1
|
Wang J, Chen B, Shi Q, Ciampa G, Zhao W, Zhang G, Weiss RM, Peng T, Hall DD, Song LS. Preventing Site-Specific Calpain Proteolysis of Junctophilin-2 Protects Against Stress-Induced Excitation-Contraction Uncoupling and Heart Failure Development. Circulation 2025; 151:171-187. [PMID: 39291390 PMCID: PMC11729472 DOI: 10.1161/circulationaha.124.069329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Excitation-contraction (E-C) coupling processes become disrupted in heart failure (HF), resulting in abnormal Ca2+ homeostasis, maladaptive structural and transcriptional remodeling, and cardiac dysfunction. Junctophilin-2 (JP2) is an essential component of the E-C coupling apparatus but becomes site-specifically cleaved by calpain, leading to disruption of E-C coupling, plasmalemmal transverse tubule degeneration, abnormal Ca2+ homeostasis, and HF. However, it is not clear whether preventing site-specific calpain cleavage of JP2 is sufficient to protect the heart against stress-induced pathological cardiac remodeling in vivo. METHODS Calpain-resistant JP2 knock-in mice (JP2CR) were generated by deleting the primary JP2 calpain cleavage site. Stress-dependent JP2 cleavage was assessed through in vitro cleavage assays and in isolated cardiomyocytes treated with 1 μmol/L isoproterenol by immunofluorescence. Cardiac outcomes were assessed in wild-type and JP2CR mice 5 weeks after transverse aortic constriction compared with sham surgery using echocardiography, histology, and RNA-sequencing methods. E-C coupling efficiency was measured by in situ confocal microscopy. E-C coupling proteins were evaluated by calpain assays and Western blotting. The effectiveness of adeno-associated virus gene therapy with JP2CR, JP2, or green fluorescent protein to slow HF progression was evaluated in mice with established cardiac dysfunction. RESULTS JP2 proteolysis by calpain and in response to transverse aortic constriction and isoproterenol was blocked in JP2CR cardiomyocytes. JP2CR hearts are more resistant to pressure-overload stress, having significantly improved Ca2+ homeostasis and transverse tubule organization with significantly attenuated cardiac dysfunction, hypertrophy, lung edema, fibrosis, and gene expression changes relative to wild-type mice. JP2CR preserves the integrity of calpain-sensitive E-C coupling-related proteins, including ryanodine receptor 2, CaV1.2, and sarcoplasmic reticulum calcium ATPase 2a, by attenuating transverse aortic constriction-induced increases in calpain activity. Furthermore, JP2CR gene therapy after the onset of cardiac dysfunction was found to be effective at slowing the progression of HF and superior to wild-type JP2. CONCLUSIONS The data presented here demonstrate that preserving JP2-dependent E-C coupling by prohibiting the site-specific calpain cleavage of JP2 offers multifaceted beneficial effects, conferring cardiac protection against stress-induced proteolysis, hypertrophy, and HF. Our data also indicate that specifically targeting the primary calpain cleavage site of JP2 by gene therapy approaches holds great therapeutic potential as a novel precision medicine for treating HF.
Collapse
Affiliation(s)
- Jinxi Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Qian Shi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Grace Ciampa
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Weiyang Zhao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Guangqin Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Robert M. Weiss
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| | - Tianqing Peng
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 4S2, Canada
| | - Duane D. Hall
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
2
|
Waight MC, Fabbricatore D, Behr ER, Sohal M, Li AC, Saba MM. The Impact of Coronary Ischemia Assessment on Outcomes in Those With Scar-Dependent Ventricular Tachycardia. J Cardiovasc Electrophysiol 2025; 36:201-211. [PMID: 39547937 PMCID: PMC11726999 DOI: 10.1111/jce.16495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/02/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Guidance and outcomes of coronary ischemia assessment (IA) in those with structural heart disease (SHD), presenting with monomorphic ventricular tachycardia (MMVT) is unclear. OBJECTIVES To assess the impact of IA on arrhythmic and non-arrhythmic outcomes in those with SHD. METHODS Patients presenting with MMVT over a 6-year period to a tertiary center were retrospectively analyzed. Propensity score-matched analysis was performed comparing those undergoing IA to those who did not. The primary endpoint was a composite of VT recurrence, appropriate ICD therapy, heart failure hospitalization, and death. Secondary analysis of the individual components of the composite was performed. Kaplan-Meier, univariate and multivariate analysis was performed to compare the two groups and derive predictors of poor outcomes. RESULTS Two hundred and seventeen patients (57.6% ICM) were analyzed. 55.8% underwent IA. Following propensity score-matching, 120 patients remained. At 12 months, freedom from the primary endpoint was 68.3% of those undergoing IA versus 43.3% who did not, p < 0.001, multivariate HR 0.56 (0.34-0.92). This was driven by a reduction in all-cause mortality, with a 12-month survival of 98.3% in those undergoing IA versus 86.5% in those not undergoing IA (p < 0.01). Coronary intervention was associated with a significantly higher event-free 12-month survival compared to those who did not undergo intervention (82.4% vs 51.5%, respectively, p = 0.01). CONCLUSIONS Patients with SHD presenting MMVT who undergo an IA have significantly improved freedom from VT recurrence, appropriate ICD therapies, HF hospitalization, and death compared to those who do not, driven by a reduction in mortality.
Collapse
Affiliation(s)
| | | | - Elijah R. Behr
- City St George's University of LondonLondonUK
- St George's University Hospitals NHS Foundation TrustLondonUK
| | - Manav Sohal
- St George's University Hospitals NHS Foundation TrustLondonUK
| | - Anthony C. Li
- City St George's University of LondonLondonUK
- St George's University Hospitals NHS Foundation TrustLondonUK
| | - Magdi M. Saba
- City St George's University of LondonLondonUK
- St George's University Hospitals NHS Foundation TrustLondonUK
- Cleveland Clinic of LondonLondonUK
| |
Collapse
|
3
|
Charest A, Nasta N, Siddiqui S, Menkes S, Thomas A, Saad D, Forman J, Huang X, Sison CP, Gerdes AM, Stout RF, Ojamaa K. Nanoscale organization of cardiac calcium channels is dependent on thyroid hormone status. Am J Physiol Heart Circ Physiol 2024; 327:H1309-H1326. [PMID: 39365674 PMCID: PMC11559645 DOI: 10.1152/ajpheart.00272.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024]
Abstract
Thyroid hormone dysfunction is frequently observed in patients with chronic illnesses including heart failure, which increases the risk of adverse events. This study examined the effects of thyroid hormones (THs) on cardiac transverse-tubule (TT) integrity, Ca2+ sparks, and nanoscale organization of ion channels in excitation-contraction (EC) coupling, including L-type calcium channel (CaV1.2), ryanodine receptor type 2 (RyR2), and junctophilin-2 (Jph2). TH deficiency was established in adult female rats by propyl-thiouracil (PTU) ingestion for 8 wk; followed by randomization to continued PTU without or with oral triiodo-l-thyronine (T3; 10 µg/kg/day) for an additional 2 wk (PTU + T3). Confocal microscopy of isolated cardiomyocytes (CMs) showed significant misalignment of TTs and increased Ca2+ sparks in thyroid-deficient CMs. Density-based spatial clustering of applications with noise (DBSCAN) analysis of stochastic optical reconstruction microscopy (STORM) images showed decreased (P < 0.0001) RyR2 cluster number per cell area in PTU CMs compared with euthyroid (EU) control myocytes, and this was normalized by T3 treatment. CaV1.2 channels and Jph2 localized within a 210 nm radius of the RyR2 clusters were significantly reduced in PTU myocytes, and these values were increased with T3 treatment. A significant percentage of the RyR2 clusters in the PTU myocytes had neither CaV1.2 nor Jph2, suggesting fewer functional clusters in EC coupling. Nearest neighbor distances between RyR2 clusters were greater (P < 0.001) in PTU cells compared with EU- and T3-treated CMs that correspond to disarray of TTs at the sarcomere z-discs. These results support a regulatory role of T3 in the nanoscale organization of RyR2 clusters and colocalization of CaV1.2 and Jph2 in optimizing EC coupling.NEW & NOTEWORTHY Thyroid hormone (TH) dysfunction exacerbates preexisting heart conditions leading to an increased risk of premature morbidity/mortality. Triiodo-l-thyronine (T3) optimizes cardiac excitation-contraction (EC) coupling by maintaining myocardial T-tubule (TT) structures and organization of calcium ion channels. Single-molecule localization microscopy shows T3 effects on the clustering of ryanodine receptors (RyR2) with colocalization of L-type calcium channels (CaV1.2) and junctophilin-2 (Jph2) at TT-SR structures. Heart disease with subclinical hypothyroidism/low T3 syndrome may benefit from TH treatment.
Collapse
Affiliation(s)
- Amanda Charest
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Nicholas Nasta
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Sumaiyah Siddiqui
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Silvia Menkes
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Anvin Thomas
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Dana Saad
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Jake Forman
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Xueqi Huang
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York, United States
| | - Cristina P Sison
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York, United States
| | - A Martin Gerdes
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Randy F Stout
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| | - Kaie Ojamaa
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, United States
| |
Collapse
|
4
|
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
|
5
|
Lee CS, Jung SY, Yee RSZ, Agha NH, Hong J, Chang T, Babcock LW, Fleischman JD, Clayton B, Hanna AD, Ward CS, Lanza D, Hurley AE, Zhang P, Wehrens XHT, Lagor WR, Rodney GG, Hamilton SL. Speg interactions that regulate the stability of excitation-contraction coupling protein complexes in triads and dyads. Commun Biol 2023; 6:942. [PMID: 37709832 PMCID: PMC10502019 DOI: 10.1038/s42003-023-05330-y] [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: 04/03/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023] Open
Abstract
Here we show that striated muscle preferentially expressed protein kinase α (Spegα) maintains cardiac function in hearts with Spegβ deficiency. Speg is required for stability of excitation-contraction coupling (ECC) complexes and interacts with esterase D (Esd), Cardiomyopathy-Associated Protein 5 (Cmya5), and Fibronectin Type III and SPRY Domain Containing 2 (Fsd2) in cardiac and skeletal muscle. Mice with a sequence encoding a V5/HA tag inserted into the first exon of the Speg gene (HA-Speg mice) display a >90% decrease in Spegβ but Spegα is expressed at ~50% of normal levels. Mice deficient in both Spegα and Speg β (Speg KO mice) develop a severe dilated cardiomyopathy and muscle weakness and atrophy, but HA-Speg mice display mild muscle weakness with no cardiac involvement. Spegα in HA-Speg mice suppresses Ca2+ leak, proteolytic cleavage of Jph2, and disruption of transverse tubules. Despite it's low levels, HA-Spegβ immunoprecipitation identified Esd, Cmya5 and Fsd2 as Spegβ binding partners that localize to triads and dyads to stabilize ECC complexes. This study suggests that Spegα and Spegβ display functional redundancy, identifies Esd, Cmya5 and Fsd2 as components of both cardiac dyads and skeletal muscle triads and lays the groundwork for the identification of new therapeutic targets for centronuclear myopathy.
Collapse
Affiliation(s)
- Chang Seok Lee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Sung Yun Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Rachel Sue Zhen Yee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Nadia H Agha
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Jin Hong
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Ting Chang
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Lyle W Babcock
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Jorie D Fleischman
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Benjamin Clayton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Amy D Hanna
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Christopher S Ward
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Denise Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Ayrea E Hurley
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Pumin Zhang
- The First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, China
| | - Xander H T Wehrens
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - William R Lagor
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - George G Rodney
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA
| | - Susan L Hamilton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77096, USA.
| |
Collapse
|
6
|
Chung J, Tilūnaitė A, Ladd D, Hunt H, Soeller C, Crampin EJ, Johnston ST, Roderick HL, Rajagopal V. IP 3R activity increases propensity of RyR-mediated sparks by elevating dyadic [Ca 2+]. Math Biosci 2023; 355:108923. [PMID: 36395827 DOI: 10.1016/j.mbs.2022.108923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/15/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022]
Abstract
Calcium (Ca2+) plays a critical role in the excitation contraction coupling (ECC) process that mediates the contraction of cardiomyocytes during each heartbeat. While ryanodine receptors (RyRs) are the primary Ca2+ channels responsible for generating the cell-wide Ca2+ transients during ECC, Ca2+ release, via inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) are also reported in cardiomyocytes to elicit ECC-modulating effects. Recent studies suggest that the localization of IP3Rs at dyads grant their ability to modify the occurrence of Ca2+ sparks (elementary Ca2+ release events that constitute cell wide Ca2+ releases associated with ECC) which may underlie their modulatory influence on ECC. Here, we aim to uncover the mechanism by which dyad-localized IP3Rs influence Ca2+ spark dynamics. To this end, we developed a mathematical model of the dyad that incorporates the behaviour of IP3Rs, in addition to RyRs, to reveal the impact of their activity on local Ca2+ handling and consequent Ca2+ spark occurrence and its properties. Consistent with published experimental data, our model predicts that the propensity for Ca2+ spark formation increases in the presence of IP3R activity. Our simulations support the hypothesis that IP3Rs elevate Ca2+ in the dyad, sensitizing proximal RyRs towards activation and hence Ca2+ spark formation. The stochasticity of IP3R gating is an important aspect of this mechanism. However, dyadic IP3R activity lowers the Ca2+ available in the junctional sarcoplasmic reticulum (JSR) for release, thus resulting in Ca2+ sparks with similar durations but lower amplitudes.
Collapse
Affiliation(s)
- Joshua Chung
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia; Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Agnė Tilūnaitė
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia; School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - David Ladd
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia; School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC 3010, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, School of Chemical and Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Hilary Hunt
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Edmund J Crampin
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia; School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC 3010, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, School of Chemical and Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Stuart T Johnston
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC 3010, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, School of Chemical and Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium.
| | - Vijay Rajagopal
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
7
|
Perera T, Pius C, Niort B, Radcliffe EJ, Dibb KM, Trafford AW, Pinali C. Serial block face scanning electron microscopy reveals region-dependent remodelling of transverse tubules post-myocardial infarction. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210331. [PMID: 36189812 PMCID: PMC9527908 DOI: 10.1098/rstb.2021.0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 11/18/2022] Open
Abstract
The highly organized transverse tubule (t-tubule) network facilitates cardiac excitation-contraction coupling and synchronous cardiac myocyte contraction. In cardiac failure secondary to myocardial infarction (MI), changes in the structure and organization of t-tubules result in impaired cardiac contractility. However, there is still little knowledge on the regional variation of t-tubule remodelling in cardiac failure post-MI. Here, we investigate post-MI t-tubule remodelling in infarct border and remote regions, using serial block face scanning electron microscopy (SBF-SEM) applied to a translationally relevant sheep ischaemia reperfusion MI model and matched controls. We performed minimally invasive coronary angioplasty of the left anterior descending artery, followed by reperfusion after 90 min to establish the MI model. Left ventricular tissues obtained from control and MI hearts eight weeks post-MI were imaged using SBF-SEM. Image analysis generated three-dimensional reconstructions of the t-tubular network in control, MI border and remote regions. Quantitative analysis revealed that the MI border region was characterized by t-tubule depletion and fragmentation, dilation of surviving t-tubules and t-tubule elongation. This study highlights region-dependent remodelling of the tubular network post-MI and may provide novel localized therapeutic targets aimed at preservation or restoration of the t-tubules to manage cardiac contractility post-MI. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Tharushi Perera
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Charlene Pius
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Barbara Niort
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Emma J. Radcliffe
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Katharine M. Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Andrew W. Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Christian Pinali
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
8
|
Kohl P, Greiner J, Rog-Zielinska EA. Electron microscopy of cardiac 3D nanodynamics: form, function, future. Nat Rev Cardiol 2022; 19:607-619. [PMID: 35396547 DOI: 10.1038/s41569-022-00677-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2022] [Indexed: 11/09/2022]
Abstract
The 3D nanostructure of the heart, its dynamic deformation during cycles of contraction and relaxation, and the effects of this deformation on cell function remain largely uncharted territory. Over the past decade, the first inroads have been made towards 3D reconstruction of heart cells, with a native resolution of around 1 nm3, and of individual molecules relevant to heart function at a near-atomic scale. These advances have provided access to a new generation of data and have driven the development of increasingly smart, artificial intelligence-based, deep-learning image-analysis algorithms. By high-pressure freezing of cardiomyocytes with millisecond accuracy after initiation of an action potential, pseudodynamic snapshots of contraction-induced deformation of intracellular organelles can now be captured. In combination with functional studies, such as fluorescence imaging, exciting insights into cardiac autoregulatory processes at nano-to-micro scales are starting to emerge. In this Review, we discuss the progress in this fascinating new field to highlight the fundamental scientific insight that has emerged, based on technological breakthroughs in biological sample preparation, 3D imaging and data analysis; to illustrate the potential clinical relevance of understanding 3D cardiac nanodynamics; and to predict further progress that we can reasonably expect to see over the next 10 years.
Collapse
Affiliation(s)
- Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Engineering, University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Joachim Greiner
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
9
|
Abstract
In mammalian cardiac myocytes, the plasma membrane includes the surface sarcolemma but also a network of membrane invaginations called transverse (t-) tubules. These structures carry the action potential deep into the cell interior, allowing efficient triggering of Ca2+ release and initiation of contraction. Once thought to serve as rather static enablers of excitation-contraction coupling, recent work has provided a newfound appreciation of the plasticity of the t-tubule network's structure and function. Indeed, t-tubules are now understood to support dynamic regulation of the heartbeat across a range of timescales, during all stages of life, in both health and disease. This review article aims to summarize these concepts, with consideration given to emerging t-tubule regulators and their targeting in future therapies.
Collapse
Affiliation(s)
- Katharine M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo Norway
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
10
|
Differential remodelling of mitochondrial subpopulations and mitochondrial dysfunction are a feature of early stage diabetes. Sci Rep 2022; 12:978. [PMID: 35046471 PMCID: PMC8770458 DOI: 10.1038/s41598-022-04929-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/22/2021] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial dysfunction is a feature of type I and type II diabetes, but there is a lack of consistency between reports and links to disease development. We aimed to investigate if mitochondrial structure–function remodelling occurs in the early stages of diabetes by employing a mouse model (GENA348) of Maturity Onset Diabetes in the Young, exhibiting hyperglycemia, but not hyperinsulinemia, with mild left ventricular dysfunction. Employing 3-D electron microscopy (SBF-SEM) we determined that compared to wild-type, WT, the GENA348 subsarcolemma mitochondria (SSM) are ~ 2-fold larger, consistent with up-regulation of fusion proteins Mfn1, Mfn2 and Opa1. Further, in comparison, GENA348 mitochondria are more irregular in shape, have more tubular projections with SSM projections being longer and wider. Mitochondrial density is also increased in the GENA348 myocardium consistent with up-regulation of PGC1-α and stalled mitophagy (down-regulation of PINK1, Parkin and Miro1). GENA348 mitochondria have more irregular cristae arrangements but cristae dimensions and density are similar to WT. GENA348 Complex activity (I, II, IV, V) activity is decreased but the OCR is increased, potentially linked to a shift towards fatty acid oxidation due to impaired glycolysis. These novel data reveal that dysregulated mitochondrial morphology, dynamics and function develop in the early stages of diabetes.
Collapse
|
11
|
Bhullar S, Shah A, Dhalla N. Mechanisms for the development of heart failure and improvement of cardiac function by angiotensin-converting enzyme inhibitors. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are well-known for the treatments of cardiovascular diseases, such as heart failure, hypertension and acute coronary syndrome. Several of these inhibitors including captopril, enalapril, ramipril, zofenopril and imidapril attenuate vasoconstriction, cardiac hypertrophy and adverse cardiac remodeling, improve clinical outcomes in patients with cardiac dysfunction and decrease mortality. Extensive experimental and clinical research over the past 35 years has revealed that the beneficial effects of ACE inhibitors in heart failure are associated with full or partial prevention of adverse cardiac remodeling. Since cardiac function is mainly determined by coordinated activities of different subcellular organelles, including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils, for regulating the intracellular concentration of Ca2+ and myocardial metabolism, there is ample evidence to suggest that adverse cardiac remodelling and cardiac dysfunction in the failing heart are the consequence of subcellular defects. In fact, the improvement of cardiac function by different ACE inhibitors has been demonstrated to be related to the attenuation of abnormalities in subcellular organelles for Ca2+-handling, metabolic alterations, signal transduction defects and gene expression changes in failing cardiomyocytes. Various ACE inhibitors have also been shown to delay the progression of heart failure by reducing the formation of angiotensin II, the development of oxidative stress, the level of inflammatory cytokines and the occurrence of subcellular defects. These observations support the view that ACE inhibitors improve cardiac function in the failing heart by multiple mechanisms including the reduction of oxidative stress, myocardial inflammation and Ca2+-handling abnormalities in cardiomyocytes.
Collapse
|
12
|
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: 2.5] [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
|
13
|
Amoni M, Dries E, Ingelaere S, Vermoortele D, Roderick HL, Claus P, Willems R, Sipido KR. Ventricular Arrhythmias in Ischemic Cardiomyopathy-New Avenues for Mechanism-Guided Treatment. Cells 2021; 10:2629. [PMID: 34685609 PMCID: PMC8534043 DOI: 10.3390/cells10102629] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic heart disease is the most common cause of lethal ventricular arrhythmias and sudden cardiac death (SCD). In patients who are at high risk after myocardial infarction, implantable cardioverter defibrillators are the most effective treatment to reduce incidence of SCD and ablation therapy can be effective for ventricular arrhythmias with identifiable culprit lesions. Yet, these approaches are not always successful and come with a considerable cost, while pharmacological management is often poor and ineffective, and occasionally proarrhythmic. Advances in mechanistic insights of arrhythmias and technological innovation have led to improved interventional approaches that are being evaluated clinically, yet pharmacological advancement has remained behind. We review the mechanistic basis for current management and provide a perspective for gaining new insights that centre on the complex tissue architecture of the arrhythmogenic infarct and border zone with surviving cardiac myocytes as the source of triggers and central players in re-entry circuits. Identification of the arrhythmia critical sites and characterisation of the molecular signature unique to these sites can open avenues for targeted therapy and reduce off-target effects that have hampered systemic pharmacotherapy. Such advances are in line with precision medicine and a patient-tailored therapy.
Collapse
Affiliation(s)
- Matthew Amoni
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
- Division of Cardiology, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
| | - Eef Dries
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
| | - Sebastian Ingelaere
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
- Division of Cardiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dylan Vermoortele
- Imaging and Cardiovascular Dynamics, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (D.V.); (P.C.)
| | - H. Llewelyn Roderick
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
| | - Piet Claus
- Imaging and Cardiovascular Dynamics, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (D.V.); (P.C.)
| | - Rik Willems
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
- Division of Cardiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Karin R. Sipido
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.A.); (E.D.); (S.I.); (H.L.R.); (R.W.)
| |
Collapse
|
14
|
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: 6.5] [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
|
15
|
Targeting JP2: A New Treatment for Pulmonary Hypertension. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2003446. [PMID: 34394822 PMCID: PMC8363443 DOI: 10.1155/2021/2003446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022]
Abstract
Pulmonary hypertension (PH) is a disease with a complex etiology and high mortality rate. Abnormal pulmonary vasoconstriction and pulmonary vascular remodeling lead to an increase in mean pulmonary arterial blood pressure for which, and there is currently no cure. Junctophilin-2 (JP2) is beneficial for the assembly of junctional membrane complexes, the structural basis for excitation-contraction coupling that tethers the plasma membrane to the sarcoplasmic reticulum/endoplasmic reticulum and is involved in maintaining intracellular calcium concentration homeostasis and normal muscle contraction function. Recent studies have shown that JP2 maintains normal contraction and relaxation of vascular smooth muscle. In some experimental studies of drug treatments for PH, JP2 expression was increased, which improved pulmonary vascular remodeling and right ventricular function. Based on JP2 research to date, this paper summarizes the current understanding of JP2 protein structure, function, and related heart diseases and mechanisms and analyzes the feasibility and possible therapeutic strategies for targeting JP2 in PH.
Collapse
|
16
|
Gilani N, Wang K, Muncan A, Peter J, An S, Bhatti S, Pandya K, Zhang Y, Tang YD, Gerdes AM, Stout RF, Ojamaa K. Triiodothyronine maintains cardiac transverse-tubule structure and function. J Mol Cell Cardiol 2021; 160:1-14. [PMID: 34175303 DOI: 10.1016/j.yjmcc.2021.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 06/03/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022]
Abstract
Subclinical hypothyroidism and low T3 syndrome are commonly associated with an increased risk of cardiovascular disease (CVD) and mortality. We examined effects of T3 on T-tubule (TT) structures, Ca2+ mobilization and contractility, and clustering of dyadic proteins. Thyroid hormone (TH) deficiency was induced in adult female rats by propyl-thiouracil (PTU; 0.025%) treatment for 8 weeks. Rats were then randomized to continued PTU or triiodo-L-thyronine (T3; 10 μg/kg/d) treatment for 2 weeks (PTU + T3). After in vivo echocardiographic and hemodynamic recordings, cardiomyocytes (CM) were isolated to record Ca2+ transients and contractility. TT organization was assessed by confocal microscopy, and STORM images were captured to measure ryanodine receptor (RyR2) cluster number and size, and L-type Ca2+ channel (LTCC, Cav1.2) co-localization. Expressed genes including two integral TT proteins, junctophilin-2 (Jph-2) and bridging integrator-1 (BIN1), were analyzed in left ventricular (LV) tissues and cultured CM using qPCR and RNA sequencing. The T3 dosage used normalized serum T3, and reversed adverse effects of TH deficiency on in vivo measures of cardiac function. Recordings of isolated CM indicated that T3 increased rates of Ca2+ release and re-uptake, resulting in increased velocities of sarcomere shortening and re-lengthening. TT periodicity was significantly decreased, with reduced transverse tubules but increased longitudinal tubules in TH-deficient CMs and LV tissue, and these structures were normalized by T3 treatment. Analysis of STORM data of PTU myocytes showed decreased RyR2 cluster numbers and RyR localizations within each cluster without significant changes in Cav1.2 localizations within RyR clusters. T3 treatment normalized RyR2 cluster size and number. qPCR and RNAseq analyses of LV and cultured CM showed that Jph2 expression was T3-responsive, and its increase with treatment may explain improved TT organization and RyR-LTCC coupling.
Collapse
Affiliation(s)
- Nimra Gilani
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Kaihao Wang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA; Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Adam Muncan
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Jerrin Peter
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Shimin An
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA; Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Simran Bhatti
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Khushbu Pandya
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Youhua Zhang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Yi-Da Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - A Martin Gerdes
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Randy F Stout
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA; NYIT Imaging Center, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| | - Kaie Ojamaa
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Blvd., Old Westbury, New York 11568, USA.
| |
Collapse
|
17
|
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: 2.0] [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
|
18
|
Fiegle DJ, Schöber M, Dittrich S, Cesnjevar R, Klingel K, Volk T, Alkassar M, Seidel T. Severe T-System Remodeling in Pediatric Viral Myocarditis. Front Cardiovasc Med 2021; 7:624776. [PMID: 33537349 PMCID: PMC7848076 DOI: 10.3389/fcvm.2020.624776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022] Open
Abstract
Chronic heart failure (HF) in adults causes remodeling of the cardiomyocyte transverse tubular system (t-system), which contributes to disease progression by impairing excitation-contraction (EC) coupling. However, it is unknown if t-system remodeling occurs in pediatric heart failure. This study investigated the t-system in pediatric viral myocarditis. The t-system and integrity of EC coupling junctions (co-localization of L-type Ca2+ channels with ryanodine receptors and junctophilin-2) were analyzed by 3D confocal microscopy in left-ventricular (LV) samples from 5 children with myocarditis (age 14 ± 3 months), undergoing ventricular assist device (VAD) implantation, and 5 children with atrioventricular septum defect (AVSD, age 17 ± 3 months), undergoing corrective surgery. LV ejection fraction (EF) was 58.4 ± 2.3% in AVSD and 12.2 ± 2.4% in acute myocarditis. Cardiomyocytes from myocarditis samples showed increased t-tubule distance (1.27 ± 0.05 μm, n = 34 cells) and dilation of t-tubules (volume-length ratio: 0.64 ± 0.02 μm2) when compared with AVSD (0.90 ± 0.02 μm, p < 0.001; 0.52 ± 0.02 μm2, n = 61, p < 0.01). Intriguingly, 4 out of 5 myocarditis samples exhibited sheet-like t-tubules (t-sheets), a characteristic feature of adult chronic heart failure. The fraction of extracellular matrix was slightly higher in myocarditis (26.6 ± 1.4%) than in AVSD samples (24.4 ± 0.8%, p < 0.05). In one case of myocarditis, a second biopsy was taken and analyzed at VAD explantation after extensive cardiac recovery (EF from 7 to 56%) and clinical remission. When compared with pre-VAD, t-tubule distance and density were unchanged, as well as volume-length ratio (0.67 ± 0.04 μm2 vs. 0.72 ± 0.05 μm2, p = 0.5), reflecting extant t-sheets. However, junctophilin-2 cluster density was considerably higher (0.12 ± 0.02 μm−3 vs. 0.05 ± 0.01 μm−3, n = 9/10, p < 0.001), approaching values of AVSD (0.13 ± 0.05 μm−3, n = 56), and the measure of intact EC coupling junctions showed a distinct increase (20.2 ± 5.0% vs. 6.8 ± 2.2%, p < 0.001). Severe t-system loss and remodeling to t-sheets can occur in acute HF in young children, resembling the structural changes of chronically failing adult hearts. T-system remodeling might contribute to cardiac dysfunction in viral myocarditis. Although t-system recovery remains elusive, recovery of EC coupling junctions may be possible and deserves further investigation.
Collapse
Affiliation(s)
- Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Schöber
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sven Dittrich
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Karin Klingel
- Cardiopathology, University Hospital Tuebingen, Tübingen, Germany
| | - Tilmann Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Muhannad Alkassar
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
19
|
Gross P, Johnson J, Romero CM, Eaton DM, Poulet C, Sanchez-Alonso J, Lucarelli C, Ross J, Gibb AA, Garbincius JF, Lambert J, Varol E, Yang Y, Wallner M, Feldsott EA, Kubo H, Berretta RM, Yu D, Rizzo V, Elrod J, Sabri A, Gorelik J, Chen X, Houser SR. Interaction of the Joining Region in Junctophilin-2 With the L-Type Ca 2+ Channel Is Pivotal for Cardiac Dyad Assembly and Intracellular Ca 2+ Dynamics. Circ Res 2021; 128:92-114. [PMID: 33092464 PMCID: PMC7790862 DOI: 10.1161/circresaha.119.315715] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/21/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Ca2+-induced Ca2+ release (CICR) in normal hearts requires close approximation of L-type calcium channels (LTCCs) within the transverse tubules (T-tubules) and RyR (ryanodine receptors) within the junctional sarcoplasmic reticulum. CICR is disrupted in cardiac hypertrophy and heart failure, which is associated with loss of T-tubules and disruption of cardiac dyads. In these conditions, LTCCs are redistributed from the T-tubules to disrupt CICR. The molecular mechanism responsible for LTCCs recruitment to and from the T-tubules is not well known. JPH (junctophilin) 2 enables close association between T-tubules and the junctional sarcoplasmic reticulum to ensure efficient CICR. JPH2 has a so-called joining region that is located near domains that interact with T-tubular plasma membrane, where LTCCs are housed. The idea that this joining region directly interacts with LTCCs and contributes to LTCC recruitment to T-tubules is unknown. OBJECTIVE To determine if the joining region in JPH2 recruits LTCCs to T-tubules through direct molecular interaction in cardiomyocytes to enable efficient CICR. METHODS AND RESULTS Modified abundance of JPH2 and redistribution of LTCC were studied in left ventricular hypertrophy in vivo and in cultured adult feline and rat ventricular myocytes. Protein-protein interaction studies showed that the joining region in JPH2 interacts with LTCC-α1C subunit and causes LTCCs distribution to the dyads, where they colocalize with RyRs. A JPH2 with induced mutations in the joining region (mutPG1JPH2) caused T-tubule remodeling and dyad loss, showing that an interaction between LTCC and JPH2 is crucial for T-tubule stabilization. mutPG1JPH2 caused asynchronous Ca2+-release with impaired excitation-contraction coupling after β-adrenergic stimulation. The disturbed Ca2+ regulation in mutPG1JPH2 overexpressing myocytes caused calcium/calmodulin-dependent kinase II activation and altered myocyte bioenergetics. CONCLUSIONS The interaction between LTCC and the joining region in JPH2 facilitates dyad assembly and maintains normal CICR in cardiomyocytes.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cats
- Cells, Cultured
- Disease Models, Animal
- Excitation Contraction Coupling
- Humans
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Kinetics
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Mutation
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Organelle Biogenesis
- Protein Binding
- Protein Interaction Domains and Motifs
- Rats, Sprague-Dawley
- Ryanodine Receptor Calcium Release Channel
- Rats
Collapse
Affiliation(s)
- Polina Gross
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Jaslyn Johnson
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Carlos M. Romero
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Deborah M. Eaton
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Claire Poulet
- Imperial College London, Department of Cardiovascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, London
| | - Jose Sanchez-Alonso
- Imperial College London, Department of Cardiovascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, London
| | - Carla Lucarelli
- Imperial College London, Department of Cardiovascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, London
| | - Jean Ross
- Bioimaging Center Research, Delaware Biotechnology Institute, Newark
| | - Andrew A. Gibb
- Lewis Katz Temple University School of Medicine, Center for Translational Medicine, Department of Pharmacology, Philadelphia
| | - Joanne F. Garbincius
- Lewis Katz Temple University School of Medicine, Center for Translational Medicine, Department of Pharmacology, Philadelphia
| | - Jonathan Lambert
- Lewis Katz Temple University School of Medicine, Center for Translational Medicine, Department of Pharmacology, Philadelphia
| | - Erdem Varol
- Columbia University, Center for Theoretical Neuroscience, Department of Statistics, New York, NY
| | - Yijun Yang
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Markus Wallner
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
- Medical University of Graz, Division of Cardiology, Graz, Austria
- Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria
| | - Eric A. Feldsott
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Hajime Kubo
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Remus M. Berretta
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Daohai Yu
- Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia
| | - Victor Rizzo
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - John Elrod
- Lewis Katz Temple University School of Medicine, Center for Translational Medicine, Department of Pharmacology, Philadelphia
| | - Abdelkarim Sabri
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Julia Gorelik
- Imperial College London, Department of Cardiovascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, London
| | - Xiongwen Chen
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| | - Steven R. Houser
- Lewis Katz Temple University School of Medicine, Cardiovascular Research Center, Department of Physiology, Philadelphia
| |
Collapse
|
20
|
Li LL, Guo QJ, Lou HY, Liang JH, Yang Y, Xing X, Li HT, Han J, Shen S, Li H, Ye H, Di Wu H, Cui B, Wang SQ. Nanobar Array Assay Revealed Complementary Roles of BIN1 Splice Isoforms in Cardiac T-Tubule Morphogenesis. NANO LETTERS 2020; 20:6387-6395. [PMID: 32787151 PMCID: PMC8486496 DOI: 10.1021/acs.nanolett.0c01957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Bridging integrator-1 (BIN1) is a family of banana-shaped molecules implicated in cell membrane tubulation. To understand the curvature sensitivity and functional roles of BIN1 splicing isoforms, we engineered vertical nanobars on a cell culture substrate to create high and low curvatures. When expressed individually, BIN1 isoforms with phosphoinositide-binding motifs (pBIN1) appeared preferentially at high-curvature nanobar ends, agreeing well with their membrane tubulation in cardiomyocytes. In contrast, the ubiquitous BIN1 isoform without phosphoinositide-binding motif (uBIN1) exhibited no affinity to membranes around nanobars but accumulated along Z-lines in cardiomyocytes. Importantly, in pBIN1-uBIN1 coexpression, pBIN1 recruited uBIN1 to high-curvature membranes at nanobar ends, and uBIN1 attached the otherwise messy pBIN1 tubules to Z-lines. The complementary cooperation of BIN1 isoforms (comboBIN1) represents a novel mechanism of T-tubule formation along Z-lines in cardiomyocytes. Dysregulation of BIN1 splicing, e.g., during myocardial infarction, underlied T-tubule disorganization, and correction of uBIN1/pBIN1 stoichiometry rescued T-tubule morphology in heart disease.
Collapse
Affiliation(s)
- Lin-Lin Li
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Qian-Jin Guo
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Hsin-Ya Lou
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Jing-Hui Liang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yang Yang
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Xin Xing
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hong-Tao Li
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jing Han
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Shan Shen
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Li
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Haihong Ye
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| | - Hao Di Wu
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Shi-Qiang Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
21
|
Vermij SH, Abriel H, Kucera JP. Modeling Depolarization Delay, Sodium Currents, and Electrical Potentials in Cardiac Transverse Tubules. Front Physiol 2020; 10:1487. [PMID: 31920695 PMCID: PMC6916517 DOI: 10.3389/fphys.2019.01487] [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: 07/19/2019] [Accepted: 11/21/2019] [Indexed: 01/22/2023] Open
Abstract
T-tubules are invaginations of the lateral membrane of striated muscle cells that provide a large surface for ion channels and signaling proteins, thereby supporting excitation–contraction coupling. T-tubules are often remodeled in heart failure. To better understand the electrical behavior of T-tubules of cardiac cells in health and disease, this study addresses two largely unanswered questions regarding their electrical properties: (1) the delay of T-tubular membrane depolarization and (2) the effects of T-tubular sodium current on T-tubular potentials. Here, we present an elementary computational model to determine the delay in depolarization of deep T-tubular membrane segments as the narrow T-tubular lumen provides resistance against the extracellular current. We compare healthy tubules to tubules with constrictions and diseased tubules from mouse and human, and conclude that constrictions greatly delay T-tubular depolarization, while diseased T-tubules depolarize faster than healthy ones due to tubule widening. Increasing the tubule length non-linearly delays the depolarization. We moreover model the effect of T-tubular sodium current on intraluminal T-tubular potentials. We observe that extracellular potentials become negative during the sodium current transient (up to −40 mV in constricted T-tubules), which feedbacks on sodium channel function (self-attenuation) in a manner resembling ephaptic effects that have been described for intercalated discs where opposing membranes are very close together. The intraluminal potential and sodium current self-attenuation however greatly depend on sodium current conductance. These results show that (1) the changes in passive electrical properties of remodeled T-tubules cannot explain the excitation–contraction coupling defects in diseased cells; and (2) the sodium current may modulate intraluminal potentials. Such extracellular potentials might also affect excitation–contraction coupling and macroscopic conduction.
Collapse
Affiliation(s)
- Sarah Helena Vermij
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | | |
Collapse
|
22
|
Adverse transverse-tubule remodeling in a rat model of heart failure is attenuated with low-dose triiodothyronine treatment. Mol Med 2019; 25:53. [PMID: 31810440 PMCID: PMC6898920 DOI: 10.1186/s10020-019-0120-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
Abstract Pre-clinical animal studies have shown that triiodothyronine (T3) replacement therapy improves cardiac contractile function after myocardial infarction (MI). We hypothesized that T3 treatment could prevent adverse post-infarction cardiomyocyte remodeling by maintaining transverse-tubule (TT) structures, thus improving calcium dynamics and contractility. Methods Myocardial infarction (MI) or sham surgeries were performed on female Sprague-Dawley rats (aged 12 wks), followed by treatment with T3 (5μg/kg/d) or vehicle in drinking water for 16 wks (n = 10–11/group). After in vivo echocardiographic and hemodynamic analyses, left ventricular myocytes were isolated by collagenase digestion and simultaneous calcium and contractile transients in single cardiomyocytes were recorded using IonOptix imaging. Live cardiomyocytes were stained with AlexaFluor-488 conjugated wheat germ agglutinin (WGA-488) or di-8-ANEPPS, and multiple z-stack images per cell were captured by confocal microscopy for analysis of TT organization. RTqPCR and immunoblot approaches determined expression of TT proteins. Results Echocardiography and in vivo hemodynamic measurements showed significant improvements in systolic and diastolic function in T3- vs vehicle-treated MI rats. Isolated cardiomyocyte analysis showed significant dysfunction in measurements of myocyte relengthening in MI hearts, and improvements with T3 treatment: max relengthening velocity (Vmax, um/s), 2.984 ± 1.410 vs 1.593 ± 0.325, p < 0.05 and time to Vmax (sec), 0.233 ± 0.037 vs 0.314 ± 0.019, p < 0.001; MI + T3 vs MI + Veh, respectively. Time to peak contraction was shortened by T3 treatment (0.161 ± 0.021 vs 0.197 ± 0.011 s., p < 0.01; MI + T3 vs MI + Veh, respectively). Analysis of TT periodicity of WGA- or ANEPPS-stained cardiomyocytes indicated significant TT disorganization in MI myocytes and improvement with T3 treatment (transverse-oriented tubules (TE%): 9.07 ± 0.39 sham, 6.94 ± 0.67 MI + Veh and 8.99 ± 0.38 MI + T3; sham vs MI + Veh, p < 0.001; MI + Veh vs MI + T3, p < 0.01). Quantitative RT-PCR showed that reduced expression of BIN1 (Bridging integrator-1), Jph2 (junctophilin-2), RyR2 (ryanodine receptor) and Cav1.2 (L-type calcium channel) in the failing myocardium were increased by T3 and immunoblot analysis further supporting a potential T3 effect on the TT-associated proteins, BIN1 and Jph2. In conclusion, low dose T3 treatment initiated immediately after myocardial infarction attenuated adverse TT remodeling, improved calcium dynamics and contractility, thus supporting the potential therapeutic utility of T3 treatment in heart failure.
Collapse
|
23
|
Glucocorticoids preserve the t-tubular system in ventricular cardiomyocytes by upregulation of autophagic flux. Basic Res Cardiol 2019; 114:47. [PMID: 31673803 DOI: 10.1007/s00395-019-0758-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Abstract
A major contributor to contractile dysfunction in heart failure is remodelling and loss of the cardiomyocyte transverse tubular system (t-system), but underlying mechanisms and signalling pathways remain elusive. It has been shown that dexamethasone promotes t-tubule development in stem cell-derived cardiomyocytes and that cardiomyocyte-specific glucocorticoid receptor (GR) knockout (GRKO) leads to heart failure. Here, we studied if the t-system is altered in GRKO hearts and if GR signalling is required for t-system preservation in adult cardiomyocytes. Confocal and 3D STED microscopy of myocardium from cardiomyocyte-specific GRKO mice revealed decreased t-system density and increased distances between ryanodine receptors (RyR) and L-type Ca2+ channels (LTCC). Because t-system remodelling and heart failure are intertwined, we investigated the underlying mechanisms in vitro. Ventricular cardiomyocytes from failing human and healthy adult rat hearts cultured in the absence of glucocorticoids (CTRL) showed distinctively lower t-system density than cells treated with dexamethasone (EC50 1.1 nM) or corticosterone. The GR antagonist mifepristone abrogated the effect of dexamethasone. Dexamethasone improved RyR-LTCC coupling and synchrony of intracellular Ca2+ release, but did not alter expression levels of t-system-associated proteins junctophilin-2 (JPH2), bridging integrator-1 (BIN1) or caveolin-3 (CAV3). Rather, dexamethasone upregulated LC3B and increased autophagic flux. The broad-spectrum protein kinase inhibitor staurosporine prevented dexamethasone-induced upregulation of autophagy and t-system preservation, and autophagy inhibitors bafilomycin A and chloroquine accelerated t-system loss. Conversely, induction of autophagy by rapamycin or amino acid starvation preserved the t-system. These findings suggest that GR signalling and autophagy are critically involved in t-system preservation and remodelling in the heart.
Collapse
|
24
|
Kitmitto A, Baudoin F, Cartwright EJ. Cardiomyocyte damage control in heart failure and the role of the sarcolemma. J Muscle Res Cell Motil 2019; 40:319-333. [PMID: 31520263 PMCID: PMC6831538 DOI: 10.1007/s10974-019-09539-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 01/07/2023]
Abstract
The cardiomyocyte plasma membrane, termed the sarcolemma, is fundamental for regulating a myriad of cellular processes. For example, the structural integrity of the cardiomyocyte sarcolemma is essential for mediating cardiac contraction by forming microdomains such as the t-tubular network, caveolae and the intercalated disc. Significantly, remodelling of these sarcolemma microdomains is a key feature in the development and progression of heart failure (HF). However, despite extensive characterisation of the associated molecular and ultrastructural events there is a lack of clarity surrounding the mechanisms driving adverse morphological rearrangements. The sarcolemma also provides protection, and is the cell's first line of defence, against external stresses such as oxygen and nutrient deprivation, inflammation and oxidative stress with a loss of sarcolemma viability shown to be a key step in cell death via necrosis. Significantly, cumulative cell death is also a feature of HF, and is linked to disease progression and loss of cardiac function. Herein, we will review the link between structural and molecular remodelling of the sarcolemma associated with the progression of HF, specifically considering the evidence for: (i) Whether intrinsic, evolutionary conserved, plasma membrane injury-repair mechanisms are in operation in the heart, and (ii) if deficits in key 'wound-healing' proteins (annexins, dysferlin, EHD2 and MG53) may play a yet to be fully appreciated role in triggering sarcolemma microdomain remodelling and/or necrosis. Cardiomyocytes are terminally differentiated with very limited regenerative capability and therefore preserving cell viability and cardiac function is crucially important. This review presents a novel perspective on sarcolemma remodelling by considering whether targeting proteins that regulate sarcolemma injury-repair may hold promise for developing new strategies to attenuate HF progression.
Collapse
Affiliation(s)
- Ashraf Kitmitto
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK.
| | - Florence Baudoin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| |
Collapse
|
25
|
Molecular determinants of homo- and heteromeric interactions of Junctophilin-1 at triads in adult skeletal muscle fibers. Proc Natl Acad Sci U S A 2019; 116:15716-15724. [PMID: 31315980 DOI: 10.1073/pnas.1820980116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In adult skeletal muscles, 2 junctophilin isoforms (JPH1 and JPH2) tether the sarcoplasmic reticulum (SR) to transverse tubule (T-tubule) membranes, generating stable membrane contact sites known as triads. JPHs are anchored to the membrane of the SR by a C-terminal transmembrane domain (TMD) and bind the T-tubule membrane through their cytosolic N-terminal region, which contains 8 lipid-binding (MORN) motifs. By combining expression of GFP-JPH1 deletion mutants in skeletal muscle fibers with in vitro biochemical experiments, we investigated the molecular determinants of JPH1 recruitment at triads in adult skeletal muscle fibers. We found that MORN motifs bind PI(4,5)P2 in the sarcolemma, but do not mediate the selective localization of JPH1 at the T-tubule compartment of triads. On the contrary, fusion proteins containing only the TMD of JPH1 were able to localize at the junctional SR compartment of the triad. Bimolecular fluorescence complementation experiments indicated that the TMD of JPH1 can form dimers, suggesting that the observed localization at triads may result from dimerization with the TMDs of resident JPH1. A second domain, capable of mediating homo- and heterodimeric interactions between JPH1 and JPH2 was identified in the cytosolic region. FRAP experiments revealed that removal of either one of these 2 domains in JPH1 decreases the association of the resulting mutant proteins with triads. Altogether, these results suggest that the ability to establish homo- and heterodimeric interactions with resident JPHs may support the recruitment and stability of newly synthesized JPHs at triads in adult skeletal muscle fibers.
Collapse
|
26
|
Jiang J, Tang M, Huang Z, Chen L. Junctophilins emerge as novel therapeutic targets. J Cell Physiol 2019; 234:16933-16943. [DOI: 10.1002/jcp.28405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Jinyong Jiang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| | - Mingzhu Tang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| | - Zhen Huang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| |
Collapse
|
27
|
Jones PP, MacQuaide N, Louch WE. Dyadic Plasticity in Cardiomyocytes. Front Physiol 2018; 9:1773. [PMID: 30618792 PMCID: PMC6298195 DOI: 10.3389/fphys.2018.01773] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/23/2018] [Indexed: 11/13/2022] Open
Abstract
Contraction of cardiomyocytes is dependent on sub-cellular structures called dyads, where invaginations of the surface membrane (t-tubules) form functional junctions with the sarcoplasmic reticulum (SR). Within each dyad, Ca2+ entry through t-tubular L-type Ca2+ channels (LTCCs) elicits Ca2+ release from closely apposed Ryanodine Receptors (RyRs) in the SR membrane. The efficiency of this process is dependent on the density and macroscale arrangement of dyads, but also on the nanoscale organization of LTCCs and RyRs within them. We presently review accumulating data demonstrating the remarkable plasticity of these structures. Dyads are known to form gradually during development, with progressive assembly of both t-tubules and junctional SR terminals, and precise trafficking of LTCCs and RyRs. While dyads can exhibit compensatory remodeling when required, dyadic degradation is believed to promote impaired contractility and arrythmogenesis in cardiac disease. Recent data indicate that this plasticity of dyadic structure/function is dependent on the regulatory proteins junctophilin-2, amphiphysin-2 (BIN1), and caveolin-3, which critically arrange dyadic membranes while stabilizing the position and activity of LTCCs and RyRs. Indeed, emerging evidence indicates that clustering of both channels enables "coupled gating", implying that nanoscale localization and function are intimately linked, and may allow fine-tuning of LTCC-RyR crosstalk. We anticipate that improved understanding of dyadic plasticity will provide greater insight into the processes of cardiac compensation and decompensation, and new opportunities to target the basic mechanisms underlying heart disease.
Collapse
Affiliation(s)
- Peter P. Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Niall MacQuaide
- Institute of Cardiovascular Sciences, University of Glasgow, Glasgow, United Kingdom
- Clyde Biosciences, Glasgow, United Kingdom
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
28
|
Daghistani HM, Rajab BS, Kitmitto A. Three-dimensional electron microscopy techniques for unravelling mitochondrial dysfunction in heart failure and identification of new pharmacological targets. Br J Pharmacol 2018; 176:4340-4359. [PMID: 30225980 PMCID: PMC6887664 DOI: 10.1111/bph.14499] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/30/2018] [Accepted: 08/18/2018] [Indexed: 12/23/2022] Open
Abstract
A hallmark of heart failure is mitochondrial dysfunction leading to a bioenergetics imbalance in the myocardium. Consequently, there is much interest in targeting mitochondrial abnormalities to attenuate the pathogenesis of heart failure. This review discusses (i) how electron microscopy (EM) techniques have been fundamental for the current understanding of mitochondrial structure–function, (ii) the paradigm shift in resolutions now achievable by 3‐D EM techniques due to the introduction of direct detection devices and phase plate technology, and (iii) the application of EM for unravelling mitochondrial pathological remodelling in heart failure. We further consider the tremendous potential of multi‐scale EM techniques for the development of therapeutics, structure‐based ligand design and for delineating how a drug elicits nanostructural effects at the molecular, organelle and cellular levels. In conclusion, 3‐D EM techniques have entered a new era of structural biology and are poised to play a pivotal role in discovering new therapies targeting mitochondria for treating heart failure. Linked Articles This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc
Collapse
Affiliation(s)
- Hussam M Daghistani
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Bodour S Rajab
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
29
|
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]
|
30
|
Seidel T, Sankarankutty AC, Sachse FB. Remodeling of the transverse tubular system after myocardial infarction in rabbit correlates with local fibrosis: A potential role of biomechanics. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 130:302-314. [PMID: 28709857 DOI: 10.1016/j.pbiomolbio.2017.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 02/03/2023]
Abstract
The transverse tubular system (t-system) of ventricular cardiomyocytes is essential for efficient excitation-contraction coupling. In cardiac diseases, such as heart failure, remodeling of the t-system contributes to reduced cardiac contractility. However, mechanisms of t-system remodeling are incompletely understood. Prior studies suggested an association with altered cardiac biomechanics and gene expression in disease. Since fibrosis may alter tissue biomechanics, we investigated the local microscopic association of t-system remodeling with fibrosis in a rabbit model of myocardial infarction (MI). Biopsies were taken from the MI border zone of 6 infarcted hearts and from 6 control hearts. Using confocal microscopy and automated image analysis, we quantified t-system integrity (ITT) and the local fraction of extracellular matrix (fECM). In control, fECM was 18 ± 0.3%. ITT was high and homogeneous (0.07 ± 0.006), and did not correlate with fECM (R2 = 0.05 ± 0.02). The MI border zone exhibited increased fECM within 3 mm from the infarct scar (30 ± 3.5%, p < 0.01 vs control), indicating fibrosis. Myocytes in the MI border zone exhibited significant t-system remodeling, with dilated, sheet-like components, resulting in low ITT (0.03 ± 0.008, p < 0.001 vs control). While both fECM and t-system remodeling decreased with infarct distance, ITT correlated better with decreasing fECM (R2 = 0.44) than with infarct distance (R2 = 0.24, p < 0.05). Our results show that t-system remodeling in the rabbit MI border zone resembles a phenotype previously described in human heart failure. T-system remodeling correlated with the amount of local fibrosis, which is known to stiffen cardiac tissue, but was not found in regions without fibrosis. Thus, locally altered tissue mechanics may contribute to t-system remodeling.
Collapse
Affiliation(s)
- T Seidel
- Institute for Cellular and Molecular Physiology, University of Erlangen-Nuremberg, Erlangen, Germany; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA.
| | - A C Sankarankutty
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA; Department of Bioengineering, University of Utah, Salt Lake City, USA
| | - F B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA; Department of Bioengineering, University of Utah, Salt Lake City, USA.
| |
Collapse
|
31
|
Gambardella J, Trimarco B, Iaccarino G, Santulli G. New Insights in Cardiac Calcium Handling and Excitation-Contraction Coupling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1067:373-385. [PMID: 28956314 DOI: 10.1007/5584_2017_106] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Excitation-contraction (EC) coupling denotes the conversion of electric stimulus in mechanic output in contractile cells. Several studies have demonstrated that calcium (Ca2+) plays a pivotal role in this process. Here we present a comprehensive and updated description of the main systems involved in cardiac Ca2+ handling that ensure a functional EC coupling and their pathological alterations, mainly related to heart failure.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy.,Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Fisciano, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Fisciano, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy. .,Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Forch 525, 10461, New York, NY, USA.
| |
Collapse
|