Huber GA, Regnier M, Amaro RE, McCammon JA, McCulloch AD. Multiscale computational modeling of the effects of 2'-deoxy-ATP on cardiac muscle calcium handling. JOURNAL OF APPLIED PHYSICS 2023; 134:074905. [PMID: 37601331 PMCID: PMC10435275 DOI: 10.1063/5.0157935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023]
Abstract
2'-Deoxy-ATP (dATP), a naturally occurring near analog of ATP, is a well-documented myosin activator that has been shown to increase contractile force, improve pump function, and enhance lusitropy in the heart. Calcium transients in cardiomyocytes with elevated levels of dATP show faster calcium decay compared with cardiomyocytes with basal levels of dATP, but the mechanisms behind this are unknown. Here, we design and utilize a multiscale computational modeling framework to test the hypothesis that dATP acts on the sarcoendoplasmic reticulum calcium-ATPase (SERCA) pump to accelerate calcium re-uptake into the sarcoplasmic reticulum during cardiac relaxation. Gaussian accelerated molecular dynamics simulations of human cardiac SERCA2A in the E1 apo, ATP-bound and dATP-bound states showed that dATP forms more stable contacts in the nucleotide binding pocket of SERCA and leads to increased closure of cytosolic domains. These structural changes ultimately lead to changes in calcium binding, which we assessed using Brownian dynamics simulations. We found that dATP increases calcium association rate constants to SERCA and that dATP binds to apo SERCA more rapidly than ATP. Using a compartmental ordinary differential equation model of human cardiomyocyte excitation-contraction coupling, we found that these increased association rate constants contributed to the accelerated rates of calcium transient decay observed experimentally. This study provides clear mechanistic evidence of enhancements in cardiac SERCA2A pump function due to interactions with dATP.
Collapse
Affiliation(s)
- Marcus T. Hock
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Abigail E. Teitgen
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Kimberly J. McCabe
- Department of Computational Physiology, Simula Resesarch Laboratory, Oslo 0164, Norway
| | - Sophia P. Hirakis
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, USA
| | - Gary A. Huber
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington 98109, USA
| | - Rommie E. Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
5
|
Nikolaienko R, Bovo E, Yuen SL, Treinen LM, Berg K, Aldrich CC, Thomas DD, Cornea RL, Zima AV. New N-aryl-N-alkyl-thiophene-2-carboxamide compound enhances intracellular Ca 2+ dynamics by increasing SERCA2a Ca 2+ pumping. Biophys J 2023; 122:386-396. [PMID: 36463408 PMCID: PMC9892616 DOI: 10.1016/j.bpj.2022.12.002] [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] [Received: 07/12/2022] [Revised: 10/31/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The type 2a sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) plays a central role in the intracellular Ca2+ homeostasis of cardiac myocytes, pumping Ca2+ from the cytoplasm into the sarcoplasmic reticulum (SR) lumen to maintain relaxation (diastole) and prepare for contraction (systole). Diminished SERCA2a function has been reported in several pathological conditions, including heart failure. Therefore, development of new drugs that improve SERCA2a Ca2+ transport is of great clinical significance. In this study, we characterized the effect of a recently identified N-aryl-N-alkyl-thiophene-2-carboxamide (or compound 1) on SERCA2a Ca2+-ATPase and Ca2+ transport activities in cardiac SR vesicles, and on Ca2+ regulation in a HEK293 cell expression system and in mouse ventricular myocytes. We found that compound 1 enhances SERCA2a Ca2+-ATPase and Ca2+ transport in SR vesicles. Fluorescence lifetime measurements of fluorescence resonance energy transfer between SERCA2a and phospholamban indicated that compound 1 interacts with the SERCA-phospholamban complex. Measurement of endoplasmic reticulum Ca2+ dynamics in HEK293 cells expressing human SERCA2a showed that compound 1 increases endoplasmic reticulum Ca2+ load by enhancing SERCA2a-mediated Ca2+ transport. Analysis of cytosolic Ca2+ dynamics in mouse ventricular myocytes revealed that compound 1 increases the action potential-induced Ca2+ transients and SR Ca2+ load, with negligible effects on L-type Ca2+ channels and Na+/Ca2+ exchanger. However, during adrenergic receptor activation, compound 1 did not further increase Ca2+ transients and SR Ca2+ load, but it decreased the propensity toward Ca2+ waves. Suggestive of concurrent desirable effects of compound 1 on RyR2, [3H]-ryanodine binding to cardiac SR vesicles shows a small decrease in nM Ca2+ and a small increase in μM Ca2+. Accordingly, compound 1 slightly decreased Ca2+ sparks in permeabilized myocytes. Thus, this novel compound shows promising characteristics to improve intracellular Ca2+ dynamics in cardiomyocytes that exhibit reduced SERCA2a Ca2+ uptake, as found in failing hearts.
Collapse
Affiliation(s)
- Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Samantha L Yuen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Levy M Treinen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Kaja Berg
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois.
| |
Collapse
|
6
|
Mechanism based therapies enable personalised treatment of hypertrophic cardiomyopathy. Sci Rep 2022; 12:22501. [PMID: 36577774 PMCID: PMC9797561 DOI: 10.1038/s41598-022-26889-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Cardiomyopathies have unresolved genotype-phenotype relationships and lack disease-specific treatments. Here we provide a framework to identify genotype-specific pathomechanisms and therapeutic targets to accelerate the development of precision medicine. We use human cardiac electromechanical in-silico modelling and simulation which we validate with experimental hiPSC-CM data and modelling in combination with clinical biomarkers. We select hypertrophic cardiomyopathy as a challenge for this approach and study genetic variations that mutate proteins of the thick (MYH7R403Q/+) and thin filaments (TNNT2R92Q/+, TNNI3R21C/+) of the cardiac sarcomere. Using in-silico techniques we show that the destabilisation of myosin super relaxation observed in hiPSC-CMs drives disease in virtual cells and ventricles carrying the MYH7R403Q/+ variant, and that secondary effects on thin filament activation are necessary to precipitate slowed relaxation of the cell and diastolic insufficiency in the chamber. In-silico modelling shows that Mavacamten corrects the MYH7R403Q/+ phenotype in agreement with hiPSC-CM experiments. Our in-silico model predicts that the thin filament variants TNNT2R92Q/+ and TNNI3R21C/+ display altered calcium regulation as central pathomechanism, for which Mavacamten provides incomplete salvage, which we have corroborated in TNNT2R92Q/+ and TNNI3R21C/+ hiPSC-CMs. We define the ideal characteristics of a novel thin filament-targeting compound and show its efficacy in-silico. We demonstrate that hybrid human-based hiPSC-CM and in-silico studies accelerate pathomechanism discovery and classification testing, improving clinical interpretation of genetic variants, and directing rational therapeutic targeting and design.
Collapse
|
7
|
Val-Blasco A, Prieto P, Jaén RI, Gil-Fernández M, Pajares M, Domenech N, Terrón V, Tamayo M, Jorge I, Vázquez J, Bueno-Sen A, Vallejo-Cremades MT, Pombo-Otero J, Sanchez-García S, Ruiz-Hurtado G, Gómez AM, Zaragoza C, Crespo-Leiro MG, López-Collazo E, Cuadrado A, Delgado C, Boscá L, Fernández-Velasco M. Specialized Proresolving Mediators Protect Against Experimental Autoimmune Myocarditis by Modulating Ca 2+ Handling and NRF2 Activation. JACC Basic Transl Sci 2022; 7:544-560. [PMID: 35818504 PMCID: PMC9270570 DOI: 10.1016/j.jacbts.2022.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/30/2022]
Abstract
Specialized proresolving mediators and, in particular, 5(S), (6)R, 7-trihydroxyheptanoic acid methyl ester (BML-111) emerge as new therapeutic tools to prevent cardiac dysfunction and deleterious cardiac damage associated with myocarditis progression. The cardioprotective role of BML-111 is mainly caused by the prevention of increased oxidative stress and nuclear factor erythroid-derived 2-like 2 (NRF2) down-regulation induced by myocarditis. At the molecular level, BML-111 activates NRF2 signaling, which prevents sarcoplasmic reticulum-adenosine triphosphatase 2A down-regulation and Ca2+ mishandling, and attenuates the cardiac dysfunction and tissue damage induced by myocarditis.
Collapse
Key Words
- 8OHdG, 8-hydroxy-2'-deoxyguanosine
- BML-111, 5(S), (6)R, 7-trihydroxyheptanoic acid methyl ester
- Ctrl, control
- Cys, cysteine
- EAM, experimental autoimmune myocarditis
- EC, excitation-contraction
- Epi, 15-epi-lipoxin A4
- LXA4, lipoxin A4
- Lut, luteolin
- NRF2
- NRF2, nuclear factor erythroid-derived 2-like 2
- SCR, spontaneous diastolic Ca2+ release
- SERCA2A
- SERCA2A, sarcoplasmic reticulum–adenosine triphosphatase 2A
- SPM, specialized proresolving mediator
- SR, sarcoplasmic reticulum
- Veh, vehicle
- calcium handling
- mRNA, messenger RNA
- myocarditis
- pro-resolving mediators
Collapse
Affiliation(s)
- Almudena Val-Blasco
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
- Signaling and Cardiovascular Pathophysiology, Unite Mixte de Recherche S 1180, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Paris, France
| | - Patricia Prieto
- Pharmacology, Pharmacognosy, and Botany Department, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Rafael Iñigo Jaén
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
| | - Marta Gil-Fernández
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
| | - Marta Pajares
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Nieves Domenech
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidad da Coruña, A Coruña, Spain
| | - Verónica Terrón
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
| | - María Tamayo
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
| | - Inmaculada Jorge
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Jesús Vázquez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Andrea Bueno-Sen
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
| | | | - Jorge Pombo-Otero
- Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidad da Coruña, A Coruña, Spain
| | - Sergio Sanchez-García
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
| | - Gema Ruiz-Hurtado
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiorenal Translational Laboratory, Institute of Research i+12, CIBERCV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ana María Gómez
- Signaling and Cardiovascular Pathophysiology, Unite Mixte de Recherche S 1180, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Paris, France
| | - Carlos Zaragoza
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departamento de Cardiología, Unidad de Investigación Mixta Universidad Francisco de Vitoria, Madrid, Spain
| | - María Generosa Crespo-Leiro
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidad da Coruña, A Coruña, Spain
| | - Eduardo López-Collazo
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Res de Enfermedades Respiratorias, Madrid, Spain
| | - Antonio Cuadrado
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Department of Biochemistry, Faculty of Medicine, UAM, Madrid, Spain
| | - Carmen Delgado
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
| | - Lisardo Boscá
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas, Autonomous University of Madrid (UAM), Madrid, Spain
| | - María Fernández-Velasco
- Innate Immune Response Group, La Paz University Hospital, Instituto de Investigación Biomédica del Hospital La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
8
|
Cleary SR, Fang X, Cho EE, Pribadi MP, Seflova J, Beach JR, Kekenes-Huskey PM, Robia SL. Inhibitory and stimulatory micropeptides preferentially bind to different conformations of the cardiac calcium pump. J Biol Chem 2022; 298:102060. [PMID: 35605666 PMCID: PMC9218510 DOI: 10.1016/j.jbc.2022.102060] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 12/04/2022] Open
Abstract
The ATP-dependent ion pump sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) sequesters Ca2+ in the endoplasmic reticulum to establish a reservoir for cell signaling. Because of its central importance in physiology, the activity of this transporter is tightly controlled via direct interactions with tissue-specific regulatory micropeptides that tune SERCA function to match changing physiological conditions. In the heart, the micropeptide phospholamban (PLB) inhibits SERCA, while dwarf open reading frame (DWORF) stimulates SERCA. These competing interactions determine cardiac performance by modulating the amplitude of Ca2+ signals that drive the contraction/relaxation cycle. We hypothesized that the functions of these peptides may relate to their reciprocal preferences for SERCA binding; SERCA binds PLB more avidly at low cytoplasmic [Ca2+] but binds DWORF better when [Ca2+] is high. In the present study, we demonstrated this opposing Ca2+ sensitivity is due to preferential binding of DWORF and PLB to different intermediate states that SERCA samples during the Ca2+ transport cycle. We show PLB binds best to the SERCA E1-ATP state, which prevails at low [Ca2+]. In contrast, DWORF binds most avidly to E1P and E2P states that are more populated when Ca2+ is elevated. Moreover, FRET microscopy revealed dynamic shifts in SERCA–micropeptide binding equilibria during cellular Ca2+ elevations. A computational model showed that DWORF exaggerates changes in PLB–SERCA binding during the cardiac cycle. These results suggest a mechanistic basis for inhibitory versus stimulatory micropeptide function, as well as a new role for DWORF as a modulator of dynamic oscillations of PLB–SERCA regulatory interactions.
Collapse
Affiliation(s)
- Sean R Cleary
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Xuan Fang
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Ellen E Cho
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Marsha P Pribadi
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Jaroslava Seflova
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Jordan R Beach
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Peter M Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, USA.
| |
Collapse
|
9
|
Changes in Thyroid Hormone Signaling Mediate Cardiac Dysfunction in the Tg197 Mouse Model of Arthritis: Potential Therapeutic Implications. J Clin Med 2021; 10:jcm10235512. [PMID: 34884213 PMCID: PMC8658216 DOI: 10.3390/jcm10235512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Background Rheumatoid Arthritis (RA) patients show a higher risk of heart failure. The present study investigated possible causes of cardiac dysfunction related to thyroid hormone (TH) signaling in a RA mouse model. Methods A TNF-driven mouse model of RA[TghuTNF (Tg197)] was used. Cardiac function was evaluated by echocardiography. SERCA2a and phospholamban protein levels in left ventricle (LV) tissue, thyroid hormone levels in serum, TH receptors in LV and TH-related kinase signaling pathways were measured. T3 hormone was administered in female Tg197 mice. Results We show LV and atrial dilatation with systolic dysfunction in Tg197 animals, accompanied by downregulated SERCA2a. We suggest an interaction of pro-inflammatory and thyroid hormone signaling indicated by increased p38 MAPK and downregulation of TRβ1 receptor in Tg197 hearts. Interestingly, female Tg197 mice showed a worse cardiac phenotype related to reduced T3 levels and Akt activation. T3 supplementation increased Akt activation, restored SERCA2a expression and improved cardiac function in female Tg197 mice. Conclusions TNF overexpression of Tg197 mice results in cardiac dysfunction via p38 MAPK activation and downregulation of TRβ1. Gender-specific reduction in T3 levels could cause the worse cardiac phenotype observed in female mice, while T3 administration improves cardiac function and calcium handling via modified Akt activation.
Collapse
|
10
|
Proteomics of Mouse Heart Ventricles Reveals Mitochondria and Metabolism as Major Targets of a Post-Infarction Short-Acting GLP1Ra-Therapy. Int J Mol Sci 2021; 22:ijms22168711. [PMID: 34445425 PMCID: PMC8395861 DOI: 10.3390/ijms22168711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease is the main cause of death worldwide, making it crucial to search for new therapies to mitigate major adverse cardiac events (MACEs) after a cardiac ischemic episode. Drugs in the class of the glucagon-like peptide-1 receptor agonists (GLP1Ra) have demonstrated benefits for heart function and reduced the incidence of MACE in patients with diabetes. Previously, we demonstrated that a short-acting GLP1Ra known as DMB (2-quinoxalinamine, 6,7-dichloro-N-[1,1-dimethylethyl]-3-[methylsulfonyl]-,6,7-dichloro-2-methylsulfonyl-3-N-tert-butylaminoquinoxaline or compound 2, Sigma) also mitigates adverse postinfarction left ventricular remodeling and cardiac dysfunction in lean mice through activation of parkin-mediated mitophagy following infarction. Here, we combined proteomics with in silico analysis to characterize the range of effects of DMB in vivo throughout the course of early postinfarction remodeling. We demonstrate that the mitochondrion is a key target of DMB and mitochondrial respiration, oxidative phosphorylation and metabolic processes such as glycolysis and fatty acid beta-oxidation are the main biological processes being regulated by this compound in the heart. Moreover, the overexpression of proteins with hub properties identified by protein–protein interaction networks, such as Atp2a2, may also be important to the mechanism of action of DMB. Data are available via ProteomeXchange with identifier PXD027867.
Collapse
|
11
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
12
|
Prakoso D, Tate M, Blasio M, Ritchie R. Adeno-associated viral (AAV) vector-mediated therapeutics for diabetic cardiomyopathy - current and future perspectives. Clin Sci (Lond) 2021; 135:1369-1387. [PMID: 34076247 PMCID: PMC8187922 DOI: 10.1042/cs20210052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Diabetes increases the prevalence of heart failure by 6-8-fold, independent of other comorbidities such as hypertension and coronary artery disease, a phenomenon termed diabetic cardiomyopathy. Several key signalling pathways have been identified that drive the pathological changes associated with diabetes-induced heart failure. This has led to the development of multiple pharmacological agents that are currently available for clinical use. While fairly effective at delaying disease progression, these treatments do not reverse the cardiac damage associated with diabetes. One potential alternative avenue for targeting diabetes-induced heart failure is the use of adeno-associated viral vector (AAV) gene therapy, which has shown great versatility in a multitude of disease settings. AAV gene therapy has the potential to target specific cells or tissues, has a low host immune response and has the possibility to represent a lifelong cure, not possible with current conventional pharmacotherapies. In this review, we will assess the therapeutic potential of AAV gene therapy as a treatment for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Darnel Prakoso
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
| | - Mitchel Tate
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
- Diabetes, Monash University, Clayton, Victoria 3800, Australia
| | - Miles J. De Blasio
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
- Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Rebecca H. Ritchie
- Departments of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Australia
- Diabetes, Monash University, Clayton, Victoria 3800, Australia
- Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
13
|
Hu Y, Zhang C, Zhu H, Wang S, Zhou Y, Zhao J, Xia Y, Li D. Luteolin modulates SERCA2a via Sp1 upregulation to attenuate myocardial ischemia/reperfusion injury in mice. Sci Rep 2020; 10:15407. [PMID: 32958799 PMCID: PMC7506543 DOI: 10.1038/s41598-020-72325-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
The sarco/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) is responsible for calcium transport during excitation-contraction coupling and is essential for maintaining myocardial systolic/diastolic function and intracellular Ca2+ levels. Therefore, it is important to investigate mechanisms whereby luteolin modulates SERCA2a expression to attenuate myocardial ischemia/reperfusion injury. C57BL/6j mice were randomly divided into eight groups. The expression and activity of SERCA2a was measured to assess interactions between the SERCA2a promoter and the Sp1 transcription factor, and the regulatory effects of luteolin. We used serum LDH release, serum cardiac troponin I level, hemodynamic data, myocardial infarction size and apoptosis-related indices to measure SERCA2a cardio-protective effects of luteolin pretreatment. Sp1 binding to SERCA2a promoter under ischemia/reperfusion conditions in the presence or absence of luteolin was analyzed by chromatin immunoprecipitation. Our experimental results indicated that during myocardial ischemia/reperfusion injury, luteolin pretreatment upregulated the expression levels of SERCA2a and Sp1. Sp1 overexpression enhanced the expression of SERCA2a at the transcriptional level. Luteolin pretreatment reversed the expression of SERCA2a through the increased expression of Sp1. Moreover, we demonstrated that luteolin pretreatment appeared to exert myocardial protective effects by upregulating the transcriptional activity of SERCA2a, via Sp1. In conclusion, during myocardial ischemia/reperfusion, Sp1 appeared to downregulate the expression of SERCA2a. Luteolin pretreatment was shown to improve SERCA2a expression via the upregulation of Sp1 to attenuate myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ya Hu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Chengmeng Zhang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Shuai Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yao Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jiaqi Zhao
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yong Xia
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
14
|
Njegic A, Wilson C, Cartwright EJ. Targeting Ca 2 + Handling Proteins for the Treatment of Heart Failure and Arrhythmias. Front Physiol 2020; 11:1068. [PMID: 33013458 PMCID: PMC7498719 DOI: 10.3389/fphys.2020.01068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Diseases of the heart, such as heart failure and cardiac arrhythmias, are a growing socio-economic burden. Calcium (Ca2+) dysregulation is key hallmark of the failing myocardium and has long been touted as a potential therapeutic target in the treatment of a variety of cardiovascular diseases (CVD). In the heart, Ca2+ is essential for maintaining normal cardiac function through the generation of the cardiac action potential and its involvement in excitation contraction coupling. As such, the proteins which regulate Ca2+ cycling and signaling play a vital role in maintaining Ca2+ homeostasis. Changes to the expression levels and function of Ca2+-channels, pumps and associated intracellular handling proteins contribute to altered Ca2+ homeostasis in CVD. The remodeling of Ca2+-handling proteins therefore results in impaired Ca2+ cycling, Ca2+ leak from the sarcoplasmic reticulum and reduced Ca2+ clearance, all of which contributes to increased intracellular Ca2+. Currently, approved treatments for targeting Ca2+ handling dysfunction in CVD are focused on Ca2+ channel blockers. However, whilst Ca2+ channel blockers have been successful in the treatment of some arrhythmic disorders, they are not universally prescribed to heart failure patients owing to their ability to depress cardiac function. Despite the progress in CVD treatments, there remains a clear need for novel therapeutic approaches which are able to reverse pathophysiology associated with heart failure and arrhythmias. Given that heart failure and cardiac arrhythmias are closely associated with altered Ca2+ homeostasis, this review will address the molecular changes to proteins associated with both Ca2+-handling and -signaling; their potential as novel therapeutic targets will be discussed in the context of pre-clinical and, where available, clinical data.
Collapse
Affiliation(s)
- Alexandra Njegic
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
15
|
Simantirakis Ε, Arkolaki E, Kontaraki J, Chlouverakis G, Mavrakis H, Kallergis E, Parthenakis F, Vardas P. The impact of paced QRS duration on the expression of genes related to contractile function of the left ventricle in chronically paced patients from the right ventricular apex. Hellenic J Cardiol 2020; 61:274-278. [DOI: 10.1016/j.hjc.2019.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 10/26/2022] Open
|
16
|
Wang R, Wang M, He S, Sun G, Sun X. Targeting Calcium Homeostasis in Myocardial Ischemia/Reperfusion Injury: An Overview of Regulatory Mechanisms and Therapeutic Reagents. Front Pharmacol 2020; 11:872. [PMID: 32581817 PMCID: PMC7296066 DOI: 10.3389/fphar.2020.00872] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Calcium homeostasis plays an essential role in maintaining excitation–contraction coupling (ECC) in cardiomyocytes, including calcium release, recapture, and storage. Disruption of calcium homeostasis may affect heart function, leading to the development of various heart diseases. Myocardial ischemia/reperfusion (MI/R) injury may occur after revascularization, which is a treatment used in coronary heart disease. MI/R injury is a complex pathological process, and the main cause of increased mortality and disability after treatment of coronary heart disease. However, current methods and drugs for treating MI/R injury are very scarce, not ideal, and have limitations. Studies have shown that MI/R injury can cause calcium overload that can further aggravate MI/R injury. Therefore, we reviewed the effects of critical calcium pathway regulators on MI/R injury and drew an intuitive diagram of the calcium homeostasis pathway. We also summarized and analyzed calcium pathway-related or MI/R drugs under research or marketing by searching Therapeutic Target and PubMed Databases. The data analysis showed that six drugs and corresponding targets are used to treat MI/R injury and involved in calcium signaling pathways. We emphasize the relevance of further detailed investigation of MI/R injury and calcium homeostasis and the therapeutic role of calcium homeostasis in MI/R injury, which bridges basic research and clinical applications of MI/R injury.
Collapse
Affiliation(s)
- Ruiying Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuaibing He
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Laforest B, Dai W, Tyan L, Lazarevic S, Shen KM, Gadek M, Broman MT, Weber CR, Moskowitz IP. Atrial fibrillation risk loci interact to modulate Ca2+-dependent atrial rhythm homeostasis. J Clin Invest 2020; 129:4937-4950. [PMID: 31609246 DOI: 10.1172/jci124231] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/16/2019] [Indexed: 12/30/2022] Open
Abstract
Atrial fibrillation (AF), defined by disorganized atrial cardiac rhythm, is the most prevalent cardiac arrhythmia worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with AF risk, including the cardiogenic transcription factor genes TBX5, GATA4, and NKX2-5. We report that Tbx5 and Gata4 interact with opposite signs for atrial rhythm controls compared with cardiac development. Using mouse genetics, we found that AF pathophysiology caused by Tbx5 haploinsufficiency, including atrial arrhythmia susceptibility, prolonged action potential duration, and ectopic cardiomyocyte depolarizations, were all rescued by Gata4 haploinsufficiency. In contrast, Nkx2-5 haploinsufficiency showed no combinatorial effect. The molecular basis of the TBX5/GATA4 interaction included normalization of intra-cardiomyocyte calcium flux and expression of calcium channel genes Atp2a2 and Ryr2. Furthermore, GATA4 and TBX5 showed antagonistic interactions on an Ryr2 enhancer. Atrial rhythm instability caused by Tbx5 haploinsufficiency was rescued by a decreased dose of phospholamban, a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, consistent with a role for decreased sarcoplasmic reticulum calcium flux in Tbx5-dependent AF susceptibility. This work defines a link between Tbx5 dose, sarcoplasmic reticulum calcium flux, and AF propensity. The unexpected interactions between Tbx5 and Gata4 in atrial rhythm control suggest that evaluating specific interactions between genetic risk loci will be necessary for ascertaining personalized risk from genetic association data.
Collapse
Affiliation(s)
| | | | - Leonid Tyan
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | | | | | - Michael T Broman
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | - Ivan P Moskowitz
- Department of Pediatrics, Pathology, and Human Genetics.,Department of Pathology, and
| |
Collapse
|
18
|
Chen X, Zhang X, Gross S, Houser SR, Soboloff J. Acetylation of SERCA2a, Another Target for Heart Failure Treatment? Circ Res 2020; 124:1285-1287. [PMID: 31021723 DOI: 10.1161/circresaha.119.315017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xiongwen Chen
- From the Cardiovascular Research Center (X.C., X.Z., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Xiaoying Zhang
- From the Cardiovascular Research Center (X.C., X.Z., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Scott Gross
- the Fels Institute for Cancer Research and Molecular Biology (S.G., J.S.), Temple University School of Medicine, Philadelphia, PA
| | - Steven R Houser
- From the Cardiovascular Research Center (X.C., X.Z., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Jonathan Soboloff
- the Fels Institute for Cancer Research and Molecular Biology (S.G., J.S.), Temple University School of Medicine, Philadelphia, PA
| |
Collapse
|
19
|
Locatelli P, Giménez CS, Vega MU, Crottogini A, Belaich MN. Targeting the Cardiomyocyte Cell Cycle for Heart Regeneration. Curr Drug Targets 2018; 20:241-254. [DOI: 10.2174/1389450119666180801122551] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023]
Abstract
Adult mammalian cardiomyocytes (CMs) exhibit limited proliferative capacity, as cell cycle
activity leads to an increase in DNA content, but mitosis and cytokinesis are infrequent. This
makes the heart highly inefficient in replacing with neoformed cardiomyocytes lost contractile cells as
occurs in diseases such as myocardial infarction and dilated cardiomyopathy. Regenerative therapies
based on the implant of stem cells of diverse origin do not warrant engraftment and electromechanical
connection of the new cells with the resident ones, a fundamental condition to restore the physiology
of the cardiac syncytium. Consequently, there is a growing interest in identifying factors playing relevant
roles in the regulation of the CM cell cycle to be targeted in order to induce the resident cardiomyocytes
to divide into daughter cells and thus achieve myocardial regeneration with preservation of
physiologic syncytial performance.
Despite the scientific progress achieved over the last decades, many questions remain unanswered, including
how cardiomyocyte proliferation is regulated during heart development in gestation and neonatal
life. This can reveal unknown cell cycle regulation mechanisms and molecules that may be manipulated
to achieve cardiac self-regeneration.
We hereby revise updated data on CM cell cycle regulation, participating molecules and pathways recently
linked with the cell cycle, as well as experimental therapies involving them.
Collapse
Affiliation(s)
- Paola Locatelli
- Laboratorio de Regeneracion Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingenieria (IMETTYB), Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET) - Universidad Favaloro, Solis 453, Buenos Aires, Argentina
| | - Carlos Sebastián Giménez
- Laboratorio de Regeneracion Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingenieria (IMETTYB), Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET) - Universidad Favaloro, Solis 453, Buenos Aires, Argentina
| | - Martín Uranga Vega
- Laboratorio de Regeneracion Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingenieria (IMETTYB), Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET) - Universidad Favaloro, Solis 453, Buenos Aires, Argentina
| | - Alberto Crottogini
- Laboratorio de Regeneracion Cardiovascular, Instituto de Medicina Traslacional, Trasplante y Bioingenieria (IMETTYB), Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET) - Universidad Favaloro, Solis 453, Buenos Aires, Argentina
| | - Mariano Nicolás Belaich
- Laboratorio de Ingenieria Genetica y Biologia Celular y Molecular, Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET) - Universidad Nacional de Quilmes (UNQ), Roque Saenz Pena 352, Bernal, Buenos Aires, Argentina
| |
Collapse
|
20
|
Liu L, Zhao W, Liu J, Gan Y, Liu L, Tian J. Epigallocatechin-3 gallate prevents pressure overload-induced heart failure by up-regulating SERCA2a via histone acetylation modification in mice. PLoS One 2018; 13:e0205123. [PMID: 30286210 PMCID: PMC6171916 DOI: 10.1371/journal.pone.0205123] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/08/2018] [Indexed: 02/07/2023] Open
Abstract
Heart failure is a common, costly, and potentially fatal condition. The cardiac sarcoplasmic reticulum Ca-ATPase (SERCA2a) plays a critical role in the regulation of cardiac function. Previously, low SERCA2a expression was revealed in mice with heart failure. Epigallocatechin-3-gallate (EGCG) can function as an epigenetic regulator and has been reported to enhance cardiac function. However, the underlying epigenetic regulatory mechanism is still unclear. In this study, we investigated whether EGCG can up-regulate SERCA2a via histone acetylation and play role in preventing heart failure. For this, we generated a mouse model of heart failure by performing a minimally invasive transverse aortic constriction (TAC) operation and used this to test the effects of EGCG. The TAC+EGCG group showed nearly normal cardiac function compared to that in the SHAM group. The expression of SERCA2a was decreased at both the mRNA and protein levels in the TAC group but was enhanced in the TAC+EGCG group. Levels of AcH3 and AcH3K9 were determined to decrease near the promoter region of Atp2a2 (the gene encoding SERCA-2a) in the TAC group, but were elevated in the TAC+EGCG group. Meanwhile, HDAC1 activity and binding near the Atp2a2 promoter were increased in the TAC group but decreased with EGCG addition. Further, binding levels of GATA4 and Mef2c near the Atp2a2 promoter region were reduced in TAC hearts, which might have been caused by histone hypoacetylation; this was reversed by EGCG. Together, upregulation of SERCA2a via the modification of histone acetylation plays a role in EGCG-mediated prevention of pressure overload-induced heart failure, and this might represent a novel pharmacological target for the treatment of heart failure.
Collapse
Affiliation(s)
- Lifei Liu
- Department of Anesthesiology, Children’s Hospital of Chongqing Medical University, Chongqing, PR China
| | - Weian Zhao
- Department of Cardiology, Heart Center, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jianxia Liu
- Department of Anesthesiology, Children’s Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yi Gan
- Ministry of Education Key Laboratory of Child Development and Disorders, PR China
| | - Lingjuan Liu
- Key Laboratory of Pediatrics in Chongqing, Chongqing, PR China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, PR China
| | - Jie Tian
- Department of Cardiology, Heart Center, Children's Hospital of Chongqing Medical University, Chongqing, PR China
- * E-mail:
| |
Collapse
|
21
|
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
|
22
|
Raguimova ON, Smolin N, Bovo E, Bhayani S, Autry JM, Zima AV, Robia SL. Redistribution of SERCA calcium pump conformers during intracellular calcium signaling. J Biol Chem 2018; 293:10843-10856. [PMID: 29764938 PMCID: PMC6052202 DOI: 10.1074/jbc.ra118.002472] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/01/2018] [Indexed: 11/06/2022] Open
Abstract
The conformational changes of a calcium transport ATPase were investigated with molecular dynamics (MD) simulations as well as fluorescence resonance energy transfer (FRET) measurements to determine the significance of a discrete structural element for regulation of the conformational dynamics of the transport cycle. Previous MD simulations indicated that a loop in the cytosolic domain of the SERCA calcium transporter facilitates an open-to-closed structural transition. To investigate the significance of this structural element, we performed additional MD simulations and new biophysical measurements of SERCA structure and function. Rationally designed in silico mutations of three acidic residues of the loop decreased SERCA domain-domain contacts and increased domain-domain separation distances. Principal component analysis of MD simulations suggested decreased sampling of compact conformations upon N-loop mutagenesis. Deficits in headpiece structural dynamics were also detected by measuring intramolecular FRET of a Cer-YFP-SERCA construct (2-color SERCA). Compared with WT, the mutated 2-color SERCA shows a partial FRET response to calcium, whereas retaining full responsiveness to the inhibitor thapsigargin. Functional measurements showed that the mutated transporter still hydrolyzes ATP and transports calcium, but that maximal enzyme activity is reduced while maintaining similar calcium affinity. In live cells, calcium elevations resulted in concomitant FRET changes as the population of WT 2-color SERCA molecules redistributed among intermediates of the transport cycle. Our results provide novel insights on how the population of SERCA pumps responds to dynamic changes in intracellular calcium.
Collapse
Affiliation(s)
- Olga N Raguimova
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Nikolai Smolin
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Elisa Bovo
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Siddharth Bhayani
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Joseph M Autry
- the Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455
| | - Aleksey V Zima
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Seth L Robia
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| |
Collapse
|
23
|
Chen H, Liu S, Zhao C, Zong Z, Ma C, Qi G. Cardiac contractility modulation improves left ventricular systolic function partially via miR-25 mediated SERCA2A expression in rabbit trans aortic constriction heart failure model. J Thorac Dis 2018; 10:3899-3908. [PMID: 30069393 DOI: 10.21037/jtd.2018.06.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The purpose of this study was to investigate the underlying mechanism of cardiac contractility modulation (CCM) in improving trans aortic constriction (TAC)-induced heart failure (HF) left ventricular (LV) systolic function. A total of 25 New Zealand white rabbits were randomly divided into 5 groups: sham operation group (SHM), TAC-induced HF group (HF), TAC-induced HF followed by CCM stimulation group (HF + CCM), TAC-induced HF followed by injection of anti-miR-25 group (HF + anti-miR-25), TAC-induced HF followed by CCM stimulation and AAV9-miR-25 injection group (HF + CCM + miR-25). CCM current was performed 6 hours a day for 4 weeks. The left ventricle ejection fraction (LVEF) was measured by ultrasound. Reverse transcription-polymerase chain reaction (RT-PCR) and Western blot were used for measuring RNA and protein levels. The sarcoplasmic reticulum calcium ATPase (SERCA2A) and LVEF were reduced, while the miR-25 expression was improved in HF group compared to SHM group. Conversely, the SERCA2A and LVEF were improved, and the miR-25 reduced in the HF + CCM and the HF + anti-miR-25 groups compared to the HF group. Moreover, the SERCA2A and LVEF were reduced, while the miR-25 was improved in the HF + CCM + miR-25 group compared to the HF + CCM group. CCM is a potentially effective procedure for improving LV systolic function, which might partially by inhibiting miR-25 expression, further improved SERCA2A expression in TAC HF models.
Collapse
Affiliation(s)
- Hongyun Chen
- Department of Geriatric Cardiology, First Hospital of China Medical University, Shenyang 110001, China
| | - Shuang Liu
- Department of Cardiovascular Ultrasound, First Hospital of China Medical University, Shenyang 110001, China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, First Hospital of China Medical University, Shenyang 110001, China
| | - Zhihong Zong
- Department of Biochemistry, China Medical University, Shenyang 110001, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, First Hospital of China Medical University, Shenyang 110001, China
| | - Guoxian Qi
- Department of Geriatric Cardiology, First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
24
|
Valenzuela V, Jackson KL, Sardi SP, Hetz C. Gene Therapy Strategies to Restore ER Proteostasis in Disease. Mol Ther 2018; 26:1404-1413. [PMID: 29728295 DOI: 10.1016/j.ymthe.2018.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/01/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Proteostasis alterations are proposed as a transversal hallmark of several pathological conditions, including metabolic disorders, mechanical injury, cardiac malfunction, neurodegeneration, and cancer. Strategies to improve proteostasis aim to reduce the accumulation of specific disease-related misfolded proteins or bolster the endogenous mechanisms to fold and degrade abnormal proteins. Endoplasmic reticulum (ER) stress is a common pathological signature of a variety of diseases, which engages the unfolded protein response (UPR) as a cellular reaction to mitigate ER stress. Pharmacological modulation of the UPR is challenging considering the physiological importance of the pathway in various organs. However, local targeting of ER stress responses in the affected tissue using gene therapy is emerging as a possible solution to overcome side effects. The delivery of ER chaperones or active UPR components using adeno-associated virus (AAV) has demonstrated outstanding beneficial effects in several disease models (e.g., neurodegenerative conditions, eye disorders, and metabolic diseases). Here, we discuss current efforts to design and optimize gene therapy strategies to improve ER proteostasis in different disease contexts.
Collapse
Affiliation(s)
- Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Kasey L Jackson
- Neuroscience Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Sergio P Sardi
- Neuroscience Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Abstract
Mitochondrial dysfunction underlines a multitude of pathologies; however, studies are scarce that rescue the mitochondria for cellular resuscitation. Exploration into the protective role of mitochondrial transcription factor A (TFAM) and its mitochondrial functions respective to cardiomyocyte death are in need of further investigation. TFAM is a gene regulator that acts to mitigate calcium mishandling and ROS production by wrapping around mitochondrial DNA (mtDNA) complexes. TFAM's regulatory functions over serca2a, NFAT, and Lon protease contribute to cardiomyocyte stability. Calcium- and ROS-dependent proteases, calpains, and matrix metalloproteinases (MMPs) are abundantly found upregulated in the failing heart. TFAM's regulatory role over ROS production and calcium mishandling leads to further investigation into the cardioprotective role of exogenous TFAM. In an effort to restabilize physiological and contractile activity of cardiomyocytes in HF models, we propose that TFAM-packed exosomes (TFAM-PE) will act therapeutically by mitigating mitochondrial dysfunction. Notably, this is the first mention of exosomal delivery of transcription factors in the literature. Here we elucidate the role of TFAM in mitochondrial rescue and focus on its therapeutic potential.
Collapse
Affiliation(s)
- George H Kunkel
- Department of Physiology and Biophysics, Health Sciences Centre, 1216, School of Medicine, University of Louisville, 500, South Preston Street, Louisville, KY, 40202, USA
| | - Pankaj Chaturvedi
- Department of Physiology and Biophysics, Health Sciences Centre, 1216, School of Medicine, University of Louisville, 500, South Preston Street, Louisville, KY, 40202, USA.
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, Health Sciences Centre, 1216, School of Medicine, University of Louisville, 500, South Preston Street, Louisville, KY, 40202, USA
| |
Collapse
|
26
|
Liu Y, Baumgardt SL, Fang J, Shi Y, Qiao S, Bosnjak ZJ, Vásquez-Vivar J, Xia Z, Warltier DC, Kersten JR, Ge ZD. Transgenic overexpression of GTP cyclohydrolase 1 in cardiomyocytes ameliorates post-infarction cardiac remodeling. Sci Rep 2017; 7:3093. [PMID: 28596578 PMCID: PMC5465102 DOI: 10.1038/s41598-017-03234-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
GTP cyclohydrolase 1 (GCH1) and its product tetrahydrobiopterin play crucial roles in cardiovascular health and disease, yet the exact regulation and role of GCH1 in adverse cardiac remodeling after myocardial infarction are still enigmatic. Here we report that cardiac GCH1 is degraded in remodeled hearts after myocardial infarction, concomitant with increases in the thickness of interventricular septum, interstitial fibrosis, and phosphorylated p38 mitogen-activated protein kinase and decreases in left ventricular anterior wall thickness, cardiac contractility, tetrahydrobiopterin, the dimers of nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and the expression of sarcoplasmic reticulum Ca2+ handling proteins. Intriguingly, transgenic overexpression of GCH1 in cardiomyocytes reduces the thickness of interventricular septum and interstitial fibrosis and increases anterior wall thickness and cardiac contractility after infarction. Moreover, we show that GCH1 overexpression decreases phosphorylated p38 mitogen-activated protein kinase and elevates tetrahydrobiopterin levels, the dimerization and phosphorylation of neuronal nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and sarcoplasmic reticulum Ca2+ handling proteins in post-infarction remodeled hearts. Our results indicate that the pivotal role of GCH1 overexpression in post-infarction cardiac remodeling is attributable to preservation of neuronal nitric oxide synthase and sarcoplasmic reticulum Ca2+ handling proteins, and identify a new therapeutic target for cardiac remodeling after infarction.
Collapse
Affiliation(s)
- Yanan Liu
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Medicine, Columbia University, 630 W. 168th Street, New York, New York, 10032, USA
| | - Shelley L Baumgardt
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, Wisconsin, 53234, USA
| | - Shigang Qiao
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zeljko J Bosnjak
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Physiology, Medical College of Wiscosin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Jeannette Vásquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, People's Republic of China
| | - David C Warltier
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Judy R Kersten
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhi-Dong Ge
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.
| |
Collapse
|
27
|
Helms AS, Alvarado FJ, Yob J, Tang VT, Pagani F, Russell MW, Valdivia HH, Day SM. Genotype-Dependent and -Independent Calcium Signaling Dysregulation in Human Hypertrophic Cardiomyopathy. Circulation 2016; 134:1738-1748. [PMID: 27688314 PMCID: PMC5127749 DOI: 10.1161/circulationaha.115.020086] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 08/05/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Aberrant calcium signaling may contribute to arrhythmias and adverse remodeling in hypertrophic cardiomyopathy (HCM). Mutations in sarcomere genes may distinctly alter calcium handling pathways. METHODS We analyzed gene expression, protein levels, and functional assays for calcium regulatory pathways in human HCM surgical samples with (n=25) and without (n=10) sarcomere mutations compared with control hearts (n=8). RESULTS Gene expression and protein levels for calsequestrin, L-type calcium channel, sodium-calcium exchanger, phospholamban, calcineurin, and calcium/calmodulin-dependent protein kinase type II (CaMKII) were similar in HCM samples compared with controls. CaMKII protein abundance was increased only in sarcomere-mutation HCM (P<0.001). The CaMKII target pT17-phospholamban was 5.5-fold increased only in sarcomere-mutation HCM (P=0.01), as was autophosphorylated CaMKII (P<0.01), suggestive of constitutive activation. Calcineurin (PPP3CB) mRNA was not increased, nor was RCAN1 mRNA level, indicating a lack of calcineurin activation. Furthermore, myocyte enhancer factor 2 and nuclear factor of activated T cell transcription factor activity was not increased in HCM, suggesting that calcineurin pathway activation is not an upstream cause of increased CAMKII protein abundance or activation. SERCA2A mRNA transcript levels were reduced in HCM regardless of genotype, as was sarcoplasmic endoplasmic reticular calcium ATPase 2/phospholamban protein ratio (45% reduced; P=0.03). 45Ca sarcoplasmic endoplasmic reticular calcium ATPaseuptake assay showed reduced uptake velocity in HCM regardless of genotype (P=0.01). The cardiac ryanodine receptor was not altered in transcript, protein, or phosphorylated (pS2808, pS2814) protein abundance, and [3H]ryanodine binding was not different in HCM, consistent with no major modification of the ryanodine receptor. CONCLUSIONS Human HCM demonstrates calcium mishandling through both genotype-specific and common pathways. Posttranslational activation of the CaMKII pathway is specific to sarcomere mutation-positive HCM, whereas sarcoplasmic endoplasmic reticular calcium ATPase 2 abundance and sarcoplasmic reticulum Ca uptake are depressed in both sarcomere mutation-positive and -negative HCM.
Collapse
Affiliation(s)
- Adam S Helms
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor.
| | - Francisco J Alvarado
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| | - Jaime Yob
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| | - Vi T Tang
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| | - Francis Pagani
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| | - Mark W Russell
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| | - Héctor H Valdivia
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| | - Sharlene M Day
- From Departments of Internal Medicine (A.S.H., J.Y., V.T.T., H.H.V., S.M.D.), Molecular and Integrative Physiology (F.J.A., H.H.V., S.M.D.), Cardiac Surgery (F.P.), and Pediatrics (M.W.R.), University of Michigan, Ann Arbor
| |
Collapse
|
28
|
Weber S, Meyer-Roxlau S, El-Armouche A. Role of protein phosphatase inhibitor-1 in cardiac beta adrenergic pathway. J Mol Cell Cardiol 2016; 101:116-126. [PMID: 27639308 DOI: 10.1016/j.yjmcc.2016.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/05/2016] [Accepted: 09/13/2016] [Indexed: 01/08/2023]
Abstract
Phosphoproteomic studies have shown that about one third of all cardiac proteins are reversibly phosphorylated, affecting virtually every cellular signaling pathway. The reversibility of this process is orchestrated by the opposing enzymatic activity of kinases and phosphatases. Conversely, imbalances in subcellular protein phosphorylation patterns are a hallmark of many cardiovascular diseases including heart failure and cardiac arrhythmias. While numerous studies have revealed excessive beta-adrenergic signaling followed by deregulated kinase expression or activity as a major driver of the latter cardiac pathologies, far less is known about the beta-adrenergic regulation of their phosphatase counterparts. In fact, most of the limited knowledge stems from the detailed analysis of the endogenous inhibitor of the protein phosphatase 1 (I-1) in cellular and animal models. I-1 acts as a nodal point between adrenergic and putatively non-adrenergic cardiac signaling pathways and is able to influence widespread cellular functions of protein phosphatase 1 which are contributing to cardiac health and disease, e.g. Ca2+ handling, sarcomere contractility and glucose metabolism. Finally, nearly all of these studies agree that I-1 is a promising drug target on the one hand but the outcome of its pharmacological regulation maybe extremely context-dependent on the other hand, thus warranting for careful interpretation of past and future experimental results. In this respect we will: 1) comprehensively review the current knowledge about structural, functional and regulatory properties of I-1 within the heart 2) highlight current working hypothesis and potential I-1 mediated disease mechanisms 3) discuss state-of-the-art knowledge and future prospects of a potential therapeutic strategy targeting I-1 by restoring the balance of cardiac protein phosphorylation.
Collapse
Affiliation(s)
- Silvio Weber
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany.
| | - Stefanie Meyer-Roxlau
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany.
| |
Collapse
|
29
|
Gan XT, Rajapurohitam V, Xue J, Huang C, Bairwa S, Tang X, Chow JTY, Liu MFW, Chiu F, Sakamoto K, Wagner KU, Karmazyn M. Myocardial Hypertrophic Remodeling and Impaired Left Ventricular Function in Mice with a Cardiac-Specific Deletion of Janus Kinase 2. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 185:3202-10. [PMID: 26475415 DOI: 10.1016/j.ajpath.2015.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/05/2015] [Accepted: 08/18/2015] [Indexed: 12/24/2022]
Abstract
The Janus kinase (JAK) system is involved in numerous cell signaling processes and is highly expressed in cardiac tissue. The JAK isoform JAK2 is activated by numerous factors known to influence cardiac function and pathologic conditions. However, although abundant, the role of JAK2 in the regulation or maintenance of cardiac homeostasis remains poorly understood. Using the Cre-loxP system, we generated a cardiac-specific deletion of Jak2 in the mouse to assess the effect on cardiac function with animals followed up for a 4-month period after birth. These animals had marked mortality during this period, although at 4 months mortality in male mice (47%) was substantially higher compared with female mice (30%). Both male and female cardiac Jak2-deleted mice had hypertrophy, dilated cardiomyopathy, and severe left ventricular dysfunction, including a marked reduction in ejection fractions as assessed by serial echocardiography, although the responses in females were somewhat less severe. Defective cardiac function was associated with altered protein levels of sarcoplasmic reticulum calcium-regulatory proteins particularly in hearts from male mice that had depressed levels of SERCA2 and phosphorylated phospholamban. In contrast, SERCA2 was unchanged in hearts of female mice, whereas phosphorylated phospholamban was increased. Our findings suggest that cardiac JAK2 is critical for maintaining normal heart function, and its ablation produces a severe pathologic phenotype composed of myocardial remodeling, heart failure, and pronounced mortality.
Collapse
Affiliation(s)
- Xiaohong T Gan
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Venkatesh Rajapurohitam
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Jenny Xue
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Cathy Huang
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Suresh Bairwa
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Xilan Tang
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Jeffrey T-Y Chow
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Melissa F W Liu
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Felix Chiu
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Kazuhito Sakamoto
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska
| | - Kay-Uwe Wagner
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska
| | - Morris Karmazyn
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
30
|
Turnbull IC, Eltoukhy AA, Fish KM, Nonnenmacher M, Ishikawa K, Chen J, Hajjar RJ, Anderson DG, Costa KD. Myocardial Delivery of Lipidoid Nanoparticle Carrying modRNA Induces Rapid and Transient Expression. Mol Ther 2016; 24:66-75. [PMID: 26471463 PMCID: PMC4754552 DOI: 10.1038/mt.2015.193] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/07/2015] [Indexed: 12/25/2022] Open
Abstract
Nanoparticle-based delivery of nucleotides offers an alternative to viral vectors for gene therapy. We report highly efficient in vivo delivery of modified mRNA (modRNA) to rat and pig myocardium using formulated lipidoid nanoparticles (FLNP). Direct myocardial injection of FLNP containing 1-10 μg eGFPmodRNA in the rat (n = 3 per group) showed dose-dependent enhanced green fluorescent protein (eGFP) mRNA levels in heart tissue 20 hours after injection, over 60-fold higher than for naked modRNA. Off-target expression, including lung, liver, and spleen, was <10% of that in heart. Expression kinetics after injecting 5 μg FLNP/eGFPmodRNA showed robust expression at 6 hours that reduced by half at 48 hours and was barely detectable at 2 weeks. Intracoronary administration of 10 μg FLNP/eGFPmodRNA also proved successful, although cardiac expression of eGFP mRNA at 20 hours was lower than direct injection, and off-target expression was correspondingly higher. Findings were confirmed in a pilot study in pigs using direct myocardial injection as well as percutaneous intracoronary delivery, in healthy and myocardial infarction models, achieving expression throughout the ventricular wall. Fluorescence microscopy revealed GFP-positive cardiomyocytes in treated hearts. This nanoparticle-enabled approach for highly efficient, rapid and short-term mRNA expression in the heart offers new opportunities to optimize gene therapies for enhancing cardiac function and regeneration.
Collapse
Affiliation(s)
- Irene C Turnbull
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ahmed A Eltoukhy
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kenneth M Fish
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mathieu Nonnenmacher
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jiqiu Chen
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kevin D Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|