1
|
Naryzhnaya NV, Logvinov SV, Kurbatov BK, Derkachev IA, Mustafina LR, Gorbunov AS, Sirotina MA, Kilin M, Gusakova SV, Maslov LN. The β 2-adrenergic receptor agonist formoterol attenuates necrosis and apoptosis in the rat myocardium under experimental stress-induced cardiac injury. Fundam Clin Pharmacol 2024; 38:1116-1130. [PMID: 38956972 DOI: 10.1111/fcp.13026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Currently, there is no effective therapy for takotsubo syndrome (stress-induced cardiac injury in humans) in the clinics. It has previously been shown that β2-adrenergic receptor (β2-AR) agonist formoterol reduces cardiomyocyte injury in experimental takotsubo syndrome. OBJECTIVES The aim of this study was to investigate whether formoterol prevents apoptosis and necrosis of cardiomyocytes and endothelial cells in stress-induced cardiomyopathy. METHODS Stress-induced cardiac injury was induced by immobilization of rats for 2, 6, and 24 hours. RESULTS The myocardium of stressed rats showed a reduction in contractility and histological manifestations of cardiomyocyte damage: karyopyknosis, perinuclear edema of cardiomyocytes and endothelial cells, and microcirculation disturbances augmented with extended exposure to stress. In addition, apoptosis of endothelial cells was detected 6 hours after the onset of stress and peaked at 24 hours. Apoptosis of cardiomyocytes significantly gained only after 24 hours of stress exposure. These morphological alterations were associated with increased levels of serum creatine kinase-MB, syndecan-1, and thrombomodulin after 24 hours of stress. Administration of β2-AR agonist formoterol (50 μg/kg) four times during 24-hour stress exposure led to the improvement in myocardial inotropy, decrease in the severity of histological signatures, reduction in the number of TUNEL-positive cardiomyocytes, serum creatine kinase-MB, syndecan-1, and thrombomodulin levels. CONCLUSION Present data suggest that apoptosis and necrosis of cardiomyocytes and necrosis of endothelial cells in stress-induced cardiac injury can be mitigated by activation of the β2-AR. However, formoterol did not eliminate completely cardiomyocyte apoptosis, histological alterations, or endothelium injury markers under stress.
Collapse
Affiliation(s)
- Natalia V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| | - Sergey V Logvinov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
- Siberian State Medical University, 2, Moskovsky tract, Tomsk, 634050, Russian Federation
| | - Boris K Kurbatov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| | - Ivan A Derkachev
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| | - Liliia R Mustafina
- Siberian State Medical University, 2, Moskovsky tract, Tomsk, 634050, Russian Federation
| | - Aleksandr S Gorbunov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| | - Maria A Sirotina
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| | - Mikhail Kilin
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| | - Svetlana V Gusakova
- Siberian State Medical University, 2, Moskovsky tract, Tomsk, 634050, Russian Federation
| | - Leonid N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia, 111a, Kievskaya str., Tomsk, 634012, Russian Federation
| |
Collapse
|
2
|
Loh YH, Lv J, Goh Y, Sun X, Zhu X, Muheyati M, Luan Y. Remodelling of T-Tubules and Associated Calcium Handling Dysfunction in Heart Failure: Mechanisms and Therapeutic Insights. Can J Cardiol 2024; 40:2569-2588. [PMID: 39455023 DOI: 10.1016/j.cjca.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
In cardiomyocytes, transverse tubules (T-tubules) are sarcolemmal invaginations that facilitate excitation-contraction coupling and diastolic function. The clinical significance of T-tubules has become evident in that their remodelling is recognised as a hallmark feature of heart failure (HF) and a key contributor to disrupted Ca2+ homeostasis, compromised cardiac function, and arrhythmogenesis. Further investigations have revealed that T-tubule remodelling is particularly pronounced in HF with reduced ejection fraction (HFrEF), but not in HF with preserved ejection fraction, implying that T-tubule remodelling may play a crucial pathophysiologic role in HFrEF. While research on the functional importance of T-tubules is ongoing, T-tubule remodelling has been found to be reversible. That finding has triggered a surge in studies aimed at identifying specific therapeutic approaches for HFrEF. This review discusses the functional importance of T-tubules and their microdomains, the pathophysiology of T-tubule remodelling, and the potential mechanisms of current HFrEF therapeutic approaches in reversing T-tubule alterations. We also highlight discrepancies regarding the roles of T-tubule proteins in the recovery process across studies to offer valuable insights for future research.
Collapse
Affiliation(s)
- Yi Hao Loh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Jingyi Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yenfang Goh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xiangjie Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xianfeng Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Critical Care Medicine, Hangzhou Ninth People's Hospital, China
| | - Muergen Muheyati
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; School of Medicine, Shaoxing University, China.
| |
Collapse
|
3
|
Caldwell JL, Clarke JD, Smith CER, Pinali C, Quinn CJ, Pearman CM, Adomaviciene A, Radcliffe EJ, Watkins A, Horn MA, Bode EF, Madders GWP, Eisner M, Eisner DA, Trafford AW, Dibb KM. Restoring Atrial T-Tubules Augments Systolic Ca Upon Recovery From Heart Failure. Circ Res 2024; 135:739-754. [PMID: 39140440 PMCID: PMC11392124 DOI: 10.1161/circresaha.124.324601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Transverse (t)-tubules drive the rapid and synchronous Ca2+ rise in cardiac myocytes. The virtual complete atrial t-tubule loss in heart failure (HF) decreases Ca2+ release. It is unknown if or how atrial t-tubules can be restored and how this affects systolic Ca2+. METHODS HF was induced in sheep by rapid ventricular pacing and recovered following termination of rapid pacing. Serial block-face scanning electron microscopy and confocal imaging were used to study t-tubule ultrastructure. Function was assessed using patch clamp, Ca2+, and confocal imaging. Candidate proteins involved in atrial t-tubule recovery were identified by western blot and expressed in rat neonatal ventricular myocytes to determine if they altered t-tubule structure. RESULTS Atrial t-tubules were lost in HF but reappeared following recovery from HF. Recovered t-tubules were disordered, adopting distinct morphologies with increased t-tubule length and branching. T-tubule disorder was associated with mitochondrial disorder. Recovered t-tubules were functional, triggering Ca2+ release in the cell interior. Systolic Ca2+, ICa-L, sarcoplasmic reticulum Ca2+ content, and sarcoendoplasmic reticulum Ca2+ ATPase function were restored following recovery from HF. Confocal microscopy showed fragmentation of ryanodine receptor staining and movement away from the z-line in HF, which was reversed following recovery from HF. Acute detubulation, to remove recovered t-tubules, confirmed their key role in restoration of the systolic Ca2+ transient, the rate of Ca2+ removal, and the peak L-type Ca2+ current. The abundance of telethonin and myotubularin decreased during HF and increased during recovery. Transfection with these proteins altered the density and structure of tubules in neonatal myocytes. Myotubularin had a greater effect, increasing tubule length and branching, replicating that seen in the recovery atria. CONCLUSIONS We show that recovery from HF restores atrial t-tubules, and this promotes recovery of ICa-L, sarcoplasmic reticulum Ca2+ content, and systolic Ca2+. We demonstrate an important role for myotubularin in t-tubule restoration. Our findings reveal a new and viable therapeutic strategy.
Collapse
Affiliation(s)
- Jessica L Caldwell
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Jessica D Clarke
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Charlotte E R Smith
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Christian Pinali
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Callum J Quinn
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Charles M Pearman
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Aiste Adomaviciene
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Emma J Radcliffe
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Amy Watkins
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Margaux A Horn
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Elizabeth F Bode
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - George W P Madders
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Mark Eisner
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - David A Eisner
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| | - Katharine M Dibb
- Unit of Cardiac Physiology, Manchester Academic Health Science Centre, University of Manchester, United Kingdom
| |
Collapse
|
4
|
Mone P, Agyapong ED, Morciano G, Jankauskas SS, De Luca A, Varzideh F, Pinton P, Santulli G. Dysfunctional mitochondria elicit bioenergetic decline in the aged heart. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:13. [PMID: 39015481 PMCID: PMC11250775 DOI: 10.20517/jca.2023.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Aging represents a complex biological progression affecting the entire body, marked by a gradual decline in tissue function, rendering organs more susceptible to stress and diseases. The human heart holds significant importance in this context, as its aging process poses life-threatening risks. It entails macroscopic morphological shifts and biochemical changes that collectively contribute to diminished cardiac function. Among the numerous pivotal factors in aging, mitochondria play a critical role, intersecting with various molecular pathways and housing several aging-related agents. In this comprehensive review, we provide an updated overview of the functional role of mitochondria in cardiac aging.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Esther Densu Agyapong
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Vanvitelli University, Naples 80100, Italy
| | - Fahimeh Varzideh
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
- Department of Advanced Biomedical Sciences, “Federico II” University, International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples 80131, Italy
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
5
|
Li J, Richmond B, Cluntun AA, Bia R, Walsh MA, Shaw K, Symons JD, Franklin S, Rutter J, Funai K, Shaw RM, Hong T. Cardiac gene therapy treats diabetic cardiomyopathy and lowers blood glucose. JCI Insight 2023; 8:e166713. [PMID: 37639557 PMCID: PMC10561727 DOI: 10.1172/jci.insight.166713] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 08/15/2023] [Indexed: 08/31/2023] Open
Abstract
Diabetic cardiomyopathy, an increasingly global epidemic and a major cause of heart failure with preserved ejection fraction (HFpEF), is associated with hyperglycemia, insulin resistance, and intracardiomyocyte calcium mishandling. Here we identify that, in db/db mice with type 2 diabetes-induced HFpEF, abnormal remodeling of cardiomyocyte transverse-tubule microdomains occurs with downregulation of the membrane scaffolding protein cardiac bridging integrator 1 (cBIN1). Transduction of cBIN1 by AAV9 gene therapy can restore transverse-tubule microdomains to normalize intracellular distribution of calcium-handling proteins and, surprisingly, glucose transporter 4 (GLUT4). Cardiac proteomics revealed that AAV9-cBIN1 normalized components of calcium handling and GLUT4 translocation machineries. Functional studies further identified that AAV9-cBIN1 normalized insulin-dependent glucose uptake in diabetic cardiomyocytes. Phenotypically, AAV9-cBIN1 rescued cardiac lusitropy, improved exercise intolerance, and ameliorated hyperglycemia in diabetic mice. Restoration of transverse-tubule microdomains can improve cardiac function in the setting of diabetic cardiomyopathy and can also improve systemic glycemic control.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology and Toxicology, College of Pharmacy
- Nora Eccles Harrison Cardiovascular Research and Training Institute
| | | | | | - Ryan Bia
- Nora Eccles Harrison Cardiovascular Research and Training Institute
| | - Maureen A. Walsh
- College of Health, Department of Nutrition and Integrative Physiology, Program in Molecular Medicine
| | - Kikuyo Shaw
- Department of Pharmacology and Toxicology, College of Pharmacy
| | - J. David Symons
- College of Health, Department of Nutrition and Integrative Physiology, Program in Molecular Medicine
- Diabetes & Metabolism Research Center, and
| | - Sarah Franklin
- Nora Eccles Harrison Cardiovascular Research and Training Institute
| | - Jared Rutter
- Department of Biochemistry
- College of Health, Department of Nutrition and Integrative Physiology, Program in Molecular Medicine
- Diabetes & Metabolism Research Center, and
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, USA
| | - Katsuhiko Funai
- College of Health, Department of Nutrition and Integrative Physiology, Program in Molecular Medicine
- Diabetes & Metabolism Research Center, and
| | - Robin M. Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute
| | - TingTing Hong
- Department of Pharmacology and Toxicology, College of Pharmacy
- Nora Eccles Harrison Cardiovascular Research and Training Institute
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
6
|
MacLeod KT. Changes in cellular Ca 2+ and Na + regulation during the progression towards heart failure. J Physiol 2023; 601:905-921. [PMID: 35946572 PMCID: PMC10952717 DOI: 10.1113/jp283082] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
In adapting to disease and loss of tissue, the heart shows great phenotypic plasticity that involves changes to its structure, composition and electrophysiology. Together with parallel whole body cardiovascular adaptations, the initial decline in cardiac function resulting from the insult is compensated. However, in the long term, the heart muscle begins to fail and patients with this condition have a very poor prognosis, with many dying from disturbances of rhythm. The surviving myocytes of these hearts gain Na+ , which is positively inotropic because of alterations to Ca2+ fluxes mediated by the Na+ /Ca2+ exchange, but compromises Ca2+ -dependent energy metabolism in mitochondria. Uptake of Ca2+ into the sarcoplasmic reticulum (SR) is reduced because of diminished function of SR Ca2+ ATPases. The result of increased Ca2+ influx and reduced SR Ca2+ uptake is an increase in the diastolic cytosolic Ca2+ concentration, which promotes spontaneous SR Ca2+ release and induces delayed afterdepolarisations. Action potential duration prolongs because of increased late Na+ current and changes in expression and function of other ion channels and transporters increasing the probability of the formation of early afterdepolarisations. There is a reduction in T-tubule density and so the normal spatial arrangements required for efficient excitation-contraction coupling are compromised and lead to temporal delays in Ca2+ release from the SR. Therefore, the structural and electrophysiological responses that occur to provide compensation do so at the expense of (1) increasing the likelihood of arrhythmogenesis; (2) activating hypertrophic, apoptotic and Ca2+ signalling pathways; and (3) decreasing the efficiency of SR Ca2+ release.
Collapse
Affiliation(s)
- Kenneth T. MacLeod
- National Heart & Lung InstituteImperial Centre for Translational and Experimental MedicineImperial CollegeHammersmith HospitalLondonUK
| |
Collapse
|
7
|
Dixon RE, Trimmer JS. Endoplasmic Reticulum-Plasma Membrane Junctions as Sites of Depolarization-Induced Ca 2+ Signaling in Excitable Cells. Annu Rev Physiol 2023; 85:217-243. [PMID: 36202100 PMCID: PMC9918718 DOI: 10.1146/annurev-physiol-032122-104610] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Membrane contact sites between endoplasmic reticulum (ER) and plasma membrane (PM), or ER-PM junctions, are found in all eukaryotic cells. In excitable cells they play unique roles in organizing diverse forms of Ca2+ signaling as triggered by membrane depolarization. ER-PM junctions underlie crucial physiological processes such as excitation-contraction coupling, smooth muscle contraction and relaxation, and various forms of activity-dependent signaling and plasticity in neurons. In many cases the structure and molecular composition of ER-PM junctions in excitable cells comprise important regulatory feedback loops linking depolarization-induced Ca2+ signaling at these sites to the regulation of membrane potential. Here, we describe recent findings on physiological roles and molecular composition of native ER-PM junctions in excitable cells. We focus on recent studies that provide new insights into canonical forms of depolarization-induced Ca2+ signaling occurring at junctional triads and dyads of striated muscle, as well as the diversity of ER-PM junctions in these cells and in smooth muscle and neurons.
Collapse
Affiliation(s)
- Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California, USA;
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California, USA;
| |
Collapse
|
8
|
Mercurio V, Ambrosio G, Correale M, Dini FL, Ghio S, Nodari S, Palazzuoli A, Ruocco G, Pedrinelli R, Mercuro G, Filardi PP, Indolfi C, Agostoni P, Tocchetti CG, Paolillo S. Innovations in medical therapy of heart failure with reduced ejection fraction. J Cardiovasc Med (Hagerstown) 2022; 24:e47-e54. [PMID: 36729606 DOI: 10.2459/jcm.0000000000001413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heart failure with reduced ejection fraction (HFrEF) is a pathological condition still characterized by high rates of mortality and disease exacerbation frequently leading to hospitalization, thus there is a continuous need for pharmacological treatments impacting on disease stability and long-term prognosis. Moreover, the phenotype of heart failure patients is continuously changing over time, and the development of new heart failure drugs is crucial to promote a personalized and targeted approach. In recent years, several therapeutic innovations have emerged in the landscape of acute and chronic HFrEF, largely changing and improving our approach to the disease. Various studies on new drugs and experimental therapeutic approaches are ongoing. The present review discusses the latest data on both recently approved drugs and developing therapeutic targets, in order to provide a critical overview for an informed and optimal approach to such a complex disease.
Collapse
Affiliation(s)
- Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples.,Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University
| | | | | | - Frank L Dini
- Cardiac, Thoracic and Vascular Department, University of Pisa, Pisa
| | - Stefano Ghio
- Division of Cardiology, Fondazione IRCCS Policlinico S.Matteo, Pavia
| | - Savina Nodari
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia
| | - Alberto Palazzuoli
- Cardiovascular Disease Unit, Department of Internal Medicine, University of Siena, Siena
| | - Gaetano Ruocco
- Cardiology Unit, Riuniti of Valdichiana Hospitals, USL Sud Est Toscana, Montepulciano
| | - Roberto Pedrinelli
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell'Area Critica, Università di Pisa
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, Federico II University, Naples.,Mediterranea Cardiocentro, Naples
| | - Ciro Indolfi
- Cardiology Unit, University Magna Graecia of Catanzaro, Catanza
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS.,Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milan
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples.,Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University.,Interdepartmental Hypertension Research Center (CIRIAPA).,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples.,Mediterranea Cardiocentro, Naples
| |
Collapse
|
9
|
Onohara D, Corporan DM, Kono T, Kumar S, Guyton RA, Padala M. Ventricular reshaping with a beating heart implant improves pump function in experimental heart failure. J Thorac Cardiovasc Surg 2022; 163:e343-e355. [PMID: 33046233 PMCID: PMC7925703 DOI: 10.1016/j.jtcvs.2020.08.097] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVE The left ventricle remodels from an ellipsoidal/conical shape to a spherical shape after a myocardial infarction. The spherical ventricle is inefficient as a pumping chamber, has higher wall stresses, and can lead to congestive heart failure. We sought to investigate if restoring physiological ventricular shape with a beating heart implant improves pump function. METHODS Rats were induced with a myocardial infarction, developing left ventricular dilatation and dysfunction, and becoming spherical over 3 weeks. Thereafter, they were randomized to undergo left ventricular reshaping with a beating heart implant (n = 19) or continue follow-up without an implant (n = 19). Biweekly echocardiography was performed until 12 weeks, with half the rats euthanized at 6 weeks and remaining at 12 weeks. At termination, invasive hemodynamic parameters and histopathology were performed. RESULTS At 3 weeks after the infarction, rats had a 22% fall in ejection fraction, 31% rise in end diastolic volume, and 23% rise in sphericity. Transventricular implant reshaping reduced the volume by 12.6% and sphericity by 21%, restoring physiologic ventricular shape and wall stress. Over the 12-week follow-up, pump function improved significantly with better ventricular-vascular coupling in the reshaped hearts. In this group, cardiomyocyte cross-section area was higher and the cells were less elongated. CONCLUSIONS Reshaping a postinfarction, failing left ventricle to restore its physiological conical shape significantly improves long-term pump function.
Collapse
Affiliation(s)
- Daisuke Onohara
- Structural Heart Research & Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, Ga
| | - Daniella M Corporan
- Structural Heart Research & Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, Ga
| | - Takanori Kono
- Structural Heart Research & Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, Ga
| | - Sandeep Kumar
- Joint Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, Ga
| | - Robert A Guyton
- Structural Heart Research & Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, Ga; Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Ga
| | - Muralidhar Padala
- Structural Heart Research & Innovation Laboratory, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, Ga; Joint Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, Ga; Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Ga.
| |
Collapse
|
10
|
Wright P, Gorelik J. Junctophillin-2: Coupling Hopes for Cardiac Gene Therapy to Gene Transcription. Circ Res 2022; 130:1318-1320. [PMID: 35482830 DOI: 10.1161/circresaha.122.321066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Peter Wright
- School of Life and Health Sciences, University of Roehampton, London, United Kingdom (P.W.)
- National Heart, and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, United Kingdom (P.W., J.G.)
| | - Julia Gorelik
- National Heart, and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, United Kingdom (P.W., J.G.)
| |
Collapse
|
11
|
Dixon RE. Nanoscale Organization, Regulation, and Dynamic Reorganization of Cardiac Calcium Channels. Front Physiol 2022; 12:810408. [PMID: 35069264 PMCID: PMC8769284 DOI: 10.3389/fphys.2021.810408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
The architectural specializations and targeted delivery pathways of cardiomyocytes ensure that L-type Ca2+ channels (CaV1.2) are concentrated on the t-tubule sarcolemma within nanometers of their intracellular partners the type 2 ryanodine receptors (RyR2) which cluster on the junctional sarcoplasmic reticulum (jSR). The organization and distribution of these two groups of cardiac calcium channel clusters critically underlies the uniform contraction of the myocardium. Ca2+ signaling between these two sets of adjacent clusters produces Ca2+ sparks that in health, cannot escalate into Ca2+ waves because there is sufficient separation of adjacent clusters so that the release of Ca2+ from one RyR2 cluster or supercluster, cannot activate and sustain the release of Ca2+ from neighboring clusters. Instead, thousands of these Ca2+ release units (CRUs) generate near simultaneous Ca2+ sparks across every cardiomyocyte during the action potential when calcium induced calcium release from RyR2 is stimulated by depolarization induced Ca2+ influx through voltage dependent CaV1.2 channel clusters. These sparks summate to generate a global Ca2+ transient that activates the myofilaments and thus the electrical signal of the action potential is transduced into a functional output, myocardial contraction. To generate more, or less contractile force to match the hemodynamic and metabolic demands of the body, the heart responds to β-adrenergic signaling by altering activity of calcium channels to tune excitation-contraction coupling accordingly. Recent accumulating evidence suggests that this tuning process also involves altered expression, and dynamic reorganization of CaV1.2 and RyR2 channels on their respective membranes to control the amplitude of Ca2+ entry, SR Ca2+ release and myocardial function. In heart failure and aging, altered distribution and reorganization of these key Ca2+ signaling proteins occurs alongside architectural remodeling and is thought to contribute to impaired contractile function. In the present review we discuss these latest developments, their implications, and future questions to be addressed.
Collapse
Affiliation(s)
- Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
12
|
Abstract
Junctophilins (JPHs) comprise a family of structural proteins that connect the plasma membrane to intracellular organelles such as the endo/sarcoplasmic reticulum. Tethering of these membrane structures results in the formation of highly organized subcellular junctions that play important signaling roles in all excitable cell types. There are four JPH isoforms, expressed primarily in muscle and neuronal cell types. Each JPH protein consists of 6 'membrane occupation and recognition nexus' (MORN) motifs, a joining region connecting these to another set of 2 MORN motifs, a putative alpha-helical region, a divergent region exhibiting low homology between JPH isoforms, and a carboxy-terminal transmembrane region anchoring into the ER/SR membrane. JPH isoforms play essential roles in developing and maintaining subcellular membrane junctions. Conversely, inherited mutations in JPH2 cause hypertrophic or dilated cardiomyopathy, while trinucleotide expansions in the JPH3 gene cause Huntington Disease-Like 2. Loss of JPH1 protein levels can cause skeletal myopathy, while loss of cardiac JPH2 levels causes heart failure and atrial fibrillation, among other disease. This review will provide a comprehensive overview of the JPH gene family, phylogeny, and evolutionary analysis of JPH genes and other MORN domain proteins. JPH biogenesis, membrane tethering, and binding partners will be discussed, as well as functional roles of JPH isoforms in excitable cells. Finally, potential roles of JPH isoform deficits in human disease pathogenesis will be reviewed.
Collapse
Affiliation(s)
- Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Department of Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States; Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Pediatrics (Cardiology), Neuroscience, and Center for Space Medicine, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
13
|
Pacheco C, Wei J, Shufelt C, C. Hitzeman T, Cook‐Wiens G, Pepine CJ, Handberg E, Anderson RD, Petersen J, Hong T, M. Shaw R, Bairey Merz CN. Association of coronary microvascular dysfunction and cardiac bridge integrator 1, a cardiomyocyte dysfunction biomarker. Clin Cardiol 2021; 44:1586-1593. [PMID: 34528718 PMCID: PMC8571552 DOI: 10.1002/clc.23726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Coronary microvascular dysfunction (CMD) is associated with heart failure with preserved ejection fraction (HFpEF); however, pathophysiology is not well described. HYPOTHESIS We hypothesized that CMD in women with suspected ischemia with no obstructive coronary artery disease (INOCA) is associated with cardiomyocyte dysfunction reflected by plasma levels of a cardiomyocyte calcium handling protein, cardiac bridge integrator 1 (cBIN1). METHODS Women with suspected INOCA undergoing coronary function testing were included. Coronary flow reserve, vasodilation to nitroglycerin, change in coronary blood flow (ΔCBF), and vasodilation to acetylcholine (ΔAch) were evaluated. cBIN1 score (CS) levels in these women (n = 39) were compared to women with HFpEF (n = 20), heart failure with reduced ejection fraction (HFrEF) (n = 36), and reference controls (RC) (n = 50). Higher CS indicates cardiomyocyte tubule dysfunction. RESULTS INOCA, HFpEF, and HFrEF women were older than RC (p < .05). Higher CS was associated with vasoconstriction to acetylcholine (r = -0.43, p = .011) with a trend towards lower ΔCBF (r = 0.30, p = .086). Higher CS was specific for ΔAch and ΔCBF but had limited sensitivity. INOCA women had higher CS than RC, but lower CS than HFpEF/HFrEF groups (p < .001). CONCLUSIONS CS, a plasma biomarker indicating poor cardiomyocyte health, was higher in women with suspected INOCA as compared to RC, but lower than in women with HFpEF. Elevated CS in suspected INOCA patients represents an intermediate group between health and disease, supporting the hypothesis that CMD may progress to HFpEF. Larger prospective cohort studies are needed to confirm the pathophysiological relationship between cBIN1, CMD, and HFpEF.
Collapse
Affiliation(s)
- Christine Pacheco
- Hôpital Pierre‐BoucherCardiology Service, Department of Medicine, Université de MontréalLongueuilQuebecCanada
- Centre Hospitalier de l'Université de Montréal (CHUM), Cardiology Service, Department of Medicine, Université de MontréalMontrealQuebecCanada
| | - Janet Wei
- Barbra Streisand Women's Heart CenterSmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Chrisandra Shufelt
- Barbra Streisand Women's Heart CenterSmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tara C. Hitzeman
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUtahUSA
| | - Galen Cook‐Wiens
- Barbra Streisand Women's Heart CenterSmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Carl J. Pepine
- Cardiovascular Division, University of FloridaGainesvilleFloridaUSA
| | - Eileen Handberg
- Cardiovascular Division, University of FloridaGainesvilleFloridaUSA
| | | | - John Petersen
- Cardiovascular Division, University of FloridaGainesvilleFloridaUSA
| | - TingTing Hong
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Robin M. Shaw
- Nora Eccles Harrison Cardiovascular Research and Training InstituteUniversity of UtahSalt Lake CityUtahUSA
| | - C. Noel Bairey Merz
- Barbra Streisand Women's Heart CenterSmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| |
Collapse
|
14
|
Calpain-2 specifically cleaves Junctophilin-2 at the same site as Calpain-1 but with less efficacy. Biochem J 2021; 478:3539-3553. [PMID: 34524407 PMCID: PMC8589432 DOI: 10.1042/bcj20210629] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022]
Abstract
Calpain proteolysis contributes to the pathogenesis of heart failure but the calpain isoforms responsible and their substrate specificities have not been rigorously defined. One substrate, Junctophilin-2 (JP2), is essential for maintaining junctional cardiac dyads and excitation-contraction coupling. We previously demonstrated that mouse JP2 is cleaved by calpain-1 (CAPN1) between Arginine 565 (R565) and Threonine 566 (T566). Recently, calpain-2 (CAPN2) was reported to cleave JP2 at a novel site between Glycine 482 (G482) and Threonine 483 (T483). We aimed to directly compare the contributions of each calpain isoform, their Ca2+ sensitivity, and their cleavage site selection for JP2. We find CAPN1, CAPN2 and their requisite CAPNS1 regulatory subunit are induced by pressure overload stress that is concurrent with JP2 cleavage. Using in vitro calpain cleavage assays, we demonstrate that CAPN1 and CAPN2 cleave JP2 into similar 75 kD N-terminal (JP2NT) and 25 kD C-terminal fragments (JP2CT) with CAPNS1 co-expression enhancing proteolysis. Deletion mutagenesis shows both CAPN1 and CAPN2 require R565/T566 but not G482/T483. When heterologously expressed, the JP2CT peptide corresponding to R565/T566 cleavage approximates the 25 kD species found during cardiac stress while the C-terminal peptide from potential cleavage at G482/T483 produces a 35 kD product. Similar results were obtained for human JP2. Finally, we show that CAPN1 has higher Ca2+ sensitivity and cleavage efficacy than CAPN2 on JP2 and other cardiac substrates including cTnT, cTnI and β2-spectrin. We conclude that CAPN2 cleaves JP2 at the same functionally conserved R565/T566 site as CAPN1 but with less efficacy and suggest heart failure may be targeted through specific inhibition of CAPN1.
Collapse
|
15
|
Abstract
Scanning ion conductance microscopy (SICM) has emerged as a versatile tool for studies of interfaces in biology and materials science with notable utility in biophysical and electrochemical measurements. The heart of the SICM is a nanometer-scale electrolyte filled glass pipette that serves as a scanning probe. In the initial conception, manipulations of ion currents through the tip of the pipette and appropriate positioning hardware provided a route to recording micro- and nanoscopic mapping of the topography of surfaces. Subsequent advances in instrumentation, probe design, and methods significantly increased opportunities for SICM beyond recording topography. Hybridization of SICM with coincident characterization techniques such as optical microscopy and faradaic electrodes have brought SICM to the forefront as a tool for nanoscale chemical measurement for a wide range of applications. Modern approaches to SICM realize an important tool in analytical, bioanalytical, biophysical, and materials measurements, where significant opportunities remain for further exploration. In this review, we chronicle the development of SICM from the perspective of both the development of instrumentation and methods and the breadth of measurements performed.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Kaixiang Huang
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Natasha P Siepser
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Lane A Baker
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
16
|
Setterberg IE, Le C, Frisk M, Li J, Louch WE. The Physiology and Pathophysiology of T-Tubules in the Heart. Front Physiol 2021; 12:718404. [PMID: 34566684 PMCID: PMC8458775 DOI: 10.3389/fphys.2021.718404] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
In cardiomyocytes, invaginations of the sarcolemmal membrane called t-tubules are critically important for triggering contraction by excitation-contraction (EC) coupling. These structures form functional junctions with the sarcoplasmic reticulum (SR), and thereby enable close contact between L-type Ca2+ channels (LTCCs) and Ryanodine Receptors (RyRs). This arrangement in turn ensures efficient triggering of Ca2+ release, and contraction. While new data indicate that t-tubules are capable of exhibiting compensatory remodeling, they are also widely reported to be structurally and functionally compromised during disease, resulting in disrupted Ca2+ homeostasis, impaired systolic and/or diastolic function, and arrhythmogenesis. This review summarizes these findings, while highlighting an emerging appreciation of the distinct roles of t-tubules in the pathophysiology of heart failure with reduced and preserved ejection fraction (HFrEF and HFpEF). In this context, we review current understanding of the processes underlying t-tubule growth, maintenance, and degradation, underscoring the involvement of a variety of regulatory proteins, including junctophilin-2 (JPH2), amphiphysin-2 (BIN1), caveolin-3 (Cav3), and newer candidate proteins. Upstream regulation of t-tubule structure/function by cardiac workload and specifically ventricular wall stress is also discussed, alongside perspectives for novel strategies which may therapeutically target these mechanisms.
Collapse
Affiliation(s)
- Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Christopher Le
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Grote Beverborg N, Später D, Knöll R, Hidalgo A, Yeh ST, Elbeck Z, Silljé HHW, Eijgenraam TR, Siga H, Zurek M, Palmér M, Pehrsson S, Albery T, Bomer N, Hoes MF, Boogerd CJ, Frisk M, van Rooij E, Damle S, Louch WE, Wang QD, Fritsche-Danielson R, Chien KR, Hansson KM, Mullick AE, de Boer RA, van der Meer P. Phospholamban antisense oligonucleotides improve cardiac function in murine cardiomyopathy. Nat Commun 2021; 12:5180. [PMID: 34462437 PMCID: PMC8405807 DOI: 10.1038/s41467-021-25439-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a major cause of morbidity and mortality worldwide, highlighting an urgent need for novel treatment options, despite recent improvements. Aberrant Ca2+ handling is a key feature of HF pathophysiology. Restoring the Ca2+ regulating machinery is an attractive therapeutic strategy supported by genetic and pharmacological proof of concept studies. Here, we study antisense oligonucleotides (ASOs) as a therapeutic modality, interfering with the PLN/SERCA2a interaction by targeting Pln mRNA for downregulation in the heart of murine HF models. Mice harboring the PLN R14del pathogenic variant recapitulate the human dilated cardiomyopathy (DCM) phenotype; subcutaneous administration of PLN-ASO prevents PLN protein aggregation, cardiac dysfunction, and leads to a 3-fold increase in survival rate. In another genetic DCM mouse model, unrelated to PLN (Cspr3/Mlp-/-), PLN-ASO also reverses the HF phenotype. Finally, in rats with myocardial infarction, PLN-ASO treatment prevents progression of left ventricular dilatation and improves left ventricular contractility. Thus, our data establish that antisense inhibition of PLN is an effective strategy in preclinical models of genetic cardiomyopathy as well as ischemia driven HF.
Collapse
Affiliation(s)
- Niels Grote Beverborg
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daniela Später
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden.
| | - Ralph Knöll
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Alejandro Hidalgo
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
- Murdoch Children's Research Institute (MCRI), Flemington, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | | | - Zaher Elbeck
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Herman H W Silljé
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tim R Eijgenraam
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Humam Siga
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
| | - Magdalena Zurek
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Malin Palmér
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Laboratory of Experimental Biomedicine, Core Facilities, Sahlgrenska Academy, Gothenburg University, Göteborg, Sweden
| | - Susanne Pehrsson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tamsin Albery
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nils Bomer
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martijn F Hoes
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cornelis J Boogerd
- Department of Molecular Cardiology, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Eva van Rooij
- Department of Molecular Cardiology, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Regina Fritsche-Danielson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kenneth R Chien
- Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Huddinge, Sweden
- Department of Cell and Molecular Biology (CMB), Karolinska Institute, Stockholm, Sweden
| | - Kenny M Hansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Rudolf A de Boer
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
18
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
19
|
Li J, Richmond B, Hong T. Cardiac T-Tubule cBIN1-Microdomain, a Diagnostic Marker and Therapeutic Target of Heart Failure. Int J Mol Sci 2021; 22:ijms22052299. [PMID: 33669042 PMCID: PMC7956774 DOI: 10.3390/ijms22052299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Since its first identification as a cardiac transverse tubule (t-tubule) protein, followed by the cloning of the cardiac isoform responsible for t-tubule membrane microdomain formation, cardiac bridging integrator 1 (cBIN1) and its organized microdomains have emerged as a key mechanism in maintaining normal beat-to-beat heart contraction and relaxation. The abnormal remodeling of cBIN1-microdomains occurs in stressed and diseased cardiomyocytes, contributing to the pathophysiology of heart failure. Due to the homeostatic turnover of t-tubule cBIN1-microdomains via microvesicle release into the peripheral circulation, plasma cBIN1 can be assayed as a liquid biopsy of cardiomyocyte health. A new blood test cBIN1 score (CS) has been developed as a dimensionless inverse index derived from plasma cBIN1 concentration with a diagnostic and prognostic power for clinical outcomes in stable ambulatory patients with heart failure with reduced or preserved ejection fraction (HFrEF or HFpEF). Recent evidence further indicates that exogenous cBIN1 introduced by adeno-associated virus 9-based gene therapy can rescue cardiac contraction and relaxation in failing hearts. The therapeutic potential of cBIN1 gene therapy is enormous given its ability to rescue cardiac inotropy and provide lusitropic protection in the meantime. These unprecedented capabilities of cBIN1 gene therapy are shifting the current paradigm of therapy development for heart failure, particularly HFpEF.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - Bradley Richmond
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - TingTing Hong
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: ; Tel.: +1-801-581-3090
| |
Collapse
|
20
|
De Jong KA, Nikolaev VO. Multifaceted remodelling of cAMP microdomains driven by different aetiologies of heart failure. FEBS J 2021; 288:6603-6622. [DOI: 10.1111/febs.15706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Kirstie A. De Jong
- Institute of Experimental Cardiovascular Research University Medical Center Hamburg‐Eppendorf Hamburg Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck D‐20246 Hamburg Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research University Medical Center Hamburg‐Eppendorf Hamburg Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck D‐20246 Hamburg Germany
| |
Collapse
|
21
|
Wang S, Zhou Y, Luo Y, Kan R, Chen J, Xuan H, Wang C, Chen J, Xu T, Li D. SERCA2a ameliorates cardiomyocyte T-tubule remodeling via the calpain/JPH2 pathway to improve cardiac function in myocardial ischemia/reperfusion mice. Sci Rep 2021; 11:2037. [PMID: 33479390 PMCID: PMC7820433 DOI: 10.1038/s41598-021-81570-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
Transverse-tubules (T-tubules) play pivotal roles in Ca2+-induced, Ca2+ release and excitation–contraction coupling in cardiomyocytes. The purpose of this study was to uncover mechanisms where sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2a) improved cardiac function through T-tubule regulation during myocardial ischemia/reperfusion (I/R). SERCA2a protein expression, cytoplasmic [Ca2+]i, calpain activity, junctophilin-2 (JPH2) protein expression and intracellular localization, cardiomyocyte T-tubules, contractility and calcium transients in single cardiomyocytes and in vivo cardiac functions were all examined after SERCA2a knockout and overexpression, and Calpain inhibitor PD150606 (PD) pretreatment, following myocardial I/R. This comprehensive approach was adopted to clarify SERCA2a mechanisms in improving cardiac function in mice. Calpain was activated during myocardial I/R, and led to the proteolytic cleavage of JPH2. This altered the T-tubule network, the contraction function/calcium transients in cardiomyocytes and in vivo cardiac functions. During myocardial I/R, PD pretreatment upregulated JPH2 expression and restored it to its intracellular location, repaired the T-tubule network, and contraction function/calcium transients of cardiomyocytes and cardiac functions in vivo. SERCA2a suppressed calpain activity via [Ca2+]i, and ameliorated these key indices. Our results suggest that SERCA2a ameliorates cardiomyocyte T-tubule remodeling via the calpain/JPH2 pathway, thereby improving cardiac function in myocardial I/R mice.
Collapse
Affiliation(s)
- Shuai Wang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - You Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Rongsheng Kan
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Haochen Xuan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Chaofan Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Junhong Chen
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Lyon AR, Babalis D, Morley-Smith AC, Hedger M, Suarez Barrientos A, Foldes G, Couch LS, Chowdhury RA, Tzortzis KN, Peters NS, Rog-Zielinska EA, Yang HY, Welch S, Bowles CT, Rahman Haley S, Bell AR, Rice A, Sasikaran T, Johnson NA, Falaschetti E, Parameshwar J, Lewis C, Tsui S, Simon A, Pepper J, Rudy JJ, Zsebo KM, Macleod KT, Terracciano CM, Hajjar RJ, Banner N, Harding SE. Investigation of the safety and feasibility of AAV1/SERCA2a gene transfer in patients with chronic heart failure supported with a left ventricular assist device - the SERCA-LVAD TRIAL. Gene Ther 2020; 27:579-590. [PMID: 32669717 PMCID: PMC7744277 DOI: 10.1038/s41434-020-0171-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/05/2020] [Accepted: 06/25/2020] [Indexed: 01/16/2023]
Abstract
The SERCA-LVAD trial was a phase 2a trial assessing the safety and feasibility of delivering an adeno-associated vector 1 carrying the cardiac isoform of the sarcoplasmic reticulum calcium ATPase (AAV1/SERCA2a) to adult chronic heart failure patients implanted with a left ventricular assist device. The SERCA-LVAD trial was one of a program of AAV1/SERCA2a cardiac gene therapy trials including CUPID1, CUPID 2 and AGENT trials. Enroled subjects were randomised to receive a single intracoronary infusion of 1 × 1013 DNase-resistant AAV1/SERCA2a particles or a placebo solution in a double-blinded design, stratified by presence of neutralising antibodies to AAV. Elective endomyocardial biopsy was performed at 6 months unless the subject had undergone cardiac transplantation, with myocardial samples assessed for the presence of exogenous viral DNA from the treatment vector. Safety assessments including ELISPOT were serially performed. Although designed as a 24 subject trial, recruitment was stopped after five subjects had been randomised and received infusion due to the neutral result from the CUPID 2 trial. Here we describe the results from the 5 patients at 3 years follow up, which confirmed that viral DNA was delivered to the failing human heart in 2 patients receiving gene therapy with vector detectable at follow up endomyocardial biopsy or cardiac transplantation. Absolute levels of detectable transgene DNA were low, and no functional benefit was observed. There were no safety concerns in this small cohort. This trial identified some of the challenges of performing gene therapy trials in this LVAD patient cohort which may help guide future trial design.
Collapse
Affiliation(s)
- A R Lyon
- National Heart and Lung Institute, Imperial College London, London, UK. .,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK.
| | - D Babalis
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - A C Morley-Smith
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - M Hedger
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - A Suarez Barrientos
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - G Foldes
- National Heart and Lung Institute, Imperial College London, London, UK
| | - L S Couch
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R A Chowdhury
- National Heart and Lung Institute, Imperial College London, London, UK
| | - K N Tzortzis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - N S Peters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - E A Rog-Zielinska
- National Heart and Lung Institute, Imperial College London, London, UK.,Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
| | - H-Y Yang
- National Heart and Lung Institute, Imperial College London, London, UK
| | - S Welch
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - C T Bowles
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - S Rahman Haley
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - A R Bell
- Department of Histopathology, Royal Brompton and Harefield Hospitals NHS Trust, Freiburg, Germany
| | - A Rice
- Department of Histopathology, Royal Brompton and Harefield Hospitals NHS Trust, Freiburg, Germany
| | - T Sasikaran
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - N A Johnson
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - E Falaschetti
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | | | - C Lewis
- Royal Papworth Hospital NHS Trust, Cambridge, UK
| | - S Tsui
- Royal Papworth Hospital NHS Trust, Cambridge, UK
| | - A Simon
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - J Pepper
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - J J Rudy
- Celladon Corporation, San Diego, CA, USA
| | - K M Zsebo
- Celladon Corporation, San Diego, CA, USA
| | - K T Macleod
- National Heart and Lung Institute, Imperial College London, London, UK
| | - C M Terracciano
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R J Hajjar
- Phospholamban Foundation, Amsterdam, Netherlands
| | - N Banner
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - S E Harding
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
23
|
Li J, Agvanian S, Zhou K, Shaw RM, Hong T. Exogenous Cardiac Bridging Integrator 1 Benefits Mouse Hearts With Pre-existing Pressure Overload-Induced Heart Failure. Front Physiol 2020; 11:708. [PMID: 32670093 PMCID: PMC7327113 DOI: 10.3389/fphys.2020.00708] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Cardiac bridging integrator 1 (cBIN1) organizes transverse tubule (t-tubule) membrane calcium handling microdomains required for normal beat-to-beat contractility. cBIN1 is transcriptionally reduced in heart failure (HF). We recently found that cBIN1 pretreatment can limit HF development in stressed mice. Here, we aim to explore whether cBIN1 replacement therapy can improve myocardial function in continuously stressed hearts with pre-existing HF. Methods: Adult male mice were subjected to sham or transverse aortic constriction (TAC) surgery at the age of 8-10 weeks old. Adeno-associated virus 9 (AAV9) transducing cBIN1-V5 or GFP-V5 (3 × 1010 vg) was administered through retro-orbital injection at 5 weeks post-TAC. Mice were followed by echocardiography to monitor cardiac function until 20 weeks after TAC. Overall survival, heart and lung weight (LW), and HF incidence were determined. In a second set of animals in which AAV9-cBIN1 pretreatment prevents HF, we recorded cardiac pressure-volume (PV) loops and obtained myocardial immunofluorescence imaging. Results: The overall Kaplan-Meir survival of AAV9-cBIN1 mice was 77.8%, indicating a significant partial rescue between AAV9-GFP (58.8%) and sham (100%) treated mice. In mice with ejection fraction (EF) ≥30% prior to AAV9 injection at 5 weeks post-TAC, AAV9-cBIN1 significantly increased survival to 93.3%, compared to 62.5% survival for AAV9-GFP treated mice. The effect of exogenous cBIN1 was to attenuate TAC-induced left ventricular (LV) dilation and prevent further HF development. Recovery of EF also occurs in AAV9-cBIN1-treated mice. We found that EF increases to a peak at 6-8 weeks post-viral injection. Furthermore, PV loop analysis identified that AAV9-cBIN1 increases both systolic and diastolic function of the post-TAC hearts. At the myocyte level, AAV9-cBIN1 normalizes cBIN1 expression, t-tubule membrane intensity, and intracellular distribution of Cav1.2 and ryanodine receptors (RyRs). Conclusions: In mice with pre-existing HF, exogenous cBIN1 can normalize t-tubule calcium handling microdomains, limit HF progression, rescue cardiac function, and improve survival.
Collapse
Affiliation(s)
- Jing Li
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sosse Agvanian
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Kang Zhou
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Robin M. Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - TingTing Hong
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology & Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
24
|
Hitzeman TC, Xie Y, Zadikany RH, Nikolova AP, Baum R, Caldaruse AM, Agvanian S, Melmed GY, McGovern DPB, Geft DR, Chang DH, Moriguchi JD, Hage A, Azarbal B, Czer LS, Kittleson MM, Patel JK, Wu AHB, Kobashigawa JA, Hamilton M, Hong T, Shaw RM. cBIN1 Score (CS) Identifies Ambulatory HFrEF Patients and Predicts Cardiovascular Events. Front Physiol 2020; 11:503. [PMID: 32670075 PMCID: PMC7326053 DOI: 10.3389/fphys.2020.00503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cardiac Bridging Integrator 1 (cBIN1) is a membrane deformation protein that generates calcium microdomains at cardiomyocyte t-tubules, whose transcription is reduced in heart failure, and is released into blood. cBIN1 score (CS), an inverse index of plasma cBIN1, measures cellular myocardial remodeling. In patients with heart failure with preserved ejection fraction (HFpEF), CS diagnoses ambulatory heart failure and prognosticates hospitalization. The performance of CS has not been tested in patients with heart failure with reduced ejection fraction (HFrEF). METHODS AND RESULTS CS was determined from plasma of patients recruited in a prospective study. Two comparative cohorts consisted of 158 ambulatory HFrEF patients (left ventricular ejection fraction (LVEF) ≤ 40%, 57 ± 10 years, 80% men) and 115 age and sex matched volunteers with no known history of HF. N-terminal pro-B-type natriuretic peptide (NT-proBNP) concentrations were also analyzed for comparison. CS follows a normal distribution with a median of 0 in the controls, which increases to a median of 1.9 (p < 0.0001) in HFrEF patients. CS correlates with clinically assessed New York Heart Association Class (p = 0.007). During 1-year follow-up, a high CS (≥ 1.9) in patients predicts increased cardiovascular events (43% vs. 26%, p = 0.01, hazard ratio 1.9). Compared to a model with demographics, clinical risk factors, and NT-proBNP, adding CS to the model improved the overall continuous net reclassification improvement (NRI 0.64; 95% CI 0.18-1.10; p = 0.006). Although performance for diagnosis and prognosis was similar to CS, NT-proBNP did not prognosticate between patients whose NT-proBNP values were > 400 pg/ml. CONCLUSION CS, which is mechanistically distinct from NT-proBNP, successfully differentiates myocardial health between patients with HFrEF and matched controls. A high CS reflects advanced NYHA stage, pathologic cardiac muscle remodeling, and predicts 1-year risk of cardiovascular events in ambulatory HFrEF patients. CS is a marker of myocardial remodeling in HFrEF patients, independent of volume status.
Collapse
Affiliation(s)
- Tara C. Hitzeman
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Yu Xie
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Ronit H. Zadikany
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Andriana P. Nikolova
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Rachel Baum
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Ana-Maria Caldaruse
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Sosse Agvanian
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Gil Y. Melmed
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dermot P. B. McGovern
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dael R. Geft
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - David H. Chang
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Jaime D. Moriguchi
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Antoine Hage
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Babak Azarbal
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Lawrence S. Czer
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Michelle M. Kittleson
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Jignesh K. Patel
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Alan H. B. Wu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jon A. Kobashigawa
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Michele Hamilton
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - TingTing Hong
- Department of Medicine, Cedars-Sinai Medical Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, United States
| | - Robin M. Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
25
|
Liu Y, Zhou K, Li J, Agvanian S, Caldaruse AM, Shaw S, Hitzeman TC, Shaw RM, Hong T. In Mice Subjected to Chronic Stress, Exogenous cBIN1 Preserves Calcium-Handling Machinery and Cardiac Function. JACC Basic Transl Sci 2020; 5:561-578. [PMID: 32613144 PMCID: PMC7315191 DOI: 10.1016/j.jacbts.2020.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Heart failure is an important, and growing, cause of morbidity and mortality. Half of patients with heart failure have preserved ejection fraction, for whom therapeutic options are limited. Here we report that cardiac bridging integrator 1 gene therapy to maintain subcellular membrane compartments within cardiomyocytes can stabilize intracellular distribution of calcium-handling machinery, preserving diastolic function in hearts stressed by chronic beta agonist stimulation and pressure overload. This study identifies that maintenance of intracellular architecture and, in particular, membrane microdomains at t-tubules, is important in the setting of sympathetic stress. Stabilization of membrane microdomains may be a pathway for future therapeutic development.
Collapse
Key Words
- AAV9, adeno-associated virus 9
- ANOVA, analysis of variance
- AR, adrenergic receptor
- ATPase, adenosine triphosphatase
- BW, body weight
- CAMKII, Ca2+/calmodulin-dependent protein kinase
- CMV, cytomegalovirus
- Di-8-ANNEPs, 4-[2-[6-(Dioctylamino)-2-naphthalenyl]ethenyl]-1-(3-sulfopropyl)-pyridinium, inner salt
- EC, excitation contraction
- EDV, end diastolic volume
- EF, ejection fraction
- GFP, green fluorescent protein
- HF, heart failure
- HR, heart rate
- HT, heterozygote
- HW, heart weight
- ISO, isoproterenol
- LSD, least significant difference
- LTCC, voltage-dependent L-type calcium channel
- LV, left ventricular
- LW, lung weight
- PBS, phosphate-buffered saline
- PKA, protein kinase A
- PLN, phospholamban
- RWT, relative wall thickness
- RyR, ryanodine receptor
- SD, standard deviation
- SEM, standard error of the mean
- SERCA2a, sarcoplasmic reticulum calcium ATPase pump 2a
- SR, sarcoplasmic reticulum
- STORM, stochastic optical reconstruction microscopy
- TAC, transverse aortic constriction
- TEM, transmission electron microscopy
- WT, wild type
- cBIN1, cardiac bridging integrator 1
- diastolic dysfunction
- heart failure
- jSR, junctional sarcoplasmic reticulum
- pressure overload
- sympathetic overdrive
- t-tubule
- t-tubule, transverse-tubule
- vg, vector genome
Collapse
Affiliation(s)
- Yan Liu
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kang Zhou
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jing Li
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sosse Agvanian
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Seiji Shaw
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Tara C Hitzeman
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Robin M Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - TingTing Hong
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, California
| |
Collapse
|
26
|
Studying signal compartmentation in adult cardiomyocytes. Biochem Soc Trans 2020; 48:61-70. [PMID: 32104883 PMCID: PMC7054744 DOI: 10.1042/bst20190247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 02/04/2023]
Abstract
Multiple intra-cellular signalling pathways rely on calcium and 3′–5′ cyclic adenosine monophosphate (cAMP) to act as secondary messengers. This is especially true in cardiomyocytes which act as the force-producing units of the cardiac muscle and are required to react rapidly to environmental stimuli. The specificity of functional responses within cardiomyocytes and other cell types is produced by the organellar compartmentation of both calcium and cAMP. In this review, we assess the role of molecular localisation and relative contribution of active and passive processes in producing compartmentation. Active processes comprise the creation and destruction of signals, whereas passive processes comprise the release or sequestration of signals. Cardiomyocytes display a highly articulated membrane structure which displays significant cell-to-cell variability. Special attention is paid to the way in which cell membrane caveolae and the transverse-axial tubule system allow molecular localisation. We explore the effects of cell maturation, pathology and regional differences in the organisation of these processes. The subject of signal compartmentation has had a significant amount of attention within the cardiovascular field and has undergone a revolution over the past two decades. Advances in the area have been driven by molecular imaging using fluorescent dyes and genetically encoded constructs based upon fluorescent proteins. We also explore the use of scanning probe microscopy in the area. These techniques allow the analysis of molecular compartmentation within specific organellar compartments which gives researchers an entirely new perspective.
Collapse
|
27
|
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: 0.8] [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
|
28
|
Nader M. The SLMAP/Striatin complex: An emerging regulator of normal and abnormal cardiac excitation-contraction coupling. Eur J Pharmacol 2019; 858:172491. [PMID: 31233748 DOI: 10.1016/j.ejphar.2019.172491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/01/2022]
Abstract
The excitation-contraction (E-C) module involves a harmonized correspondence between the sarcolemma and the sarcoplasmic reticulum. This is provided by membrane proteins, which primarily shape the caveolae, the T-tubule/Sarcoplasmic reticulum (TT/SR) junction, and the intercalated discs (ICDs). Distortion of either one of these structures impairs myocardial contraction, and subsequently translates into cardiac failure. Thus, detailed studies on the molecular cues of the E-C module are becoming increasingly necessary to pharmacologically eradicate cardiac failure Herein we reviewed the organization of caveolae, TT/SR junctions, and the ICDs in the heart, with special attention to the Sarcolemma Membrane Associated Protein (SLMAP) and striatin (STRN) in cardiac membranes biology and cardiomyocyte contraction. We emphasized on their in vivo and in vitro signaling in cardiac function/dysfunction. SLMAP is a cardiac membrane protein that plays an important role in E-C coupling and the adrenergic response of the heart. Similarly, STRN is a dynamic protein that is also involved in cardiac E-C coupling and ICD-related cardiomyopathies. Both SLMAP and STRN are linked to cardiac conditions, including heart failure, and their role in cardiomyocyte function was elucidated in our laboratory. They interact together in a protein complex that holds therapeutic potentials for cardiac dysfunction. This review is the first of its kind to conceptualize the role of the SLMAP/STRN complex in cardiac function and failure. It provides in depth information on the signaling of these two proteins and projects their interaction as a novel therapeutic target for cardiac failure.
Collapse
Affiliation(s)
- Moni Nader
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, 11533, P.O. Box 50927, Saudi Arabia; Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
29
|
Lagarto JL, Dyer BT, Peters NS, French PMW, Dunsby C, Lyon AR. In vivo label-free optical monitoring of structural and metabolic remodeling of myocardium following infarction. BIOMEDICAL OPTICS EXPRESS 2019; 10:3506-3521. [PMID: 31360603 PMCID: PMC6640823 DOI: 10.1364/boe.10.003506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/15/2019] [Accepted: 05/29/2019] [Indexed: 05/14/2023]
Abstract
Cardiac remodeling following myocardial infarction (MI) involves structural and functional alterations in the infarcted and remote viable myocardium that can ultimately lead to heart failure. The underlying mechanisms are not fully understood and, following our previous study of the autofluorescence lifetime and diffuse reflectance signatures of the myocardium in vivo at 16 weeks post MI in rats [Biomed. Opt. Express6(2), 324 (2015)], we here present data obtained at 1, 2 and 4 weeks post myocardial infarction that help follow the temporal progression of these changes. Our results demonstrate that both structural and metabolic changes in the heart can be monitored from the earliest time points following MI using label-free optical readouts, not only in the region of infarction but also in the remote non-infarcted myocardium. Changes in the autofluorescence intensity and lifetime parameters associated with collagen type I autofluorescence were indicative of progressive collagen deposition in tissue that was most pronounced at earlier time points and in the region of infarction. In addition to significant collagen deposition in infarcted and non-infarcted myocardium, we also report changes in the autofluorescence parameters associated with reduced nicotinamide adenine (phosphate) dinucleotide (NAD(P)H) and flavin adenine dinucleotide (FAD), which we associate with metabolic alterations throughout the heart. Parallel measurements of the diffuse reflectance spectra indicated an increased contribution of reduced cytochrome c. Our findings suggest that combining time-resolved spectrofluorometry and diffuse reflectance spectroscopy could provide a useful means to monitor cardiac function in vivo at the time of surgery.
Collapse
Affiliation(s)
- João L. Lagarto
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
- Authors contributed equally to this work
| | - Benjamin T. Dyer
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, United Kingdom
- Authors contributed equally to this work
| | - Nicholas S. Peters
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, United Kingdom
- Centre for Cardiac Engineering, Imperial College London, Du Cane Road, London, W12 0NN, United Kingdom
| | - Paul M. W. French
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
| | - Chris Dunsby
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London, SW7 2AZ, United Kingdom
- Centre for Pathology, Imperial College London Du Cane Road, London W12 0NN, United Kingdom
- Authors contributed equally to this work
| | - Alexander R. Lyon
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, United Kingdom
- Authors contributed equally to this work
| |
Collapse
|
30
|
Watson SA, Perbellini F, Terracciano CM. Cardiac t-tubules: where structural plasticity meets functional adaptation. Cardiovasc Res 2019; 112:423-5. [PMID: 27659500 DOI: 10.1093/cvr/cvw198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Samuel A Watson
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, DuCane Road, W120NN, London, UK
| | - Filippo Perbellini
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, DuCane Road, W120NN, London, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, DuCane Road, W120NN, London, UK
| |
Collapse
|
31
|
Affiliation(s)
- Ying Fu
- From the Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., R.M.S); and Department of Medicine, University of California Los Angeles (R.M.S.)
| | - Robin M Shaw
- From the Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., R.M.S); and Department of Medicine, University of California Los Angeles (R.M.S.).
| |
Collapse
|
32
|
Watson SA, Duff J, Bardi I, Zabielska M, Atanur SS, Jabbour RJ, Simon A, Tomas A, Smolenski RT, Harding SE, Perbellini F, Terracciano CM. Biomimetic electromechanical stimulation to maintain adult myocardial slices in vitro. Nat Commun 2019; 10:2168. [PMID: 31092830 PMCID: PMC6520377 DOI: 10.1038/s41467-019-10175-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Adult cardiac tissue undergoes a rapid process of dedifferentiation when cultured outside the body. The in vivo environment, particularly constant electromechanical stimulation, is fundamental to the regulation of cardiac structure and function. We investigated the role of electromechanical stimulation in preventing culture-induced dedifferentiation of adult cardiac tissue using rat, rabbit and human heart failure myocardial slices. Here we report that the application of a preload equivalent to sarcomere length (SL) = 2.2 μm is optimal for the maintenance of rat myocardial slice structural, functional and transcriptional properties at 24 h. Gene sets associated with the preservation of structure and function are activated, while gene sets involved in dedifferentiation are suppressed. The maximum contractility of human heart failure myocardial slices at 24 h is also optimally maintained at SL = 2.2 μm. Rabbit myocardial slices cultured at SL = 2.2 μm remain stable for 5 days. This approach substantially prolongs the culture of adult cardiac tissue in vitro.
Collapse
Affiliation(s)
- Samuel A Watson
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - James Duff
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ifigeneia Bardi
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Magdalena Zabielska
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Santosh S Atanur
- Faculty of Medicine, Department of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Richard J Jabbour
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - André Simon
- Department of Cardiothoracic Transplantation & Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield, UB9 6JH, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Ryszard T Smolenski
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Sian E Harding
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Filippo Perbellini
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK.
| | | |
Collapse
|
33
|
Lawless M, Caldwell JL, Radcliffe EJ, Smith CER, Madders GWP, Hutchings DC, Woods LS, Church SJ, Unwin RD, Kirkwood GJ, Becker LK, Pearman CM, Taylor RF, Eisner DA, Dibb KM, Trafford AW. Phosphodiesterase 5 inhibition improves contractile function and restores transverse tubule loss and catecholamine responsiveness in heart failure. Sci Rep 2019; 9:6801. [PMID: 31043634 PMCID: PMC6494852 DOI: 10.1038/s41598-019-42592-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is characterized by poor survival, a loss of catecholamine reserve and cellular structural remodeling in the form of disorganization and loss of the transverse tubule network. Indeed, survival rates for HF are worse than many common cancers and have not improved over time. Tadalafil is a clinically relevant drug that blocks phosphodiesterase 5 with high specificity and is used to treat erectile dysfunction. Using a sheep model of advanced HF, we show that tadalafil treatment improves contractile function, reverses transverse tubule loss, restores calcium transient amplitude and the heart's response to catecholamines. Accompanying these effects, tadalafil treatment normalized BNP mRNA and prevented development of subjective signs of HF. These effects were independent of changes in myocardial cGMP content and were associated with upregulation of both monomeric and dimerized forms of protein kinase G and of the cGMP hydrolyzing phosphodiesterases 2 and 3. We propose that the molecular switch for the loss of transverse tubules in HF and their restoration following tadalafil treatment involves the BAR domain protein Amphiphysin II (BIN1) and the restoration of catecholamine sensitivity is through reductions in G-protein receptor kinase 2, protein phosphatase 1 and protein phosphatase 2 A abundance following phosphodiesterase 5 inhibition.
Collapse
Affiliation(s)
- Michael Lawless
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Jessica L Caldwell
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Emma J Radcliffe
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Charlotte E R Smith
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - George W P Madders
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - David C Hutchings
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Lori S Woods
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Stephanie J Church
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Richard D Unwin
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Graeme J Kirkwood
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Lorenz K Becker
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Charles M Pearman
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Rebecca F Taylor
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - David A Eisner
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Katharine M Dibb
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom.
| |
Collapse
|
34
|
Lipsett DB, Frisk M, Aronsen JM, Nordén ES, Buonarati OR, Cataliotti A, Hell JW, Sjaastad I, Christensen G, Louch WE. Cardiomyocyte substructure reverts to an immature phenotype during heart failure. J Physiol 2019; 597:1833-1853. [PMID: 30707448 PMCID: PMC6441900 DOI: 10.1113/jp277273] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Key points As reactivation of the fetal gene program has been implicated in pathological remodelling during heart failure (HF), we examined whether cardiomyocyte subcellular structure and function revert to an immature phenotype during this disease. Surface and internal membrane structures appeared gradually during development, and returned to a juvenile state during HF. Similarly, dyadic junctions between the cell membrane and sarcoplasmic reticulum were progressively ‘packed’ with L‐type Ca2+ channels and ryanodine receptors during development, and ‘unpacked’ during HF. Despite similarities in subcellular structure, dyads were observed to be functional from early developmental stages, but exhibited an impaired ability to release Ca2+ in failing cardiomyocytes. Thus, while immature and failing cardiomyocytes share similarities in subcellular structure, these do not fully account for the marked impairment of Ca2+ homeostasis observed in HF.
Abstract Reactivation of the fetal gene programme has been implicated as a driver of pathological cardiac remodelling. Here we examined whether pathological remodelling of cardiomyocyte substructure and function during heart failure (HF) reflects a reversion to an immature phenotype. Using scanning electron microscopy, we observed that Z‐grooves and t‐tubule openings at the cell surface appeared gradually during cardiac development, and disappeared during HF. Confocal and super‐resolution imaging within the cell interior revealed similar structural parallels; disorganization of t‐tubules in failing cells was strikingly reminiscent of the late stages of postnatal development, with fewer transverse elements and a high proportion of longitudinal tubules. Ryanodine receptors (RyRs) were observed to be laid down in advance of developing t‐tubules and similarly ‘orphaned’ in HF, although RyR distribution along Z‐lines was relatively sparse. Indeed, nanoscale imaging revealed coordinated packing of L‐type Ca2+ channels and RyRs into dyadic junctions during development, and orderly unpacking during HF. These findings support a ‘last in, first out’ paradigm, as the latest stages of dyadic structural development are reversed during disease. Paired imaging of t‐tubules and Ca2+ showed that the disorganized arrangement of dyads in immature and failing cells promoted desynchronized and slowed Ca2+ release in these two states. However, while developing cells exhibited efficient triggering of Ca2+ release at newly formed dyads, dyadic function was impaired in failing cells despite similar organization of Ca2+ handling proteins. Thus, pathologically deficient Ca2+ homeostasis during HF is only partly linked to the re‐emergence of immature subcellular structure, and additionally reflects lost dyadic functionality. As reactivation of the fetal gene program has been implicated in pathological remodelling during heart failure (HF), we examined whether cardiomyocyte subcellular structure and function revert to an immature phenotype during this disease. Surface and internal membrane structures appeared gradually during development, and returned to a juvenile state during HF. Similarly, dyadic junctions between the cell membrane and sarcoplasmic reticulum were progressively ‘packed’ with L‐type Ca2+ channels and ryanodine receptors during development, and ‘unpacked’ during HF. Despite similarities in subcellular structure, dyads were observed to be functional from early developmental stages, but exhibited an impaired ability to release Ca2+ in failing cardiomyocytes. Thus, while immature and failing cardiomyocytes share similarities in subcellular structure, these do not fully account for the marked impairment of Ca2+ homeostasis observed in HF.
Collapse
Affiliation(s)
- D B Lipsett
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - M Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J M Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - E S Nordén
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - O R Buonarati
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - A Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J W Hell
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - I Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - G Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - W E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Kolstad TR, van den Brink J, MacQuaide N, Lunde PK, Frisk M, Aronsen JM, Norden ES, Cataliotti A, Sjaastad I, Sejersted OM, Edwards AG, Lines GT, Louch WE. Ryanodine receptor dispersion disrupts Ca 2+ release in failing cardiac myocytes. eLife 2018; 7:39427. [PMID: 30375974 PMCID: PMC6245731 DOI: 10.7554/elife.39427] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
Reduced cardiac contractility during heart failure (HF) is linked to impaired Ca2+ release from Ryanodine Receptors (RyRs). We investigated whether this deficit can be traced to nanoscale RyR reorganization. Using super-resolution imaging, we observed dispersion of RyR clusters in cardiomyocytes from post-infarction HF rats, resulting in more numerous, smaller clusters. Functional groupings of RyR clusters which produce Ca2+ sparks (Ca2+ release units, CRUs) also became less solid. An increased fraction of small CRUs in HF was linked to augmented ‘silent’ Ca2+ leak, not visible as sparks. Larger multi-cluster CRUs common in HF also exhibited low fidelity spark generation. When successfully triggered, sparks in failing cells displayed slow kinetics as Ca2+ spread across dispersed CRUs. During the action potential, these slow sparks protracted and desynchronized the overall Ca2+ transient. Thus, nanoscale RyR reorganization during HF augments Ca2+ leak and slows Ca2+ release kinetics, leading to weakened contraction in this disease. The muscle cells of the heart coordinate how they contract and relax in order to produce the heartbeat. During heart failure, these cells become less able to contract. As a result the heart becomes inefficient, pumping less blood around the body. For the cardiac muscle cells to contract, the levels of calcium ions in the cells needs to rapidly increase. In failing hearts, these increases in calcium ion levels are smaller, slower and less well coordinated. It was not known what causes these changes, making it difficult to treat heart failure. Calcium ions are released in cardiac muscle cells through protein channels called ryanodine receptors. These receptors form clusters that allow them to synchronize when they open and close. Could the reorganization of ryanodine receptors account for the problems seen in failing hearts? To investigate, Kolstad et al. examined rat hearts using a technique called super-resolution microscopy. This showed that the clusters of ryanodine receptors break apart during heart failure to form smaller clusters. Further experiments showed that calcium ions ‘leak’ from these smaller clusters, reducing the amount of calcium that can be released into cardiac muscle cells during each heartbeat. Released calcium also spreads between the dispersed clusters, resulting in a slower rise of the calcium levels in the cells. Both changes contribute to weakened contractions of cells in failing hearts. Therefore, heart failure can be traced back to very small rearrangements of the ryanodine receptors. This understanding will help researchers as they investigate new ways to treat heart failure.
Collapse
Affiliation(s)
- Terje R Kolstad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | | | - Niall MacQuaide
- Institute of Cardiovascular Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - Einar S Norden
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ole M Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | | | | | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
36
|
Jayasinghe I, Clowsley AH, de Langen O, Sali SS, Crossman DJ, Soeller C. Shining New Light on the Structural Determinants of Cardiac Couplon Function: Insights From Ten Years of Nanoscale Microscopy. Front Physiol 2018; 9:1472. [PMID: 30405432 PMCID: PMC6204384 DOI: 10.3389/fphys.2018.01472] [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: 07/25/2018] [Accepted: 09/28/2018] [Indexed: 12/12/2022] Open
Abstract
Remodelling of the membranes and protein clustering patterns during the pathogenesis of cardiomyopathies has renewed the interest in spatial visualisation of these structures in cardiomyocytes. Coincidental emergence of single molecule (super-resolution) imaging and tomographic electron microscopy tools in the last decade have led to a number of new observations on the structural features of the couplons, the primary sites of excitation-contraction coupling in the heart. In particular, super-resolution and tomographic electron micrographs have revised and refined the classical views of the nanoscale geometries of couplons, t-tubules and the organisation of the principal calcium handling proteins in both healthy and failing hearts. These methods have also allowed the visualisation of some features which were too small to be detected with conventional microscopy tools. With new analytical capabilities such as single-protein mapping, in situ protein quantification, correlative and live cell imaging we are now observing an unprecedented interest in adapting these research tools across the cardiac biophysical research discipline. In this article, we review the depth of the new insights that have been enabled by these tools toward understanding the structure and function of the cardiac couplon. We outline the major challenges that remain in these experiments and emerging avenues of research which will be enabled by these technologies.
Collapse
Affiliation(s)
- Izzy Jayasinghe
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Oscar de Langen
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sonali S Sali
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - David J Crossman
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christian Soeller
- Living Systems Institute, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
37
|
Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat Rev Mol Cell Biol 2018; 19:638-653. [DOI: 10.1038/s41580-018-0049-3] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Basheer WA, Shaw RM. Connexin 43 and CaV1.2 Ion Channel Trafficking in Healthy and Diseased Myocardium. Circ Arrhythm Electrophysiol 2018; 9:e001357. [PMID: 27266274 DOI: 10.1161/circep.115.001357] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/29/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Wassim A Basheer
- From the Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (W.A.B., R.M.S.); and Department of Medicine, University of California Los Angeles (R.M.S.)
| | - Robin M Shaw
- From the Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (W.A.B., R.M.S.); and Department of Medicine, University of California Los Angeles (R.M.S.).
| |
Collapse
|
39
|
Schobesberger S, Wright P, Tokar S, Bhargava A, Mansfield C, Glukhov AV, Poulet C, Buzuk A, Monszpart A, Sikkel M, Harding SE, Nikolaev VO, Lyon AR, Gorelik J. T-tubule remodelling disturbs localized β2-adrenergic signalling in rat ventricular myocytes during the progression of heart failure. Cardiovasc Res 2018; 113:770-782. [PMID: 28505272 PMCID: PMC5437368 DOI: 10.1093/cvr/cvx074] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/11/2017] [Indexed: 11/12/2022] Open
Abstract
Aims Cardiomyocyte β2-adrenergic receptor (β2AR) cyclic adenosine monophosphate (cAMP) signalling is regulated by the receptors' subcellular location within transverse tubules (T-tubules), via interaction with structural and regulatory proteins, which form a signalosome. In chronic heart failure (HF), β2ARs redistribute from T-tubules to the cell surface, which disrupts functional signalosomes and leads to diffuse cAMP signalling. However, the functional consequences of structural changes upon β2AR-cAMP signalling during progression from hypertrophy to advanced HF are unknown. Methods and results Rat left ventricular myocytes were isolated at 4-, 8-, and 16-week post-myocardial infarction (MI), β2ARs were stimulated either via whole-cell perfusion or locally through the nanopipette of the scanning ion conductance microscope. cAMP release was measured via a Förster Resonance Energy Transfer-based sensor Epac2-camps. Confocal imaging of di-8-ANNEPS-stained cells and immunoblotting were used to determine structural alterations. At 4-week post-MI, T-tubule regularity, density and junctophilin-2 (JPH2) expression were significantly decreased. The amplitude of local β2AR-mediated cAMP in T-tubules was reduced and cAMP diffused throughout the cytosol instead of being locally confined. This was accompanied by partial caveolin-3 (Cav-3) dissociation from the membrane. At 8-week post-MI, the β2AR-mediated cAMP response was observed at the T-tubules and the sarcolemma (crest). Finally, at 16-week post-MI, the whole cell β2AR-mediated cAMP signal was depressed due to adenylate cyclase dysfunction, while overall Cav-3 levels were significantly increased and a substantial portion of Cav-3 dissociated into the cytosol. Overexpression of JPH2 in failing cells in vitro or AAV9.SERCA2a gene therapy in vivo did not improve β2AR-mediated signal compartmentation or reduce cAMP diffusion. Conclusion Although changes in T-tubule structure and β2AR-mediated cAMP signalling are significant even at 4-week post-MI, progression to the HF phenotype is not linear. At 8-week post-MI the loss of β2AR-mediated cAMP is temporarily reversed. Complete disorganization of β2AR-mediated cAMP signalling due to changes in functional receptor localization and cellular structure occurs at 16-week post-MI.
Collapse
Affiliation(s)
- Sophie Schobesberger
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martinistraße, Hamburg D-20246, Germany
| | - Peter Wright
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Sergiy Tokar
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Anamika Bhargava
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,Department of Biotechnology, Indian Institute of Technology Hyderabad, Ordnance Factory Estate, Yeddumailaram, 502205 Telangana, India
| | - Catherine Mansfield
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Alexey V Glukhov
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Claire Poulet
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Andrey Buzuk
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Aron Monszpart
- Department of Computer Science, University College London, Gower Street, London WC1E 6BT, UK
| | - Markus Sikkel
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Sian E Harding
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martinistraße, Hamburg D-20246, Germany
| | - Alexander R Lyon
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,NIHR Cardiovascular Biomedical Research Unit, Department of Cardiology, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Julia Gorelik
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| |
Collapse
|
40
|
Smith CER, Trafford AW, Caldwell JL, Dibb KM. Physiology and patho-physiology of the cardiac transverse tubular system. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Abstract
PURPOSE OF REVIEW Membrane invaginations called t-tubules play an integral role in triggering cardiomyocyte contraction, and their disruption during diseases such as heart failure critically impairs cardiac performance. In this review, we outline the growing understanding of the malleability of t-tubule structure and function, and highlight emerging t-tubule regulators which may be exploited for novel therapies. RECENT FINDINGS New technologies are revealing the nanometer scale organization of t-tubules, and their functional junctions with the sarcoplasmic reticulum called dyads, which generate Ca2+ sparks. Recent data have indicated that the dyadic anchoring protein junctophilin-2, and the membrane-bending protein BIN1 are key regulators of dyadic formation and maintenance. While the underlying signals which control expression and localization of these proteins remain unclear, accumulating data support an important role of myocardial workload. Although t-tubule alterations are believed to be a key cause of heart failure, the plasticity of these structures also creates an opportunity for therapy. Promising recent data suggest that such therapies may specifically target junctophilin-2, BIN1, and/or mechanotransduction.
Collapse
|
42
|
Bryant SM, Kong CHT, Cannell MB, Orchard CH, James AF. Loss of caveolin-3-dependent regulation of I Ca in rat ventricular myocytes in heart failure. Am J Physiol Heart Circ Physiol 2017; 314:H521-H529. [PMID: 29101175 PMCID: PMC5899261 DOI: 10.1152/ajpheart.00458.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
β2-Adrenoceptors and L-type Ca2+ current (ICa) redistribute from the t-tubules to the surface membrane of ventricular myocytes from failing hearts. The present study investigated the role of changes in caveolin-3 and PKA signaling, both of which have previously been implicated in this redistribution. ICa was recorded using the whole cell patch-clamp technique from ventricular myocytes isolated from the hearts of rats that had undergone either coronary artery ligation (CAL) or equivalent sham operation 18 wk earlier. ICa distribution between the surface and t-tubule membranes was determined using formamide-induced detubulation (DT). In sham myocytes, β2-adrenoceptor stimulation increased ICa in intact but not DT myocytes; however, forskolin (to increase cAMP directly) and H-89 (to inhibit PKA) increased and decreased, respectively, ICa at both the surface and t-tubule membranes. C3SD peptide (which decreases binding to caveolin-3) inhibited ICa in intact but not DT myocytes but had no effect in the presence of H-89. In contrast, in CAL myocytes, β2-adrenoceptor stimulation increased ICa in both intact and DT myocytes, but C3SD had no effect on ICa; forskolin and H-89 had similar effects as in sham myocytes. These data show the redistribution of β2-adrenoceptor activity and ICa in CAL myocytes and suggest constitutive stimulation of ICa by PKA in sham myocytes via concurrent caveolin-3-dependent (at the t-tubules) and caveolin-3-independent mechanisms, with the former being lost in CAL myocytes. NEW & NOTEWORTHY In ventricular myocytes from normal hearts, regulation of the L-type Ca2+ current by β2-adrenoceptors and the constitutive regulation by caveolin-3 is localized to the t-tubules. In heart failure, the regulation of L-type Ca2+ current by β2-adrenoceptors is redistributed to the surface membrane, and the constitutive regulation by caveolin-3 is lost.
Collapse
Affiliation(s)
- Simon M Bryant
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Cherrie H T Kong
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Mark B Cannell
- 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
| | - Andrew F James
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| |
Collapse
|
43
|
Growth hormone-releasing hormone attenuates cardiac hypertrophy and improves heart function in pressure overload-induced heart failure. Proc Natl Acad Sci U S A 2017; 114:12033-12038. [PMID: 29078377 PMCID: PMC5692579 DOI: 10.1073/pnas.1712612114] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pathological cardiac hypertrophy, characterized by heart growth in response to pressure or volume overload, such as in the setting of hypertension, is the main risk factor for heart failure (HF). The identification of therapeutic strategies to prevent or reverse cardiac hypertrophy is therefore a priority for curing HF. It is known that growth hormone-releasing hormone (GHRH) displays cardioprotective functions; however, its therapeutic potential in hypertrophy and HF is unknown. Here we show that GHRH reduces cardiomyocyte hypertrophy in vitro through inhibition of hypertrophic pathways. In vivo, the GHRH analog MR-409 attenuates cardiac hypertrophy in mice subjected to transverse aortic constriction and improves cardiac function. These findings suggest therapeutic use of GHRH analogs for treatment of pathological cardiac hypertrophy and HF. It has been shown that growth hormone-releasing hormone (GHRH) reduces cardiomyocyte (CM) apoptosis, prevents ischemia/reperfusion injury, and improves cardiac function in ischemic rat hearts. However, it is still not known whether GHRH would be beneficial for life-threatening pathological conditions, like cardiac hypertrophy and heart failure (HF). Thus, we tested the myocardial therapeutic potential of GHRH stimulation in vitro and in vivo, using GHRH or its agonistic analog MR-409. We show that in vitro, GHRH(1-44)NH2 attenuates phenylephrine-induced hypertrophy in H9c2 cardiac cells, adult rat ventricular myocytes, and human induced pluripotent stem cell-derived CMs, decreasing expression of hypertrophic genes and regulating hypertrophic pathways. Underlying mechanisms included blockade of Gq signaling and its downstream components phospholipase Cβ, protein kinase Cε, calcineurin, and phospholamban. The receptor-dependent effects of GHRH also involved activation of Gαs and cAMP/PKA, and inhibition of increase in exchange protein directly activated by cAMP1 (Epac1). In vivo, MR-409 mitigated cardiac hypertrophy in mice subjected to transverse aortic constriction and improved cardiac function. Moreover, CMs isolated from transverse aortic constriction mice treated with MR-409 showed improved contractility and reversal of sarcolemmal structure. Overall, these results identify GHRH as an antihypertrophic regulator, underlying its therapeutic potential for HF, and suggest possible beneficial use of its analogs for treatment of pathological cardiac hypertrophy.
Collapse
|
44
|
The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles. PLoS Biol 2017; 15:e2002354. [PMID: 28806752 PMCID: PMC5570487 DOI: 10.1371/journal.pbio.2002354] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/24/2017] [Accepted: 07/26/2017] [Indexed: 12/17/2022] Open
Abstract
Microparticles (MPs) are cell–cell communication vesicles derived from the cell surface plasma membrane, although they are not known to originate from cardiac ventricular muscle. In ventricular cardiomyocytes, the membrane deformation protein cardiac bridging integrator 1 (cBIN1 or BIN1+13+17) creates transverse-tubule (t-tubule) membrane microfolds, which facilitate ion channel trafficking and modulate local ionic concentrations. The microfold-generated microdomains continuously reorganize, adapting in response to stress to modulate the calcium signaling apparatus. We explored the possibility that cBIN1-microfolds are externally released from cardiomyocytes. Using electron microscopy imaging with immunogold labeling, we found in mouse plasma that cBIN1 exists in membrane vesicles about 200 nm in size, which is consistent with the size of MPs. In mice with cardiac-specific heterozygous Bin1 deletion, flow cytometry identified 47% less cBIN1-MPs in plasma, supporting cardiac origin. Cardiac release was also evidenced by the detection of cBIN1-MPs in medium bathing a pure population of isolated adult mouse cardiomyocytes. In human plasma, osmotic shock increased cBIN1 detection by enzyme-linked immunosorbent assay (ELISA), and cBIN1 level decreased in humans with heart failure, a condition with reduced cardiac muscle cBIN1, both of which support cBIN1 release in MPs from human hearts. Exploring putative mechanisms of MP release, we found that the membrane fission complex endosomal sorting complexes required for transport (ESCRT)-III subunit charged multivesicular body protein 4B (CHMP4B) colocalizes and coimmunoprecipitates with cBIN1, an interaction enhanced by actin stabilization. In HeLa cells with cBIN1 overexpression, knockdown of CHMP4B reduced the release of cBIN1-MPs. Using truncation mutants, we identified that the N-terminal BAR (N-BAR) domain in cBIN1 is required for CHMP4B binding and MP release. This study links the BAR protein superfamily to the ESCRT pathway for MP biogenesis in mammalian cardiac ventricular cells, identifying elements of a pathway by which cytoplasmic cBIN1 is released into blood. Microparticles are small vesicles generated from the cell surface membrane and externally released for communication with other cells. We now show that heart ventricular muscle cells, which form the main pumping chambers of the heart, release microparticles in both mouse and human. Ventricular microparticles arise from surface membrane microdomains organized by cardiac bridging integrator 1 (cBIN1), a membrane deformation protein that has been shown to be reduced during human heart failure. Here we identify microparticles containing cBIN1 in blood, which were reduced in mutant mice lacking heart cBIN1 expression. Furthermore, the process leading to microparticle release involves the recruitment of CHMP4B protein to snip the cBIN1 membrane. In humans, cBIN1 is present in blood and within microparticles. Upon osmotic shock, human microparticles burst, allowing for the quantification of cBIN1 in plasma by enzyme-linked immunosorbent assay (ELISA). The measured cBIN1 level was greatly reduced in patients with heart failure. Thus, we introduce the biology for a new blood-based diagnostic tool that can assess cardiac muscle health and identify failing heart in human patients.
Collapse
|
45
|
Abstract
Unique to striated muscle cells, transverse tubules (t-tubules) are membrane organelles that consist of sarcolemma penetrating into the myocyte interior, forming a highly branched and interconnected network. Mature t-tubule networks are found in mammalian ventricular cardiomyocytes, with the transverse components of t-tubules occurring near sarcomeric z-discs. Cardiac t-tubules contain membrane microdomains enriched with ion channels and signaling molecules. The microdomains serve as key signaling hubs in regulation of cardiomyocyte function. Dyad microdomains formed at the junctional contact between t-tubule membrane and neighboring sarcoplasmic reticulum are critical in calcium signaling and excitation-contraction coupling necessary for beat-to-beat heart contraction. In this review, we provide an overview of the current knowledge in gross morphology and structure, membrane and protein composition, and function of the cardiac t-tubule network. We also review in detail current knowledge on the formation of functional membrane subdomains within t-tubules, with a particular focus on the cardiac dyad microdomain. Lastly, we discuss the dynamic nature of t-tubules including membrane turnover, trafficking of transmembrane proteins, and the life cycles of membrane subdomains such as the cardiac BIN1-microdomain, as well as t-tubule remodeling and alteration in diseased hearts. Understanding cardiac t-tubule biology in normal and failing hearts is providing novel diagnostic and therapeutic opportunities to better treat patients with failing hearts.
Collapse
Affiliation(s)
- TingTing Hong
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California; and Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Robin M Shaw
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California; and Department of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
46
|
Zhou K, Hong T. Cardiac BIN1 (cBIN1) is a regulator of cardiac contractile function and an emerging biomarker of heart muscle health. SCIENCE CHINA-LIFE SCIENCES 2016; 60:257-263. [DOI: 10.1007/s11427-016-0249-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/06/2016] [Indexed: 12/01/2022]
|
47
|
Frisk M, Ruud M, Espe EKS, Aronsen JM, Røe ÅT, Zhang L, Norseng PA, Sejersted OM, Christensen GA, Sjaastad I, Louch WE. Elevated ventricular wall stress disrupts cardiomyocyte t-tubule structure and calcium homeostasis. Cardiovasc Res 2016; 112:443-51. [PMID: 27226008 PMCID: PMC5031949 DOI: 10.1093/cvr/cvw111] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/16/2016] [Indexed: 11/13/2022] Open
Abstract
AIMS Invaginations of the cellular membrane called t-tubules are essential for maintaining efficient excitation-contraction coupling in ventricular cardiomyocytes. Disruption of t-tubule structure during heart failure has been linked to dyssynchronous, slowed Ca(2+) release and reduced power of the heartbeat. The underlying mechanism is, however, unknown. We presently investigated whether elevated ventricular wall stress triggers remodelling of t-tubule structure and function. METHODS AND RESULTS MRI and blood pressure measurements were employed to examine regional wall stress across the left ventricle of sham-operated and failing, post-infarction rat hearts. In failing hearts, elevated left ventricular diastolic pressure and ventricular dilation resulted in markedly increased wall stress, particularly in the thin-walled region proximal to the infarct. High wall stress in this proximal zone was associated with reduced expression of the dyadic anchor junctophilin-2 and disrupted cardiomyocyte t-tubular structure. Indeed, local wall stress measurements predicted t-tubule density across sham and failing hearts. Elevated wall stress and disrupted cardiomyocyte structure in the proximal zone were also associated with desynchronized Ca(2+) release in cardiomyocytes and markedly reduced local contractility in vivo. A causative role of wall stress in promoting t-tubule remodelling was established by applying stretch to papillary muscles ex vivo under culture conditions. Loads comparable to wall stress levels observed in vivo in the proximal zone reduced expression of junctophilin-2 and promoted t-tubule loss. CONCLUSION Elevated wall stress reduces junctophilin-2 expression and disrupts t-tubule integrity, Ca(2+) release, and contractile function. These findings provide new insight into the role of wall stress in promoting heart failure progression.
Collapse
Affiliation(s)
- Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Emil K S Espe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | | | - Åsmund T Røe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Ole M Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Geir A Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
48
|
Jiang M, Zhang M, Howren M, Wang Y, Tan A, Balijepalli RC, Huizar JF, Tseng GN. JPH-2 interacts with Cai-handling proteins and ion channels in dyads: Contribution to premature ventricular contraction-induced cardiomyopathy. Heart Rhythm 2016; 13:743-52. [PMID: 26538326 PMCID: PMC4762763 DOI: 10.1016/j.hrthm.2015.10.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND In a canine model of premature ventricular contraction-induced cardiomyopathy (PVC-CM), Cav1.2 is downregulated and misplaced from transverse tubules (T tubules). Junctophilin-2 (JPH-2) is also downregulated. OBJECTIVES The objectives of this study were to understand the role of JPH-2 in PVC-CM and to probe changes in other proteins involved in dyad structure and function. METHODS We quantify T-tubule contents (di-8-ANEPPS fluorescence in live myocytes), examine myocyte ultrastructures (electron microscopy), probe JPH-2-interacting proteins (co-immunoprecipitation), quantify dyad and nondyad protein levels (immunoblotting), and examine subcellular distributions of dyad proteins (immunofluorescence/confocal microscopy). We also test direct JPH-2 modulation of channel function (vs indirect modulation through dyad formation) using heterologous expression. RESULTS PVC myocytes have reduced T-tubule contents but otherwise normal ultrastructures. Among 19 proteins examined, only JPH-2, bridging integrator-1 (BIN-1), and Cav1.2 are highly downregulated in PVC hearts. However, statistical analysis indicates a general reduction in dyad protein levels when JPH-2 is downregulated. Furthermore, several dyad proteins, including Na/Ca exchanger, are missing or shifted from dyads to the peripheral surface in PVC myocytes. JPH-2 directly or indirectly interacts with Cai-handling proteins, Cav1.2 and KCNQ1, although not BIN-1 or other scaffolding proteins tested. Expression in mammalian cells that do not have dyads confirms direct JPH-2 modulation of the L-type Ca channel current (Cav1.2/voltage-gated Ca channel β subunit 2) and slow delayed rectifier current (KCNQ1/KCNE1). CONCLUSION JPH-2 is more than a "dyad glue": it can modulate Cai handling and ion channel function in the dyad region. Downregulation of JPH-2, BIN-1, and Cav1.2 plays a deterministic role in PVC-CM. Dissecting the hierarchical relationship among the three is necessary for the design of therapeutic interventions to prevent the progression of PVC-CM.
Collapse
Affiliation(s)
- Min Jiang
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Mei Zhang
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Maureen Howren
- Department of Medicine/Cardiology Division, McGuire VA Medical Center, Richmond, Virginia
| | - Yuhong Wang
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Alex Tan
- Department of Medicine/Cardiology Division, McGuire VA Medical Center, Richmond, Virginia
| | - Ravi C Balijepalli
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison, Wisconsin
| | - Jose F Huizar
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Gea-Ny Tseng
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
49
|
Fu Y, Shaw SA, Naami R, Vuong CL, Basheer WA, Guo X, Hong T. Isoproterenol Promotes Rapid Ryanodine Receptor Movement to Bridging Integrator 1 (BIN1)-Organized Dyads. Circulation 2016; 133:388-97. [PMID: 26733606 DOI: 10.1161/circulationaha.115.018535] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/21/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The key pathophysiology of human acquired heart failure is impaired calcium transient, which is initiated at dyads consisting of ryanodine receptors (RyRs) at sarcoplasmic reticulum apposing CaV1.2 channels at t-tubules. Sympathetic tone regulates myocardial calcium transients through β-adrenergic receptor (β-AR)-mediated phosphorylation of dyadic proteins. Phosphorylated RyRs (P-RyR) have increased calcium sensitivity and open probability, amplifying calcium transient at a cost of receptor instability. Given that bridging integrator 1 (BIN1) organizes t-tubule microfolds and facilitates CaV1.2 delivery, we explored whether β-AR-regulated RyRs are also affected by BIN1. METHODS AND RESULTS Isolated adult mouse hearts or cardiomyocytes were perfused for 5 minutes with the β-AR agonist isoproterenol (1 µmol/L) or the blockers CGP+ICI (baseline). Using biochemistry and superresolution fluorescent imaging, we identified that BIN1 clusters P-RyR and CaV1.2. Acute β-AR activation increases coimmunoprecipitation between P-RyR and cardiac spliced BIN1+13+17 (with exons 13 and 17). Isoproterenol redistributes BIN1 to t-tubules, recruiting P-RyRs and improving the calcium transient. In cardiac-specific Bin1 heterozygote mice, isoproterenol fails to concentrate BIN1 to t-tubules, impairing P-RyR recruitment. The resultant accumulation of uncoupled P-RyRs increases the incidence of spontaneous calcium release. In human hearts with end-stage ischemic cardiomyopathy, we find that BIN1 is also 50% reduced, with diminished P-RyR association with BIN1. CONCLUSIONS On β-AR activation, reorganization of BIN1-induced microdomains recruits P-RyR into dyads, increasing the calcium transient while preserving electric stability. When BIN1 is reduced as in human acquired heart failure, acute stress impairs microdomain formation, limiting contractility and promoting arrhythmias.
Collapse
Affiliation(s)
- Ying Fu
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.)
| | - Seiji A Shaw
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.)
| | - Robert Naami
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.)
| | - Caresse L Vuong
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.)
| | - Wassim A Basheer
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.)
| | - Xiuqing Guo
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.)
| | - TingTing Hong
- From Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.A.S., R.N., C.L.V., W.A.B., T.H.); Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (X.G.); and Departments of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, CA (T.H.).
| |
Collapse
|
50
|
Miragoli M, Sanchez-Alonso JL, Bhargava A, Wright PT, Sikkel M, Schobesberger S, Diakonov I, Novak P, Castaldi A, Cattaneo P, Lyon AR, Lab MJ, Gorelik J. Microtubule-Dependent Mitochondria Alignment Regulates Calcium Release in Response to Nanomechanical Stimulus in Heart Myocytes. Cell Rep 2015; 14:140-151. [PMID: 26725114 PMCID: PMC4983655 DOI: 10.1016/j.celrep.2015.12.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/07/2015] [Accepted: 11/23/2015] [Indexed: 12/01/2022] Open
Abstract
Arrhythmogenesis during heart failure is a major clinical problem. Regional electrical gradients produce arrhythmias, and cellular ionic transmembrane gradients are its originators. We investigated whether the nanoscale mechanosensitive properties of cardiomyocytes from failing hearts have a bearing upon the initiation of abnormal electrical activity. Hydrojets through a nanopipette indent specific locations on the sarcolemma and initiate intracellular calcium release in both healthy and heart failure cardiomyocytes, as well as in human failing cardiomyocytes. In healthy cells, calcium is locally confined, whereas in failing cardiomyocytes, calcium propagates. Heart failure progressively stiffens the membrane and displaces sub-sarcolemmal mitochondria. Colchicine in healthy cells mimics the failing condition by stiffening the cells, disrupting microtubules, shifting mitochondria, and causing calcium release. Uncoupling the mitochondrial proton gradient abolished calcium initiation in both failing and colchicine-treated cells. We propose the disruption of microtubule-dependent mitochondrial mechanosensor microdomains as a mechanism for abnormal calcium release in failing heart. Nanomechanical pressure application changes mechanosensitivity in failing heart cells Microtubular network disorganization mediates the change in mechanosensitivity Mitochondria are displaced from their original location and trigger calcium release Uncoupling the mitochondrial proton gradient completely abolishes the phenomena
Collapse
Affiliation(s)
- Michele Miragoli
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; Humanitas Clinical and Research Center, via Manzoni 56, Rozzano, 20090 Milan, Italy; Center of Excellence for Toxicological Research, INAIL exISPESL, University of Parma, via Gramsci 14, 43126 Parma, Italy.
| | - Jose L Sanchez-Alonso
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Anamika Bhargava
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; Department of Biotechnology, Indian Institute of Technology Hyderabad, Ordnance Factory Estate, Yeddumailaram, 502205 Telangana, India
| | - Peter T Wright
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Markus Sikkel
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Sophie Schobesberger
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Ivan Diakonov
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Pavel Novak
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - Alessandra Castaldi
- Humanitas Clinical and Research Center, via Manzoni 56, Rozzano, 20090 Milan, Italy
| | - Paola Cattaneo
- Humanitas Clinical and Research Center, via Manzoni 56, Rozzano, 20090 Milan, Italy
| | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital, London SW36NP, UK
| | - Max J Lab
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK.
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK.
| |
Collapse
|