51
|
Liew LC, Ho BX, Soh BS. Mending a broken heart: current strategies and limitations of cell-based therapy. Stem Cell Res Ther 2020; 11:138. [PMID: 32216837 PMCID: PMC7098097 DOI: 10.1186/s13287-020-01648-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
The versatility of pluripotent stem cells, attributable to their unlimited self-renewal capacity and plasticity, has sparked a considerable interest for potential application in regenerative medicine. Over the past decade, the concept of replenishing the lost cardiomyocytes, the crux of the matter in ischemic heart disease, with pluripotent stem cell-derived cardiomyocytes (PSC-CM) has been validated with promising pre-clinical results. Nevertheless, clinical translation was hemmed in by limitations such as immature cardiac properties, long-term engraftment, graft-associated arrhythmias, immunogenicity, and risk of tumorigenicity. The continuous progress of stem cell-based cardiac therapy, incorporated with tissue engineering strategies and delivery of cardio-protective exosomes, provides an optimistic outlook on the development of curative treatment for heart failure. This review provides an overview and current status of stem cell-based therapy for heart regeneration, with particular focus on the use of PSC-CM. In addition, we also highlight the associated challenges in clinical application and discuss the potential strategies in developing successful cardiac-regenerative therapy.
Collapse
Affiliation(s)
- Lee Chuen Liew
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Beatrice Xuan Ho
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore. .,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore. .,Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
52
|
Gilbert G, Demydenko K, Dries E, Puertas RD, Jin X, Sipido K, Roderick HL. Calcium Signaling in Cardiomyocyte Function. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035428. [PMID: 31308143 DOI: 10.1101/cshperspect.a035428] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Rhythmic increases in intracellular Ca2+ concentration underlie the contractile function of the heart. These heart muscle-wide changes in intracellular Ca2+ are induced and coordinated by electrical depolarization of the cardiomyocyte sarcolemma by the action potential. Originating at the sinoatrial node, conduction of this electrical signal throughout the heart ensures synchronization of individual myocytes into an effective cardiac pump. Ca2+ signaling pathways also regulate gene expression and cardiomyocyte growth during development and in pathology. These fundamental roles of Ca2+ in the heart are illustrated by the prevalence of altered Ca2+ homeostasis in cardiovascular diseases. Indeed, heart failure (an inability of the heart to support hemodynamic needs), rhythmic disturbances, and inappropriate cardiac growth all share an involvement of altered Ca2+ handling. The prevalence of these pathologies, contributing to a third of all deaths in the developed world as well as to substantial morbidity makes understanding the mechanisms of Ca2+ handling and dysregulation in cardiomyocytes of great importance.
Collapse
Affiliation(s)
- Guillaume Gilbert
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Kateryna Demydenko
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Eef Dries
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Rosa Doñate Puertas
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Xin Jin
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Karin Sipido
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| |
Collapse
|
53
|
Zhao Y, Rafatian N, Wang EY, Wu Q, Lai BFL, Lu RX, Savoji H, Radisic M. Towards chamber specific heart-on-a-chip for drug testing applications. Adv Drug Deliv Rev 2020; 165-166:60-76. [PMID: 31917972 PMCID: PMC7338250 DOI: 10.1016/j.addr.2019.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
Modeling of human organs has long been a task for scientists in order to lower the costs of therapeutic development and understand the pathological onset of human disease. For decades, despite marked differences in genetics and etiology, animal models remained the norm for drug discovery and disease modeling. Innovative biofabrication techniques have facilitated the development of organ-on-a-chip technology that has great potential to complement conventional animal models. However, human organ as a whole, more specifically the human heart, is difficult to regenerate in vitro, in terms of its chamber specific orientation and its electrical functional complexity. Recent progress with the development of induced pluripotent stem cell differentiation protocols, made recapitulating the complexity of the human heart possible through the generation of cells representative of atrial & ventricular tissue, the sinoatrial node, atrioventricular node and Purkinje fibers. Current heart-on-a-chip approaches incorporate biological, electrical, mechanical, and topographical cues to facilitate tissue maturation, therefore improving the predictive power for the chamber-specific therapeutic effects targeting adult human. In this review, we will give a summary of current advances in heart-on-a-chip technology and provide a comprehensive outlook on the challenges involved in the development of human physiologically relevant heart-on-a-chip.
Collapse
Affiliation(s)
- Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Naimeh Rafatian
- Division of Cardiology and Peter Munk Cardiac Center, University of Health Network, Toronto, Ontario M5G 2N2, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Benjamin F L Lai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xingze Lu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
54
|
Swift LM, Burke M, Guerrelli D, Reilly M, Ramadan M, McCullough D, Prudencio T, Mulvany C, Chaluvadi A, Jaimes R, Posnack NG. Age-dependent changes in electrophysiology and calcium handling: implications for pediatric cardiac research. Am J Physiol Heart Circ Physiol 2019; 318:H354-H365. [PMID: 31886723 DOI: 10.1152/ajpheart.00521.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Rodent models are frequently employed in cardiovascular research, yet our understanding of pediatric cardiac physiology has largely been deduced from more simplified two-dimensional cell studies. Previous studies have shown that postnatal development includes an alteration in the expression of genes and proteins involved in cell coupling, ion channels, and intracellular calcium handling. Accordingly, we hypothesized that postnatal cell maturation is likely to lead to dynamic alterations in whole heart electrophysiology and calcium handling. To test this hypothesis, we employed multiparametric imaging and electrophysiological techniques to quantify developmental changes from neonate to adult. In vivo electrocardiograms were collected to assess changes in heart rate, variability, and atrioventricular conduction (Sprague-Dawley rats). Intact, whole hearts were transferred to a Langendorff-perfusion system for multiparametric imaging (voltage, calcium). Optical mapping was performed in conjunction with an electrophysiology study to assess cardiac dynamics throughout development. Postnatal age was associated with an increase in the heart rate (181 ± 34 vs. 429 ± 13 beats/min), faster atrioventricular conduction (94 ± 13 vs. 46 ± 3 ms), shortened action potentials (APD80: 113 ± 18 vs. 60 ± 17 ms), and decreased ventricular refractoriness (VERP: 157 ± 45 vs. 57 ± 14 ms; neonatal vs. adults, means ± SD, P < 0.05). Calcium handling matured with development, resulting in shortened calcium transient durations (168 ± 18 vs. 117 ± 14 ms) and decreased propensity for calcium transient alternans (160 ± 18- vs. 99 ± 11-ms cycle length threshold; neonatal vs. adults, mean ± SD, P < 0.05). Results of this study can serve as a comprehensive baseline for future studies focused on pediatric disease modeling and/or preclinical testing.NEW & NOTEWORTHY This is the first study to assess cardiac electrophysiology and calcium handling throughout postnatal development, using both in vivo and whole heart models.
Collapse
Affiliation(s)
- Luther M Swift
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Morgan Burke
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Devon Guerrelli
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Marissa Reilly
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Manelle Ramadan
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Damon McCullough
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Tomas Prudencio
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Colm Mulvany
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Ashika Chaluvadi
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Rafael Jaimes
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia.,Department of Pediatrics and Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia
| |
Collapse
|
55
|
Espejo MS, Orlowski A, Ibañez AM, Di Mattía RA, Velásquez FC, Rossetti NS, Ciancio MC, De Giusti VC, Aiello EA. The functional association between the sodium/bicarbonate cotransporter (NBC) and the soluble adenylyl cyclase (sAC) modulates cardiac contractility. Pflugers Arch 2019; 472:103-115. [PMID: 31754830 DOI: 10.1007/s00424-019-02331-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/15/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022]
Abstract
The soluble adenylyl cyclase (sAC) was identified in the heart as another source of cyclic AMP (cAMP). However, its cardiac physiological function is unknown. On the other hand, the cardiac Na+/HCO3- cotransporter (NBC) promotes the cellular co-influx of HCO3- and Na+. Since sAC activity is regulated by HCO3-, our purpose was to investigate the potential functional relationship between NBC and sAC in the cardiomyocyte. Rat ventricular myocytes were loaded with Fura-2, Fluo-3, or BCECF to measure Ca2+ transient (Ca2+i) by epifluorescence, Ca2+ sparks frequency (CaSF) by confocal microscopy, or intracellular pH (pHi) by epifluorescence, respectively. Sarcomere or cell shortening was measured with a video camera as an index of contractility. The NBC blocker S0859 (10 μM), the selective inhibitor of sAC KH7 (1 μM), and the PKA inhibitor H89 (0.1 μM) induced a negative inotropic effect which was associated with a decrease in Ca2+i. Since PKA increases Ca2+ release through sarcoplasmic reticulum RyR channels, CaSF was measured as an index of RyR open probability. The generation of CaSF was prevented by KH7. Finally, we investigated the potential role of sAC activation on NBC activity. NBC-mediated recovery from acidosis was faster in the presence of KH7 or H89, suggesting that the pathway sAC-PKA is negatively regulating NBC function, consistent with a negative feedback modulation of the HCO3- influx that activates sAC. In summary, the results demonstrated that the complex NBC-sAC-PKA plays a relevant role in Ca2+ handling and basal cardiac contractility.
Collapse
Affiliation(s)
- María S Espejo
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Alejandro Orlowski
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Alejandro M Ibañez
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Romina A Di Mattía
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Fernanda Carrizo Velásquez
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Noelia S Rossetti
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - María C Ciancio
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina
| | - Verónica C De Giusti
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina.
| | - Ernesto A Aiello
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata-CONICET, Calle 60 y 120, 1900, La Plata, Argentina.
| |
Collapse
|
56
|
Chan BYH, Roczkowsky A, Cho WJ, Poirier M, Lee TYT, Mahmud Z, Schulz R. Junctophilin-2 is a target of matrix metalloproteinase-2 in myocardial ischemia-reperfusion injury. Basic Res Cardiol 2019; 114:42. [PMID: 31506724 DOI: 10.1007/s00395-019-0749-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/02/2019] [Indexed: 11/30/2022]
Abstract
Junctophilin-2 is a structural membrane protein that tethers T-tubules to the sarcoplasmic reticulum to allow for coordinated calcium-induced calcium release in cardiomyocytes. Defective excitation-contraction coupling in myocardial ischemia-reperfusion (IR) injury is associated with junctophilin-2 proteolysis. However, it remains unclear whether preventing junctophilin-2 proteolysis improves the recovery of cardiac contractile dysfunction in IR injury. Matrix metalloproteinase-2 (MMP-2) is a zinc and calcium-dependent protease that is activated by oxidative stress in myocardial IR injury and cleaves both intracellular and extracellular substrates. To determine whether junctophilin-2 is targeted by MMP-2, isolated rat hearts were perfused in working mode aerobically or subjected to IR injury with the selective MMP inhibitor ARP-100. IR injury impaired the recovery of cardiac contractile function which was associated with increased degradation of junctophilin-2 and damaged cardiac dyads. In IR hearts, ARP-100 improved the recovery of cardiac contractile function, attenuated junctophilin-2 proteolysis, and prevented ultrastructural damage to the dyad. MMP-2 was co-localized with junctophilin-2 in aerobic and IR hearts by immunoprecipitation and immunohistochemistry. In situ zymography showed that MMP activity was localized to the Z-disc and sarcomere in aerobic hearts and accumulated at sites where the striated JPH-2 staining was disrupted in IR hearts. In vitro proteolysis assays determined that junctophilin-2 is susceptible to proteolysis by MMP-2 and in silico analysis predicted multiple MMP-2 cleavage sites between the membrane occupation and recognition nexus repeats and within the divergent region of junctophilin-2. Degradation of junctophilin-2 by MMP-2 is an early consequence of myocardial IR injury which may initiate a cascade of sequelae leading to impaired contractile function.
Collapse
Affiliation(s)
- Brandon Y H Chan
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Andrej Roczkowsky
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Woo Jung Cho
- Faculty of Medicine and Dentistry Cell Imaging Centre, University of Alberta, Edmonton, AB, Canada
| | - Mathieu Poirier
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Tim Y T Lee
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Zabed Mahmud
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
57
|
Liu C, Spinozzi S, Chen JY, Fang X, Feng W, Perkins G, Cattaneo P, Guimarães-Camboa N, Dalton ND, Peterson KL, Wu T, Ouyang K, Fu XD, Evans SM, Chen J. Nexilin Is a New Component of Junctional Membrane Complexes Required for Cardiac T-Tubule Formation. Circulation 2019; 140:55-66. [PMID: 30982350 PMCID: PMC6889818 DOI: 10.1161/circulationaha.119.039751] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/02/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Membrane contact sites are fundamental for transmission and translation of signals in multicellular organisms. The junctional membrane complexes in the cardiac dyads, where transverse (T) tubules are juxtaposed to the sarcoplasmic reticulum, are a prime example. T-tubule uncoupling and remodeling are well-known features of cardiac disease and heart failure. Even subtle alterations in the association between T-tubules and the junctional sarcoplasmic reticulum can cause serious cardiac disorders. NEXN (nexilin) has been identified as an actin-binding protein, and multiple mutations in the NEXN gene are associated with cardiac diseases, but the precise role of NEXN in heart function and disease is still unknown. METHODS Nexn global and cardiomyocyte-specific knockout mice were generated. Comprehensive phenotypic and RNA sequencing and mass spectrometry analyses were performed. Heart tissue samples and isolated single cardiomyocytes were analyzed by electron and confocal microscopy. RESULTS Global and cardiomyocyte-specific loss of Nexn in mice resulted in a rapidly progressive dilated cardiomyopathy. In vivo and in vitro analyses revealed that NEXN interacted with junctional sarcoplasmic reticulum proteins, was essential for optimal calcium transients, and was required for initiation of T-tubule invagination and formation. CONCLUSIONS These results demonstrated that NEXN is a pivotal component of the junctional membrane complex and is required for initiation and formation of T-tubules, thus providing insight into mechanisms underlying cardiomyopathy in patients with mutations in NEXN.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/metabolism
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cell Membrane/genetics
- Cell Membrane/metabolism
- Cell Membrane/pathology
- Cells, Cultured
- Intercellular Junctions/genetics
- Intercellular Junctions/metabolism
- Intercellular Junctions/pathology
- Mice
- Mice, Knockout
- Mice, Transgenic
- Microfilament Proteins/deficiency
- Microfilament Proteins/genetics
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
Collapse
Affiliation(s)
- Canzhao Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Simone Spinozzi
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia-Yu Chen
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Wei Feng
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Paola Cattaneo
- National Research Council, Institute of Genetics and Biomedical Research, Milan 20138, Italy
- Humanitas Clinical and Research Center, Rozzano 20089, Italy
| | - Nuno Guimarães-Camboa
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Berlin 13347, Germany
| | - Nancy D. Dalton
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kirk L. Peterson
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Sylvia M. Evans
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
58
|
Kong CHT, Bryant SM, Watson JJ, Gadeberg HC, Roth DM, Patel HH, Cannell MB, Orchard CH, James AF. The Effects of Aging on the Regulation of T-Tubular ICa by Caveolin in Mouse Ventricular Myocytes. J Gerontol A Biol Sci Med Sci 2019; 73:711-719. [PMID: 29236992 PMCID: PMC5946816 DOI: 10.1093/gerona/glx242] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 12/07/2017] [Indexed: 11/25/2022] Open
Abstract
Aging is associated with diminished cardiac function in males. Cardiac excitation-contraction coupling in ventricular myocytes involves Ca influx via the Ca current (ICa) and Ca release from the sarcoplasmic reticulum, which occur predominantly at t-tubules. Caveolin-3 regulates t-tubular ICa, partly through protein kinase A (PKA), and both ICa and caveolin-3 decrease with age. We therefore investigated ICa and t-tubule structure and function in cardiomyocytes from male wild-type (WT) and caveolin-3-overexpressing (Cav-3OE) mice at 3 and 24 months of age. In WT cardiomyocytes, t-tubular ICa-density was reduced by ~50% with age while surface ICa density was unchanged. Although regulation by PKA was unaffected by age, inhibition of caveolin-3-binding reduced t-tubular ICa at 3 months, but not at 24 months. While Cav-3OE increased cardiac caveolin-3 protein expression ~2.5-fold at both ages, the age-dependent reduction in caveolin-3 (WT ~35%) was preserved in transgenic mice. Overexpression of caveolin-3 reduced t-tubular ICa density at 3 months but prevented further ICa loss with age. Measurement of Ca release at the t-tubules revealed that the triggering of local Ca release by t-tubular ICa was unaffected by age. In conclusion, the data suggest that the reduction in ICa density with age is associated with the loss of a caveolin-3-dependent mechanism that augments t-tubular ICa density.
Collapse
Affiliation(s)
- Cherrie H T Kong
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| | - Simon M Bryant
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| | - Judy J Watson
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| | - Hanne C Gadeberg
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| | - David M Roth
- VA San Diego Healthcare System and Department of Anesthesiology, University of California, San Diego
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California, San Diego
| | - Mark B Cannell
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| | - Clive H Orchard
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| | - Andrew F James
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK
| |
Collapse
|
59
|
Lipsett DB, Frisk M, Aronsen JM, Nordén ES, Buonarati OR, Cataliotti A, Hell JW, Sjaastad I, Christensen G, Louch WE. Cardiomyocyte substructure reverts to an immature phenotype during heart failure. J Physiol 2019; 597:1833-1853. [PMID: 30707448 PMCID: PMC6441900 DOI: 10.1113/jp277273] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Key points As reactivation of the fetal gene program has been implicated in pathological remodelling during heart failure (HF), we examined whether cardiomyocyte subcellular structure and function revert to an immature phenotype during this disease. Surface and internal membrane structures appeared gradually during development, and returned to a juvenile state during HF. Similarly, dyadic junctions between the cell membrane and sarcoplasmic reticulum were progressively ‘packed’ with L‐type Ca2+ channels and ryanodine receptors during development, and ‘unpacked’ during HF. Despite similarities in subcellular structure, dyads were observed to be functional from early developmental stages, but exhibited an impaired ability to release Ca2+ in failing cardiomyocytes. Thus, while immature and failing cardiomyocytes share similarities in subcellular structure, these do not fully account for the marked impairment of Ca2+ homeostasis observed in HF.
Abstract Reactivation of the fetal gene programme has been implicated as a driver of pathological cardiac remodelling. Here we examined whether pathological remodelling of cardiomyocyte substructure and function during heart failure (HF) reflects a reversion to an immature phenotype. Using scanning electron microscopy, we observed that Z‐grooves and t‐tubule openings at the cell surface appeared gradually during cardiac development, and disappeared during HF. Confocal and super‐resolution imaging within the cell interior revealed similar structural parallels; disorganization of t‐tubules in failing cells was strikingly reminiscent of the late stages of postnatal development, with fewer transverse elements and a high proportion of longitudinal tubules. Ryanodine receptors (RyRs) were observed to be laid down in advance of developing t‐tubules and similarly ‘orphaned’ in HF, although RyR distribution along Z‐lines was relatively sparse. Indeed, nanoscale imaging revealed coordinated packing of L‐type Ca2+ channels and RyRs into dyadic junctions during development, and orderly unpacking during HF. These findings support a ‘last in, first out’ paradigm, as the latest stages of dyadic structural development are reversed during disease. Paired imaging of t‐tubules and Ca2+ showed that the disorganized arrangement of dyads in immature and failing cells promoted desynchronized and slowed Ca2+ release in these two states. However, while developing cells exhibited efficient triggering of Ca2+ release at newly formed dyads, dyadic function was impaired in failing cells despite similar organization of Ca2+ handling proteins. Thus, pathologically deficient Ca2+ homeostasis during HF is only partly linked to the re‐emergence of immature subcellular structure, and additionally reflects lost dyadic functionality. As reactivation of the fetal gene program has been implicated in pathological remodelling during heart failure (HF), we examined whether cardiomyocyte subcellular structure and function revert to an immature phenotype during this disease. Surface and internal membrane structures appeared gradually during development, and returned to a juvenile state during HF. Similarly, dyadic junctions between the cell membrane and sarcoplasmic reticulum were progressively ‘packed’ with L‐type Ca2+ channels and ryanodine receptors during development, and ‘unpacked’ during HF. Despite similarities in subcellular structure, dyads were observed to be functional from early developmental stages, but exhibited an impaired ability to release Ca2+ in failing cardiomyocytes. Thus, while immature and failing cardiomyocytes share similarities in subcellular structure, these do not fully account for the marked impairment of Ca2+ homeostasis observed in HF.
Collapse
Affiliation(s)
- D B Lipsett
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - M Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J M Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - E S Nordén
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - O R Buonarati
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - A Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J W Hell
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - I Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - G Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - W E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
60
|
Fernández-Morales JC, Hua W, Yao Y, Morad M. Regulation of Ca 2+ signaling by acute hypoxia and acidosis in cardiomyocytes derived from human induced pluripotent stem cells. Cell Calcium 2018; 78:1-14. [PMID: 30579812 DOI: 10.1016/j.ceca.2018.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 12/20/2022]
Abstract
AIMS The effects of acute (100 s) hypoxia and/or acidosis on Ca2+ signaling parameters of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are explored here for the first time. METHODS AND RESULTS 1) hiPSC-CMs express two cell populations: rapidly-inactivating ICa myocytes (τi<40 ms, in 4-5 day cultures) and slowly-inactivating ICa (τi ≥ 40 ms, in 6-8 day cultures). 2) Hypoxia suppressed ICa by 10-20% in rapidly- and 40-55% in slowly-inactivating ICa cells. 3) Isoproterenol enhanced ICa in hiPSC-CMs, but either enhanced or did not alter the hypoxic suppression. 4) Hypoxia had no differential suppressive effects in the two cell-types when Ba2+ was the charge carrier through the calcium channels, implicating Ca2+-dependent inactivation in O2 sensing. 5) Acidosis suppressed ICa by ∼35% and ∼25% in rapidly and slowly inactivating ICa cells, respectively. 6) Hypoxia and acidosis suppressive effects on Ca-transients depended on whether global or RyR2-microdomain were measured: with acidosis suppression was ∼25% in global and ∼37% in RyR2 Ca2+-microdomains in either cell type, whereas with hypoxia suppression was ∼20% and ∼25% respectively in global and RyR2-microdomaine in rapidly and ∼35% and ∼45% respectively in global and RyR2-microdomaine in slowly-inactivating cells. CONCLUSIONS Variability in ICa inactivation kinetics rather than cellular ancestry seems to underlie the action potential morphology differences generally attributed to mixed atrial and ventricular cell populations in hiPSC-CMs cultures. The differential hypoxic regulation of Ca2+-signaling in the two-cell types arises from differential Ca2+-dependent inactivation of the Ca2+-channel caused by proximity of Ca2+-release stores to the Ca2+ channels.
Collapse
Affiliation(s)
| | - Wei Hua
- Cardiac Signaling Center of MUSC, USC and Clemson, Charleston, SC, USA
| | - Yuyu Yao
- Cardiac Signaling Center of MUSC, USC and Clemson, Charleston, SC, USA
| | - Martin Morad
- Cardiac Signaling Center of MUSC, USC and Clemson, Charleston, SC, USA; Department of Pharmacology,Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
61
|
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
|
62
|
Jackman C, Li H, Bursac N. Long-term contractile activity and thyroid hormone supplementation produce engineered rat myocardium with adult-like structure and function. Acta Biomater 2018; 78:98-110. [PMID: 30086384 DOI: 10.1016/j.actbio.2018.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
The field of cardiac tissue engineering has developed rapidly, but structural and functional immaturity of engineered heart tissues hinder their widespread use. Here, we show that a combination of low-rate (0.2 Hz) contractile activity and thyroid hormone (T3) supplementation significantly promote structural and functional maturation of engineered rat cardiac tissues ("cardiobundles"). The progressive maturation of cardiobundles during first 2 weeks of culture resulted in cell cycle exit and loss of spontaneous activity, which in longer culture yielded decreased contractile function. Maintaining a low level of contractile activity by 0.2 Hz pacing between culture weeks 3 and 5, combined with T3 treatment, yielded significant growth of cardiobundle and myocyte cross-sectional areas (by 68% and 32%, respectively), increased nuclei numbers (by 22%), improved twitch force (by 39%), shortened action potential duration (by 32%), polarized N-cadherin distribution, and switch from immature (slow skeletal) to mature (fast) cardiac troponin I isoform expression. Along with advanced functional output (conduction velocity 53.7 ± 0.8 cm/s, specific force 70.1 ± 5.8 mN/mm2), quantitative ultrastructural analyses revealed similar metrics and abundance of sarcomeres, T-tubules, M-bands, and intercalated disks compared to native age-matched (5-week) and adult (3-month) ventricular myocytes. Unlike 0.2 Hz regime, chronic 1 Hz pacing resulted in significant cardiomyocyte loss and formation of necrotic core despite the use of dynamic culture. Overall, our results demonstrate remarkable ultrastructural and functional maturation of neonatal rat cardiomyocytes in 3D culture and reveal importance of combined biophysical and hormonal inputs for in vitro engineering of adult-like myocardium. STATEMENT OF SIGNIFICANCE Compared to human stem cell-derived cardiomyocytes, neonatal rat ventricular myocytes show advanced maturation state which makes them suitable for in vitro studies of postnatal cardiac development. Still, maturation process from a neonatal to an adult cardiomyocyte has not been recapitulated in rodent cell cultures. Here, we show that low-frequency pacing and thyroid hormone supplementation of 3D engineered neonatal rat cardiac tissues synergistically yield significant increase in cell and tissue volume, robust formation of T-tubules and M-lines, improved sarcomere organization, and faster and more forceful contractions. To the best of our knowledge, 5-week old engineered cardiac tissues described in this study are the first that exhibit both ultrastructural and functional characteristics approaching or matching those of adult ventricular myocardium.
Collapse
|
63
|
Jiang Y, Park P, Hong SM, Ban K. Maturation of Cardiomyocytes Derived from Human Pluripotent Stem Cells: Current Strategies and Limitations. Mol Cells 2018; 41:613-621. [PMID: 29890820 PMCID: PMC6078855 DOI: 10.14348/molcells.2018.0143] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 01/06/2023] Open
Abstract
The capacity of differentiation of human pluripotent stem cells (hPSCs), which include both embryonic stem cells and induced pluripotent stem cells, into cardiomyocytes (CMs) in vitro provides an unlimited resource for human CMs for a wide range of applications such as cell based cardiac repair, cardiac drug toxicology screening, and human cardiac disease modeling. However, their applicability is significantly limited by immature phenotypes. It has been well known that currently available CMs derived from hPSCs (hPSC-CMs) represent immature embryonic or fetal stage CMs and are functionally and structurally different from mature human CMs. To overcome this critical issue, several new approaches aiming to generate more mature hPSC-CMs have been developed. This review describes recent approaches to generate more mature hPSC-CMs including their scientific principles, advantages, and limitations.
Collapse
Affiliation(s)
- Yanqing Jiang
- University of Toronto, Hospital of Sick Children, Toronto,
Canada
| | - Peter Park
- Emory University, Department of Biology, Atlanta, Georgia,
USA
| | - Sang-Min Hong
- Department of Physical Education, Dongguk University Seoul, Seoul 04620,
Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong,
Hong Kong
| |
Collapse
|
64
|
Bryant SM, Kong CHT, Watson JJ, Gadeberg HC, James AF, Cannell MB, Orchard CH. Caveolin 3-dependent loss of t-tubular I Ca during hypertrophy and heart failure in mice. Exp Physiol 2018; 103:652-665. [PMID: 29473235 PMCID: PMC6099270 DOI: 10.1113/ep086731] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/15/2018] [Indexed: 12/29/2022]
Abstract
NEW FINDINGS What is the central question of this study? Heart failure is associated with redistribution of L-type Ca2+ current (ICa ) away from the t-tubule membrane to the surface membrane of cardiac ventricular myocytes. However, the underlying mechanism and its dependence on severity of pathology (hypertrophy versus failure) are unclear. What is the main finding and its importance? Increasing severity of response to transverse aortic constriction, from hypertrophy to failure, was accompanied by graded loss of t-tubular ICa and loss of regulation of ICa by caveolin 3. Thus, the pathological loss of t-tubular ICa , which contributes to impaired excitation-contraction coupling and thereby cardiac function in vivo, appears to be attributable to loss of caveolin 3-dependent stimulation of t-tubular ICa . ABSTRACT Previous work has shown redistribution of L-type Ca2+ current (ICa ) from the t-tubules to the surface membrane of rat ventricular myocytes after myocardial infarction. However, whether this occurs in all species and in response to other insults, the relationship of this redistribution to the severity of the pathology, and the underlying mechanism, are unknown. We have therefore investigated the response of mouse hearts and myocytes to pressure overload induced by transverse aortic constriction (TAC). Male C57BL/6 mice underwent TAC or equivalent sham operation 8 weeks before use. ICa and Ca2+ transients were measured in isolated myocytes, and expression of caveolin 3 (Cav3), junctophilin 2 (Jph2) and bridging integrator 1 (Bin1) was determined. C3SD peptide was used to disrupt Cav3 binding to its protein partners. Some animals showed cardiac hypertrophy in response to TAC with little evidence of heart failure, whereas others showed greater hypertrophy and pulmonary congestion. These graded changes were accompanied by graded cellular hypertrophy, t-tubule disruption, decreased expression of Jph2 and Cav3, and decreased t-tubular ICa density, with no change at the cell surface, and graded impairment of Ca2+ release at t-tubules. C3SD decreased ICa density in control but not in TAC myocytes. These data suggest that the graded changes in cardiac function and size that occur in response to TAC are paralleled by graded changes in cell structure and function, which will contribute to the impaired function observed in vivo. They also suggest that loss of t-tubular ICa is attributable to loss of Cav3-dependent stimulation of ICa .
Collapse
Affiliation(s)
- Simon M Bryant
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Cherrie H T Kong
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Judy J Watson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Hanne C Gadeberg
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Andrew F James
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Mark B Cannell
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Clive H Orchard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
65
|
Schobesberger S, Wright P, Tokar S, Bhargava A, Mansfield C, Glukhov AV, Poulet C, Buzuk A, Monszpart A, Sikkel M, Harding SE, Nikolaev VO, Lyon AR, Gorelik J. T-tubule remodelling disturbs localized β2-adrenergic signalling in rat ventricular myocytes during the progression of heart failure. Cardiovasc Res 2018; 113:770-782. [PMID: 28505272 PMCID: PMC5437368 DOI: 10.1093/cvr/cvx074] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/11/2017] [Indexed: 11/12/2022] Open
Abstract
Aims Cardiomyocyte β2-adrenergic receptor (β2AR) cyclic adenosine monophosphate (cAMP) signalling is regulated by the receptors' subcellular location within transverse tubules (T-tubules), via interaction with structural and regulatory proteins, which form a signalosome. In chronic heart failure (HF), β2ARs redistribute from T-tubules to the cell surface, which disrupts functional signalosomes and leads to diffuse cAMP signalling. However, the functional consequences of structural changes upon β2AR-cAMP signalling during progression from hypertrophy to advanced HF are unknown. Methods and results Rat left ventricular myocytes were isolated at 4-, 8-, and 16-week post-myocardial infarction (MI), β2ARs were stimulated either via whole-cell perfusion or locally through the nanopipette of the scanning ion conductance microscope. cAMP release was measured via a Förster Resonance Energy Transfer-based sensor Epac2-camps. Confocal imaging of di-8-ANNEPS-stained cells and immunoblotting were used to determine structural alterations. At 4-week post-MI, T-tubule regularity, density and junctophilin-2 (JPH2) expression were significantly decreased. The amplitude of local β2AR-mediated cAMP in T-tubules was reduced and cAMP diffused throughout the cytosol instead of being locally confined. This was accompanied by partial caveolin-3 (Cav-3) dissociation from the membrane. At 8-week post-MI, the β2AR-mediated cAMP response was observed at the T-tubules and the sarcolemma (crest). Finally, at 16-week post-MI, the whole cell β2AR-mediated cAMP signal was depressed due to adenylate cyclase dysfunction, while overall Cav-3 levels were significantly increased and a substantial portion of Cav-3 dissociated into the cytosol. Overexpression of JPH2 in failing cells in vitro or AAV9.SERCA2a gene therapy in vivo did not improve β2AR-mediated signal compartmentation or reduce cAMP diffusion. Conclusion Although changes in T-tubule structure and β2AR-mediated cAMP signalling are significant even at 4-week post-MI, progression to the HF phenotype is not linear. At 8-week post-MI the loss of β2AR-mediated cAMP is temporarily reversed. Complete disorganization of β2AR-mediated cAMP signalling due to changes in functional receptor localization and cellular structure occurs at 16-week post-MI.
Collapse
Affiliation(s)
- Sophie Schobesberger
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martinistraße, Hamburg D-20246, Germany
| | - Peter Wright
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Sergiy Tokar
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Anamika Bhargava
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,Department of Biotechnology, Indian Institute of Technology Hyderabad, Ordnance Factory Estate, Yeddumailaram, 502205 Telangana, India
| | - Catherine Mansfield
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Alexey V Glukhov
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Claire Poulet
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Andrey Buzuk
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Aron Monszpart
- Department of Computer Science, University College London, Gower Street, London WC1E 6BT, UK
| | - Markus Sikkel
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Sian E Harding
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Martinistraße, Hamburg D-20246, Germany
| | - Alexander R Lyon
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12 0NN, UK.,NIHR Cardiovascular Biomedical Research Unit, Department of Cardiology, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Julia Gorelik
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College, Du Cane Road, London W12?0NN, UK
| |
Collapse
|
66
|
Human ISL1 + Ventricular Progenitors Self-Assemble into an In Vivo Functional Heart Patch and Preserve Cardiac Function Post Infarction. Mol Ther 2018; 26:1644-1659. [PMID: 29606507 PMCID: PMC6035340 DOI: 10.1016/j.ymthe.2018.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/09/2018] [Accepted: 02/11/2018] [Indexed: 11/26/2022] Open
Abstract
The generation of human pluripotent stem cell (hPSC)-derived ventricular progenitors and their assembly into a 3-dimensional in vivo functional ventricular heart patch has remained an elusive goal. Herein, we report the generation of an enriched pool of hPSC-derived ventricular progenitors (HVPs), which can expand, differentiate, self-assemble, and mature into a functional ventricular patch in vivo without the aid of any gel or matrix. We documented a specific temporal window, in which the HVPs will engraft in vivo. On day 6 of differentiation, HVPs were enriched by depleting cells positive for pluripotency marker TRA-1-60 with magnetic-activated cell sorting (MACS), and 3 million sorted cells were sub-capsularly transplanted onto kidneys of NSG mice where, after 2 months, they formed a 7 mm × 3 mm × 4 mm myocardial patch resembling the ventricular wall. The graft acquired several features of maturation: expression of ventricular marker (MLC2v), desmosomes, appearance of T-tubule-like structures, and electrophysiological action potential signature consistent with maturation, all this in a non-cardiac environment. We further demonstrated that HVPs transplanted into un-injured hearts of NSG mice remain viable for up to 8 months. Moreover, transplantation of 2 million HVPs largely preserved myocardial contractile function following myocardial infarction. Taken together, our study reaffirms the promising idea of using progenitor cells for regenerative therapy.
Collapse
|
67
|
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]
|
68
|
Schilling JM, Head BP, Patel HH. Caveolins as Regulators of Stress Adaptation. Mol Pharmacol 2018; 93:277-285. [PMID: 29358220 DOI: 10.1124/mol.117.111237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/19/2018] [Indexed: 12/21/2022] Open
Abstract
Caveolins have been recognized over the past few decades as key regulators of cell physiology. They are ubiquitously expressed and regulate a number of processes that ultimately impact efficiency of cellular processes. Though not critical to life, they are central to stress adaptation in a number of organs. The following review will focus specifically on the role of caveolin in stress adaptation in the heart, brain, and eye, three organs that are susceptible to acute and chronic stress and that show as well declining function with age. In addition, we consider some novel molecular mechanisms that may account for this stress adaptation and also offer potential to drive the future of caveolin research.
Collapse
Affiliation(s)
- Jan M Schilling
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| | - Brian P Head
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| | - Hemal H Patel
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| |
Collapse
|
69
|
Besser RR, Ishahak M, Mayo V, Carbonero D, Claure I, Agarwal A. Engineered Microenvironments for Maturation of Stem Cell Derived Cardiac Myocytes. Am J Cancer Res 2018; 8:124-140. [PMID: 29290797 PMCID: PMC5743464 DOI: 10.7150/thno.19441] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/19/2017] [Indexed: 01/11/2023] Open
Abstract
Through the use of stem cell-derived cardiac myocytes, tissue-engineered human myocardial constructs are poised for modeling normal and diseased physiology of the heart, as well as discovery of novel drugs and therapeutic targets in a human relevant manner. This review highlights the recent bioengineering efforts to recapitulate microenvironmental cues to further the maturation state of newly differentiated cardiac myocytes. These techniques include long-term culture, co-culture, exposure to mechanical stimuli, 3D culture, cell-matrix interactions, and electrical stimulation. Each of these methods has produced various degrees of maturation; however, a standardized measure for cardiomyocyte maturation is not yet widely accepted by the scientific community.
Collapse
|
70
|
Park M, Yoon YS. Cardiac Regeneration with Human Pluripotent Stem Cell-Derived Cardiomyocytes. Korean Circ J 2018; 48:974-988. [PMID: 30334384 PMCID: PMC6196153 DOI: 10.4070/kcj.2018.0312] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/27/2018] [Indexed: 12/29/2022] Open
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), which are collectively called pluripotent stem cells (PSCs), have emerged as a promising source for regenerative medicine. Particularly, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have shown robust potential for regenerating injured heart. Over the past two decades, protocols to differentiate hPSCs into CMs at high efficiency have been developed, opening the door for clinical application. Studies further demonstrated therapeutic effects of hPSC-CMs in small and large animal models and the underlying mechanisms of cardiac repair. However, gaps remain in explanations of the therapeutic effects of engrafted hPSC-CMs. In addition, bioengineering technologies improved survival and therapeutic effects of hPSC-CMs in vivo. While most of the original concerns associated with the use of hPSCs have been addressed, several issues remain to be resolved such as immaturity of transplanted cells, lack of electrical integration leading to arrhythmogenic risk, and tumorigenicity. Cell therapy with hPSC-CMs has shown great potential for biological therapy of injured heart; however, more studies are needed to ensure the therapeutic effects, underlying mechanisms, and safety, before this technology can be applied clinically.
Collapse
Affiliation(s)
- Misun Park
- Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Young Sup Yoon
- Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
71
|
Abstract
PURPOSE OF REVIEW Membrane invaginations called t-tubules play an integral role in triggering cardiomyocyte contraction, and their disruption during diseases such as heart failure critically impairs cardiac performance. In this review, we outline the growing understanding of the malleability of t-tubule structure and function, and highlight emerging t-tubule regulators which may be exploited for novel therapies. RECENT FINDINGS New technologies are revealing the nanometer scale organization of t-tubules, and their functional junctions with the sarcoplasmic reticulum called dyads, which generate Ca2+ sparks. Recent data have indicated that the dyadic anchoring protein junctophilin-2, and the membrane-bending protein BIN1 are key regulators of dyadic formation and maintenance. While the underlying signals which control expression and localization of these proteins remain unclear, accumulating data support an important role of myocardial workload. Although t-tubule alterations are believed to be a key cause of heart failure, the plasticity of these structures also creates an opportunity for therapy. Promising recent data suggest that such therapies may specifically target junctophilin-2, BIN1, and/or mechanotransduction.
Collapse
|
72
|
Cho GS, Lee DI, Tampakakis E, Murphy S, Andersen P, Uosaki H, Chelko S, Chakir K, Hong I, Seo K, Chen HSV, Chen X, Basso C, Houser SR, Tomaselli GF, O'Rourke B, Judge DP, Kass DA, Kwon C. Neonatal Transplantation Confers Maturation of PSC-Derived Cardiomyocytes Conducive to Modeling Cardiomyopathy. Cell Rep 2017; 18:571-582. [PMID: 28076798 DOI: 10.1016/j.celrep.2016.12.040] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 12/05/2016] [Accepted: 12/13/2016] [Indexed: 12/31/2022] Open
Abstract
Pluripotent stem cells (PSCs) offer unprecedented opportunities for disease modeling and personalized medicine. However, PSC-derived cells exhibit fetal-like characteristics and remain immature in a dish. This has emerged as a major obstacle for their application for late-onset diseases. We previously showed that there is a neonatal arrest of long-term cultured PSC-derived cardiomyocytes (PSC-CMs). Here, we demonstrate that PSC-CMs mature into adult CMs when transplanted into neonatal hearts. PSC-CMs became similar to adult CMs in morphology, structure, and function within a month of transplantation into rats. The similarity was further supported by single-cell RNA-sequencing analysis. Moreover, this in vivo maturation allowed patient-derived PSC-CMs to reveal the disease phenotype of arrhythmogenic right ventricular cardiomyopathy, which manifests predominantly in adults. This study lays a foundation for understanding human CM maturation and pathogenesis and can be instrumental in PSC-based modeling of adult heart diseases.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dong I Lee
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emmanouil Tampakakis
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hideki Uosaki
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen Chelko
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Khalid Chakir
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kinya Seo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Huei-Sheng Vincent Chen
- Del E. Webb Neuroscience, Aging & Stem Cell Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Xiongwen Chen
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Cristina Basso
- Department of Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140 USA; Department of Cardiac, Thoracic, and Vascular Sciences, University of Padua, Padova, Italy
| | - Steven R Houser
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Gordon F Tomaselli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel P Judge
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
73
|
Fernández-Morales JC, Morad M. Regulation of Ca 2+ signaling by acute hypoxia and acidosis in rat neonatal cardiomyocytes. J Mol Cell Cardiol 2017; 114:58-71. [PMID: 29032102 DOI: 10.1016/j.yjmcc.2017.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/20/2017] [Accepted: 10/08/2017] [Indexed: 11/25/2022]
Abstract
Ischemic heart disease is an arrhythmogenic condition, accompanied by hypoxia, acidosis, and impaired Ca2+ signaling. Here we report on effects of acute hypoxia and acidification in rat neonatal cardiomyocytes cultures. RESULTS Two populations of neonatal cardiomyocyte were identified based on inactivation kinetics of L-type ICa: rapidly-inactivating ICa (τ~20ms) myocytes (prevalent in 3-4-day cultures), and slow-inactivating ICa (τ≥40ms) myocytes (dominant in 7-day cultures). Acute hypoxia (pO2<5mmHg for 50-100s) suppressed ICa reversibly in both cell-types to different extent and with different kinetics. This disparity disappeared when Ba2+ was the channel charge carrier, or when the intracellular Ca2+ buffering capacity was increased by dialysis of high concentrations of EGTA and BAPTA, suggesting critical role for calcium-dependent inactivation. Suppressive effect of acute acidosis on ICa (~40%, pH6.7), on the other hand, was not cell-type dependent. Isoproterenol enhanced ICa in both cell-types, but protected only against suppressive effects of acidosis and not hypoxia. Hypoxia and acidosis suppressed global Ca2+ transients by ~20%, but suppression was larger, ~35%, at the RyR2 microdomains, using GCaMP6-FKBP targeted probe. Hypoxia and acidosis also suppressed mitochondrial Ca2+ uptake by 40% and 10%, respectively, using mitochondrial targeted Ca2+ biosensor (mito-GCaMP6). CONCLUSION Our studies suggest that acute hypoxia suppresses ICa in rapidly inactivating cell population by a mechanism involving Ca2+-dependent inactivation, while compromised mitochondrial Ca2+ uptake seems also to contribute to ICa suppression in slowly inactivating cell population. Proximity of cellular Ca2+ pools to sarcolemmal Ca2+ channels may contribute to the variability of inactivation kinetics of ICa in the two cell populations, while acidosis suppression of ICa appears mediated by proton-induced block of the calcium channel.
Collapse
Affiliation(s)
| | - Martin Morad
- Cardiac Signaling Center of MUSC, USC and Clemson, Charleston, SC, USA; Department of Pharmacology, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
74
|
Parikh SS, Blackwell DJ, Gomez-Hurtado N, Frisk M, Wang L, Kim K, Dahl CP, Fiane A, Tønnessen T, Kryshtal DO, Louch WE, Knollmann BC. Thyroid and Glucocorticoid Hormones Promote Functional T-Tubule Development in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Circ Res 2017; 121:1323-1330. [PMID: 28974554 DOI: 10.1161/circresaha.117.311920] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/26/2017] [Accepted: 10/02/2017] [Indexed: 12/16/2022]
Abstract
RATIONALE Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are increasingly being used for modeling heart disease and are under development for regeneration of the injured heart. However, incomplete structural and functional maturation of hiPSC-CM, including lack of T-tubules, immature excitation-contraction coupling, and inefficient Ca-induced Ca release remain major limitations. OBJECTIVE Thyroid and glucocorticoid hormones are critical for heart maturation. We hypothesized that their addition to standard protocols would promote T-tubule development and mature excitation-contraction coupling of hiPSC-CM when cultured on extracellular matrix with physiological stiffness (Matrigel mattress). METHODS AND RESULTS hiPSC-CM were generated using a standard chemical differentiation method supplemented with T3 (triiodothyronine) and/or Dex (dexamethasone) during days 16 to 30 followed by single-cell culture for 5 days on Matrigel mattress. hiPSC-CM treated with T3+Dex, but not with either T3 or Dex alone, developed an extensive T-tubule network. Notably, Matrigel mattress was necessary for T-tubule formation. Compared with adult human ventricular cardiomyocytes, T-tubules in T3+Dex-treated hiPSC-CM were less organized and had more longitudinal elements. Confocal line scans demonstrated spatially and temporally uniform Ca release that is characteristic of excitation-contraction coupling in the heart ventricle. T3+Dex enhanced elementary Ca release measured by Ca sparks and promoted RyR2 (ryanodine receptor) structural organization. Simultaneous measurements of L-type Ca current and intracellular Ca release confirmed enhanced functional coupling between L-type Ca channels and RyR2 in T3+Dex-treated cells. CONCLUSIONS Our results suggest a permissive role of combined thyroid and glucocorticoid hormones during the cardiac differentiation process, which when coupled with further maturation on Matrigel mattress, is sufficient for T-tubule development, enhanced Ca-induced Ca release, and more ventricular-like excitation-contraction coupling. This new hormone maturation method could advance the use of hiPSC-CM for disease modeling and cell-based therapy.
Collapse
Affiliation(s)
- Shan S Parikh
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Daniel J Blackwell
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Nieves Gomez-Hurtado
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Michael Frisk
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Lili Wang
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Kyungsoo Kim
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Christen P Dahl
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Arnt Fiane
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Theis Tønnessen
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Dmytro O Kryshtal
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - William E Louch
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.)
| | - Bjorn C Knollmann
- From the Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine (S.S.P., D.J.B., N.G.-H., L.W., K.K., D.O.K., B.C.K.), Department of Pharmacology (S.S.P., B.C.K.), Vanderbilt University Medical School, Nashville, TN; Institute for Experimental Medical Research, Oslo University Hospital, Norway (M.F., T.T., W.E.L.); University of Oslo, Norway (M.F., T.T., W.E.L.); Department of Cardiology (C.P.D.), and Department of Cardiothoracic Surgery (A.F.), Oslo University Hospital Rikshospitalet, Norway; and Oslo University Hospital Ullevål, Norway (T.T.).
| |
Collapse
|
75
|
Macková K, Zahradníková A, Hoťka M, Hoffmannová B, Zahradník I, Zahradníková A. Calcium release-dependent inactivation precedes formation of the tubular system in developing rat cardiac myocytes. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:691-703. [PMID: 28913625 DOI: 10.1007/s00249-017-1249-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/21/2017] [Accepted: 08/17/2017] [Indexed: 01/06/2023]
Abstract
Developing cardiac myocytes undergo substantial structural and functional changes transforming the mechanism of excitation-contraction coupling from the embryonic form, based on calcium influx through sarcolemmal DHPR calcium channels, to the adult form, relying on local calcium release through RYR calcium channels of sarcoplasmic reticulum stimulated by calcium influx. We characterized day-by-day the postnatal development of the structure of sarcolemma, using techniques of confocal fluorescence microscopy, and the development of the calcium current, measured by the whole-cell patch-clamp in isolated rat ventricular myocytes. We characterized the appearance and expansion of the t-tubule system and compared it with the appearance and progress of the calcium current inactivation induced by the release of calcium ions from sarcoplasmic reticulum as structural and functional measures of direct DHPR-RYR interaction. The release-dependent inactivation of calcium current preceded the development of the t-tubular system by several days, indicating formation of the first DHPR-RYR couplons at the surface sarcolemma and their later spreading close to contractile myofibrils with the growing t-tubules. Large variability of both of the measured parameters among individual myocytes indicates uneven maturation of myocytes within the growing myocardium.
Collapse
Affiliation(s)
- Katarina Macková
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05, Bratislava, Slovakia
| | - Alexandra Zahradníková
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05, Bratislava, Slovakia
| | - Matej Hoťka
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05, Bratislava, Slovakia
| | - Barbora Hoffmannová
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05, Bratislava, Slovakia
| | - Ivan Zahradník
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05, Bratislava, Slovakia
| | - Alexandra Zahradníková
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05, Bratislava, Slovakia.
| |
Collapse
|
76
|
Kumar TKS, Mathis C, Sathanandam S, Zurakowski D, Subramanian S, Allen J, Solimine M, Berrios L, Jackson S, Landers M, Sullivan R, Barnett S, Rayburn M, Loftis C, Price L, Tansey JB, Hoskoppal D, Knott-Craig C. Effect of thyroid hormone on cardiac function following orthotopic heart transplantation in piglets. Pediatr Transplant 2017; 21. [PMID: 28710785 DOI: 10.1111/petr.13002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2017] [Indexed: 11/28/2022]
Abstract
Studies in adult HT have demonstrated improved cardiac function in the recipient following administration of T3 to the donor. The purpose of this experiment was to assess the effects of T3 on the function of the immature donor heart following HT in a piglet model. A total of 32 piglets were divided into 16 donors and 16 recipients. Following creation of brain death, half of the donor piglets were randomized to receive three doses of T3 (0.2 μg/kg) along with hydrocortisone (1 mg/kg). The donor hearts were then transplanted into the recipient piglets on CPB. Duration of survival off CPB, inotrope score, and EF of heart following CPB were evaluated. There were no differences between the two groups in age, weight, pre-brain death EF, T3 levels, and CPB times. Post-CPB survival times were inversely related to the ischemic times in both groups (Pearson r=-0.80, P<.001), and this relationship was not influenced by T3. There was no difference in inotrope score, EF, or biochemical assessment between the two groups. Administration of T3 in combination with hydrocortisone to the brain-dead donor confers no beneficial effect on myocardial function or survival following HT in a piglet model.
Collapse
Affiliation(s)
- T K Susheel Kumar
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Craig Mathis
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Shyam Sathanandam
- Department of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - David Zurakowski
- Departments of Anesthesia and Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Saradha Subramanian
- Department of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Jerry Allen
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Michael Solimine
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Lindsay Berrios
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Scott Jackson
- Department of Comparative Medicine, University of Tennessee, Memphis, TN, USA
| | - Mark Landers
- Department of Comparative Medicine, University of Tennessee, Memphis, TN, USA
| | - Ryan Sullivan
- Department of Comparative Medicine, University of Tennessee, Memphis, TN, USA
| | - Stacey Barnett
- Department of Comparative Medicine, University of Tennessee, Memphis, TN, USA
| | - Mark Rayburn
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Christopher Loftis
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - Lauren Price
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | - James B Tansey
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| | | | - Christopher Knott-Craig
- Department of Pediatric Cardiothoracic Surgery, Le Bonheur Children's Hospital, University of Tennessee, Memphis, TN, USA
| |
Collapse
|
77
|
Nader M, Alotaibi S, Alsolme E, Khalil B, Abu-Zaid A, Alsomali R, Bakheet D, Dzimiri N. Cardiac striatin interacts with caveolin-3 and calmodulin in a calcium sensitive manner and regulates cardiomyocyte spontaneous contraction rate. Can J Physiol Pharmacol 2017; 95:1306-1312. [PMID: 28825318 DOI: 10.1139/cjpp-2017-0155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Impaired cardiomyocyte contraction rate is detrimental to cardiac function and often lethal. Despite advancements in the field, there is a paucity of information regarding the coordination of molecules implicated in regulating the heart rate. Striatin (STRN) is a dynamic protein with binding domains to calmodulin (CaM) and caveolin (Cav), both of which are regulators of myocardial function. However, its role in cardiomyocyte contraction is not yet determined. Herein, we show that STRN is expressed in cardiomyocytes and is more abundant in atrial myocardium than in ventricles. Cardiac expression of STRN (protein and mRNA) was developmentally regulated with the highest expression being at neonatal stage (day one) and the lowest in adult rats (13 weeks). CaM pulldown assay indicated that the interaction of cardiac STRN with CaM and caveolin-3 (Cav-3) was calcium sensitive. Interestingly, the overexpression of STRN induced an increase (∼2-fold) in the rate of the spontaneous contraction of cultured cardiomyocytes, while the knockdown of STRN reduced their contraction rate (∼40%). The expression level of STRN was inversely proportional to the interaction of Cav-3 with the CaM/STRN complex. Collectively, our data delineate a novel role for STRN in regulating cardiomyocyte spontaneous contraction rate and the dynamics of the STRN/Cav-3/CaM complex.
Collapse
Affiliation(s)
- Moni Nader
- a Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh 11533, Kingdom of Saudi Arabia.,b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Shahd Alotaibi
- a Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh 11533, Kingdom of Saudi Arabia.,b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Ebtehal Alsolme
- a Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh 11533, Kingdom of Saudi Arabia.,b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Bariaa Khalil
- a Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh 11533, Kingdom of Saudi Arabia.,b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Abu-Zaid
- a Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh 11533, Kingdom of Saudi Arabia.,b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Rahmah Alsomali
- b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Dana Bakheet
- b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Nduna Dzimiri
- b Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
78
|
Affiliation(s)
- Yoshinori Yoshida
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| | - Shinya Yamanaka
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| |
Collapse
|
79
|
Moon SH, Bae D, Jung TH, Chung EB, Jeong YH, Park SJ, Chung HM. From Bench to Market: Preparing Human Pluripotent Stem Cells Derived Cardiomyocytes for Various Applications. Int J Stem Cells 2017; 10:1-11. [PMID: 28531912 PMCID: PMC5488771 DOI: 10.15283/ijsc17024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Human cardiomyocytes (CMs) cease to proliferate and remain terminally differentiated thereafter, when humans reach the mid-20s. Thus, any damages sustained by myocardium tissue are irreversible, and they require medical interventions to regain functionality. To date, new surgical procedures and drugs have been developed, albeit with limited success, to treat various heart diseases including myocardial infarction. Hence, there is a pressing need to develop more effective treatment methods to address the increasing mortality rate of the heart diseases. Functional CMs are not only an important in vitro cellular tool to model various types of heart diseases for drug development, but they are also a promising therapeutic agent for cell therapy. However, the limited proliferative capacity entails difficulties in acquiring functional CMs in the scale that is required for pathological studies and cell therapy development. Stem cells, human pluripotent stem cells (hPSCs) in particular, have been considered as an unlimited cellular source for providing functional CMs for various applications. Notable progress has already been made: the first clinical trials of hPSCs derived CMs (hPSC-CMs) for treating myocardial infarction was approved in 2015, and their potential use in disease modeling and drug discovery is being fully explored. This concise review gives an account of current development of differentiation, purification and maturation techniques for hPSC-CMs, and their application in cell therapy development and pharmaceutical industries will be discussed with the latest experimental evidence.
Collapse
Affiliation(s)
- Sung-Hwan Moon
- Department of Medicine, School of Medicine, Konkuk University, Seoul, Korea
| | | | - Taek-Hee Jung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Eun-Bin Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Young-Hoon Jeong
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Soon-Jung Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
80
|
Scuderi GJ, Butcher J. Naturally Engineered Maturation of Cardiomyocytes. Front Cell Dev Biol 2017; 5:50. [PMID: 28529939 PMCID: PMC5418234 DOI: 10.3389/fcell.2017.00050] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022] Open
Abstract
Ischemic heart disease remains one of the most prominent causes of mortalities worldwide with heart transplantation being the gold-standard treatment option. However, due to the major limitations associated with heart transplants, such as an inadequate supply and heart rejection, there remains a significant clinical need for a viable cardiac regenerative therapy to restore native myocardial function. Over the course of the previous several decades, researchers have made prominent advances in the field of cardiac regeneration with the creation of in vitro human pluripotent stem cell-derived cardiomyocyte tissue engineered constructs. However, these engineered constructs exhibit a functionally immature, disorganized, fetal-like phenotype that is not equivalent physiologically to native adult cardiac tissue. Due to this major limitation, many recent studies have investigated approaches to improve pluripotent stem cell-derived cardiomyocyte maturation to close this large functionality gap between engineered and native cardiac tissue. This review integrates the natural developmental mechanisms of cardiomyocyte structural and functional maturation. The variety of ways researchers have attempted to improve cardiomyocyte maturation in vitro by mimicking natural development, known as natural engineering, is readily discussed. The main focus of this review involves the synergistic role of electrical and mechanical stimulation, extracellular matrix interactions, and non-cardiomyocyte interactions in facilitating cardiomyocyte maturation. Overall, even with these current natural engineering approaches, pluripotent stem cell-derived cardiomyocytes within three-dimensional engineered heart tissue still remain mostly within the early to late fetal stages of cardiomyocyte maturity. Therefore, although the end goal is to achieve adult phenotypic maturity, more emphasis must be placed on elucidating how the in vivo fetal microenvironment drives cardiomyocyte maturation. This information can then be utilized to develop natural engineering approaches that can emulate this fetal microenvironment and thus make prominent progress in pluripotent stem cell-derived maturity toward a more clinically relevant model for cardiac regeneration.
Collapse
Affiliation(s)
- Gaetano J Scuderi
- Meinig School of Biomedical Engineering, Cornell UniversityIthaca, NY, USA
| | - Jonathan Butcher
- Meinig School of Biomedical Engineering, Cornell UniversityIthaca, NY, USA
| |
Collapse
|
81
|
Parra V, Rothermel BA. Calcineurin signaling in the heart: The importance of time and place. J Mol Cell Cardiol 2017; 103:121-136. [PMID: 28007541 PMCID: PMC5778886 DOI: 10.1016/j.yjmcc.2016.12.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
The calcium-activated protein phosphatase, calcineurin, lies at the intersection of protein phosphorylation and calcium signaling cascades, where it provides an essential nodal point for coordination between these two fundamental modes of intracellular communication. In excitatory cells, such as neurons and cardiomyocytes, that experience rapid and frequent changes in cytoplasmic calcium, calcineurin protein levels are exceptionally high, suggesting that these cells require high levels of calcineurin activity. Yet, it is widely recognized that excessive activation of calcineurin in the heart contributes to pathological hypertrophic remodeling and the progression to failure. How does a calcium activated enzyme function in the calcium-rich environment of the continuously contracting heart without pathological consequences? This review will discuss the wide range of calcineurin substrates relevant to cardiovascular health and the mechanisms calcineurin uses to find and act on appropriate substrates in the appropriate location while potentially avoiding others. Fundamental differences in calcineurin signaling in neonatal verses adult cardiomyocytes will be addressed as well as the importance of maintaining heterogeneity in calcineurin activity across the myocardium. Finally, we will discuss how circadian oscillations in calcineurin activity may facilitate integration with other essential but conflicting processes, allowing a healthy heart to reap the benefits of calcineurin signaling while avoiding the detrimental consequences of sustained calcineurin activity that can culminate in heart failure.
Collapse
Affiliation(s)
- Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago,Chile; Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chie, Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
82
|
Early transverse tubule development begins in utero in the sheep heart. J Muscle Res Cell Motil 2017; 37:195-202. [PMID: 28062939 DOI: 10.1007/s10974-016-9462-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/10/2016] [Indexed: 10/20/2022]
Abstract
The ventricular cardiomyocytes of adult mammals contain invaginations of the plasma membrane known as transverse (t)-tubules. These regular structures are essential for the synchronisation of excitation-contraction (EC) coupling throughout the cell, which is a vital process for cardiac function. T-tubules form a close association with the sarcoplasmic reticulum (SR) to form junctions, where several key proteins involved in EC coupling are localised, including the SR calcium release channels-the ryanodine receptors (RyR). The lipophilic SR protein junctophilin-2 (JPH2) has been implicated in the development of both the junctions and t-tubules. Several studies have identified that t-tubules develop only postnatally in rodents, while historical electron microscopy data indicate that this is not the case in larger mammals, including humans. We have performed, to our knowledge, the first fluorescent, target-specific study to characterise t-tubule development in the large mammalian fetal heart, focussing on the sheep. T-tubules were present in fetal sheep hearts from 114 days gestation (with term being 145 days), with occurrence progressively increasing with gestational age, and further maturation after birth. This was accompanied by an increasing intracellular localisation of JPH2, which progressively increased its association with RyR within the cardiomyocytes as they undergo hypertrophy. These findings indicate that large mammalian hearts exhibit a significantly different temporal pattern of development compared to that of the rodent. Our findings have potential implications for human cardiac development, including the future investigation of congenital heart disease.
Collapse
|
83
|
Reynolds JO, Quick AP, Wang Q, Beavers DL, Philippen LE, Showell J, Barreto-Torres G, Thuerauf DJ, Doroudgar S, Glembotski CC, Wehrens XHT. Junctophilin-2 gene therapy rescues heart failure by normalizing RyR2-mediated Ca 2+ release. Int J Cardiol 2016; 225:371-380. [PMID: 27760414 DOI: 10.1016/j.ijcard.2016.10.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Junctophilin-2 (JPH2) is the primary structural protein for the coupling of transverse (T)-tubule associated cardiac L-type Ca channels and type-2 ryanodine receptors on the sarcoplasmic reticulum within junctional membrane complexes (JMCs) in cardiomyocytes. Effective signaling between these channels ensures adequate Ca-induced Ca release required for normal cardiac contractility. Disruption of JMC subcellular domains, a common feature of failing hearts, has been attributed to JPH2 downregulation. Here, we tested the hypothesis that adeno-associated virus type 9 (AAV9) mediated overexpression of JPH2 could halt the development of heart failure in a mouse model of transverse aortic constriction (TAC). METHODS AND RESULTS Following TAC, a progressive decrease in ejection fraction was paralleled by a progressive decrease of cardiac JPH2 levels. AAV9-mediated expression of JPH2 rescued cardiac contractility in mice subjected to TAC. AAV9-JPH2 also preserved T-tubule structure. Moreover, the Ca2+ spark frequency was reduced and the Ca2+ transient amplitude was increased in AAV9-JPH2 mice following TAC, consistent with JPH2-mediated normalization of SR Ca2+ handling. CONCLUSIONS This study demonstrates that AAV9-mediated JPH2 gene therapy maintained cardiac function in mice with early stage heart failure. Moreover, restoration of JPH2 levels prevented loss of T-tubules and suppressed abnormal SR Ca2+ leak associated with contractile failure following TAC. These findings suggest that targeting JPH2 might be an attractive therapeutic approach for treating pathological cardiac remodeling during heart failure.
Collapse
Affiliation(s)
- Julia O Reynolds
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ann P Quick
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qiongling Wang
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - David L Beavers
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Leonne E Philippen
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jordan Showell
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Giselle Barreto-Torres
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Donna J Thuerauf
- San Diego State University Heart Institute and Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Shirin Doroudgar
- Department of Cardiology, Angiology, and Pneumology, University Hospital Heidelberg, Innere Medizin III, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Christopher C Glembotski
- San Diego State University Heart Institute and Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Medicine/Cardiology, Baylor College of Medicine, Houston, TX 77030, USA; Dept. of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
84
|
Uosaki H, Taguchi YH. Comparative Gene Expression Analysis of Mouse and Human Cardiac Maturation. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:207-15. [PMID: 27431744 PMCID: PMC4996857 DOI: 10.1016/j.gpb.2016.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 04/07/2016] [Accepted: 04/10/2016] [Indexed: 12/12/2022]
Abstract
Understanding how human cardiomyocytes mature is crucial to realizing stem cell-based heart regeneration, modeling adult heart diseases, and facilitating drug discovery. However, it is not feasible to analyze human samples for maturation due to inaccessibility to samples while cardiomyocytes mature during fetal development and childhood, as well as difficulty in avoiding variations among individuals. Using model animals such as mice can be a useful strategy; nonetheless, it is not well-understood whether and to what degree gene expression profiles during maturation are shared between humans and mice. Therefore, we performed a comparative gene expression analysis of mice and human samples. First, we examined two distinct mice microarray platforms for shared gene expression profiles, aiming to increase reliability of the analysis. We identified a set of genes displaying progressive changes during maturation based on principal component analysis. Second, we demonstrated that the genes identified had a differential expression pattern between adult and earlier stages (e.g., fetus) common in mice and humans. Our findings provide a foundation for further genetic studies of cardiomyocyte maturation.
Collapse
Affiliation(s)
- Hideki Uosaki
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Y-H Taguchi
- Department of Physics, Chuo University, Tokyo 112-8551, Japan
| |
Collapse
|
85
|
Aigha I, Raynaud C. Maturation of pluripotent stem cell derived cardiomyocytes: The new challenge. Glob Cardiol Sci Pract 2016; 2016:e201606. [PMID: 29043256 PMCID: PMC5642835 DOI: 10.21542/gcsp.2016.6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stem cell therapy appears to be a promising area of research for cardiac regeneration following ischemic heart failure. However, in vitro differentiation of cardiomyocytes from pluripotent stem cells, or directly from somatic cells, leads to generation of "immature" cardiomyocytes that differ from their adult counterparts in various ways. This immaturity triggers some challenges for their potential clinical use, and multiple techniques reviewed here have been developed for in vitro maturation of those cells. Nevertheless, full maturity of cardiomyocytes remains elusive and will remain the main challenge for stem cell therapy in the near future.
Collapse
Affiliation(s)
- Idil Aigha
- Qatar Cardiovascular Research Center, Qatar Foundation, Education City, Doha, Qatar
| | - Christophe Raynaud
- Qatar Cardiovascular Research Center, Qatar Foundation, Education City, Doha, Qatar
| |
Collapse
|
86
|
Schultz F, Hasan A, Alvarez-Laviada A, Miragoli M, Bhogal N, Wells S, Poulet C, Chambers J, Williamson C, Gorelik J. The protective effect of ursodeoxycholic acid in an in vitro model of the human fetal heart occurs via targeting cardiac fibroblasts. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:149-63. [PMID: 26777584 DOI: 10.1016/j.pbiomolbio.2016.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 12/27/2022]
Abstract
Bile acids are elevated in the blood of women with intrahepatic cholestasis of pregnancy (ICP) and this may lead to fetal arrhythmia, fetal hypoxia and potentially fetal death in utero. The bile acid taurocholic acid (TC) causes abnormal calcium dynamics and contraction in neonatal rat cardiomyocytes. Ursodeoxycholic acid (UDCA), a drug clinically used to treat ICP, prevents adverse effects of TC. During development, the fetus is in a state of relative hypoxia. Although this is essential for the development of the heart and vasculature, resident fibroblasts can transiently differentiate into myofibroblasts and form gap junctions with cardiomyocytes in vitro, resulting in cardiomyocyte depolarization. We expanded on previously published work using an in vitro hypoxia model to investigate the differentiation of human fetal fibroblasts into myofibroblasts. Recent evidence shows that potassium channels are involved in maintaining the membrane potential of ventricular fibroblasts and that ATP-dependent potassium (KATP) channel subunits are expressed in cultured fibroblasts. KATP channels are a valuable target as they are thought to have a cardioprotective role during ischaemic and hypoxic conditions. We investigated whether UDCA could modulate fibroblast membrane potential. We established the isolation and culture of human fetal cardiomyocytes and fibroblasts to investigate the effect of hypoxia, TC and UDCA on human fetal cardiac cells. UDCA hyperpolarized myofibroblasts and prevented TC-induced depolarisation, possibly through the activation of KATP channels that are expressed in cultured fibroblasts. Also, similar to the rat model, UDCA can counteract TC-induced calcium abnormalities in human fetal cultures of cardiomyocytes and myofibroblasts. Under normoxic conditions, we found a higher number of myofibroblasts in cultures derived from human fetal hearts compared to cells isolated from neonatal rat hearts, indicating a possible increased number of myofibroblasts in human fetal hearts. Hypoxia further increased the number of human fetal and rat neonatal myofibroblasts. However, chronically administered UDCA reduced the number of myofibroblasts and prevented hypoxia-induced depolarisation. In conclusion, our results show that the protective effect of UDCA involves both the reduction of fibroblast differentiation into myofibroblasts, and hyperpolarisation of myofibroblasts, most likely through the stimulation of potassium channels, i.e. KATP channels. This could be important in validating UDCA as an antifibrotic and antiarrhythmic drug for treatment of failing hearts and fetal arrhythmia.
Collapse
Affiliation(s)
- Francisca Schultz
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK; Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Alveera Hasan
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Anita Alvarez-Laviada
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Michele Miragoli
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK; Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Navneet Bhogal
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Sarah Wells
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Claire Poulet
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Jenny Chambers
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK; Women's Health Academic Centre, King's College London, London, United Kingdom
| | - Catherine Williamson
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK; Women's Health Academic Centre, King's College London, London, United Kingdom
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK.
| |
Collapse
|
87
|
Uosaki H, Cahan P, Lee DI, Wang S, Miyamoto M, Fernandez L, Kass DA, Kwon C. Transcriptional Landscape of Cardiomyocyte Maturation. Cell Rep 2015; 13:1705-16. [PMID: 26586429 DOI: 10.1016/j.celrep.2015.10.032] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/19/2015] [Accepted: 10/09/2015] [Indexed: 01/06/2023] Open
Abstract
Decades of progress in developmental cardiology has advanced our understanding of the early aspects of heart development, including cardiomyocyte (CM) differentiation. However, control of the CM maturation that is subsequently required to generate adult myocytes remains elusive. Here, we analyzed over 200 microarray datasets from early embryonic to adult hearts and identified a large number of genes whose expression shifts gradually and continuously during maturation. We generated an atlas of integrated gene expression, biological pathways, transcriptional regulators, and gene regulatory networks (GRNs), which show discrete sets of key transcriptional regulators and pathways activated or suppressed during CM maturation. We developed a GRN-based program named MatStat(CM) that indexes CM maturation status. MatStat(CM) reveals that pluripotent-stem-cell-derived CMs mature early in culture but are arrested at the late embryonic stage with aberrant regulation of key transcription factors. Our study provides a foundation for understanding CM maturation.
Collapse
Affiliation(s)
- Hideki Uosaki
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Johns Hopkins Institute for Cell Engineering, Baltimore, MD 21205, USA
| | - Patrick Cahan
- Stem Cell Transplantation Program, Division of Pediatric Hematology and Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Dong I Lee
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Songnan Wang
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Johns Hopkins Institute for Cell Engineering, Baltimore, MD 21205, USA
| | - Matthew Miyamoto
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Johns Hopkins Institute for Cell Engineering, Baltimore, MD 21205, USA
| | - Laviel Fernandez
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Johns Hopkins Institute for Cell Engineering, Baltimore, MD 21205, USA
| | - David A Kass
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Johns Hopkins Institute for Cell Engineering, Baltimore, MD 21205, USA.
| |
Collapse
|
88
|
Organization of junctional sarcoplasmic reticulum proteins in skeletal muscle fibers. J Muscle Res Cell Motil 2015; 36:501-15. [DOI: 10.1007/s10974-015-9421-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/08/2015] [Indexed: 01/24/2023]
|
89
|
Defined MicroRNAs Induce Aspects of Maturation in Mouse and Human Embryonic-Stem-Cell-Derived Cardiomyocytes. Cell Rep 2015; 12:1960-7. [PMID: 26365191 DOI: 10.1016/j.celrep.2015.08.042] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 06/22/2015] [Accepted: 08/12/2015] [Indexed: 11/22/2022] Open
Abstract
Pluripotent-cell-derived cardiomyocytes have great potential for use in research and medicine, but limitations in their maturity currently constrain their usefulness. Here, we report a method for improving features of maturation in murine and human embryonic-stem-cell-derived cardiomyocytes (m/hESC-CMs). We found that coculturing m/hESC-CMs with endothelial cells improves their maturity and upregulates several microRNAs. Delivering four of these microRNAs, miR-125b-5p, miR-199a-5p, miR-221, and miR-222 (miR-combo), to m/hESC-CMs resulted in improved sarcomere alignment and calcium handling, a more negative resting membrane potential, and increased expression of cardiomyocyte maturation markers. Although this could not fully phenocopy all adult cardiomyocyte characteristics, these effects persisted for two months following delivery of miR-combo. A luciferase assay demonstrated that all four miRNAs target ErbB4, and siRNA knockdown of ErbB4 partially recapitulated the effects of miR-combo. In summary, a combination of miRNAs induced via endothelial coculture improved ESC-CM maturity, in part through suppression of ErbB4 signaling.
Collapse
|
90
|
Cai Y, Tu J, Pan S, Jiang J, Shou Q, Ling Y, Chen Y, Wang D, Yang W, Shan L, Chen M. Medicinal effect and its JP2/RyR2-based mechanism of Smilax glabra flavonoids on angiotensin II-induced hypertrophy model of cardiomyocytes. JOURNAL OF ETHNOPHARMACOLOGY 2015; 169:435-440. [PMID: 25926285 DOI: 10.1016/j.jep.2015.04.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 04/08/2015] [Accepted: 04/18/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhizome and root of Smilax glabra Roxb (Liliaceae family) is a widely used traditional Chinese medicine (TCM) named Tu-fu-ling (TFL) for cardiac disease therapy. The TFL flavonoids (TFLF) has been extracted and proven to possess the anti-cardiac hypertrophy effect in our previous reports. Such effect could be mediated by the modulation of intracellular Ca(2+) flux in myocardial cells, in which junctophilin-2 (JP2) and ryanodine receptor 2 (RyR2) play an important role. However, its mechanism of the anti-cardiac hypertrophy effect remains unclarified. MATERIALS AND METHODS 2μmol/L Ang II was applied to induce hypertrophy model of rat primary cardiomyocytes. After treatment of TFLF at 0.25, 0.5 and 1.0mg/ml, the cell size was microscopic measured, and the protein and mRNA expressions of JP2 and RyR2 in cardiomyocytes were estimated by immunofluorescence imaging, ELISA and real-time PCR assay. RESULTS Obvious hypertrophy of cardiomyocytes was induced by Ang II but reversed by TFLF from 0.5 to 1.0mg/ml. The protein and mRNA expressions of JP2 and RyR2 in cardiomyocytes were also inhibited by Ang II but restored by TFLF at its dose range. Such effect of TFLF was exerted at a dose dependent manner, which was even better than that of verapamil. CONCLUSIONS Our findings may evidence the correlation between JP2/RyR2 and myocardiac hypertrophy, and indicate the JP2/RyR2-mediated anti-hypertrophy mechanism of TFLF for the first time. It deserves to be developed as a promising TCM candidate of new drug for myocardial hypertrophy treatment.
Collapse
Affiliation(s)
- Yueqin Cai
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jue Tu
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shuizhen Pan
- Zhejiang Academy of Medical Sciences, Hangzhou 310007, China
| | - Jianping Jiang
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiyang Shou
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yun Ling
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yunxiang Chen
- Zhejiang Academy of Medical Sciences, Hangzhou 310007, China
| | - Dejun Wang
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Weiji Yang
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Letian Shan
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Minli Chen
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
91
|
Physiologic force-frequency response in engineered heart muscle by electromechanical stimulation. Biomaterials 2015; 60:82-91. [PMID: 25985155 DOI: 10.1016/j.biomaterials.2015.03.055] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/28/2015] [Indexed: 12/30/2022]
Abstract
A hallmark of mature mammalian ventricular myocardium is a positive force-frequency relationship (FFR). Despite evidence of organotypic structural and molecular maturation, a positive FFR has not been observed in mammalian tissue engineered heart muscle. We hypothesized that concurrent mechanical and electrical stimulation at frequencies matching physiological heart rate will result in functional maturation. We investigated the role of biomimetic mechanical and electrical stimulation in functional maturation in engineered heart muscle (EHM). Following tissue consolidation, EHM were subjected to electrical field stimulation at 0, 2, 4, or 6 Hz for 5 days, while strained on flexible poles to facilitate auxotonic contractions. EHM stimulated at 2 and 4 Hz displayed a similarly enhanced inotropic reserve, but a clearly diverging FFR. The positive FFR in 4 Hz stimulated EHM was associated with reduced calcium sensitivity, frequency-dependent acceleration of relaxation, and enhanced post-rest potentiation. This was paralleled on the cellular level with improved calcium storage and release capacity of the sarcoplasmic reticulum and enhanced T-tubulation. We conclude that electro-mechanical stimulation at a physiological frequency supports functional maturation in mammalian EHM. The observed positive FFR in EHM has important implications for the applicability of EHM in cardiovascular research.
Collapse
|
92
|
Chen B, Zhang C, Guo A, Song LS. In situ single photon confocal imaging of cardiomyocyte T-tubule system from Langendorff-perfused hearts. Front Physiol 2015; 6:134. [PMID: 25999861 PMCID: PMC4422017 DOI: 10.3389/fphys.2015.00134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/15/2015] [Indexed: 12/24/2022] Open
Abstract
Transverse tubules (T-tubules) are orderly invaginations of the sarcolemma in mammalian cardiomyocytes. The integrity of T-tubule architecture is critical for cardiac excitation–contraction coupling function. T-tubule remodeling is recognized as a key player in cardiac dysfunction. Early studies on T-tubule structure were based on electron microscopy, which uncovered important information about the T-tubule architecture. The advent of fluorescent membrane probes allowed the application of confocal microscopy to investigations of T-tubule structure. Studies have now been extended beyond single cardiomyocytes to examine the T-tubule network in intact hearts through in situ confocal imaging of Langendorff-perfused hearts. This technique has allowed visualization of T-tubule organization in their natural habitat, avoiding the damage induced by isolation of cardiomyocytes. Additionally, it is possible to obtain T-tubule images in different subepicardial regions in a single intact heart. We review how this state-of-the-art imaging technique has provided important mechanistic insights into maturation of T-tubules in developing hearts and defined the role of T-tubule remodeling in development and progression of heart failure.
Collapse
Affiliation(s)
- Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| | - Caimei Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| | - Ang Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| |
Collapse
|
93
|
Targeted disruption of PDE3B, but not PDE3A, protects murine heart from ischemia/reperfusion injury. Proc Natl Acad Sci U S A 2015; 112:E2253-62. [PMID: 25877153 DOI: 10.1073/pnas.1416230112] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although inhibition of cyclic nucleotide phosphodiesterase type 3 (PDE3) has been reported to protect rodent heart against ischemia/reperfusion (I/R) injury, neither the specific PDE3 isoform involved nor the underlying mechanisms have been identified. Targeted disruption of PDE3 subfamily B (PDE3B), but not of PDE3 subfamily A (PDE3A), protected mouse heart from I/R injury in vivo and in vitro, with reduced infarct size and improved cardiac function. The cardioprotective effect in PDE3B(-/-) heart was reversed by blocking cAMP-dependent PKA and by paxilline, an inhibitor of mitochondrial calcium-activated K channels, the opening of which is potentiated by cAMP/PKA signaling. Compared with WT mitochondria, PDE3B(-/-) mitochondria were enriched in antiapoptotic Bcl-2, produced less reactive oxygen species, and more frequently contacted transverse tubules where PDE3B was localized with caveolin-3. Moreover, a PDE3B(-/-) mitochondrial fraction containing connexin-43 and caveolin-3 was more resistant to Ca(2+)-induced opening of the mitochondrial permeability transition pore. Proteomics analyses indicated that PDE3B(-/-) heart mitochondria fractions were enriched in buoyant ischemia-induced caveolin-3-enriched fractions (ICEFs) containing cardioprotective proteins. Accumulation of proteins into ICEFs was PKA dependent and was achieved by ischemic preconditioning or treatment of WT heart with the PDE3 inhibitor cilostamide. Taken together, these findings indicate that PDE3B deletion confers cardioprotective effects because of cAMP/PKA-induced preconditioning, which is associated with the accumulation of proteins with cardioprotective function in ICEFs. To our knowledge, our study is the first to define a role for PDE3B in cardioprotection against I/R injury and suggests PDE3B as a target for cardiovascular therapies.
Collapse
|
94
|
Louch WE, Koivumäki JT, Tavi P. Calcium signalling in developing cardiomyocytes: implications for model systems and disease. J Physiol 2015; 593:1047-63. [PMID: 25641733 PMCID: PMC4358669 DOI: 10.1113/jphysiol.2014.274712] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 12/28/2014] [Indexed: 12/15/2022] Open
Abstract
Adult cardiomyocytes exhibit complex Ca(2+) homeostasis, enabling tight control of contraction and relaxation. This intricate regulatory system develops gradually, with progressive maturation of specialized structures and increasing capacity of Ca(2+) sources and sinks. In this review, we outline current understanding of these developmental processes, and draw parallels to pathophysiological conditions where cardiomyocytes exhibit a striking regression to an immature state of Ca(2+) homeostasis. We further highlight the importance of understanding developmental physiology when employing immature cardiomyocyte models such as cultured neonatal cells and stem cells.
Collapse
Affiliation(s)
- William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo0424, Oslo, Norway
- K. G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo0316, Oslo, Norway
| | - Jussi T Koivumäki
- Simula Research Laboratory, Center for Cardiological Innovation and Center for Biomedical ComputingOslo, Norway
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern FinlandKuopio, Finland
| |
Collapse
|
95
|
Takeshima H, Hoshijima M, Song LS. Ca²⁺ microdomains organized by junctophilins. Cell Calcium 2015; 58:349-56. [PMID: 25659516 PMCID: PMC5159448 DOI: 10.1016/j.ceca.2015.01.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/15/2015] [Accepted: 01/16/2015] [Indexed: 11/21/2022]
Abstract
Excitable cells typically possess junctional membrane complexes (JMCs) constructed by the plasma membrane and the endo/sarcoplasmic reticulum (ER/SR) for channel crosstalk. These JMCs are termed triads in skeletal muscle, dyads in cardiac muscle, peripheral couplings in smooth and developing striated muscles, and subsurface cisterns in neurons. Junctophilin subtypes contribute to the formation and maintenance of JMCs by serving as a physical bridge between the plasma membrane and ER/SR membrane in different cell types. In muscle cells, junctophilin deficiency prevents JMC formation and functional crosstalk between cell-surface Ca2+ channels and ER/SR Ca2+ release channels. Human genetic mutations in junctophilin subtypes are linked to congenital hypertrophic cardiomyopathy and neurodegenerative diseases. Furthermore, growing evidence suggests that dysregulation of junctophilins induces pathological alterations in skeletal and cardiac muscle.
Collapse
Affiliation(s)
- Hiroshi Takeshima
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| | - Masahiko Hoshijima
- Department of Medicine and Center for Research in Biological Systems, University of California, San Diego, CA 92093, USA.
| | - Long-Sheng Song
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
96
|
Pham TLB, Nguyen TT, Van Bui A, Nguyen MT, Van Pham P. Fetal heart extract facilitates the differentiation of human umbilical cord blood-derived mesenchymal stem cells into heart muscle precursor cells. Cytotechnology 2014; 68:645-58. [PMID: 25377264 DOI: 10.1007/s10616-014-9812-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/27/2014] [Indexed: 02/07/2023] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) are a promising stem cell source with the potential to modulate the immune system as well as the capacity to differentiate into osteoblasts, chondrocytes, and adipocytes. In previous publications, UCB-MSCs have been successfully differentiated into cardiomyocytes. This study aimed to improve the efficacy of differentiation of UCB-MSCs into cardiomyocytes by combining 5-azacytidine (Aza) with mouse fetal heart extract (HE) in the induction medium. UCB-MSCs were isolated from umbilical cord blood according to a published protocol. Murine fetal hearts were used to produce fetal HE using a rapid freeze-thaw procedure. MSCs at the 3rd to 5th passage were differentiated into cardiomyocytes in two kinds of induction medium: complete culture medium plus Aza (Aza group) and complete culture medium plus Aza and fetal HE (Aza + HE group). The results showed that the cells in both kinds of induction medium exhibited the phenotype of cardiomyocytes. At the transcriptional level, the cells expressed a number of cardiac muscle-specific genes such as Nkx2.5, Gata 4, Mef2c, HCN2, hBNP, α-Ca, cTnT, Desmin, and β-MHC on day 27 in the Aza group and on day 18 in the Aza + HE group. At the translational level, sarcomic α-actin was expressed on day 27 in the Aza group and day 18 in the Aza + HE group. Although they expressed specific genes and proteins of cardiac muscle cells, the induced cells in both groups did not contract and beat spontaneously. These properties are similar to properties of heart muscle precursor cells in vivo. These results demonstrated that the fetal HE facilitates the differentiation process of human UCB-MSCs into heart muscle precursor cells.
Collapse
Affiliation(s)
- Truc Le-Buu Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Tam Thanh Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Anh Van Bui
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - My Thu Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
97
|
Beavers DL, Landstrom AP, Chiang DY, Wehrens XHT. Emerging roles of junctophilin-2 in the heart and implications for cardiac diseases. Cardiovasc Res 2014; 103:198-205. [PMID: 24935431 DOI: 10.1093/cvr/cvu151] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cardiomyocytes rely on a highly specialized subcellular architecture to maintain normal cardiac function. In a little over a decade, junctophilin-2 (JPH2) has become recognized as a cardiac structural protein critical in forming junctional membrane complexes (JMCs), which are subcellular domains essential for excitation-contraction coupling within the heart. While initial studies described the structure of JPH2 and its role in anchoring junctional sarcoplasmic reticulum and transverse-tubule (T-tubule) membrane invaginations, recent research has an expanded role of JPH2 in JMC structure and function. For example, JPH2 is necessary for the development of postnatal T-tubule in mammals. It is also critical for the maintenance of the complex JMC architecture and stabilization of local ion channels in mature cardiomyocytes. Loss of this function by mutations or down-regulation of protein expression has been linked to hypertrophic cardiomyopathy, arrhythmias, and progression of disease in failing hearts. In this review, we summarize current views on the roles of JPH2 within the heart and how JPH2 dysregulation may contribute to a variety of cardiac diseases.
Collapse
Affiliation(s)
- David L Beavers
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX, USA
| | - Andrew P Landstrom
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - David Y Chiang
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA Deptartment of Medicine (Cardiology), Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
98
|
Landstrom AP, Beavers DL, Wehrens XHT. The junctophilin family of proteins: from bench to bedside. Trends Mol Med 2014; 20:353-62. [PMID: 24636942 PMCID: PMC4041816 DOI: 10.1016/j.molmed.2014.02.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 02/10/2014] [Accepted: 02/14/2014] [Indexed: 12/25/2022]
Abstract
Excitable tissues rely on junctional membrane complexes to couple cell surface signals to intracellular channels. The junctophilins have emerged as a family of proteins critical in coordinating the maturation and maintenance of this cellular ultrastructure. Within skeletal and cardiac muscle, junctophilin 1 and junctophilin 2, respectively, couple sarcolemmal and intracellular calcium channels. In neuronal tissue, junctophilin 3 and junctophilin 4 may have an emerging role in coupling membrane neurotransmitter receptors and intracellular calcium channels. These important physiological roles are highlighted by the pathophysiology which results when these proteins are perturbed, and a growing body of literature has associated junctophilins with the pathogenesis of human disease.
Collapse
Affiliation(s)
- Andrew P Landstrom
- Department of Pediatrics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - David L Beavers
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xander H T Wehrens
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine (Cardiology), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
99
|
Abstract
OPINION STATEMENT Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) represent a powerful new model system to study the basic mechanisms of inherited cardiomyopathies. hiPSC-CMs have been utilized to model several cardiovascular diseases, achieving the most success in the inherited arrhythmias, including long QT and Timothy syndromes (Moretti et al. N Engl J Med. 363:1397-409, 2010; Yazawa et al. Nature. 471:230-4, 2011) and arrhythmogenic right ventricular dysplasia (ARVD) (Ma et al. Eur Heart J. 34:1122-33, 2013). Recently, studies have applied hiPSC-CMs to the study of both dilated (DCM) (Sun et al. Sci Transl Med. 4:130ra47, 2012) and hypertrophic (HCM) cardiomyopathies (Lan et al. Cell Stem Cell. 12:101-13, 2013; Carvajal-Vergara et al. Nature. 465:808-12, 2010), providing new insights into basic mechanisms of disease. However, hiPSC-CMs do not recapitulate many of the structural and functional aspects of mature human cardiomyocytes, instead mirroring an immature - embryonic or fetal - phenotype. Much work remains in order to better understand these differences, as well as to develop methods to induce hiPSC-CMs into a fully mature phenotype. Despite these limitations, hiPSC-CMs represent the best current in vitro correlate of the human heart and an invaluable tool in the search for mechanisms underlying cardiomyopathy and for screening new pharmacologic therapies.
Collapse
|
100
|
Alternative splicing regulates vesicular trafficking genes in cardiomyocytes during postnatal heart development. Nat Commun 2014; 5:3603. [PMID: 24752171 PMCID: PMC4018662 DOI: 10.1038/ncomms4603] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 03/10/2014] [Indexed: 12/22/2022] Open
Abstract
During postnatal development the heart undergoes a rapid and dramatic transition to adult function through transcriptional and post-transcriptional mechanisms, including alternative splicing (AS). Here we perform deep RNA-sequencing on RNA from cardiomyocytes and cardiac fibroblasts to conduct a high-resolution analysis of transcriptome changes during postnatal mouse heart development. We reveal extensive changes in gene expression and AS that occur primarily between postnatal days 1 and 28. Cardiomyocytes and cardiac fibroblasts show reciprocal regulation of gene expression reflecting differences in proliferative capacity, cell adhesion functions, and mitochondrial metabolism. We further demonstrate that AS plays a role in vesicular trafficking and membrane organization, These AS transitions are enriched among targets of two RNA-binding proteins, Celf1 and Mbnl1, which undergo developmentally regulated changes in expression. Vesicular trafficking genes affected by AS during normal development (when Celf1 is down-regulated) show a reversion to neonatal splicing patterns after Celf1 re-expression in adults. Short-term Celf1 induction in adult animals results in disrupted transverse tubule organization and calcium handling. These results identify potential roles for AS in multiple aspects of postnatal heart maturation, including vesicular trafficking and intracellular membrane dynamics.
Collapse
|