1
|
Crispino SP, Segreti A, Nafisio V, Valente D, Crisci F, Ferro A, Cavallari I, Nusca A, Ussia GP, Grigioni F. The Role of SGLT2-Inhibitors Across All Stages of Heart Failure and Mechanisms of Early Clinical Benefit: From Prevention to Advanced Heart Failure. Biomedicines 2025; 13:608. [PMID: 40149587 PMCID: PMC11940307 DOI: 10.3390/biomedicines13030608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i), initially developed as antihyperglycemic agents, have revolutionized heart failure (HF) management, offering substantial benefits across all stages and phenotypes of the disease. Regardless of left ventricular ejection fraction (LVEF), these agents have proven efficacy in both chronic and acute HF presentations. This review explores SGLT2i applications spanning the HF continuum, from early stages (Stage A) in at-risk individuals to the mitigation of progression in advanced HF (Stage D). Evidence from numerous trials has shown that SGLT2i significantly lower rates of HF hospitalization, improve renal function, and decreases cardiovascular mortality, highlighting their multifaced mechanisms of action in HF care. This review also highlights the potential mechanisms by which SGLT2i exert their beneficial effects on the cardiovascular and renal systems, each contributing to early and sustained clinical improvements. However, the integration of SGLT2i into guideline-directed medical therapy poses practical challenges, including initiation timing, dosing, and monitoring, which are addressed to support effective treatment adaptation across patient populations. Ultimately, this review provides a comprehensive assessment of SGLT2i as a foundational therapy in HF, emphasizing their role as an intervention across multiple stages aimed at improving outcomes across the entire HF spectrum.
Collapse
Affiliation(s)
- Simone Pasquale Crispino
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Andrea Segreti
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy
| | - Vincenzo Nafisio
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Daniele Valente
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Filippo Crisci
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Aurora Ferro
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Ilaria Cavallari
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Annunziata Nusca
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Gian Paolo Ussia
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| | - Francesco Grigioni
- Department of Cardiovascular Sciences, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy; (S.P.C.); (V.N.); (D.V.); (F.C.); (A.F.); (I.C.); (A.N.); (G.P.U.); (F.G.)
| |
Collapse
|
2
|
Panday N, Sigdel D, Adam I, Ramirez J, Verma A, Eranki AN, Wang W, Wang D, Ping P. Data-Driven Insights into the Association Between Oxidative Stress and Calcium-Regulating Proteins in Cardiovascular Disease. Antioxidants (Basel) 2024; 13:1420. [PMID: 39594561 PMCID: PMC11590986 DOI: 10.3390/antiox13111420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
A growing body of biomedical literature suggests a bidirectional regulatory relationship between cardiac calcium (Ca2+)-regulating proteins and reactive oxygen species (ROS) that is integral to the pathogenesis of various cardiac disorders via oxidative stress (OS) signaling. To address the challenge of finding hidden connections within the growing volume of biomedical research, we developed a data science pipeline for efficient data extraction, transformation, and loading. Employing the CaseOLAP (Context-Aware Semantic Analytic Processing) algorithm, our pipeline quantifies interactions between 128 human cardiomyocyte Ca2+-regulating proteins and eight cardiovascular disease (CVD) categories. Our machine-learning analysis of CaseOLAP scores reveals that the molecular interfaces of Ca2+-regulating proteins uniquely associate with cardiac arrhythmias and diseases of the cardiac conduction system, distinguishing them from other CVDs. Additionally, a knowledge graph analysis identified 59 of the 128 Ca2+-regulating proteins as involved in OS-related cardiac diseases, with cardiomyopathy emerging as the predominant category. By leveraging a link prediction algorithm, our research illuminates the interactions between Ca2+-regulating proteins, OS, and CVDs. The insights gained from our study provide a deeper understanding of the molecular interplay between cardiac ROS and Ca2+-regulating proteins in the context of CVDs. Such an understanding is essential for the innovation and development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Namuna Panday
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Dibakar Sigdel
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
| | - Irsyad Adam
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Joseph Ramirez
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Aarushi Verma
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Anirudh N. Eranki
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Wei Wang
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA;
- Department of Computational Medicine, University of California, Los Angeles, CA 90095, USA
- Scalable Analytics Institute (ScAi), University of California, Los Angeles, CA 90095, USA
- Department of Bioinformatics and Biomedical Informatics, University of California, Los Angeles, CA 90095, USA
| | - Ding Wang
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
| | - Peipei Ping
- Department of Physiology, School of Medicine, University of California, Los Angeles, CA 90095, USA; (N.P.); (D.S.)
- NHLBI Integrated Cardiovascular Data Science Training Program (iDISCOVER), University of California, Los Angeles, CA 90095, USA; (I.A.); (J.R.); (A.V.); (A.N.E.)
- Scalable Analytics Institute (ScAi), University of California, Los Angeles, CA 90095, USA
- Department of Bioinformatics and Biomedical Informatics, University of California, Los Angeles, CA 90095, USA
- Department of Medicine/Cardiology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Alvarez JAE, Jafri MS, Ullah A. Using a Failing Human Ventricular Cardiomyocyte Model to Re-Evaluate Ca 2+ Cycling, Voltage Dependence, and Spark Characteristics. Biomolecules 2024; 14:1371. [PMID: 39595549 PMCID: PMC11591732 DOI: 10.3390/biom14111371] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Previous studies have observed alterations in excitation-contraction (EC) coupling during end-stage heart failure that include action potential and calcium (Ca2+) transient prolongation and a reduction of the Ca2+ transient amplitude. Underlying these phenomena are the downregulation of potassium (K+) currents, downregulation of the sarcoplasmic reticulum Ca2+ ATPase (SERCA), increase Ca2+ sensitivity of the ryanodine receptor, and the upregulation of the sodium-calcium (Na=-Ca2+) exchanger. However, in human heart failure (HF), debate continues about the relative contributions of the changes in calcium handling vs. the changes in the membrane currents. To understand the consequences of the above changes, they are incorporated into a computational human ventricular myocyte HF model that can explore the contributions of the spontaneous Ca2+ release from the sarcoplasmic reticulum (SR). The reduction of transient outward K+ current (Ito) is the main membrane current contributor to the decrease in RyR2 open probability and L-type calcium channel (LCC) density which emphasizes its importance to phase 1 of the action potential (AP) shape and duration (APD). During current-clamp conditions, RyR2 hyperphosphorylation exhibits the least amount of Ca2+ release from the SR into the cytosol and SR Ca2+ fractional release during a dynamic slow-rapid-slow (0.5-2.5-0.5 Hz) pacing, but it displays the most abundant and more lasting Ca2+ sparks two-fold longer than a normal cell. On the other hand, under voltage-clamp conditions, HF by decreased SERCA and upregulated INCX show the least SR Ca2+ uptake and EC coupling gain, as compared to HF by hyperphosphorylated RyR2s. Overall, this study demonstrates that the (a) combined effect of SERCA and NCX, and the (b) RyR2 dysfunction, along with the downregulation of the cardiomyocyte's potassium currents, could substantially contribute to Ca2+ mishandling at the spark level that leads to heart failure.
Collapse
Affiliation(s)
- Jerome Anthony E. Alvarez
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
- US Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC 20375, USA
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
| |
Collapse
|
4
|
Cheng S, Li H, Hu Y, Jin H, Weng S, He P, Huang H, Liu X, Gu M, Niu H, Cai M, Pei J, Chen L, Ding L, Hua W. Left Bundle Branch Area Pacing With or Without Conduction System Capture in Heart Failure Models. JACC Clin Electrophysiol 2024; 10:2234-2246. [PMID: 38970598 DOI: 10.1016/j.jacep.2024.05.007] [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: 11/09/2023] [Revised: 04/10/2024] [Accepted: 05/01/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Left bundle branch area pacing includes left bundle branch pacing (LBBP) and left ventricular septal pacing (LVSP), which is effective in patients with dyssynchronous heart failure (DHF). However, the basic mechanisms are unknown. OBJECTIVES This study aimed to compare LBBP with LVSP and explore potential mechanisms underlying the better clinical outcomes of LBBP. METHODS A total of 24 beagles were assigned to the following groups: 1) control group; 2) DHF group, left bundle branch ablation followed by 6 weeks of AOO pacing at 200 ppm; 3) LBBP group, DHF for 3 weeks followed by 3 weeks of DOO pacing at 200 ppm; and 4) LVSP with the same interventions in the LBBP group. Metrics of electrocardiogram, echocardiography, hemodynamics, and expression of left ventricular proteins were evaluated. RESULTS Compared with LVSP, LBBP had better peak strain dispersion (44.67 ± 1.75 ms vs 55.50 ± 4.85 ms; P < 0.001) and hemodynamic effect (dP/dtmax improvement: 27.16% ± 7.79% vs 11.37% ± 4.73%; P < 0.001), whereas no significant differences in cardiac function were shown. The altered expressions of proteins in the lateral wall vs septum in the DHF group were partially reversed by LBBP and LVSP, which was associated with the contraction and adhesion process, separately. CONCLUSIONS The animal study demonstrated that LBBP offered better mechanical synchrony and improved hemodynamics than LVSP, which might be explained by the reversed expression of contraction proteins. These results supported the potential superiority of left bundle branch area pacing with the capture of the conduction system in DHF model.
Collapse
Affiliation(s)
- Sijing Cheng
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Li
- Department of Ultrasound, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiran Hu
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology and Macrovascular Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Han Jin
- Cardiology department, Peking University First Hospital, Beijing, China
| | - Sixian Weng
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengkang He
- Cardiology department, Peking University First Hospital, Beijing, China
| | - Hao Huang
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi Liu
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Gu
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongxia Niu
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minsi Cai
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiu Pei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Liang Chen
- Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College. Beijing, China
| | - Ligang Ding
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Hua
- Department of Cardiology, The Cardiac Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Wegener JW, Mitronova GY, ElShareif L, Quentin C, Belov V, Pochechueva T, Hasenfuss G, Ackermann L, Lehnart SE. A dual-targeted drug inhibits cardiac ryanodine receptor Ca 2+ leak but activates SERCA2a Ca 2+ uptake. Life Sci Alliance 2024; 7:e202302278. [PMID: 38012000 PMCID: PMC10681910 DOI: 10.26508/lsa.202302278] [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: 07/17/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
In the heart, genetic or acquired mishandling of diastolic [Ca2+] by ryanodine receptor type 2 (RyR2) overactivity correlates with risks of arrhythmia and sudden cardiac death. Strategies to avoid these risks include decrease of Ca2+ release by drugs modulating RyR2 activity or increase in Ca2+ uptake by drugs modulating SR Ca2+ ATPase (SERCA2a) activity. Here, we combine these strategies by developing experimental compounds that act simultaneously on both processes. Our screening efforts identified the new 1,4-benzothiazepine derivative GM1869 as a promising compound. Consequently, we comparatively studied the effects of the known RyR2 modulators Dantrolene and S36 together with GM1869 on RyR2 and SERCA2a activity in cardiomyocytes from wild type and arrhythmia-susceptible RyR2R2474S/+ mice by confocal live-cell imaging. All drugs reduced RyR2-mediated Ca2+ spark frequency but only GM1869 accelerated SERCA2a-mediated decay of Ca2+ transients in murine and human cardiomyocytes. Our data indicate that S36 and GM1869 are more suitable than dantrolene to directly modulate RyR2 activity, especially in RyR2R2474S/+ mice. Remarkably, GM1869 may represent a new dual-acting lead compound for maintenance of diastolic [Ca2+].
Collapse
Affiliation(s)
- Jörg W Wegener
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gyuzel Y Mitronova
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Lina ElShareif
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
| | - Christine Quentin
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Vladimir Belov
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Tatiana Pochechueva
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Lutz Ackermann
- Georg-August University of Göttingen, Institute of Organic and Biomolecular Chemistry, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Stephan E Lehnart
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
6
|
Xiong H, Zheng Z, Zhao C, Zhao M, Wang Q, Zhang P, Li Y, Zhu Y, Zhu S, Li J. Insight into the underlying molecular mechanism of dilated cardiomyopathy through integrative analysis of data mining, iTRAQ-PRM proteomics and bioinformatics. Proteome Sci 2023; 21:13. [PMID: 37740197 PMCID: PMC10517512 DOI: 10.1186/s12953-023-00214-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND DCM is a common cardiomyopathy worldwide, which is characterized by ventricular dilatation and systolic dysfunction. DCM is one of the most widespread diseases contributing to sudden death and heart failure. However, our understanding of its molecular mechanisms is limited because of its etiology and underlying mechanisms. Hence, this study explored the underlying molecular mechanism of dilated cardiomyopathy through integrative analysis of data mining, iTRAQ-PRM proteomics and bioinformatics METHODS: DCM target genes were downloaded from the public databases. Next, DCM was induced in 20 rats by 8 weeks doxorubicin treatment (2.5 mg/kg/week). We applied isobaric tags for a relative and absolute quantification (iTRAQ) coupled with proteomics approach to identify differentially expressed proteins (DEPs) in myocardial tissue. After association analysis of the DEPs and the key target genes, subsequent analyses, including functional annotation, pathway enrichment, validation, were performed. RESULTS Nine hundred thirty-five genes were identified as key target genes from public databases. Meanwhile, a total of 782 DEPs, including 348 up-regulated and 434 down-regulated proteins, were identified in our animal experiment. The functional annotation of these DEPs revealed complicated molecular mechanisms including TCA cycle, Oxidative phosphorylation, Cardiac muscle contraction. Moreover, the DEPs were analyzed for association with the key target genes screened in the public dataset. We further determined the importance of these three pathways. CONCLUSION Our results demonstrate that TCA cycle, Oxidative phosphorylation, Cardiac muscle contraction played important roles in the detailed molecular mechanisms of DCM.
Collapse
Affiliation(s)
- Hongli Xiong
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Zhe Zheng
- Department of Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, Henan, China
| | - Congcong Zhao
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Minzhu Zhao
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Qi Wang
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Peng Zhang
- Department of Forensic Medicine, Hainan Medical University, Haikou, 571100, China
| | - Yongguo Li
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Ying Zhu
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Shisheng Zhu
- Faculty of Basic Medical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China.
| | - Jianbo Li
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
7
|
Prosperi S, D’Amato A, Severino P, Myftari V, Monosilio S, Marchiori L, Zagordi LM, Filomena D, Di Pietro G, Birtolo LI, Badagliacca R, Mancone M, Maestrini V, Vizza CD. Sizing SGLT2 Inhibitors Up: From a Molecular to a Morpho-Functional Point of View. Int J Mol Sci 2023; 24:13848. [PMID: 37762152 PMCID: PMC10530908 DOI: 10.3390/ijms241813848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), or gliflozins, have recently been shown to reduce cardiovascular death and hospitalization in patients with heart failure, representing a revolutionary therapeutic tool. The purpose of this review is to explore their multifaceted mechanisms of actions, beyond their known glucose reduction power. The cardioprotective effects of gliflozins seem to be linked to the maintenance of cellular homeostasis and to an action on the main metabolic pathways. They improve the oxygen supply for cardiomyocytes with a considerable impact on both functional and morphological myocardial aspects. Moreover, multiple molecular actions of SGLT2i are being discovered, such as the reduction of both inflammation, oxidative stress and cellular apoptosis, all responsible for myocardial damage. Various studies showed controversial results concerning the role of SGLT2i in reverse cardiac remodeling and the lowering of natriuretic peptides, suggesting that their overall effect has yet to be fully understood. In addition to this, advanced imaging studies evaluating the effect on all four cardiac chambers are lacking. Further studies will be needed to better understand the real impact of their administration, their use in daily practice and how they can contribute to benefits in terms of reverse cardiac remodeling.
Collapse
Affiliation(s)
| | - Andrea D’Amato
- Correspondence: ; Tel.: +39-06-49979021; Fax: +39-06-49979060
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Su SA, Zhang Y, Li W, Xi Y, Lu Y, Shen J, Ma Y, Wang Y, Shen Y, Xie L, Ma H, Xie Y, Xiang M. Cardiac Piezo1 Exacerbates Lethal Ventricular Arrhythmogenesis by Linking Mechanical Stress with Ca 2+ Handling After Myocardial Infarction. RESEARCH (WASHINGTON, D.C.) 2023; 6:0165. [PMID: 37303604 PMCID: PMC10255393 DOI: 10.34133/research.0165] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/17/2023] [Indexed: 06/13/2023]
Abstract
Ventricular arrhythmogenesis is a key cause of sudden cardiac death following myocardial infarction (MI). Accumulating data show that ischemia, sympathetic activation, and inflammation contribute to arrhythmogenesis. However, the role and mechanisms of abnormal mechanical stress in ventricular arrhythmia following MI remain undefined. We aimed to examine the impact of increased mechanical stress and identify the role of the key sensor Piezo1 in ventricular arrhythmogenesis in MI. Concomitant with increased ventricular pressure, Piezo1, as a newly recognized mechano-sensitive cation channel, was the most up-regulated mechanosensor in the myocardium of patients with advanced heart failure. Piezo1 was mainly located at the intercalated discs and T-tubules of cardiomyocytes, which are responsible for intracellular calcium homeostasis and intercellular communication. Cardiomyocyte-conditional Piezo1 knockout mice (Piezo1Cko) exhibited preserved cardiac function after MI. Piezo1Cko mice also displayed a dramatically decreased mortality in response to the programmed electrical stimulation after MI with a markedly reduced incidence of ventricular tachycardia. In contrast, activation of Piezo1 in mouse myocardium increased the electrical instability as indicated by prolonged QT interval and sagging ST segment. Mechanistically, Piezo1 impaired intracellular calcium cycling dynamics by mediating the intracellular Ca2+ overload and increasing the activation of Ca2+-modulated signaling, CaMKII, and calpain, which led to the enhancement of phosphorylation of RyR2 and further increment of Ca2+ leaking, finally provoking cardiac arrhythmias. Furthermore, in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), Piezo1 activation remarkably triggered cellular arrhythmogenic remodeling by significantly shortening the duration of the action potential, inducing early afterdepolarization, and enhancing triggered activity.This study uncovered a proarrhythmic role of Piezo1 during cardiac remodeling, which is achieved by regulating Ca2+ handling, implying a promising therapeutic target in sudden cardiac death and heart failure.
Collapse
Affiliation(s)
- Sheng-an Su
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yuhao Zhang
- Department of Endocrinology, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wudi Li
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yutao Xi
- Texas Heart Institute, Houston, TX 77030, USA
| | - Yunrui Lu
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jian Shen
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yuankun Ma
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yaping Wang
- Department of Endocrinology, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yimin Shen
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lan Xie
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Hong Ma
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yao Xie
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Meixiang Xiang
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
9
|
Keefe JA, Moore OM, Ho KS, Wehrens XHT. Role of Ca 2+ in healthy and pathologic cardiac function: from normal excitation-contraction coupling to mutations that cause inherited arrhythmia. Arch Toxicol 2023; 97:73-92. [PMID: 36214829 PMCID: PMC10122835 DOI: 10.1007/s00204-022-03385-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/15/2022] [Indexed: 01/19/2023]
Abstract
Calcium (Ca2+) ions are a key second messenger involved in the rhythmic excitation and contraction of cardiomyocytes throughout the heart. Proper function of Ca2+-handling proteins is required for healthy cardiac function, whereas disruption in any of these can cause cardiac arrhythmias. This comprehensive review provides a broad overview of the roles of Ca2+-handling proteins and their regulators in healthy cardiac function and the mechanisms by which mutations in these proteins contribute to inherited arrhythmias. Major Ca2+ channels and Ca2+-sensitive regulatory proteins involved in cardiac excitation-contraction coupling are discussed, with special emphasis on the function of the RyR2 macromolecular complex. Inherited arrhythmia disorders including catecholaminergic polymorphic ventricular tachycardia, long QT syndrome, Brugada syndrome, short QT syndrome, and arrhythmogenic right-ventricular cardiomyopathy are discussed with particular emphasis on subtypes caused by mutations in Ca2+-handling proteins.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Oliver M Moore
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin S Ho
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA. .,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Rizwan T, Kothidar A, Meghwani H, Sharma V, Shobhawat R, Saini R, Vaishnav HK, Singh V, Pratap M, Sihag H, Kumar S, Dey JK, Dey SK. Comparative analysis of SARS-CoV-2 envelope viroporin mutations from COVID-19 deceased and surviving patients revealed implications on its ion-channel activities and correlation with patient mortality. J Biomol Struct Dyn 2022; 40:10454-10469. [PMID: 34229570 DOI: 10.1080/07391102.2021.1944319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One major obstacle in designing a successful therapeutic regimen to combat COVID-19 pandemic is the frequent occurrence of mutations in the SARS-CoV-2 resulting in patient to patient variations. Out of the four structural proteins of SARS-CoV-2 namely, spike, envelope, nucleocapsid and membrane, envelope protein governs the virus pathogenicity and induction of acute-respiratory-distress-syndrome which is the major cause of death in COVID-19 patients. These effects are facilitated by the viroporin (ion-channel) like activities of the envelope protein. Our current work reports metagenomic analysis of envelope protein at the amino acid sequence level through mining all the available SARS-CoV-2 genomes from the GISAID and coronapp servers. We found majority of mutations in envelope protein were localized at or near PDZ binding motif. Our analysis also demonstrates that the acquired mutations might have important implications on its structure and ion-channel activity. A statistical correlation between specific mutations (e.g. F4F, R69I, P71L, L73F) with patient mortalities were also observed, based on the patient data available for 18,691 SARS-CoV-2-genomes in the GISAID database till 30 April 2021. Albeit, whether these mutations exist as the cause or the effect of co-infections and/or co-morbid disorders within COVID-19 patients is still unclear. Moreover, most of the current vaccine and therapeutic interventions are revolving around spike protein. However, emphasizing on envelope protein's (1) conserved epitopes, (2) pathogenicity attenuating mutations, and (3) mutations present in the deceased patients, as reported in our present study, new directions to the ongoing efforts of therapeutic developments against COVID-19 can be achieved by targeting envelope viroporin.
Collapse
Affiliation(s)
- Tayyeba Rizwan
- Department of Biochemistry, University of Delhi South Campus, New Delhi, Delhi, India
| | - Akansha Kothidar
- Centre for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Himanshu Meghwani
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Vaibhav Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Rahul Shobhawat
- Department of Bioscience and Bioengineering, Indian Institute of Technology-Bombay, Mumbai, Maharashtra, India
| | - Rajpal Saini
- Department of Statistics, Faculty of Mathematical Sciences, University of Delhi, New Delhi, Delhi, India
| | - Hemendra Kumar Vaishnav
- Operations Management, Quantitative Methods and Information Systems Area, Indian Institute of Management Udaipur, Udaipur, Rajasthan, India
| | - Vikramaditya Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, Delhi, India
| | - Mukut Pratap
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Hitaishi Sihag
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Shakti Kumar
- Centre for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of AYUSH, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, Delhi, India
| |
Collapse
|
11
|
Zheng J, Dooge HC, Pérez-Hernández M, Zhao YT, Chen X, Hernandez JJ, Valdivia CR, Palomeque J, Rothenberg E, Delmar M, Valdivia HH, Alvarado FJ. Preserved cardiac performance and adrenergic response in a rabbit model with decreased ryanodine receptor 2 expression. J Mol Cell Cardiol 2022; 167:118-128. [PMID: 35413295 PMCID: PMC9610860 DOI: 10.1016/j.yjmcc.2022.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/11/2022] [Accepted: 04/06/2022] [Indexed: 11/19/2022]
Abstract
Ryanodine receptor 2 (RyR2) is an ion channel in the heart responsible for releasing into the cytosol most of the Ca2+ required for contraction. Proper regulation of RyR2 is critical, as highlighted by the association between channel dysfunction and cardiac arrhythmia. Lower RyR2 expression is also observed in some forms of heart disease; however, there is limited information on the impact of this change on excitation-contraction (e-c) coupling, Ca2+-dependent arrhythmias, and cardiac performance. We used a constitutive knock-out of RyR2 in rabbits (RyR2-KO) to assess the extent to which a stable decrease in RyR2 expression modulates Ca2+ handling in the heart. We found that homozygous knock-out of RyR2 in rabbits is embryonic lethal. Remarkably, heterozygotes (KO+/-) show ~50% loss of RyR2 protein without developing an overt phenotype at the intact animal and whole heart levels. Instead, we found that KO+/- myocytes show (1) remodeling of RyR2 clusters, favoring smaller groups in which channels are more densely arranged; (2) lower Ca2+ spark frequency and amplitude; (3) slower rate of Ca2+ release and mild but significant desynchronization of the Ca2+ transient; and (4) a significant decrease in the basal phosphorylation of S2031, likely due to increased association between RyR2 and PP2A. Our data show that RyR2 deficiency, although remarkable at the molecular and subcellular level, has only a modest impact on global Ca2+ release and is fully compensated at the whole-heart level. This highlights the redundancy of RyR2 protein expression and the plasticity of the e-c coupling apparatus.
Collapse
Affiliation(s)
- Jingjing Zheng
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Holly C Dooge
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Marta Pérez-Hernández
- Leon H Charney Division of Cardiology, New York University Grossman School of Medicine,. New York, NY, United States of America
| | - Yan-Ting Zhao
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, United States of America
| | - Xi Chen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Jonathan J Hernandez
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States of America
| | - Carmen R Valdivia
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Ciencias Médicas, UNLP, La Plata, Argentina
| | - Eli Rothenberg
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Mario Delmar
- Leon H Charney Division of Cardiology, New York University Grossman School of Medicine,. New York, NY, United States of America
| | - Héctor H Valdivia
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Francisco J Alvarado
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
12
|
Non-Coding RNAs in the Therapeutic Landscape of Pathological Cardiac Hypertrophy. Cells 2022; 11:cells11111805. [PMID: 35681500 PMCID: PMC9180404 DOI: 10.3390/cells11111805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are a major health problem, and long-term survival for people diagnosed with heart failure is, still, unrealistic. Pathological cardiac hypertrophy largely contributes to morbidity and mortality, as effective therapeutic approaches are lacking. Non-coding RNAs (ncRNAs) arise as active regulators of the signaling pathways and mechanisms that govern this pathology, and their therapeutic potential has received great attention in the last decades. Preclinical studies in large animal models have been successful in ameliorating cardiac hypertrophy, and an antisense drug for the treatment of heart failure has, already, entered clinical trials. In this review, we provide an overview of the molecular mechanisms underlying cardiac hypertrophy, the involvement of ncRNAs, and the current therapeutic landscape of oligonucleotides targeting these regulators. Strategies to improve the delivery of such therapeutics and overcome the actual challenges are, also, defined and discussed. With the fast advance in the improvement of oligonucleotide drug delivery, the inclusion of ncRNAs-targeting therapies for cardiac hypertrophy seems, increasingly, a closer reality.
Collapse
|
13
|
Bhullar S, Shah A, Dhalla N. Mechanisms for the development of heart failure and improvement of cardiac function by angiotensin-converting enzyme inhibitors. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are well-known for the treatments of cardiovascular diseases, such as heart failure, hypertension and acute coronary syndrome. Several of these inhibitors including captopril, enalapril, ramipril, zofenopril and imidapril attenuate vasoconstriction, cardiac hypertrophy and adverse cardiac remodeling, improve clinical outcomes in patients with cardiac dysfunction and decrease mortality. Extensive experimental and clinical research over the past 35 years has revealed that the beneficial effects of ACE inhibitors in heart failure are associated with full or partial prevention of adverse cardiac remodeling. Since cardiac function is mainly determined by coordinated activities of different subcellular organelles, including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils, for regulating the intracellular concentration of Ca2+ and myocardial metabolism, there is ample evidence to suggest that adverse cardiac remodelling and cardiac dysfunction in the failing heart are the consequence of subcellular defects. In fact, the improvement of cardiac function by different ACE inhibitors has been demonstrated to be related to the attenuation of abnormalities in subcellular organelles for Ca2+-handling, metabolic alterations, signal transduction defects and gene expression changes in failing cardiomyocytes. Various ACE inhibitors have also been shown to delay the progression of heart failure by reducing the formation of angiotensin II, the development of oxidative stress, the level of inflammatory cytokines and the occurrence of subcellular defects. These observations support the view that ACE inhibitors improve cardiac function in the failing heart by multiple mechanisms including the reduction of oxidative stress, myocardial inflammation and Ca2+-handling abnormalities in cardiomyocytes.
Collapse
|
14
|
Du C, Chen X, Su Q, Lu W, Wang Q, Yuan H, Zhang Z, Wang X, Wu H, Qi Y. The Function of SUMOylation and Its Critical Roles in Cardiovascular Diseases and Potential Clinical Implications. Int J Mol Sci 2021; 22:10618. [PMID: 34638970 PMCID: PMC8509021 DOI: 10.3390/ijms221910618] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular disease (CVD) is a common disease caused by many factors, including atherosclerosis, congenital heart disease, heart failure, and ischemic cardiomyopathy. CVD has been regarded as one of the most common diseases and has a severe impact on the life quality of patients. The main features of CVD include high morbidity and mortality, which seriously threaten human health. SUMO proteins covalently conjugate lysine residues with a large number of substrate proteins, and SUMOylation regulates the function of target proteins and participates in cellular activities. Under certain pathological conditions, SUMOylation of proteins related to cardiovascular development and function are greatly changed. Numerous studies have suggested that SUMOylation of substrates plays critical roles in normal cardiovascular development and function. We reviewed the research progress of SUMOylation in cardiovascular development and function, and the regulation of protein SUMOylation may be applied as a potential therapeutic strategy for CVD treatment.
Collapse
Affiliation(s)
- Congcong Du
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Xu Chen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Qi Su
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Wenbin Lu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Qiqi Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Hong Yuan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Zhenzhen Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Xiaotong Wang
- School of Agriculture, Ludong University, Yantai 246011, China;
| | - Hongmei Wu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| | - Yitao Qi
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an 710119, China; (C.D.); (X.C.); (Q.S.); (W.L.); (Q.W.); (H.Y.); (Z.Z.)
| |
Collapse
|
15
|
Bhullar SK, Shah AK, Dhalla NS. Role of angiotensin II in the development of subcellular remodeling
in heart failure. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of heart failure under various pathological conditions such as myocardial infarction (MI), hypertension and diabetes are accompanied by adverse cardiac remodeling and cardiac dysfunction. Since heart function is mainly determined by coordinated activities of different subcellular organelles including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils for regulating the intracellular concentration of Ca2+, it has been suggested that the occurrence of heart failure is a consequence of subcellular remodeling, metabolic alterations and Ca2+-handling abnormalities in cardiomyocytes. Because of the elevated plasma levels of angiotensin II (ANG II) due to activation of the renin-angiotensin system (RAS) in heart failure, we have evaluated the effectiveness of treatments with angiotensin converting enzyme (ACE) inhibitors and ANG II type 1 receptor (AT1R) antagonists in different experimental models of heart failure. Attenuation of marked alterations in subcellular activities, protein content and gene expression were associated with improvement in cardiac function in MI-induced heart failure by treatment with enalapril (an ACE inhibitor) or losartan (an AT1R antagonist). Similar beneficial effects of ANG II blockade on subcellular remodeling and cardiac performance were also observed in failing hearts due to pressure overload, volume overload or chronic diabetes. Treatments with enalapril and losartan were seen to reduce the degree of RAS activation as well as the level of oxidative stress in failing hearts. These observations provide evidence which further substantiate to support the view that activation of RAS and high level of plasma ANG II play a critical role in inducing subcellular defects and cardiac dys-function during the progression of heart failure.
Collapse
Affiliation(s)
- Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada
| | - Anureet K. Shah
- School of Kinesiology, Nutrition and Food Science, California State University, Los Angeles, CA 90032, USA
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada; Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 3P5, Canada
| |
Collapse
|
16
|
Abstract
Heart failure (HF) continues to be a serious public health challenge despite significant advancements in therapeutics and is often complicated by multiple other comorbidities. Of particular concern is type 2 diabetes mellitus (T2DM) which not only amplifies the risk, but also limits the treatment options available to patients. The sodium-glucose linked cotransporter subtype 2 (SGLT2)-inhibitor class, which was initially developed as a treatment for T2DM, has shown great promise in reducing cardiovascular risk, particularly around HF outcomes - regardless of diabetes status.There are ongoing efforts to elucidate the true mechanism of action of this novel drug class. Its primary mechanism of inducing glycosuria and diuresis from receptor blockade in the renal nephron seems unlikely to be responsible for the rapid and striking benefits seen in clinical trials. Early mechanistic work around conventional therapeutic targets seem to be inconclusive. There are some emerging theories around its effect on myocardial energetics and calcium balance as well as on renal physiology. In this review, we discuss some of the cutting-edge hypotheses and concepts currently being explored around this drug class in an attempt better understand the molecular mechanics of this novel agent.
Collapse
Affiliation(s)
- Amir Fathi
- Department of Neuroanaesthesia and Critical Care, National Hospital for Neurology and Neurosurgery, University College London, London, UK
| | - Keeran Vickneson
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Jagdeep S Singh
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK.
- Department of Cardiology, The Edinburgh Heart Center, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, EH16 4SA, UK.
| |
Collapse
|
17
|
Teixeira VP, Miranda K, Scalzo S, Rocha-Resende C, Silva MM, Tezini GCSV, Melo MB, Souza-Neto FP, Silva KSC, Jesus ICG, Santos AK, de Oliveira M, Szawka RE, Salgado HC, Prado MAM, Poletini MO, Guatimosim S. Increased cholinergic activity under conditions of low estrogen leads to adverse cardiac remodeling. Am J Physiol Cell Physiol 2021; 320:C602-C612. [PMID: 33296286 DOI: 10.1152/ajpcell.00142.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Cholinesterase inhibitors are used in postmenopausal women for the treatment of neurodegenerative diseases. Despite their widespread use in the clinical practice, little is known about the impact of augmented cholinergic signaling on cardiac function under reduced estrogen conditions. To address this gap, we subjected a genetically engineered murine model of systemic vesicular acetylcholine transporter overexpression (Chat-ChR2) to ovariectomy and evaluated cardiac parameters. Left-ventricular function was similar between Chat-ChR2 and wild-type (WT) mice. Following ovariectomy, WT mice showed signs of cardiac hypertrophy. Conversely, ovariectomized (OVX) Chat-ChR2 mice evolved to cardiac dilation and failure. Transcript levels for cardiac stress markers atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) were similarly upregulated in WT/OVX and Chat-ChR2/OVX mice. 17β-Estradiol (E2) treatment normalized cardiac parameters in Chat-ChR2/OVX to the Chat-ChR2/SHAM levels, providing a link between E2 status and the aggravated cardiac response in this model. To investigate the cellular basis underlying the cardiac alterations, ventricular myocytes were isolated and their cellular area and contractility were assessed. Myocytes from WT/OVX mice were wider than WT/SHAM, an indicative of concentric hypertrophy, but their fractional shortening was similar. Conversely, Chat-ChR2/OVX myocytes were elongated and presented contractile dysfunction. E2 treatment again prevented the structural and functional changes in Chat-ChR2/OVX myocytes. We conclude that hypercholinergic mice under reduced estrogen conditions do not develop concentric hypertrophy, a critical compensatory adaptation, evolving toward cardiac dilation and failure. This study emphasizes the importance of understanding the consequences of cholinesterase inhibition, used clinically to treat dementia, for cardiac function in postmenopausal women.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Animals
- Cholinergic Fibers/metabolism
- Estradiol/pharmacology
- Estrogen Replacement Therapy
- Estrogens/deficiency
- Female
- Heart/innervation
- Heart Rate
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Mice, Inbred C57BL
- Mice, Transgenic
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Ovariectomy
- Signal Transduction
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Vesicular Acetylcholine Transport Proteins/genetics
- Vesicular Acetylcholine Transport Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Vanessa P Teixeira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kiany Miranda
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio Scalzo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cibele Rocha-Resende
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mário Morais Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geisa C S V Tezini
- Ribeirão Preto Medical School, Universidade de São Paulo, Riberão Preto, São Paulo, Brazil
| | - Marcos B Melo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando Pedro Souza-Neto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kaoma S C Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Itamar C G Jesus
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anderson K Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro de Oliveira
- Ribeirão Preto Medical School, Universidade de São Paulo, Riberão Preto, São Paulo, Brazil
| | - Raphael E Szawka
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Helio C Salgado
- Ribeirão Preto Medical School, Universidade de São Paulo, Riberão Preto, São Paulo, Brazil
| | - Marco Antonio Máximo Prado
- Robarts Research Institute, Department of Physiology and Pharmacology and Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Maristela O Poletini
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
18
|
Lee JM, Mayall JR, Chevalier A, McCarthy H, Van Helden D, Hansbro PM, Horvat JC, Jobling P. Chlamydia muridarum infection differentially alters smooth muscle function in mouse uterine horn and cervix. Am J Physiol Endocrinol Metab 2020; 318:E981-E994. [PMID: 32315215 DOI: 10.1152/ajpendo.00513.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chlamydia trachomatis infection is a primary cause of reproductive tract diseases including infertility. Previous studies showed that this infection alters physiological activities in mouse oviducts. Whether this occurs in the uterus and cervix has never been investigated. This study characterized the physiological activities of the uterine horn and the cervix in a Chlamydia muridarum (Cmu)-infected mouse model at three infection time points of 7, 14, and 21 days postinfection (dpi). Cmu infection significantly decreased contractile force of spontaneous contraction in the cervix (7 and 14 dpi; P < 0.001 and P < 0.05, respectively), but this effect was not observed in the uterine horn. The responses of the uterine horn and cervix to oxytocin were significantly altered by Cmu infection at 7 dpi (P < 0.0001), but such responses were attenuated at 14 and 21 dpi. Cmu infection increased contractile force to prostaglandin (PGF2α) by 53-83% in the uterine horn. This corresponded with the increased messenger ribonucleic acid (mRNA) expression of Ptgfr that encodes for its receptor. However, Cmu infection did not affect contractions of the uterine horn and cervix to PGE2 and histamine. The mRNA expression of Otr and Ptger4 was inversely correlated with the mRNA expression of Il1b, Il6 in the uterine horn of Cmu-inoculated mice (P < 0.01 to P < 0.001), suggesting that the changes in the Otr and Ptger4 mRNA expression might be linked to the changes in inflammatory cytokines. Lastly, this study also showed a novel physiological finding of the differential response to PGE2 in mouse uterine horn and cervix.
Collapse
Affiliation(s)
- Jia Ming Lee
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Anne Chevalier
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Huw McCarthy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Dirk Van Helden
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
- Centenary Institute and the University of Technology Sydney, Sydney, New South Wales, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Phillip Jobling
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
19
|
Association with SERCA2a directs phospholamban trafficking to sarcoplasmic reticulum from a nuclear envelope pool. J Mol Cell Cardiol 2020; 143:107-119. [DOI: 10.1016/j.yjmcc.2020.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
|
20
|
Oldfield CJ, Duhamel TA, Dhalla NS. Mechanisms for the transition from physiological to pathological cardiac hypertrophy. Can J Physiol Pharmacol 2020; 98:74-84. [DOI: 10.1139/cjpp-2019-0566] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The heart is capable of responding to stressful situations by increasing muscle mass, which is broadly defined as cardiac hypertrophy. This phenomenon minimizes ventricular wall stress for the heart undergoing a greater than normal workload. At initial stages, cardiac hypertrophy is associated with normal or enhanced cardiac function and is considered to be adaptive or physiological; however, at later stages, if the stimulus is not removed, it is associated with contractile dysfunction and is termed as pathological cardiac hypertrophy. It is during physiological cardiac hypertrophy where the function of subcellular organelles, including the sarcolemma, sarcoplasmic reticulum, mitochondria, and myofibrils, may be upregulated, while pathological cardiac hypertrophy is associated with downregulation of these subcellular activities. The transition of physiological cardiac hypertrophy to pathological cardiac hypertrophy may be due to the reduction in blood supply to hypertrophied myocardium as a consequence of reduced capillary density. Oxidative stress, inflammatory processes, Ca2+-handling abnormalities, and apoptosis in cardiomyocytes are suggested to play a critical role in the depression of contractile function during the development of pathological hypertrophy.
Collapse
Affiliation(s)
- Christopher J. Oldfield
- Faculty of Kinesiology & Recreation Management, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Todd A. Duhamel
- Faculty of Kinesiology & Recreation Management, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
21
|
Kaur S, Shen X, Power A, Ward ML. Stretch modulation of cardiac contractility: importance of myocyte calcium during the slow force response. Biophys Rev 2020; 12:135-142. [PMID: 31939110 DOI: 10.1007/s12551-020-00615-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical response of the heart to myocardial stretch has been understood since the work of muscle physiologists more than 100 years ago, whereby an increase in ventricular chamber filling during diastole increases the subsequent force of contraction. The stretch-induced increase in contraction is biphasic. There is an abrupt increase in the force that coincides with the stretch (the rapid response), which is then followed by a slower response that develops over several minutes (the slow force response, or SFR). The SFR is associated with a progressive increase in the magnitude of the Ca2+ transient, the event that initiates myocyte cross-bridge cycling and force development. However, the mechanisms underlying the stretch-dependent increase in the Ca2+ transient are still debated. This review outlines recent literature on the SFR and summarizes the different stretch-activated Ca2+ entry pathways. The SFR might result from a combination of several different cellular mechanisms initiated in response to activation of different cellular stretch sensors.
Collapse
Affiliation(s)
- Sarbjot Kaur
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G.Jebsen Center for Cardiac Research, Oslo, Norway
| | - Amelia Power
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Marie-Louise Ward
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
22
|
Díaz HS, Toledo C, Andrade DC, Marcus NJ, Del Rio R. Neuroinflammation in heart failure: new insights for an old disease. J Physiol 2020; 598:33-59. [PMID: 31671478 DOI: 10.1113/jp278864] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 08/25/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome affecting roughly 26 million people worldwide. Increased sympathetic drive is a hallmark of HF and is associated with disease progression and higher mortality risk. Several mechanisms contribute to enhanced sympathetic activity in HF, but these pathways are still incompletely understood. Previous work suggests that inflammation and activation of the renin-angiotensin system (RAS) increases sympathetic drive. Importantly, chronic inflammation in several brain regions is commonly observed in aged populations, and a growing body of evidence suggests neuroinflammation plays a crucial role in HF. In animal models of HF, central inhibition of RAS and pro-inflammatory cytokines normalizes sympathetic drive and improves cardiac function. The precise molecular and cellular mechanisms that lead to neuroinflammation and its effect on HF progression remain undetermined. This review summarizes the most recent advances in the field of neuroinflammation and autonomic control in HF. In addition, it focuses on cellular and molecular mediators of neuroinflammation in HF and in particular on brain regions involved in sympathetic control. Finally, we will comment on what is known about neuroinflammation in the context of preserved vs. reduced ejection fraction HF.
Collapse
Affiliation(s)
- Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Noah J Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, USA
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
23
|
Leong CN, Dokos S, Andriyana A, Liew YM, Chan BT, Abdul Aziz YF, Chee KH, Sridhar GS, Lim E. The role of end-diastolic myocardial fibre stretch on infarct extension. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2020; 36:e3291. [PMID: 31799767 DOI: 10.1002/cnm.3291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/11/2019] [Accepted: 11/20/2019] [Indexed: 06/10/2023]
Abstract
Myocardial infarct extension, a process involving the enlargement of infarct and border zone, leads to progressive degeneration of left ventricular (LV) function and eventually gives rise to heart failure. Despite carrying a high risk, the causation of infarct extension is still a subject of much speculation. In this study, patient-specific LV models were developed to investigate the correlation between infarct extension and impaired regional mechanics. Subsequently, sensitivity analysis was performed to examine the causal factors responsible for the impaired regional mechanics observed in regions surrounding the infarct and border zone. From our simulations, fibre strain, fibre stress and fibre stress-strain loop (FSSL) were the key biomechanical variables affected in these regions. Among these variables, only FSSL was correlated with infarct extension, as reflected in its work density dissipation (WDD) index value, with high WDD indices recorded at regions with infarct extension. Impaired FSSL is caused by inadequate contraction force generation during the isovolumic contraction and ejection phases. Our further analysis revealed that the inadequacy in contraction force generation is not necessarily due to impaired myocardial intrinsic contractility, but at least in part, due to inadequate muscle fibre stretch at end-diastole, which depresses the ability of myocardium to generate adequate contraction force in the subsequent systole (according to the Frank-Starling law). Moreover, an excessively stiff infarct may cause its neighbouring myocardium to be understretched at end-diastole, subsequently depressing the systolic contractile force of the neighbouring myocardium, which was found to be correlated with infarct extension.
Collapse
Affiliation(s)
- Chin Neng Leong
- Department of Biomedical Engineering, University of Malaya, Kuala Lumpur, Malaysia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Socrates Dokos
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Andri Andriyana
- Department of Mechanical Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Yih Miin Liew
- Department of Biomedical Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Bee Ting Chan
- Mechanical Engineering, UCSI University, Kuala Lumpur, Malaysia
| | | | - Kok-Han Chee
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Einly Lim
- Department of Biomedical Engineering, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
Adverse transverse-tubule remodeling in a rat model of heart failure is attenuated with low-dose triiodothyronine treatment. Mol Med 2019; 25:53. [PMID: 31810440 PMCID: PMC6898920 DOI: 10.1186/s10020-019-0120-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
Abstract Pre-clinical animal studies have shown that triiodothyronine (T3) replacement therapy improves cardiac contractile function after myocardial infarction (MI). We hypothesized that T3 treatment could prevent adverse post-infarction cardiomyocyte remodeling by maintaining transverse-tubule (TT) structures, thus improving calcium dynamics and contractility. Methods Myocardial infarction (MI) or sham surgeries were performed on female Sprague-Dawley rats (aged 12 wks), followed by treatment with T3 (5μg/kg/d) or vehicle in drinking water for 16 wks (n = 10–11/group). After in vivo echocardiographic and hemodynamic analyses, left ventricular myocytes were isolated by collagenase digestion and simultaneous calcium and contractile transients in single cardiomyocytes were recorded using IonOptix imaging. Live cardiomyocytes were stained with AlexaFluor-488 conjugated wheat germ agglutinin (WGA-488) or di-8-ANEPPS, and multiple z-stack images per cell were captured by confocal microscopy for analysis of TT organization. RTqPCR and immunoblot approaches determined expression of TT proteins. Results Echocardiography and in vivo hemodynamic measurements showed significant improvements in systolic and diastolic function in T3- vs vehicle-treated MI rats. Isolated cardiomyocyte analysis showed significant dysfunction in measurements of myocyte relengthening in MI hearts, and improvements with T3 treatment: max relengthening velocity (Vmax, um/s), 2.984 ± 1.410 vs 1.593 ± 0.325, p < 0.05 and time to Vmax (sec), 0.233 ± 0.037 vs 0.314 ± 0.019, p < 0.001; MI + T3 vs MI + Veh, respectively. Time to peak contraction was shortened by T3 treatment (0.161 ± 0.021 vs 0.197 ± 0.011 s., p < 0.01; MI + T3 vs MI + Veh, respectively). Analysis of TT periodicity of WGA- or ANEPPS-stained cardiomyocytes indicated significant TT disorganization in MI myocytes and improvement with T3 treatment (transverse-oriented tubules (TE%): 9.07 ± 0.39 sham, 6.94 ± 0.67 MI + Veh and 8.99 ± 0.38 MI + T3; sham vs MI + Veh, p < 0.001; MI + Veh vs MI + T3, p < 0.01). Quantitative RT-PCR showed that reduced expression of BIN1 (Bridging integrator-1), Jph2 (junctophilin-2), RyR2 (ryanodine receptor) and Cav1.2 (L-type calcium channel) in the failing myocardium were increased by T3 and immunoblot analysis further supporting a potential T3 effect on the TT-associated proteins, BIN1 and Jph2. In conclusion, low dose T3 treatment initiated immediately after myocardial infarction attenuated adverse TT remodeling, improved calcium dynamics and contractility, thus supporting the potential therapeutic utility of T3 treatment in heart failure.
Collapse
|
25
|
Meng T, Ren X, Chen X, Yu J, Agrimi J, Paolocci N, Gao WD. Anesthetic Agents Isoflurane and Propofol Decrease Maximal Ca 2+-Activated Force and Thus Contractility in the Failing Myocardium. J Pharmacol Exp Ther 2019; 371:615-623. [PMID: 31515443 PMCID: PMC6863458 DOI: 10.1124/jpet.119.259556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/11/2019] [Indexed: 01/16/2023] Open
Abstract
In the normal heart, frequently used anesthetics such as isoflurane and propofol can reduce inotropy. However, the impact of these agents on the failing myocardium is unclear. Here, we examined whether and how isoflurane and propofol influence cardiac contractility in intact cardiac muscles from rats treated with monocrotaline to induce heart failure. We measured force and intracellular Ca2+ ([Ca2 +]i) in trabeculae from the right ventricles of the rats in the absence or presence of propofol or isoflurane. At low to moderate concentrations, both propofol and isoflurane dose-dependently depressed cardiac force generation in failing trabeculae without altering [Ca2+]i At high doses, propofol (but not isoflurane) also decreased amplitude of [Ca2+]i transients. During steady-state activation, both propofol and isoflurane impaired maximal Ca2+-activated force (Fmax) while increasing the amount of [Ca2+]i required for 50% of maximal activation (Ca50). These events occurred without apparent change in the Hill coefficient, suggesting no impairment of cooperativity. Exposing these same muscles to the anesthetics after fiber skinning resulted in a similar decrement in Fmax and rise in Ca50 but no change in the myofibrillar ATPase-Ca2+ relationship. Thus, our study demonstrates that challenging the failing myocardium with commonly used anesthetic agents such as propofol and isoflurane leads to reduced force development as a result of lowered myofilament responsiveness to Ca2+ SIGNIFICANCE STATEMENT: Commonly used anesthetics such as isoflurane and propofol can impair myocardial contractility in subjects with heart failure by lowering myofilament responsiveness to Ca2+. High doses of propofol can also reduce the overall amplitude of the intracellular Ca2+ transient. These findings may have important implications for the safety and quality of intra- and perioperative care of patients with heart failure and other cardiac disorders.
Collapse
Affiliation(s)
- Tao Meng
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Xianfeng Ren
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Xinzhong Chen
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jingui Yu
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jacopo Agrimi
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Nazareno Paolocci
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Wei Dong Gao
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shangdong, China (T.M., J.Y.); Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China (X.R.); Department of Cardiac Surgery, Tongji University Medical Center, Wuhan, China (X.C.); Division of Cardiology (J.A., N.P.) and Department of Anesthesiology and Critical Care Medicine (W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| |
Collapse
|
26
|
Chen L, Wang L, Li X, Wang C, Hong M, Li Y, Cao J, Fu L. The role of desmin alterations in mechanical electrical feedback in heart failure. Life Sci 2019; 241:117119. [PMID: 31794771 DOI: 10.1016/j.lfs.2019.117119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 10/25/2022]
Abstract
AIM Mechanoelectric feedback (MEF) was related to malignant arrhythmias in heart failure (HF). Desmin is a cytoskeleton protein and could be involved in MEF as a mechanoelectrical transducer. In this study, we will discuss the role of desmin alterations in mechanical electrical feedback in heart failure and its mechanisms. METHODS We used both an in vivo rat model and an in vitro cardiomyocyte model to address this issue. For the in vivo experiments, we establish a sham group, an HF group, streptomycin (SM) group, and an MDL-28170 group. The occurrence of ventricular arrhythmias (VA) was recorded in each group. For the in vitro cardiomyocyte model, we established an NC group, a si-desmin group, and a si-desmin + NBD IKK group. The expression of desmin, IKKβ, p-IKKβ, IKBα, p-NF-κB, and SERCA2 were detected in both in vivo and in vitro experiments. The content of Ca2+ in cytoplasm and sarcoplasmic were detected by confocal imaging in vitro experiments. RESULTS An increased number of VAs were found in the HF group. SM and MDL-28170 can reduce desmin breakdown and the number of VAs in heart failure. The knockdown of desmin in the cardiomyocyte can activate the NF-κB pathway, decrease the level of SERCA2, and result in abnormal distribution of Ca2+. While treatment with NF-κB inhibitor can elevate the level of SERCA2 and alleviate the abnormal distribution of Ca2+. SIGNIFICANCE Overall, desmin may participate in MEF through the NF-κB pathway. This study provides a potential therapeutic target for VA in HF.
Collapse
Affiliation(s)
- Lin Chen
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Li Wang
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xingyi Li
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Can Wang
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Mingyang Hong
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yuanshi Li
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Junxian Cao
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Lu Fu
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
27
|
Lomivorotov VV, Leonova EA, Belletti A, Shmyrev VA, Landoni G. Calcium Administration During Weaning From Cardiopulmonary Bypass: A Narrative Literature Review. J Cardiothorac Vasc Anesth 2019; 34:235-244. [PMID: 31350149 DOI: 10.1053/j.jvca.2019.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
The search for safe and effective patient management strategies during weaning from cardiopulmonary bypass is ongoing; intravenous calcium is occasionally used as a first-line drug. The physiologic role of calcium suggests that it can support the function of the cardiovascular system during this critical period. Patients may be mildly hypocalcemic after cardiopulmonary bypass; however, this degree of hypocalcemia does not significantly impair the cardiovascular system. The transient beneficial effects of calcium administration (increase in arterial blood pressure, systemic vascular resistance, cardiac index, stroke volume, and coronary perfusion pressure) might be helpful in cases of moderate contractility reduction or vasoplegia. Nonetheless, effects on clinically relevant endpoints are unknown, and possible systemic side effects, such as transient reduction in internal mammary artery graft flow, attenuation of the effects of β-sympathomimetics, "stone heart" phenomenon, and pancreatic cellular injury, may limit the use of calcium salts. Further studies are needed to expand the understanding of the effects of calcium administration on patient outcomes.
Collapse
Affiliation(s)
- Vladimir V Lomivorotov
- Department of Anaesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia; Novosibirsk State University, Novosibirsk, Russia
| | - Elizaveta A Leonova
- Department of Anaesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Alessandro Belletti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vladimir A Shmyrev
- Department of Anaesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
28
|
Verkaik M, Oranje M, Abdurrachim D, Goebel M, Gam Z, Prompers JJ, Helmes M, Ter Wee PM, van der Velden J, Kuster DW, Vervloet MG, Eringa EC. High Fibroblast Growth Factor 23 concentrations in experimental renal failure impair calcium handling in cardiomyocytes. Physiol Rep 2019; 6:e13591. [PMID: 29611320 PMCID: PMC5880876 DOI: 10.14814/phy2.13591] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
The overwhelming majority of patients with chronic kidney disease (CKD) die prematurely before reaching end‐stage renal disease, mainly due to cardiovascular causes, of which heart failure is the predominant clinical presentation. We hypothesized that CKD‐induced increases of plasma FGF23 impair cardiac diastolic and systolic function. To test this, mice were subjected to 5/6 nephrectomy (5/6Nx) or were injected with FGF23 for seven consecutive days. Six weeks after surgery, plasma FGF23 was higher in 5/6Nx mice compared to sham mice (720 ± 31 vs. 256 ± 3 pg/mL, respectively, P = 0.034). In cardiomyocytes isolated from both 5/6Nx and FGF23 injected animals the rise of cytosolic calcium during systole was slowed (−13% and −19%, respectively) as was the decay of cytosolic calcium during diastole (−15% and −21%, respectively) compared to controls. Furthermore, both groups had similarly decreased peak cytosolic calcium content during systole. Despite lower cytosolic calcium contents in CKD or FGF23 pretreated animals, no changes were observed in contractile parameters of cardiomyocytes between the groups. Expression of calcium handling proteins and cardiac troponin I phosphorylation were similar between groups. Blood pressure, the heart weight:tibia length ratio, α‐MHC/β‐MHC ratio and ANF mRNA expression, and systolic and diastolic function as measured by MRI did not differ between groups. In conclusion, the rapid, CKD‐induced rise in plasma FGF23 and the similar decrease in cardiomyocyte calcium transients in modeled kidney disease and following 1‐week treatment with FGF23 indicate that FGF23 partly mediates cardiomyocyte dysfunction in CKD.
Collapse
Affiliation(s)
- Melissa Verkaik
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Maarten Oranje
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Max Goebel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Zeineb Gam
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michiel Helmes
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Pieter M Ter Wee
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Diederik W Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc G Vervloet
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
29
|
Curran J, Burkhoff D, Kloner RA. Beyond Reperfusion: Acute Ventricular Unloading and Cardioprotection During Myocardial Infarction. J Cardiovasc Transl Res 2019; 12:95-106. [PMID: 30671717 PMCID: PMC6497619 DOI: 10.1007/s12265-019-9863-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022]
Abstract
Heart failure is a major cause of morbidity and mortality around the world, and myocardial infarction is its leading cause. Myocardial infarction destroys viable myocardium, and this dead tissue is replaced by a non-contractile scar that results in impaired cardiac function and a significantly increased likelihood of the patient developing heart failure. Limiting infarct scar size has been the target of pre-clinical and clinical investigations for decades. However, beyond reperfusion, few therapies have translated into the clinic that limit its formation. New approaches are needed. This review will focus on new clinical and pre-clinical data demonstrating that acute ventricular unloading prior to reperfusion by means of percutaneous left ventricular support devices reduces ischemia-reperfusion injury and limits infarct scar size. Emphasis will be given to summarizing our current mechanistic understanding of this new therapeutic approach to treating myocardial infarction.
Collapse
Affiliation(s)
| | | | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, USA
- University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
30
|
Salin Raj P, Swapna SUS, Raghu KG. High glucose induced calcium overload via impairment of SERCA/PLN pathway and mitochondrial dysfunction leads to oxidative stress in H9c2 cells and amelioration with ferulic acid. Fundam Clin Pharmacol 2019; 33:412-425. [PMID: 30739350 DOI: 10.1111/fcp.12452] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/15/2019] [Accepted: 01/31/2019] [Indexed: 01/05/2023]
Abstract
Oxidative stress and associated complications are the major pathological concerns of diabetic cardiomyopathy (DC). We aim to elucidate the mechanisms by which high glucose (HG) induced alteration in calcium homeostasis and evaluation of the beneficial effect of two concentrations (10 and 25 μm) of ferulic acid (FA). HG was induced in H9c2 cardiomyoblast by treating with glucose (33 mm) for 48 h, and FA was co-treated. Intracellular calcium ([Ca2+ ]i) overload was found increased significantly with HG. For elucidation of mechanism, the SERCA pathway and mitochondrial integrity (transmembrane potential and permeability transition pore) were explored. Then, we assessed oxidative stress, and cell injury with brain natriuretic peptide (BNP), atrial natriuretic peptide (ANP), and lactate dehydrogenase (LDH) release. HG caused significant [Ca2+ ]i overload through downregulation of SERCA2/1, pPLN, and pPKA C-α; and upregulation of PLN and PKA C-α and alteration in the integrity of mitochondria with HG. The [Ca2+ ]i overload in turn caused oxidative stress via generation of reactive oxygen species, lipid peroxidation, and protein carbonylation. This resulted in cell injury which was evident with significant release of BNP, ANP, and LDH. FA co-treatment was effective to mitigate all pathological changes caused by HG. From the overall results, we conclude that [Ca2+ ]i overload via SERCA pathway and altered mitochondrial integrity is the main cause for oxidative stress during HG. Based on our result, we report that FA could be an attractive nutraceutical for DC.
Collapse
Affiliation(s)
- Palayyan Salin Raj
- Biochemistry and Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus Ghaziabad, Uttar Pradesh, 201 002, India
| | - Sasi U S Swapna
- Biochemistry and Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus Ghaziabad, Uttar Pradesh, 201 002, India
| | - Kozhiparambil G Raghu
- Biochemistry and Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus Ghaziabad, Uttar Pradesh, 201 002, India
| |
Collapse
|
31
|
Ke HY, Yang HY, Francis AJ, Collins TP, Surendran H, Alvarez-Laviada A, Firth JM, MacLeod KT. Changes in cellular Ca 2+ and Na + regulation during the progression towards heart failure in the guinea pig. J Physiol 2019; 598:1339-1359. [PMID: 30811606 PMCID: PMC7187457 DOI: 10.1113/jp277038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/26/2019] [Indexed: 12/19/2022] Open
Abstract
Key points During compensated hypertrophy in vivo fractional shortening (FS) remains constant until heart failure (HF) develops, when FS decreases from 70% to 39%. Compensated hypertrophy is accompanied by an increase in INa,late and a decrease in Na+,K+‐ATPase current. These changes persist as HF develops. SR Ca2+ content increases during compensated hypertrophy then decreases in HF. In healthy cells, increases in SR Ca2+ content and Ca2+ transients can be achieved by the same amount of inhibition of the Na+,K+‐ATPase as measured in the diseased cells. SERCA function remains constant during compensated hypertrophy then decreases in HF, when there is also an increase in spark frequency and spark‐mediated Ca2+ leak. We suggest an increase in INa,late and a decrease in Na+,K+‐ATPase current and function alters the balance of Ca2+ flux mediated by the Na+/Ca2+ exchange that limits early contractile impairment.
Abstract We followed changes in cardiac myocyte Ca2+ and Na+ regulation from the formation of compensated hypertrophy (CH) until signs of heart failure (HF) are apparent using a trans‐aortic pressure overload (TAC) model. In this model, in vivo fractional shortening (FS) remained constant despite HW:BW ratio increasing by 39% (CH) until HF developed 150 days post‐TAC when FS decreased from 70% to 39%. Using live and fixed fluorescence imaging and electrophysiological techniques, we found an increase in INa,late from –0.34 to –0.59 A F−1 and a decrease in Na+,K+‐ATPase current from 1.09 A F−1 to 0.54 A F−1 during CH. These changes persisted as HF developed (INa,late increased to –0.82 A F−1 and Na+,K+‐ATPase current decreased to 0.51 A F−1). Sarcoplasmic reticulum (SR) Ca2+ content increased during CH then decreased in HF (from 32 to 15 μm l−1) potentially supporting the maintenance of FS in the whole heart and Ca2+ transients in single myocytes during the former stage. We showed using glycoside blockade in healthy myocytes that increases in SR Ca2+ content and Ca2+ transients can be driven by the same amount of inhibition of the Na+,K+‐ATPase as measured in the diseased cells. SERCA function remains constant in CH but decreases (τ for SERCA‐mediated Ca2+ removal changed from 6.3 to 3.0 s−1) in HF. In HF there was an increase in spark frequency and spark‐mediated Ca2+ leak. We suggest an increase in INa,late and a decrease in Na+,K+‐ATPase current and function alters the balance of Ca2+ flux mediated by the Na+/Ca2+ exchange that limits early contractile impairment. During compensated hypertrophy in vivo fractional shortening (FS) remains constant until heart failure (HF) develops, when FS decreases from 70% to 39%. Compensated hypertrophy is accompanied by an increase in INa,late and a decrease in Na+,K+‐ATPase current. These changes persist as HF develops. SR Ca2+ content increases during compensated hypertrophy then decreases in HF. In healthy cells, increases in SR Ca2+ content and Ca2+ transients can be achieved by the same amount of inhibition of the Na+,K+‐ATPase as measured in the diseased cells. SERCA function remains constant during compensated hypertrophy then decreases in HF, when there is also an increase in spark frequency and spark‐mediated Ca2+ leak. We suggest an increase in INa,late and a decrease in Na+,K+‐ATPase current and function alters the balance of Ca2+ flux mediated by the Na+/Ca2+ exchange that limits early contractile impairment.
Collapse
Affiliation(s)
- H-Y Ke
- Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (ROC)
| | - H-Y Yang
- Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (ROC)
| | - A J Francis
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - T P Collins
- The Wellcome Trust, Gibbs Building, 215 Euston Road, London, NW1 2BE, UK
| | - H Surendran
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - A Alvarez-Laviada
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - J M Firth
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - K T MacLeod
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
32
|
Targeting protein-protein interactions for therapeutic discovery via FRET-based high-throughput screening in living cells. Sci Rep 2018; 8:12560. [PMID: 30135432 PMCID: PMC6105598 DOI: 10.1038/s41598-018-29685-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/16/2018] [Indexed: 01/16/2023] Open
Abstract
We have developed a structure-based high-throughput screening (HTS) method, using time-resolved fluorescence resonance energy transfer (TR-FRET) that is sensitive to protein-protein interactions in living cells. The membrane protein complex between the cardiac sarcoplasmic reticulum Ca-ATPase (SERCA2a) and phospholamban (PLB), its Ca-dependent regulator, is a validated therapeutic target for reversing cardiac contractile dysfunction caused by aberrant calcium handling. However, efforts to develop compounds with SERCA2a-PLB specificity have yet to yield an effective drug. We co-expressed GFP-SERCA2a (donor) in the endoplasmic reticulum membrane of HEK293 cells with RFP-PLB (acceptor), and measured FRET using a fluorescence lifetime microplate reader. We screened a small-molecule library and identified 21 compounds (Hits) that changed FRET by >3SD. 10 of these Hits reproducibly alter SERCA2a-PLB structure and function. One compound increases SERCA2a calcium affinity in cardiac membranes but not in skeletal, suggesting that the compound is acting specifically on the SERCA2a-PLB complex, as needed for a drug to mitigate deficient calcium transport in heart failure. The excellent assay quality and correlation between structural and functional assays validate this method for large-scale HTS campaigns. This approach offers a powerful pathway to drug discovery for a wide range of protein-protein interaction targets that were previously considered “undruggable”.
Collapse
|
33
|
Vinogradova TM, Tagirova Sirenko S, Lakatta EG. Unique Ca 2+-Cycling Protein Abundance and Regulation Sustains Local Ca 2+ Releases and Spontaneous Firing of Rabbit Sinoatrial Node Cells. Int J Mol Sci 2018; 19:ijms19082173. [PMID: 30044420 PMCID: PMC6121616 DOI: 10.3390/ijms19082173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 11/16/2022] Open
Abstract
Spontaneous beating of the heart pacemaker, the sinoatrial node, is generated by sinoatrial node cells (SANC) and caused by gradual change of the membrane potential called diastolic depolarization (DD). Submembrane local Ca2+ releases (LCR) from sarcoplasmic reticulum (SR) occur during late DD and activate an inward Na+/Ca2+ exchange current, which accelerates the DD rate leading to earlier occurrence of an action potential. A comparison of intrinsic SR Ca2+ cycling revealed that, at similar physiological Ca2+ concentrations, LCRs are large and rhythmic in permeabilized SANC, but small and random in permeabilized ventricular myocytes (VM). Permeabilized SANC spontaneously released more Ca2+ from SR than VM, despite comparable SR Ca2+ content in both cell types. In this review we discuss specific patterns of expression and distribution of SR Ca2+ cycling proteins (SR Ca2+ ATPase (SERCA2), phospholamban (PLB) and ryanodine receptors (RyR)) in SANC and ventricular myocytes. We link ability of SANC to generate larger and rhythmic LCRs with increased abundance of SERCA2, reduced abundance of the SERCA inhibitor PLB. In addition, an increase in intracellular [Ca2+] increases phosphorylation of both PLB and RyR exclusively in SANC. The differences in SR Ca2+ cycling protein expression between SANC and VM provide insights into diverse regulation of intrinsic SR Ca2+ cycling that drives automaticity of SANC.
Collapse
Affiliation(s)
- Tatiana M Vinogradova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Blvd, Room 8B-123, Baltimore, MD 21224, USA.
| | - Syevda Tagirova Sirenko
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Blvd, Room 8B-123, Baltimore, MD 21224, USA.
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Blvd, Room 8B-123, Baltimore, MD 21224, USA.
| |
Collapse
|
34
|
Daniels LJ, Wallace RS, Nicholson OM, Wilson GA, McDonald FJ, Jones PP, Baldi JC, Lamberts RR, Erickson JR. Inhibition of calcium/calmodulin-dependent kinase II restores contraction and relaxation in isolated cardiac muscle from type 2 diabetic rats. Cardiovasc Diabetol 2018; 17:89. [PMID: 29903013 PMCID: PMC6001139 DOI: 10.1186/s12933-018-0732-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/06/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Calcium/calmodulin-dependent kinase II-delta (CaMKIIδ) activity is enhanced during hyperglycemia and has been shown to alter intracellular calcium handling in cardiomyocytes, ultimately leading to reduced cardiac performance. However, the effects of CaMKIIδ on cardiac contractility during type 2 diabetes are undefined. METHODS We examined the expression and activation of CaMKIIδ in right atrial appendages from non-diabetic and type 2 diabetic patients (n = 7 patients per group) with preserved ejection fraction, and also in right ventricular tissue from Zucker Diabetic Fatty rats (ZDF) (n = 5-10 animals per group) during early diabetic cardiac dysfunction, using immunoblot. We also measured whole heart function of ZDF and control rats using echocardiography. Then we measured contraction and relaxation parameters of isolated trabeculae from ZDF to control rats in the presence and absence of CaMKII inhibitors. RESULTS CaMKIIδ phosphorylation (at Thr287) was increased in both the diabetic human and animal tissue, indicating increased CaMKIIδ activation in the type 2 diabetic heart. Basal cardiac contractility and relaxation were impaired in the cardiac muscles from the diabetic rats, and CaMKII inhibition with KN93 partially restored contractility and relaxation. Autocamtide-2-related-inhibitor peptide (AIP), another CaMKII inhibitor that acts via a different mechanism than KN93, fully restored cardiac contractility and relaxation. CONCLUSIONS Our results indicate that CaMKIIδ plays a key role in modulating performance of the diabetic heart, and moreover, suggest a potential therapeutic role for CaMKII inhibitors in improving myocardial function during type 2 diabetes.
Collapse
Affiliation(s)
- Lorna J Daniels
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| | - Rachel S Wallace
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| | - Olivia M Nicholson
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| | - Genevieve A Wilson
- Otago School of Medical Sciences, Department of Medicine and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Fiona J McDonald
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| | - Peter P Jones
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| | - J Chris Baldi
- Otago School of Medical Sciences, Department of Medicine and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Regis R Lamberts
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| | - Jeffrey R Erickson
- Otago School of Medical Sciences, Department of Physiology and HeartOtago, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand.
| |
Collapse
|
35
|
Li T, Shen Y, Su L, Fan X, Lin F, Ye X, Ding D, Tang Y, Yang Y, Lei C, Hu S. Cardiac adenovirus-associated viral Presenilin 1 gene delivery protects the left ventricular function of the heart via regulating RyR2 function in post-ischaemic heart failure. J Drug Target 2018. [PMID: 29521549 DOI: 10.1080/1061186x.2018.1450412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Post-ischaemic heart failure is a major cause of death worldwide. Reperfusion of infarcted heart tissue after myocardial infarction has been an important medical intervention to improve outcomes. However, disturbances in Ca2+ and redox homeostasis at the cellular level caused by ischaemia/reperfusion remain major clinical challenges. In this study, we investigated the potential of adeno-associated virus (AAV)-9-mediated cardiac expression of a Type-2 ryanodine receptor (RyR2) degradation-associated gene, Presenilin 1 (PSEN1), to combat post-ischaemic heart failure. Adeno-associated viral PSEN1 gene delivery elevated PSEN1 protein expression in a post-infarction rat heart failure model, and this administration normalised the contractile dysfunction of the failing myocardium in vivo and in vitro by reversing myocardial Ca2+ handling and function. Moreover, PSEN1 gene transfer to failing cardiomyocytes reduced sarcoplasmic reticulum (SR) Ca2+ leak, thereby restoring the diminished intracellular Ca2+ transients and SR Ca2+ load. Moreover, PSEN1 gene transfer reversed the phosphorylation of RyR2 in failing cardiomyocytes. However, selective autophagy inhibition did not prevent the PSEN1-induced blockade of RyR2 degradation, making the participation of autophagy in PSEN1-associated RyR2 degradation unlikely. Our results established a role of the cardiac expression of PSEN1 with AAV9 vectors as a promising therapeutic approach for post-ischaemic heart failure.
Collapse
Affiliation(s)
- Tian Li
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Yafeng Shen
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Li Su
- b School of Pharmacy , Second Military Medical University , Shanghai , China
| | - Xiaoyan Fan
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Fangxing Lin
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Xuting Ye
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Dianer Ding
- c Pharchoice Therapeutics Inc , Shanghai , China
| | - Ying Tang
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Yongji Yang
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Changhai Lei
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| | - Shi Hu
- a Department of Biophysics, College of Basic Medical Sciences , Second Military Medical University , Shanghai , China
| |
Collapse
|
36
|
Gattoni S, Røe ÅT, Aronsen JM, Sjaastad I, Louch WE, Smith NP, Niederer SA. Compensatory and decompensatory alterations in cardiomyocyte Ca 2+ dynamics in hearts with diastolic dysfunction following aortic banding. J Physiol 2017; 595:3867-3889. [PMID: 28542952 PMCID: PMC5471387 DOI: 10.1113/jp273879] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 01/06/2017] [Indexed: 01/20/2023] Open
Abstract
Key points At the cellular level cardiac hypertrophy causes remodelling, leading to changes in ionic channel, pump and exchanger densities and kinetics. Previous studies have focused on quantifying changes in channels, pumps and exchangers without quantitatively linking these changes with emergent cellular scale functionality. Two biophysical cardiac cell models were created, parameterized and validated and are able to simulate electrophysiology and calcium dynamics in myocytes from control sham operated rats and aortic‐banded rats exhibiting diastolic dysfunction. The contribution of each ionic pathway to the calcium kinetics was calculated, identifying the L‐type Ca2+ channel and sarco/endoplasmic reticulum Ca2+ATPase as the principal regulators of systolic and diastolic Ca2+, respectively. Results show that the ability to dynamically change systolic Ca2+, through changes in expression of key Ca2+ modelling protein densities, is drastically reduced following the aortic banding procedure; however the cells are able to compensate Ca2+ homeostasis in an efficient way to minimize systolic dysfunction.
Abstract Elevated left ventricular afterload leads to myocardial hypertrophy, diastolic dysfunction, cellular remodelling and compromised calcium dynamics. At the cellular scale this remodelling of the ionic channels, pumps and exchangers gives rise to changes in the Ca2+ transient. However, the relative roles of the underlying subcellular processes and the positive or negative impact of each remodelling mechanism are not fully understood. Biophysical cardiac cell models were created to simulate electrophysiology and calcium dynamics in myocytes from control rats (SHAM) and aortic‐banded rats exhibiting diastolic dysfunction. The model parameters and framework were validated and the fitted parameters demonstrated to be unique for explaining our experimental data. The contribution of each ionic pathway to the calcium kinetics was calculated, identifying the L‐type Ca2+ channel (LCC) and the sarco/endoplasmic reticulum Ca2+‐ATPase (SERCA) as the principal regulators of systolic and diastolic Ca2+, respectively. In the aortic banding model, the sensitivity of systolic Ca2+ to LCC density and diastolic Ca2+ to SERCA density decreased by 16‐fold and increased by 23%, respectively, relative to the SHAM model. The energy cost of ionic homeostasis is maintained across the two models. The models predict that changes in ionic pathway densities in compensated aortic banding rats maintain Ca2+ function and efficiency. The ability to dynamically alter systolic function is significantly diminished, while the capacity to maintain diastolic Ca2+ is moderately increased. At the cellular level cardiac hypertrophy causes remodelling, leading to changes in ionic channel, pump and exchanger densities and kinetics. Previous studies have focused on quantifying changes in channels, pumps and exchangers without quantitatively linking these changes with emergent cellular scale functionality. Two biophysical cardiac cell models were created, parameterized and validated and are able to simulate electrophysiology and calcium dynamics in myocytes from control sham operated rats and aortic‐banded rats exhibiting diastolic dysfunction. The contribution of each ionic pathway to the calcium kinetics was calculated, identifying the L‐type Ca2+ channel and sarco/endoplasmic reticulum Ca2+ATPase as the principal regulators of systolic and diastolic Ca2+, respectively. Results show that the ability to dynamically change systolic Ca2+, through changes in expression of key Ca2+ modelling protein densities, is drastically reduced following the aortic banding procedure; however the cells are able to compensate Ca2+ homeostasis in an efficient way to minimize systolic dysfunction.
Collapse
Affiliation(s)
- Sara Gattoni
- King's College London, Department of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, 4th floor North Wing, The Rayne Institute, London, SE1 7EH, UK
| | - Åsmund Treu Røe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | | | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Nicolas P Smith
- King's College London, Department of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, 4th floor North Wing, The Rayne Institute, London, SE1 7EH, UK.,University of Auckland, Engineering School Block 1, Level 5, 20 Symonds St., Auckland, 101, New Zealand
| | - Steven A Niederer
- King's College London, Department of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, 4th floor North Wing, The Rayne Institute, London, SE1 7EH, UK
| |
Collapse
|
37
|
Alvarado FJ, Chen X, Valdivia HH. Ablation of the cardiac ryanodine receptor phospho-site Ser2808 does not alter the adrenergic response or the progression to heart failure in mice. Elimination of the genetic background as critical variable. J Mol Cell Cardiol 2017; 103:40-47. [PMID: 28065668 DOI: 10.1016/j.yjmcc.2017.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/04/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Phosphorylation of the cardiac ryanodine receptor (RyR2) phospho-site S2808 has been touted by the Marks group as a hallmark of heart failure (HF) and a critical mediator of the physiological fight-or-flight response of the heart. In support of this hypothesis, mice unable to undergo phosphorylation at RyR2-S2808 (S2808A) were significantly protected against HF and displayed a blunted response to adrenergic stimulation. However, the issue remains highly controversial because several groups have been unable to reproduce these findings. An important variable not considered before is the genetic background of the mice used to obtain these divergent results. METHODS AND RESULTS We backcrossed a RyR2-S2808A mouse into a congenic C57Bl/6 strain, the same strain used by the Marks group to conduct their experiments. We then performed several key experiments to confirm or discard the genetic background of the mouse as a relevant variable, including induction of HF by myocardial infarction and tests of integrity of adrenergic response. Congenic C57Bl/6 harboring the S2808A mutation showed similar echocardiographic parameters that indicated identical progression towards HF compared to wild type controls, and had a normal response to adrenergic stimulation in whole animal and cellular experiments. CONCLUSIONS The genetic background of the different mouse models is unlikely to be the source of the divergent results obtained by the Marks group in comparison to several other groups. Cardiac adrenergic response and progression towards HF proceed unaltered in mice harboring the RyR2-S2808A mutation. Preventing RyR2-S2808 phosphorylation does not preclude a normal sympathetic response nor mitigates the pathophysiological consequences of MI.
Collapse
Affiliation(s)
- Francisco J Alvarado
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States.
| | - Xi Chen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Héctor H Valdivia
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
38
|
Booij HG, Koning AM, van Goor H, de Boer RA, Westenbrink BD. Selecting heart failure patients for metabolic interventions. Expert Rev Mol Diagn 2016; 17:141-152. [DOI: 10.1080/14737159.2017.1266939] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Harmen G. Booij
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anne M. Koning
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Harry van Goor
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rudolf A. de Boer
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - B. Daan Westenbrink
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
39
|
Elsherif L, Jiang Y, Saari JT, Kang YJ. Dietary Copper Restriction-Induced Changes in Myocardial Gene Expression and the Effect of Copper Repletion. Exp Biol Med (Maywood) 2016. [DOI: 10.1177/153537020422900705] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Dietary copper (Cu) restriction leads to cardiac hypertrophy and failure in mice, and Cu repletion (CuR) reverses the hypertrophy and prevents the transition to heart failure. The present study was undertaken to determine changes in myocardial gene expression involved in Cu deficient (CuD) cardiomyopathy and its reversal by CuR. Analysis was performed on three groups of mice: 4-week-old CuD mice that exhibited signs of cardiac failure, their age-matched copper-adequate (CuA) controls, and the CuD mice that were re-fed adequate Cu for 2 weeks. Total RNA was isolated from hearts and subjected to cDNA microarray and real-time reverse transcription-polymerase chain reaction analysis. Dietary CuD caused a decrease in cardiac mRNA of β-MHC, L-type Ca2+ channel, K-dependent NCX, MMP-2, -8, and -13, NF-κB, and VEGF. The mRNA levels of ET-1, TGF-β, TNF-α, and procollagen-l-α1 and III-α1 were increased in the CuD cardiac tissue. Copper repletion resulted in cardiac mRNA levels of most of the genes examined returning to control levels, although the K-dependent NCX and MMP-2 values did not reach those of the CuA control. In addition, CuR caused an increase in β-MHC, L-type Ca2+channel, MMP-13 to levels surpassing those of CuA control, and a decrease in ET-1, and TNF-at mRNA levels. In summary, changes in gene expression of elements involved in contractility, Ca2+ cycling, and inflammation and fibrosis may account for the altered cardiac function found in CuD mice. The return to normal cardiac function by CuR may be a result of the favorable regression in gene expression of these critical components in myocardial tissue.
Collapse
Affiliation(s)
| | - Youchun Jiang
- Departments of Medicine, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Jack T. Saari
- U.S. Department of Agriculture, Human Nutrition Research Center, Grand Forks, North Dakota 58202
| | - Y. James Kang
- Departments of Pharmacology and Toxicology
- Departments of Medicine, University of Louisville School of Medicine, Louisville, Kentucky 40202
| |
Collapse
|
40
|
Jang SP, Oh JG, Kang DH, Kang JY, Kang SW, Hajjar RJ, Park WJ. A Decoy Peptide Targeted to Protein Phosphatase 1 Attenuates Degradation of SERCA2a in Vascular Smooth Muscle Cells. PLoS One 2016; 11:e0165569. [PMID: 27792751 PMCID: PMC5085086 DOI: 10.1371/journal.pone.0165569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/13/2016] [Indexed: 01/14/2023] Open
Abstract
Neointimal growth in the injured vasculature is largely facilitated by the proliferation of vascular smooth muscle cells (VSMC), which associates with reduced sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) activity. The gene transfer-mediated restoration of the SERCA2a level thus attenuates neointimal growth and VSMC proliferation. We previously reported that a peptide targeted to protein phosphatase 1, ψPLB-SE, normalizes SERCA2a activity in cardiomyocytes. In this study, we found that ψPLB-SE attenuated neointimal growth in balloon-injured rat carotid arteries, and the proliferation and migration of VSMC cultured in high-serum media (synthetic conditions). In parallel, ψPLB-SE inhibited the degradation of SERCA2a in the injured carotid arteries and VSMC under synthetic conditions. The calpain inhibitor MDL28170 also attenuated SERCA2a degradation and VSMC proliferation under synthetic conditions, indicating that calpain degrades SERCA2a. The Ca2+ ionophore A23187 induced SERCA2a degradation in VSMC, which was blocked by either ψPLB-SE or MDL28170. Additionally, ψPLB-SE normalized the cytosolic Ca2+ level in VSMC that was increased by either A23187 or synthetic stimulation. Collectively, these data indicate that ψPLB-SE corrects the abnormal Ca2+ handling by activating SERCA2a, which further protects SERCA2a from calpain-dependent degradation in VSMC. We conclude that ψPLB-SE may form the basis of a therapeutic strategy for vascular proliferative disorders.
Collapse
Affiliation(s)
- Seung Pil Jang
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jae Gyun Oh
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States of America
| | - Dong Hoon Kang
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Ju Young Kang
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Sang Won Kang
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Roger J. Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States of America
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
- * E-mail:
| |
Collapse
|
41
|
Bostan HB, Rezaee R, Valokala MG, Tsarouhas K, Golokhvast K, Tsatsakis AM, Karimi G. Cardiotoxicity of nano-particles. Life Sci 2016; 165:91-99. [PMID: 27686832 DOI: 10.1016/j.lfs.2016.09.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/14/2016] [Accepted: 09/23/2016] [Indexed: 01/08/2023]
Abstract
Nano-particles (NPs) are used in industrial and biomedical fields such as cosmetics, food additives and biosensors. Beside their favorable properties, nanoparticles are responsible for toxic effects. Local adverse effects and/or systemic toxicity are described with nanoparticle delivery to target organs of the human body. Animal studies provide evidence for the aforementioned toxicity. Cardiac function is a specific target of nanoparticles. Thus, reviewing the current bibliography on cardiotoxicity of nanoparticles and specifically of titanium, zinc, silver, carbon, silica and iron oxide nano-materials is the aim of this study.
Collapse
Affiliation(s)
- Hasan Badie Bostan
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Scientific Educational Center of Nanotechnology, Far Eastern Federal University, 10 Pushkinskaya Street, Vladivostok 690950, Russia
| | - Mahmoud Gorji Valokala
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Kirill Golokhvast
- Scientific Educational Center of Nanotechnology, Far Eastern Federal University, 10 Pushkinskaya Street, Vladivostok 690950, Russia
| | - Aristidis M Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion 71003, Greece.
| | - Gholamreza Karimi
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
42
|
Singh RB, Dandekar SP, Elimban V, Gupta SK, Dhalla NS. Role of proteases in the pathophysiology of cardiac disease. Mol Cell Biochem 2016; 263:241-56. [PMID: 27520682 DOI: 10.1023/b:mcbi.0000041865.63445.40] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is a major cause of death and thus a great deal of effort has been made in salvaging the diseased myocardium. Although various factors have been identified as possible causes of different cardiac diseases such as heart failure and ischemic heart disease, there is a real need to elucidate their role for the better understanding of the cardiac disease pathology and formulation of strategies for developing newer therapeutic interventions. In view of the intimate involvement of different types of proteases in maintaining cellular structure, the role of proteases in various cardiac diseases has become the focus of recent research. Proteases are present in the cytosol as well as are localized in a number of subcellular organelles in the cell. These are known to use extracellular matrix, cytoskeletal, sarcolemmal, sarcoplasmic reticular, mitochondrial and myofibrillar proteins as substrates. Work from different laboratories using a wide variety of techniques has shown that the activation of proteases causes alterations of a number of specific proteins leading to subcellular remodeling and cardiac dysfunction. Inhibition of protease action by different drugs and agents, therefore, has a clinical relevance and is expected to form a part of new treatment paradigm for improving heart function. This review examines the biochemistry and localization of some of the proteases in the cardiac tissue in addition to identification of the sites of action of some protease inhibitors. (Mol Cell Biochem 263: 241-256, 2004).
Collapse
Affiliation(s)
- Raja B Singh
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Sucheta P Dandekar
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Vijayan Elimban
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Suresh K Gupta
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada R2H 2A6
| |
Collapse
|
43
|
Abstract
The aim of this review is to provide the reader with a synopsis of some of the emerging ideas and experimental findings in cardiac physiology and pathophysiology that were published in 2015. To provide context for the non-specialist, a brief summary of cardiac contraction and calcium (Ca) regulation in the heart in health and disease is provided. Thereafter, some recently published articles are introduced that indicate the current thinking on (1) the Ca regulatory pathways modulated by Ca/calmodulin-dependent protein kinase II, (2) the potential influences of nitrosylation by nitric oxide or S-nitrosated proteins, (3) newly observed effects of reactive oxygen species (ROS) on contraction and Ca regulation following myocardial infarction and a possible link with changes in mitochondrial Ca, and (4) the effects of some of these signaling pathways on late Na current and pro-arrhythmic afterdepolarizations as well as the effects of transverse tubule disturbances.
Collapse
Affiliation(s)
- Ken T MacLeod
- Faculty of Medicine, National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
44
|
Affiliation(s)
- James E. Udelson
- From Division of Cardiology and the Cardiovascular Center, Tufts Medical Center, Boston, MA (J.E.U.); and Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston MA (L.W.S.)
| | - Lynne Warner Stevenson
- From Division of Cardiology and the Cardiovascular Center, Tufts Medical Center, Boston, MA (J.E.U.); and Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston MA (L.W.S.)
| |
Collapse
|
45
|
Daniels L, Bell JR, Delbridge LMD, McDonald FJ, Lamberts RR, Erickson JR. The role of CaMKII in diabetic heart dysfunction. Heart Fail Rev 2016. [PMID: 26198034 DOI: 10.1007/s10741-015-9498-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) is an increasing epidemic that places a significant burden on health services worldwide. The incidence of heart failure (HF) is significantly higher in diabetic patients compared to non-diabetic patients. One underlying mechanism proposed for the link between DM and HF is activation of calmodulin-dependent protein kinase (CaMKIIδ). CaMKIIδ mediates ion channel function and Ca(2+) handling during excitation-contraction and excitation-transcription coupling in the myocardium. CaMKIIδ activity is up-regulated in the myocardium of diabetic patients and mouse models of diabetes, where it promotes pathological signaling that includes hypertrophy, fibrosis and apoptosis. Pharmacological inhibition and knockout models of CaMKIIδ have shown some promise of a potential therapeutic benefit of CaMKIIδ inhibition, with protection against cardiac hypertrophy and apoptosis reported. This review will highlight the pathological role of CaMKIIδ in diabetes and discuss CaMKIIδ as a therapeutic target in DM, and also the effects of exercise on CaMKIIδ.
Collapse
Affiliation(s)
- Lorna Daniels
- Department of Physiology, University of Otago, PO Box 56, Dunedin, New Zealand
| | | | | | | | | | | |
Collapse
|
46
|
Lee A, Oh JG, Gorski PA, Hajjar RJ, Kho C. Post-translational Modifications in Heart Failure: Small Changes, Big Impact. Heart Lung Circ 2016; 25:319-24. [PMID: 26795636 PMCID: PMC4775300 DOI: 10.1016/j.hlc.2015.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 12/31/2022]
Abstract
Heart failure is a complex disease process with various aetiologies and is a significant cause of morbidity and death world-wide. Post-translational modifications (PTMs) alter protein structure and provide functional diversity in terms of physiological functions of the heart. In addition, alterations in protein PTMs have been implicated in human disease pathogenesis. Small ubiquitin-like modifier mediated modification (SUMOylation) pathway was found to play essential roles in cardiac development and function. Abnormal SUMOylation has emerged as a new feature of heart failure pathology. In this review, we will highlight the importance of SUMOylation as a regulatory mechanism of SERCA2a function, and its therapeutic potential for the treatment of heart failure.
Collapse
Affiliation(s)
- Ahyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jae Gyun Oh
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Przemek A Gorski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Changwon Kho
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
47
|
Hafver TL, Hodne K, Wanichawan P, Aronsen JM, Dalhus B, Lunde PK, Lunde M, Martinsen M, Enger UH, Fuller W, Sjaastad I, Louch WE, Sejersted OM, Carlson CR. Protein Phosphatase 1c Associated with the Cardiac Sodium Calcium Exchanger 1 Regulates Its Activity by Dephosphorylating Serine 68-phosphorylated Phospholemman. J Biol Chem 2015; 291:4561-79. [PMID: 26668322 DOI: 10.1074/jbc.m115.677898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Indexed: 11/06/2022] Open
Abstract
The sodium (Na(+))-calcium (Ca(2+)) exchanger 1 (NCX1) is an important regulator of intracellular Ca(2+) homeostasis. Serine 68-phosphorylated phospholemman (pSer-68-PLM) inhibits NCX1 activity. In the context of Na(+)/K(+)-ATPase (NKA) regulation, pSer-68-PLM is dephosphorylated by protein phosphatase 1 (PP1). PP1 also associates with NCX1; however, the molecular basis of this association is unknown. In this study, we aimed to analyze the mechanisms of PP1 targeting to the NCX1-pSer-68-PLM complex and hypothesized that a direct and functional NCX1-PP1 interaction is a prerequisite for pSer-68-PLM dephosphorylation. Using a variety of molecular techniques, we show that PP1 catalytic subunit (PP1c) co-localized, co-fractionated, and co-immunoprecipitated with NCX1 in rat cardiomyocytes, left ventricle lysates, and HEK293 cells. Bioinformatic analysis, immunoprecipitations, mutagenesis, pulldown experiments, and peptide arrays constrained PP1c anchoring to the K(I/V)FF motif in the first Ca(2+) binding domain (CBD) 1 in NCX1. This binding site is also partially in agreement with the extended PP1-binding motif K(V/I)FF-X5-8Φ1Φ2-X8-9-R. The cytosolic loop of NCX1, containing the K(I/V)FF motif, had no effect on PP1 activity in an in vitro assay. Dephosphorylation of pSer-68-PLM in HEK293 cells was not observed when NCX1 was absent, when the K(I/V)FF motif was mutated, or when the PLM- and PP1c-binding sites were separated (mimicking calpain cleavage of NCX1). Co-expression of PLM and NCX1 inhibited NCX1 current (both modes). Moreover, co-expression of PLM with NCX1(F407P) (mutated K(I/V)FF motif) resulted in the current being completely abolished. In conclusion, NCX1 is a substrate-specifying PP1c regulator protein, indirectly regulating NCX1 activity through pSer-68-PLM dephosphorylation.
Collapse
Affiliation(s)
- Tandekile Lubelwana Hafver
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Kjetil Hodne
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway, the Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences (NMBU), 0454 Oslo, Norway
| | - Pimthanya Wanichawan
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Jan Magnus Aronsen
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the Bjørknes College, Oslo, Norway
| | - Bjørn Dalhus
- the Department of Microbiology, Oslo University Hospital, Rikshospitalet, 0424 Oslo, Norway, the Department of Medical Biochemistry, Institute for Clinical Medicine, University of Oslo, 0424 Oslo, Norway and
| | - Per Kristian Lunde
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Marianne Lunde
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Marita Martinsen
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Ulla Helene Enger
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - William Fuller
- the Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Dundee, Scotland, United Kingdom DD1 9SY
| | - Ivar Sjaastad
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - William Edward Louch
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Ole Mathias Sejersted
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Cathrine Rein Carlson
- From the Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway, the KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway,
| |
Collapse
|
48
|
Wang G, Tang C, Yan G, Feng B. Gene Expression Profiling of H9c2 Cells Subjected to H2O2-Induced Apoptosis with/without AF-HF001. Biol Pharm Bull 2015; 39:207-14. [PMID: 26607605 DOI: 10.1248/bpb.b15-00601] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heart failure represents a major health problem. The development of new drugs to treat this condition is essential. We previously discovered that AF-001 attenuates the cardiac defects caused by heart failure in zebrafish. In this paper, we report the identification of AF-HF001, an AF-001 derivative, and its effects on live cardiomyocytes subjected to oxidative damage. The in vitro results demonstrated that AF-HF001 attenuates the production of reactive oxygen species (ROS) and the myocardial cell apoptosis. A DNA microarray was performed to broadly analyze gene expression after H2O2 treatment with or without AF-HF001. Hierarchical clustering analysis revealed that AF-HF001 modifies the expression of certain genes (Ndufs2, Ndufb6, Ndufb8, Ndufa13, Ndufs3, Ndufs5, TPM1, MYH14, RyR1, and TIMP4) related to ROS production, cardiac contractility and extracellular matrix remodeling. AF-HF001 ameliorates oxidative damage, which may be related to the mitogen-activated protein kinase (MAPK) family and the intrinsic mitochondrial pathway. Altogether, this study suggests that AF-HF001 exhibits potential as a clinical drug candidate for the treatment of heart failure.
Collapse
Affiliation(s)
- Guping Wang
- School of Pharmaceutical Science, Jiangnan University
| | | | | | | |
Collapse
|
49
|
Gadeberg HC, Bryant SM, James AF, Orchard CH. Altered Na/Ca exchange distribution in ventricular myocytes from failing hearts. Am J Physiol Heart Circ Physiol 2015; 310:H262-8. [PMID: 26566728 PMCID: PMC4796630 DOI: 10.1152/ajpheart.00597.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/11/2015] [Indexed: 11/22/2022]
Abstract
In mammalian cardiac ventricular myocytes, Ca efflux via Na/Ca exchange (NCX) occurs predominantly at T tubules. Heart failure is associated with disrupted t-tubular structure, but its effect on t-tubular function is less clear. We therefore investigated t-tubular NCX activity in ventricular myocytes isolated from rat hearts ∼18 wk after coronary artery ligation (CAL) or corresponding sham operation (Sham). NCX current (INCX) and l-type Ca current (ICa) were recorded using the whole cell, voltage-clamp technique in intact and detubulated (DT) myocytes; intracellular free Ca concentration ([Ca]i) was monitored simultaneously using fluo-4. INCX was activated and measured during application of caffeine to release Ca from sarcoplasmic reticulum (SR). Whole cell INCX was not significantly different in Sham and CAL myocytes and occurred predominantly in the T tubules in Sham myocytes. CAL was associated with redistribution of INCX and ICa away from the T tubules to the cell surface and an increase in t-tubular INCX/ICa density from 0.12 in Sham to 0.30 in CAL myocytes. The decrease in t-tubular INCX in CAL myocytes was accompanied by an increase in the fraction of Ca sequestered by SR. However, SR Ca content was not significantly different in Sham, Sham DT, and CAL myocytes but was significantly increased by DT of CAL myocytes. In Sham myocytes, there was hysteresis between INCX and [Ca]i, which was absent in DT Sham but present in CAL and DT CAL myocytes. These data suggest altered distribution of NCX in CAL myocytes.
Collapse
Affiliation(s)
- Hanne C Gadeberg
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Simon M Bryant
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Andrew F James
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Clive H Orchard
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
50
|
Fischer TH, Herting J, Eiringhaus J, Pabel S, Hartmann NH, Ellenberger D, Friedrich M, Renner A, Gummert J, Maier LS, Zabel M, Hasenfuss G, Sossalla S. Sex-dependent alterations of Ca2+ cycling in human cardiac hypertrophy and heart failure. Europace 2015; 18:1440-8. [PMID: 26493982 DOI: 10.1093/europace/euv313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 08/19/2015] [Indexed: 11/12/2022] Open
Abstract
AIMS Clinical studies have shown differences in the propensity for malignant ventricular arrhythmias between women and men suffering from cardiomyopathies and heart failure (HF). This is clinically relevant as it impacts therapies like prophylactic implantable cardioverter-defibrillator implantation but the pathomechanisms are unknown. As an increased sarcoplasmic reticulum (SR) Ca(2+) leak is arrhythmogenic, it could represent a cellular basis for this paradox. METHODS/RESULTS We evaluated the SR Ca(2+) leak with respect to sex differences in (i) afterload-induced cardiac hypertrophy (Hy) with preserved left ventricular (LV) function and (ii) end-stage HF. Cardiac function did not differ between sexes in both cardiac pathologies. Human cardiomyocytes isolated from female patients with Hy showed a significantly lower Ca(2+) spark frequency (CaSpF, confocal microscopy, Fluo3-AM) compared with men (P < 0.05). As Ca(2+) spark width and duration were similar in women and men, this difference in CaSpF did not yet translate into a significant difference of the calculated SR Ca(2+) leak between both sexes at this stage of disease (P = 0.14). Epifluorescence measurements (Fura2-AM) revealed comparable Ca(2+) cycling properties (diastolic Ca(2+) levels, amplitude of systolic Ca(2+) transients, SR Ca(2+) load) in patients of both sexes suffering from Hy. Additionally, the increased diastolic CaSpF in male patients with Hy did not yet translate into an elevated ratio of cells showing arrhythmic events (Ca(2+) waves, spontaneous Ca(2+) transients) (P = 0.77). In the transition to HF, both sexes showed an increase of the CaSpF (P < 0.05) and the sex dependence was even more pronounced. Female patients had a 69 ± 10% lower SR Ca(2+) leak (P < 0.05), which now even translated into a lower ratio of arrhythmic cells in female HF patients compared with men (P < 0.001). CONCLUSION These data show that the SR Ca(2+) leak is lower in women than in men with comparable cardiac impairment. Since the SR Ca(2+) leak triggers delayed afterdepolarizations, our findings may explain why women are less prone to ventricular arrhythmias and confirm the rationale of therapeutic measures reducing the SR Ca(2+) leak.
Collapse
Affiliation(s)
- Thomas H Fischer
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Jonas Herting
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Jörg Eiringhaus
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Steffen Pabel
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Nico H Hartmann
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - David Ellenberger
- Institut für Medizinische Statistik, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Martin Friedrich
- Abt. Thorax-, Herz- und Gefäßchirurgie, Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - André Renner
- Abt. Thorax-, Herz-, Gefäßchirurgie, Herz- und Diabeteszentrum Nordrheinwestfalen, Bad Oeynhausen, Germany
| | - Jan Gummert
- Abt. Thorax-, Herz-, Gefäßchirurgie, Herz- und Diabeteszentrum Nordrheinwestfalen, Bad Oeynhausen, Germany
| | - Lars S Maier
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Markus Zabel
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Göttingen, Germany
| | - Gerd Hasenfuss
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Göttingen, Germany
| | - Samuel Sossalla
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Göttingen, Germany
| |
Collapse
|