101
|
Wang L, Myles RC, Lee IJ, Bers DM, Ripplinger CM. Role of Reduced Sarco-Endoplasmic Reticulum Ca 2+-ATPase Function on Sarcoplasmic Reticulum Ca 2+ Alternans in the Intact Rabbit Heart. Front Physiol 2021; 12:656516. [PMID: 34045974 PMCID: PMC8144333 DOI: 10.3389/fphys.2021.656516] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/29/2021] [Indexed: 01/16/2023] Open
Abstract
Sarcoplasmic reticulum (SR) Ca2+ cycling is tightly regulated by ryanodine receptor (RyR) Ca2+ release and sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) Ca2+ uptake during each excitation–contraction coupling cycle. We previously showed that RyR refractoriness plays a key role in the onset of SR Ca2+ alternans in the intact rabbit heart, which contributes to arrhythmogenic action potential duration (APD) alternans. Recent studies have also implicated impaired SERCA function, a key feature of heart failure, in cardiac alternans and arrhythmias. However, the relationship between reduced SERCA function and SR Ca2+ alternans is not well understood. Simultaneous optical mapping of transmembrane potential (Vm) and SR Ca2+ was performed in isolated rabbit hearts (n = 10) using the voltage-sensitive dye RH237 and the low-affinity Ca2+ indicator Fluo-5N-AM. Alternans was induced by rapid ventricular pacing. SERCA was inhibited with cyclopiazonic acid (CPA; 1–10 μM). SERCA inhibition (1, 5, and 10 μM of CPA) resulted in dose-dependent slowing of SR Ca2+ reuptake, with the time constant (tau) increasing from 70.8 ± 3.5 ms at baseline to 85.5 ± 6.6, 129.9 ± 20.7, and 271.3 ± 37.6 ms, respectively (p < 0.05 vs. baseline for all doses). At fast pacing frequencies, CPA significantly increased the magnitude of SR Ca2+ and APD alternans, most strongly at 10 μM (pacing cycle length = 220 ms: SR Ca2+ alternans magnitude: 57.1 ± 4.7 vs. 13.4 ± 8.9 AU; APD alternans magnitude 3.8 ± 1.9 vs. 0.2 ± 0.19 AU; p < 0.05 10 μM of CPA vs. baseline for both). SERCA inhibition also promoted the emergence of spatially discordant alternans. Notably, at all CPA doses, alternation of SR Ca2+ release occurred prior to alternation of diastolic SR Ca2+ load as pacing frequency increased. Simultaneous optical mapping of SR Ca2+ and Vm in the intact rabbit heart revealed that SERCA inhibition exacerbates pacing-induced SR Ca2+ and APD alternans magnitude, particularly at fast pacing frequencies. Importantly, SR Ca2+ release alternans always occurred before the onset of SR Ca2+ load alternans. These findings suggest that even in settings of diminished SERCA function, relative refractoriness of RyR Ca2+ release governs the onset of intracellular Ca2+ alternans.
Collapse
Affiliation(s)
- Lianguo Wang
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Rachel C Myles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - I-Ju Lee
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Donald M Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
102
|
Henderson MJ, Trychta KA, Yang SM, Bäck S, Yasgar A, Wires ES, Danchik C, Yan X, Yano H, Shi L, Wu KJ, Wang AQ, Tao D, Zahoránszky-Kőhalmi G, Hu X, Xu X, Maloney D, Zakharov AV, Rai G, Urano F, Airavaara M, Gavrilova O, Jadhav A, Wang Y, Simeonov A, Harvey BK. A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome. Cell Rep 2021; 35:109040. [PMID: 33910017 DOI: 10.1016/j.celrep.2021.109040] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Endoplasmic reticulum (ER) dysregulation is associated with pathologies including neurodegenerative, muscular, and diabetic conditions. Depletion of ER calcium can lead to the loss of resident proteins in a process termed exodosis. To identify compounds that attenuate the redistribution of ER proteins under pathological conditions, we performed a quantitative high-throughput screen using the Gaussia luciferase (GLuc)-secreted ER calcium modulated protein (SERCaMP) assay, which monitors secretion of ER-resident proteins triggered by calcium depletion. We identify several clinically used drugs, including bromocriptine, and further characterize them using assays to measure effects on ER calcium, ER stress, and ER exodosis. Bromocriptine elicits protective effects in cell-based models of exodosis as well as in vivo models of stroke and diabetes. Bromocriptine analogs with reduced dopamine receptor activity retain similar efficacy in stabilizing the ER proteome, indicating a non-canonical mechanism of action. This study describes a strategic approach to identify small-molecule drugs capable of improving ER proteostasis in human disease conditions.
Collapse
Affiliation(s)
- Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA.
| | - Kathleen A Trychta
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Shyh-Ming Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Susanne Bäck
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carina Danchik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xiaokang Yan
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Hideaki Yano
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lei Shi
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Amy Q Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Gergely Zahoránszky-Kőhalmi
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xin Hu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - David Maloney
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Mikko Airavaara
- Neuroscience Center, HiLIFE & Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Oksana Gavrilova
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
103
|
Zhihao L, Jingyu N, Lan L, Michael S, Rui G, Xiyun B, Xiaozhi L, Guanwei F. SERCA2a: a key protein in the Ca 2+ cycle of the heart failure. Heart Fail Rev 2021; 25:523-535. [PMID: 31701344 DOI: 10.1007/s10741-019-09873-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Calcium ion (Ca2+) cycle plays a crucial role in the contraction and relaxation of cardiomyocytes. The sarcoplasmic reticulum (SR) acts as an organelle for storing Ca2+, which mediated the release and re-uptake of Ca2+ during contraction and relaxation. Disorders of SR function lead to the dysfunction of Ca2+ cycle and myocardial cell function. The sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) acts as a subtype of SERCA expressed in the heart, which mediates the contraction of cardiomyocytes and Ca2+ in the cytoplasm to re-enter into the SR. The rate of uptake of Ca2+ by the SR determines the rate of myocardial relaxation. The regulation of SERCA2a activity controls the contractility and relaxation of the heart, affecting cardiac function. The expression and activity of SERCA2a are reduced in failing hearts. Gene therapy by increasing the expression of SERCA2a in the heart has been proven effective. In addition, SERCA2a is regulated by a variety of factors, including transmembrane micropeptides, protein kinases, and post-translational modifications (PTMs). In this review, we discuss the regulatory factors of SERCA2a and provide new insights into future treatments and the direction of heart failure research. In addition, gene therapy for SERCA2a has recently emerged as therapeutic option and hence will be discussed in this review.
Collapse
Affiliation(s)
- Liu Zhihao
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Ni Jingyu
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Li Lan
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Sarhene Michael
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Guo Rui
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Bian Xiyun
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, 300450, People's Republic of China
| | - Liu Xiaozhi
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, 300450, People's Republic of China
| | - Fan Guanwei
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, 300193, People's Republic of China. .,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
104
|
Lei Z, Wahlquist C, El Azzouzi H, Deddens JC, Kuster D, van Mil A, Rojas-Munoz A, Huibers MM, Mercola M, de Weger R, Van der Velden J, Xiao J, Doevendans PA, Sluijter JPG. miR-132/212 Impairs Cardiomyocytes Contractility in the Failing Heart by Suppressing SERCA2a. Front Cardiovasc Med 2021; 8:592362. [PMID: 33816571 PMCID: PMC8017124 DOI: 10.3389/fcvm.2021.592362] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/15/2021] [Indexed: 12/21/2022] Open
Abstract
Compromised cardiac function is a hallmark for heart failure, mostly appearing as decreased contractile capacity due to dysregulated calcium handling. Unfortunately, the underlying mechanism causing impaired calcium handling is still not fully understood. Previously the miR-132/212 family was identified as a regulator of cardiac function in the failing mouse heart, and pharmaceutically inhibition of miR-132 is beneficial for heart failure. In this study, we further investigated the molecular mechanisms of miR-132/212 in modulating cardiomyocyte contractility in the context of the pathological progression of heart failure. We found that upregulated miR-132/212 expressions in all examined hypertrophic heart failure mice models. The overexpression of miR-132/212 prolongs calcium decay in isolated neonatal rat cardiomyocytes, whereas cardiomyocytes isolated from miR-132/212 KO mice display enhanced contractility in comparison to wild type controls. In response to chronic pressure-overload, miR-132/212 KO mice exhibited a blunted deterioration of cardiac function. Using a combination of biochemical approaches and in vitro assays, we confirmed that miR-132/212 regulates SERCA2a by targeting the 3′-end untranslated region of SERCA2a. Additionally, we also confirmed PTEN as a direct target of miR-132/212 and potentially participates in the cardiac response to miR132/212. In end-stage heart failure patients, miR-132/212 is upregulated and correlates with reduced SERCA2a expression. The up-regulation of miR-132/212 in heart failure impairs cardiac contractile function by targeting SERCA2a, suggesting that pharmaceutical inhibition of miR-132/212 might be a promising therapeutic approach to promote cardiac function in heart failure patients.
Collapse
Affiliation(s)
- Zhiyong Lei
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,Division Lab, Central Diagnosis Laboratory Research, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christine Wahlquist
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Hamid El Azzouzi
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Janine C Deddens
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Diederik Kuster
- Department of Physiology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Alain van Mil
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,University Medical Center Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | - Agustin Rojas-Munoz
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Manon M Huibers
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mark Mercola
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Roel de Weger
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jolanda Van der Velden
- Department of Physiology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Junjie Xiao
- Regeneration and Ageing Lab, School of Life Science, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China
| | - Pieter A Doevendans
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands.,Central Military Hospital Utrecht, Utrecht, Netherlands
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.,University Medical Center Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
105
|
Li J, Richmond B, Hong T. Cardiac T-Tubule cBIN1-Microdomain, a Diagnostic Marker and Therapeutic Target of Heart Failure. Int J Mol Sci 2021; 22:ijms22052299. [PMID: 33669042 PMCID: PMC7956774 DOI: 10.3390/ijms22052299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Since its first identification as a cardiac transverse tubule (t-tubule) protein, followed by the cloning of the cardiac isoform responsible for t-tubule membrane microdomain formation, cardiac bridging integrator 1 (cBIN1) and its organized microdomains have emerged as a key mechanism in maintaining normal beat-to-beat heart contraction and relaxation. The abnormal remodeling of cBIN1-microdomains occurs in stressed and diseased cardiomyocytes, contributing to the pathophysiology of heart failure. Due to the homeostatic turnover of t-tubule cBIN1-microdomains via microvesicle release into the peripheral circulation, plasma cBIN1 can be assayed as a liquid biopsy of cardiomyocyte health. A new blood test cBIN1 score (CS) has been developed as a dimensionless inverse index derived from plasma cBIN1 concentration with a diagnostic and prognostic power for clinical outcomes in stable ambulatory patients with heart failure with reduced or preserved ejection fraction (HFrEF or HFpEF). Recent evidence further indicates that exogenous cBIN1 introduced by adeno-associated virus 9-based gene therapy can rescue cardiac contraction and relaxation in failing hearts. The therapeutic potential of cBIN1 gene therapy is enormous given its ability to rescue cardiac inotropy and provide lusitropic protection in the meantime. These unprecedented capabilities of cBIN1 gene therapy are shifting the current paradigm of therapy development for heart failure, particularly HFpEF.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - Bradley Richmond
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
| | - TingTing Hong
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; (J.L.); (B.R.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: ; Tel.: +1-801-581-3090
| |
Collapse
|
106
|
Salazar-Ramírez F, Ramos-Mondragón R, García-Rivas G. Mitochondrial and Sarcoplasmic Reticulum Interconnection in Cardiac Arrhythmia. Front Cell Dev Biol 2021; 8:623381. [PMID: 33585462 PMCID: PMC7876262 DOI: 10.3389/fcell.2020.623381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Ca2+ plays a pivotal role in mitochondrial energy production, contraction, and apoptosis. Mitochondrial Ca2+-targeted fluorescent probes have demonstrated that mitochondria Ca2+ transients are synchronized with Ca2+ fluxes occurring in the sarcoplasmic reticulum (SR). The presence of specialized proteins tethering SR to mitochondria ensures the local Ca2+ flux between these organelles. Furthermore, communication between SR and mitochondria impacts their functionality in a bidirectional manner. Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniplex is essential for ATP production and controlled reactive oxygen species levels for proper cellular signaling. Conversely, mitochondrial ATP ensures the proper functioning of SR Ca2+-handling proteins, which ensures that mitochondria receive an adequate supply of Ca2+. Recent evidence suggests that altered SR Ca2+ proteins, such as ryanodine receptors and the sarco/endoplasmic reticulum Ca2+ ATPase pump, play an important role in maintaining proper cardiac membrane excitability, which may be initiated and potentiated when mitochondria are dysfunctional. This recognized mitochondrial role offers the opportunity to develop new therapeutic approaches aimed at preventing cardiac arrhythmias in cardiac disease.
Collapse
Affiliation(s)
- Felipe Salazar-Ramírez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico
| | - Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico.,TecSalud, Centro de Investigación Biomédica, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico.,TecSalud, Centro de Medicina Funcional, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico
| |
Collapse
|
107
|
De Jong KA, Nikolaev VO. Multifaceted remodelling of cAMP microdomains driven by different aetiologies of heart failure. FEBS J 2021; 288:6603-6622. [DOI: 10.1111/febs.15706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Kirstie A. De Jong
- Institute of Experimental Cardiovascular Research University Medical Center Hamburg‐Eppendorf Hamburg Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck D‐20246 Hamburg Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research University Medical Center Hamburg‐Eppendorf Hamburg Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck D‐20246 Hamburg Germany
| |
Collapse
|
108
|
Goodman JB, Qin F, Morgan RJ, Chambers JM, Croteau D, Siwik DA, Hobai I, Panagia M, Luptak I, Bachschmid M, Tong X, Pimentel DR, Cohen RA, Colucci WS. Redox-Resistant SERCA [Sarco(endo)plasmic Reticulum Calcium ATPase] Attenuates Oxidant-Stimulated Mitochondrial Calcium and Apoptosis in Cardiac Myocytes and Pressure Overload-Induced Myocardial Failure in Mice. Circulation 2020; 142:2459-2469. [PMID: 33076678 PMCID: PMC7752816 DOI: 10.1161/circulationaha.120.048183] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND SERCA [sarco(endo)plasmic reticulum calcium ATPase] is regulated by oxidative posttranslational modifications at cysteine 674 (C674). Because sarcoplasmic reticulum (SR) calcium has been shown to play a critical role in mediating mitochondrial dysfunction in response to reactive oxygen species, we hypothesized that SERCA oxidation at C674 would modulate the effects of reactive oxygen species on mitochondrial calcium and mitochondria-dependent apoptosis in cardiac myocytes. METHODS Adult rat ventricular myocytes expressing wild-type SERCA2b or a redox-insensitive mutant in which C674 is replaced by serine (C674S) were exposed to H2O2 (100 µmol/Lμ). Free mitochondrial calcium concentration was measured in adult rat ventricular myocytes with a genetically targeted fluorescent probe, and SR calcium content was assessed by measuring caffeine-stimulated release. Mice with heterozygous knock-in of the SERCA C674S mutation were subjected to chronic ascending aortic constriction. RESULTS In adult rat ventricular myocytes expressing wild-type SERCA, H2O2 caused a 25% increase in mitochondrial calcium concentration that was associated with a 50% decrease in SR calcium content, both of which were prevented by the ryanodine receptor inhibitor tetracaine. In cells expressing the C674S mutant, basal SR calcium content was decreased by 31% and the H2O2-stimulated rise in mitochondrial calcium concentration was attenuated by 40%. In wild-type cells, H2O2 caused cytochrome c release and apoptosis, both of which were prevented in C674S-expressing cells. In myocytes from SERCA knock-in mice, basal SERCA activity and SR calcium content were decreased. To test the effect of C674 oxidation on apoptosis in vivo, SERCA knock-in mice were subjected to chronic ascending aortic constriction. In wild-type mice, ascending aortic constriction caused myocyte apoptosis, LV dilation, and systolic failure, all of which were inhibited in SERCA knock-in mice. CONCLUSIONS Redox activation of SERCA C674 regulates basal SR calcium content, thereby mediating the pathologic reactive oxygen species-stimulated rise in mitochondrial calcium required for myocyte apoptosis and myocardial failure.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Calcium/metabolism
- Calcium Signaling
- Cells, Cultured
- Disease Models, Animal
- Heart Failure/enzymology
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Failure/physiopathology
- Hydrogen Peroxide/toxicity
- Male
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/genetics
- Mitochondria, Heart/pathology
- Mutation
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxidants/toxicity
- Oxidation-Reduction
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Jena B. Goodman
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Fuzhong Qin
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Robert J. Morgan
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Jordan M. Chambers
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Dominique Croteau
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Deborah A. Siwik
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Ion Hobai
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Marcello Panagia
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Ivan Luptak
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Markus Bachschmid
- Vascular Biology Unit, Boston University School of
Medicine, Boston, MA
| | - XiaoYong Tong
- Vascular Biology Unit, Boston University School of
Medicine, Boston, MA
| | - David R. Pimentel
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Richard A. Cohen
- Vascular Biology Unit, Boston University School of
Medicine, Boston, MA
| | - Wilson S. Colucci
- Cardiovascular Medicine Section, Boston University School
of Medicine, Boston, MA
- Myocardial Biology Unit, Boston University School of
Medicine, Boston, MA
| |
Collapse
|
109
|
Brun MJ, Song K, Kang B, Lueck C, Chen W, Thatcher K, Gao E, Koch WJ, Lincoln J, Rajan S, Suh J. Constructing and evaluating caspase-activatable adeno-associated virus vector for gene delivery to the injured heart. J Control Release 2020; 328:834-845. [PMID: 33157191 PMCID: PMC7770761 DOI: 10.1016/j.jconrel.2020.10.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/11/2020] [Accepted: 10/29/2020] [Indexed: 01/16/2023]
Abstract
Adeno-associated virus (AAV) is a promising vector for gene therapy, but its broad tropism can be detrimental if the transgene being delivered is harmful when expressed ubiquitously in the body, i.e. in non-target tissues. Delivering the transgene of interest to target cells at levels high enough to be therapeutically effective while maintaining safety by minimizing delivery to off-target cells is a prevalent challenge in the field of gene therapy. We have developed a protease activatable vector (provector) platform based on AAV9 that can be injected systemically to deliver therapeutic transgenes site-specifically to diseased cells by responding to extracellular proteases present at the disease site. The provector platform consists of a peptide insertion into the virus capsid which disrupts the virus' ability to bind to cell surface receptors. This peptide contains a blocking motif (aspartic acid residues) flanked on either side by cleavage sequences that are recognized by certain proteases. Exposure to proteases cleaves the peptides off the capsid, activating or "switching ON" the provector. In response to the activation, the provectors regain their ability to bind and transduce cells. Here, we have designed a provector that is activated by cysteine aspartic proteases (caspases), which have roles in inflammation and apoptosis and thus are elevated at sites of diseases such as heart failure, neurodegenerative diseases, and ischemic stroke. This provector demonstrates a 200-fold reduction in transduction ability in the OFF state compared to AAV9, reducing the virus' ability to transduce off-target healthy tissue. Following exposure to and proteolysis by caspase-3, the provector shows a 95-fold increase in transduction compared to the OFF state. The switchable transduction behavior was found to be a direct result of the peptide insertion ablating the ability of the virus to bind to cells. In vivo studies were conducted to characterize the biodistribution, blood circulation time, neutralizing antibody formation, and targeted delivery ability of the caspase-activatable provector in a model of heart failure.
Collapse
Affiliation(s)
- Mitchell J Brun
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main St., Houston, TX 77005, United States
| | - Kefan Song
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, United States
| | - Byunguk Kang
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, United States
| | - Cooper Lueck
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, United States
| | - Weitong Chen
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main St., Houston, TX 77005, United States
| | - Kaitlyn Thatcher
- Pediatric Cardiology, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, United States
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, United States
| | - Joy Lincoln
- Pediatric Cardiology, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Sudarsan Rajan
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, United States
| | - Junghae Suh
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main St., Houston, TX 77005, United States; Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, United States.
| |
Collapse
|
110
|
van der Pol A, Hoes MF, de Boer RA, van der Meer P. Cardiac foetal reprogramming: a tool to exploit novel treatment targets for the failing heart. J Intern Med 2020; 288:491-506. [PMID: 32557939 PMCID: PMC7687159 DOI: 10.1111/joim.13094] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/26/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
As the heart matures during embryogenesis from its foetal stages, several structural and functional modifications take place to form the adult heart. This process of maturation is in large part due to an increased volume and work load of the heart to maintain proper circulation throughout the growing body. In recent years, it has been observed that these changes are reversed to some extent as a result of cardiac disease. The process by which this occurs has been characterized as cardiac foetal reprogramming and is defined as the suppression of adult and re-activation of a foetal genes profile in the diseased myocardium. The reasons as to why this process occurs in the diseased myocardium are unknown; however, it has been suggested to be an adaptive process to counteract deleterious events taking place during cardiac remodelling. Although still in its infancy, several studies have demonstrated that targeting foetal reprogramming in heart failure can lead to substantial improvement in cardiac functionality. This is highlighted by a recent study which found that by modulating the expression of 5-oxoprolinase (OPLAH, a novel cardiac foetal gene), cardiac function can be significantly improved in mice exposed to cardiac injury. Additionally, the utilization of angiotensin receptor neprilysin inhibitors (ARNI) has demonstrated clear benefits, providing important clinical proof that drugs that increase natriuretic peptide levels (part of the foetal gene programme) indeed improve heart failure outcomes. In this review, we will highlight the most important aspects of cardiac foetal reprogramming and will discuss whether this process is a cause or consequence of heart failure. Based on this, we will also explain how a deeper understanding of this process may result in the development of novel therapeutic strategies in heart failure.
Collapse
Affiliation(s)
- A van der Pol
- From the, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Perioperative Inflammation and Infection Group, Department of Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - M F Hoes
- From the, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - R A de Boer
- From the, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - P van der Meer
- From the, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
111
|
Cardiac Optogenetics in Atrial Fibrillation: Current Challenges and Future Opportunities. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8814092. [PMID: 33195698 PMCID: PMC7641281 DOI: 10.1155/2020/8814092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/07/2020] [Indexed: 12/23/2022]
Abstract
Although rarely life-threatening on short term, atrial fibrillation leads to increased mortality and decreased quality of life through its complications, including heart failure and stroke. Recent studies highlight the benefits of maintaining sinus rhythm. However, pharmacological long-term rhythm control strategies may be shadowed by associated proarrhythmic effects. At the same time, electrical cardioversion is limited to hospitals, while catheter ablation therapy, although effective, is invasive and is dedicated to specific patients, usually with low amounts of atrial fibrosis (preferably Utah I-II). Cardiac optogenetics allows influencing the heart's electrical activity by applying specific wavelength light pulses to previously engineered cardiomyocytes into expressing microbial derived light-sensitive proteins called opsins. The resulting ion influx may give rise to either hyperpolarizing or depolarizing currents, thus offering a therapeutic potential in cardiac electrophysiology, including pacing, resynchronization, and arrhythmia termination. Optogenetic atrial fibrillation cardioversion might be achieved by inducing a conduction block or filling of the excitable gap. The authors agree that transmural opsin expression and appropriate illumination with an exposure time longer than the arrhythmia cycle length are necessary to achieve successful arrhythmia termination. However, the efficiency and safety of biological cardioversion in humans remain to be seen, as well as side effects such as immune reactions and loss of opsin expression. The possibility of delivering pain-free shocks with out-of-hospital biological cardioversion is tempting; however, there are several issues that need to be addressed first: applicability and safety in humans, long-term behaviour, anticoagulation requirements, and fibrosis interactions.
Collapse
|
112
|
Circular RNAs are a novel type of non-coding RNAs in ROS regulation, cardiovascular metabolic inflammations and cancers. Pharmacol Ther 2020; 220:107715. [PMID: 33141028 DOI: 10.1016/j.pharmthera.2020.107715] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
Circular RNAs (circRNAs) are a novel class of endogenous non-coding RNAs characterized by a covalently closed-loop structure generated through a special type of alternative splicing termed back-splicing. Currently, an increasing body of evidence has demonstrated that 1) majority of circRNAs are evolutionarily conserved across species, stable, and resistant to RNase R degradation, and often exhibit cell-specific, and tissue-specific/developmental-stage-specific expression and can be largely independent of the expression levels of the linear host gene-encoded linear RNAs; 2) the biogenesis of circRNAs via back-splicing is different from the canonical splicing of linear RNAs; 3) circRNA biogenesis is regulated by specific cis-acting elements and trans-acting factors; 4) circRNAs regulate biological and pathological processes by sponging miRNAs, binding to RNA-binding protein (RBP), regulators of splicing and transcription, modifiers of parental gene expression, and regulators of protein translation or being translated into peptides in various diseases; 5) circRNAs have been identified for their enrichment and stability in exosomes and detected in body fluids such as human blood, saliva, and cerebrospinal fluids, suggesting that these exo-circRNAs have potential applications as disease biomarkers and novel therapeutic targets; 6) several circRNAs are regulated by oxidative stress and mediate reactive oxygen species (ROS) production as well as promote ROS-induced cellular death, cell apoptosis, and inflammation; 7) circRNAs have also emerged as important regulators in atherosclerotic cardiovascular disease, metabolic disease, and cancers; 8) the potential mechanisms of several circRNAs have been described in diseases, hinting at their potential applications as novel therapeutic targets. In this highlight, we summarized the current understandings of the biogenesis and functions of circRNAs and their roles in ROS regulation and vascular inflammation-associated with cardiovascular and metabolic disease. (Word count: 272).
Collapse
|
113
|
Njegic A, Wilson C, Cartwright EJ. Targeting Ca 2 + Handling Proteins for the Treatment of Heart Failure and Arrhythmias. Front Physiol 2020; 11:1068. [PMID: 33013458 PMCID: PMC7498719 DOI: 10.3389/fphys.2020.01068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Diseases of the heart, such as heart failure and cardiac arrhythmias, are a growing socio-economic burden. Calcium (Ca2+) dysregulation is key hallmark of the failing myocardium and has long been touted as a potential therapeutic target in the treatment of a variety of cardiovascular diseases (CVD). In the heart, Ca2+ is essential for maintaining normal cardiac function through the generation of the cardiac action potential and its involvement in excitation contraction coupling. As such, the proteins which regulate Ca2+ cycling and signaling play a vital role in maintaining Ca2+ homeostasis. Changes to the expression levels and function of Ca2+-channels, pumps and associated intracellular handling proteins contribute to altered Ca2+ homeostasis in CVD. The remodeling of Ca2+-handling proteins therefore results in impaired Ca2+ cycling, Ca2+ leak from the sarcoplasmic reticulum and reduced Ca2+ clearance, all of which contributes to increased intracellular Ca2+. Currently, approved treatments for targeting Ca2+ handling dysfunction in CVD are focused on Ca2+ channel blockers. However, whilst Ca2+ channel blockers have been successful in the treatment of some arrhythmic disorders, they are not universally prescribed to heart failure patients owing to their ability to depress cardiac function. Despite the progress in CVD treatments, there remains a clear need for novel therapeutic approaches which are able to reverse pathophysiology associated with heart failure and arrhythmias. Given that heart failure and cardiac arrhythmias are closely associated with altered Ca2+ homeostasis, this review will address the molecular changes to proteins associated with both Ca2+-handling and -signaling; their potential as novel therapeutic targets will be discussed in the context of pre-clinical and, where available, clinical data.
Collapse
Affiliation(s)
- Alexandra Njegic
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
114
|
Hawkins LJ, Storey KB. MicroRNA expression in the heart of Xenopus laevis facilitates metabolic adaptation to dehydration. Genomics 2020; 112:3525-3536. [DOI: 10.1016/j.ygeno.2020.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/24/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022]
|
115
|
Berry-Kravis E, Horrigan JP, Tartaglia N, Hagerman R, Kolevzon A, Erickson CA, Hatti S, Snape M, Yaroshinsky A, Stoms G, Glass L, Jones NE. A Double-Blind, Randomized, Placebo-Controlled Clinical Study of Trofinetide in the Treatment of Fragile X Syndrome. Pediatr Neurol 2020; 110:30-41. [PMID: 32660869 DOI: 10.1016/j.pediatrneurol.2020.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/10/2020] [Accepted: 04/25/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND We analyze the safety and tolerability of trofinetide and provide a preliminary evaluation of its efficacy in adolescent and adult males with fragile X syndrome. METHODS This study was an exploratory, phase 2, multicenter, double-blind, placebo-controlled, parallel group study of the safety and tolerability of orally administered trofinetide in 72 adolescent and adult males with fragile X syndrome. Subjects were randomly assigned in a 1:1:1 ratio to 35 or 70 mg/kg twice daily trofinetide or placebo for 28 days. Safety assessments included adverse events, clinical laboratory tests, vital signs, electrocardiograms, physical examinations, and concomitant medications. Efficacy measurements were categorized into four efficacy domains, which related to clinically relevant phenotypic dimensions of impairment associated with fragile X syndrome. RESULTS Both 35 and 70 mg/kg dose levels of trofinetide were well tolerated and appeared to be generally safe. Trofinetide at the 70 mg/kg dose level demonstrated efficacy compared with placebo based on prespecified criteria. On the basis of a permutation test, the probability of a false-positive outcome for the achieved prespecified success was 0.045. In the group analysis, improvement from treatment baseline was demonstrated on three fragile X syndrome-specific outcome measures. CONCLUSIONS Trofinetide was well tolerated in adolescent and adult males with fragile X syndrome. Despite the relatively short duration of the study, a consistent signal of efficacy at the higher dose was observed in both caregiver and clinician assessments, based on a novel analytical model incorporating evaluation of multiple key symptom areas of fragile X syndrome. This finding suggests a potential for trofinetide treatment to provide clinically meaningful improvement in core fragile X syndrome symptoms.
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, Illinois; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois; Department of Biochemistry, Rush University Medical Center, Chicago, Illinois.
| | - Joseph P Horrigan
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
| | - Nicole Tartaglia
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Randi Hagerman
- Department of Pediatrics, University of California Davis MIND Institute, Sacramento, California
| | - Alexander Kolevzon
- Division of Child and Adolescent Psychiatry, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Child and Adolescent Psychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Craig A Erickson
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | | | | | | | | - Larry Glass
- Neuren Pharmaceuticals, Ltd., Melbourne, Victoria, Australia
| | - Nancy E Jones
- Neuren Pharmaceuticals, Ltd., Melbourne, Victoria, Australia
| |
Collapse
|
116
|
Dashwood A, Cheesman E, Beard N, Haqqani H, Wong YW, Molenaar P. Understanding How Phosphorylation and Redox Modifications Regulate Cardiac Ryanodine Receptor Type 2 Activity to Produce an Arrhythmogenic Phenotype in Advanced Heart Failure. ACS Pharmacol Transl Sci 2020; 3:563-582. [PMID: 32832863 DOI: 10.1021/acsptsci.0c00003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Indexed: 12/17/2022]
Abstract
Heart failure (HF) is a global pandemic with significant mortality and morbidity. Despite current medications, 50% of individuals die within 5 years of diagnosis. Of these deaths, 30-50% will be a result of sudden cardiac death from ventricular arrhythmias. This review discusses two stress-induced mechanisms, phosphorylation from chronic β-adrenoceptor (β-AR) stimulation and thiol modifications from oxidative stress, and how they modulate the cardiac ryanodine receptor type 2 (RyR2) and foster an arrhythmogenic phenotype. Calcium (Ca2+) is the ubiquitous secondary messenger of excitation-contraction coupling and provides a common pathway for contractile dysfunction and arrhythmia genesis. In a healthy heart, Ca2+ is released from the sarcoplasmic reticulum (SR) by RyR2. The open probability of RyR2 is under the dynamic influence of co-proteins, ions, and kinases that are in strict balance to ensure normal physiological functioning. In HF, chronic β-AR activity and production of reactive oxygen species and reactive nitrogen species provide two stress-induced mechanisms uncoupling RyR2 control, resulting in pathological diastolic SR Ca2+ leak. This increased cytosolic [Ca2+] promotes Ca2+ extrusion via the local Na+/Ca2+ exchanger, resulting in net sarcolemmal depolarization, delayed after depolarization and ventricular arrhythmia. Experimental models researching oxidative stress and phosphorylation have aimed to identify how post-translational modifications to the RyR2 macromolecular complex, and the associated Na+/Ca2+ cycling proteins, result in pathological Ca2+ handling and diastolic leak. However, the causative molecular changes remain controversial and undefined. Through understanding the molecular mechanisms that produce an arrhythmic phenotype, novel therapeutic targets to treat HF and prevent its malignant course can be identified.
Collapse
Affiliation(s)
- Alexander Dashwood
- Heart Lung Institute, The Prince Charles Hospital, Chermside, Brisbane, Queensland 4032, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4032, Australia.,Griffith University, Southport, Queensland 4215, Australia
| | - Elizabeth Cheesman
- Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4032, Australia
| | - Nicole Beard
- Queensland University of Technology (QUT), School of Biomedical Sciences, Institute of Health and Biomedical Innovation, 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia.,Faculty of Science and Technology, University of Canberra, Bruce, Australian Capital Territory 2617, Australia
| | - Haris Haqqani
- Heart Lung Institute, The Prince Charles Hospital, Chermside, Brisbane, Queensland 4032, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4032, Australia
| | - Yee Weng Wong
- Heart Lung Institute, The Prince Charles Hospital, Chermside, Brisbane, Queensland 4032, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4032, Australia
| | - Peter Molenaar
- Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4032, Australia.,Queensland University of Technology (QUT), School of Biomedical Sciences, Institute of Health and Biomedical Innovation, 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia
| |
Collapse
|
117
|
Yamada KP, Tharakan S, Ishikawa K. Consideration of clinical translation of cardiac AAV gene therapy. ACTA ACUST UNITED AC 2020; 6:609-615. [PMID: 32774893 DOI: 10.18609/cgti.2020.073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Advancements in conventional cardiac care have significantly reduced mortality from coronary heart disease and acute myocardial infarction. However, the prevalence of heart failure continues to increase in an aging population with profound social and economic consequences. Cardiac gene therapy with adeno-associated virus (AAV) vectors is emerging as a potential modality for addressing this desperate clinical need. After showing initial promise in extensive preclinical studies and an early clinical trial, disappointing results of large-scale clinical trial derailed the progress of AAV-mediated cardiac gene therapy. However, it appears that knowledge gained from previous failures coupled with developments in targeted gene delivery have set the stage for a new frontier in cardiac AAV gene therapy.
Collapse
Affiliation(s)
- Kelly P Yamada
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Serena Tharakan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
118
|
Collins L, Binder P, Chen H, Wang X. Regulation of Long Non-coding RNAs and MicroRNAs in Heart Disease: Insight Into Mechanisms and Therapeutic Approaches. Front Physiol 2020; 11:798. [PMID: 32754048 PMCID: PMC7365882 DOI: 10.3389/fphys.2020.00798] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide and there is an increasing need to identify new therapeutic targets that could be used to prevent or treat these diseases. Due to recent scientific advances, non-coding RNAs are widely accepted as important regulators of cellular processes, and the identification of an axis of interaction between long non-coding RNAs (lncRNAs) and micro RNAs (miRNAs) has provided another platform through which cardiovascular disease could be targeted therapeutically. Increasing evidence has detailed the importance of these non-coding RNAs, both individually and in an axis of regulation, in the processes and diseases involving the heart. However, further investigation into the consequences of targeting this mechanism, as well as refinement of how the system is targeted, are required before a treatment can be provided in clinic. This level of genomic regulation provides an exciting potential novel therapeutic strategy for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Lucy Collins
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Pablo Binder
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Wang
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
119
|
García-Manzanares M, Tarazón E, Ortega A, Gil-Cayuela C, Martínez-Dolz L, González-Juanatey JR, Lago F, Portolés M, Roselló-Lletí E, Rivera M. XPO1 Gene Therapy Attenuates Cardiac Dysfunction in Rats with Chronic Induced Myocardial Infarction. J Cardiovasc Transl Res 2020; 13:593-600. [PMID: 31768947 PMCID: PMC7423868 DOI: 10.1007/s12265-019-09932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Transcriptomic signature of XPO1 was highly expressed and inversely related to left ventricular function in ischemic cardiomyopathy patients. We hypothesized that treatment with AAV9-shXPO1 attenuates left ventricular dysfunction and remodeling in a myocardial infarction rat model. We induced myocardial infarction by coronary ligation in Sprague-Dawley rats (n = 10), which received AAV9-shXPO1 (n = 5) or placebo AAV9-scramble (n = 5) treatment. Serial echocardiographic assessment was performed throughout the study. After myocardial infarction, AAV9-shXPO1-treated rats showed partial recovery of left ventricular fractional shortening (16.8 ± 2.8 vs 24.6 ± 4.1%, P < 0.05) and a maintained left ventricular dimension (6.17 ± 0.95 vs 4.70 ± 0.93 mm, P < 0.05), which was not observed in non-treated rats. Furthermore, lower levels of EXP-1 (P < 0.05) and lower collagen fibers and fibrosis in cardiac tissue were observed. However, no differences were found in the IL-6 or TNFR1 plasma levels of the myocardium of AAV9-shXPO1 rats. AAV9-shXPO1 administration attenuates cardiac dysfunction and remodeling in rats after myocardial infarction, producing the gene silencing of XPO1.
Collapse
Affiliation(s)
- María García-Manzanares
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Estefanía Tarazón
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Ana Ortega
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Carolina Gil-Cayuela
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Luis Martínez-Dolz
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Heart Failure and Transplantation Unit, Cardiology Department, University Hospital La Fe, Valencia, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Manuel Portolés
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Esther Roselló-Lletí
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology, Veterinary Faculty, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Miguel Rivera
- Myocardial Dysfunction and Cardiac Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
120
|
Jolicoeur EM, Verheye S, Henry TD, Joseph L, Doucet S, White CJ, Edelman E, Banai S. A novel method to interpret early phase trials shows how the narrowing of the coronary sinus concordantly improves symptoms, functional status and quality of life in refractory angina. Heart 2020; 107:41-46. [PMID: 32719097 DOI: 10.1136/heartjnl-2020-316644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Reduction of the coronary sinus was shown to improve angina in patients unsuitable for revascularisation. We assessed whether a percutaneous device that reduces the diameter of the coronary sinus improved outcomes across multiple endpoints in a phase II trial. METHODS We conducted a novel analysis performed as a post hoc efficacy analysis of the COSIRA (Coronary Sinus Reducer for Treatment of Refractory Angina) trial, which enrolled patients with Canadian Cardiovascular Society (CCS) class 3-4 refractory angina. We used four domains: symptoms (CCS Angina Scale), functionality (total exercise duration), ischaemia (imaging) and health-related quality of life. For all domains, we specified a meaningful threshold for change. The primary endpoint was defined as a probability of ≥80% that the reducer exceeded the meaningful threshold on two or more domains (group-level analysis) or that the average efficacy score in the reducer group exceeded the sham control group by at least two points (patient-level analysis). RESULTS We randomised 104 participants to either a device that narrows to coronary sinus (n=52) or a sham implantation (n=52). The reducer group met the prespecified criteria for concordance at the group level and demonstrated improvement in symptoms (0.59 CCS grade, 95% credible interval (CrI)=0.22 to 0.95), total exercise duration (+27.9%, 95% CrI=2.8% to 59.8%) and quality of life (stability +11.2 points, 95% CrI=3.3 to 19.1; perception +11.0, 95% CrI=3.3 to 18.7). CONCLUSIONS The reducer concordantly improved symptoms, functionality and quality of life compared with a sham intervention in patients with angina unsuitable for coronary revascularisation. Concordant analysis such as this one can help interpret early phase trials and guide the decision to pursue a clinical programme into a larger confirmatory trial. TRAIL REGISTRATION NUMBER ClinicalTrials.gov identifier: NCT01205893.
Collapse
Affiliation(s)
- E Marc Jolicoeur
- Department of Medicine, Interventional Cardiology, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Stefan Verheye
- Antwerp Cardiovascular Institute, ZNA Middelheim Hospital, Antwerpen, Belgium
| | - Timothy D Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital, Cincinnati, Ohio, USA
| | - Lawrence Joseph
- Department of Epidemiology and Biostatistics, McGill University, Montreal, Quebec, Canada
| | - Serge Doucet
- Department of Medicine, Interventional Cardiology, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Christopher J White
- The John Ochsner Heart & Vascular Institute, Ochsner Clinical School, University of Queensland, New Orleans, Louisiana, USA
| | - Elazer Edelman
- Institute for Medical Engineering and Science, Cambridge, Massachusetts, USA.,Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shmuel Banai
- The Tel Aviv Medical Center, The Tel Aviv University Medical School, Tel Aviv, Israel
| |
Collapse
|
121
|
Bezzerides VJ, Prondzynski M, Carrier L, Pu WT. Gene therapy for inherited arrhythmias. Cardiovasc Res 2020; 116:1635-1650. [PMID: 32321160 PMCID: PMC7341167 DOI: 10.1093/cvr/cvaa107] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/13/2020] [Accepted: 04/15/2020] [Indexed: 01/16/2023] Open
Abstract
Inherited arrhythmias are disorders caused by one or more genetic mutations that increase the risk of arrhythmia, which result in life-long risk of sudden death. These mutations either primarily perturb electrophysiological homeostasis (e.g. long QT syndrome and catecholaminergic polymorphic ventricular tachycardia), cause structural disease that is closely associated with severe arrhythmias (e.g. hypertrophic cardiomyopathy), or cause a high propensity for arrhythmia in combination with altered myocardial structure and function (e.g. arrhythmogenic cardiomyopathy). Currently available therapies offer incomplete protection from arrhythmia and fail to alter disease progression. Recent studies suggest that gene therapies may provide potent, molecularly targeted options for at least a subset of inherited arrhythmias. Here, we provide an overview of gene therapy strategies, and review recent studies on gene therapies for catecholaminergic polymorphic ventricular tachycardia and hypertrophic cardiomyopathy caused by MYBPC3 mutations.
Collapse
Affiliation(s)
- Vassilios J Bezzerides
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Lucie Carrier
- Institute of Experimental and Clinical Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site, Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - William T Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
122
|
Lyon AR, Babalis D, Morley-Smith AC, Hedger M, Suarez Barrientos A, Foldes G, Couch LS, Chowdhury RA, Tzortzis KN, Peters NS, Rog-Zielinska EA, Yang HY, Welch S, Bowles CT, Rahman Haley S, Bell AR, Rice A, Sasikaran T, Johnson NA, Falaschetti E, Parameshwar J, Lewis C, Tsui S, Simon A, Pepper J, Rudy JJ, Zsebo KM, Macleod KT, Terracciano CM, Hajjar RJ, Banner N, Harding SE. Investigation of the safety and feasibility of AAV1/SERCA2a gene transfer in patients with chronic heart failure supported with a left ventricular assist device - the SERCA-LVAD TRIAL. Gene Ther 2020; 27:579-590. [PMID: 32669717 PMCID: PMC7744277 DOI: 10.1038/s41434-020-0171-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/05/2020] [Accepted: 06/25/2020] [Indexed: 01/16/2023]
Abstract
The SERCA-LVAD trial was a phase 2a trial assessing the safety and feasibility of delivering an adeno-associated vector 1 carrying the cardiac isoform of the sarcoplasmic reticulum calcium ATPase (AAV1/SERCA2a) to adult chronic heart failure patients implanted with a left ventricular assist device. The SERCA-LVAD trial was one of a program of AAV1/SERCA2a cardiac gene therapy trials including CUPID1, CUPID 2 and AGENT trials. Enroled subjects were randomised to receive a single intracoronary infusion of 1 × 1013 DNase-resistant AAV1/SERCA2a particles or a placebo solution in a double-blinded design, stratified by presence of neutralising antibodies to AAV. Elective endomyocardial biopsy was performed at 6 months unless the subject had undergone cardiac transplantation, with myocardial samples assessed for the presence of exogenous viral DNA from the treatment vector. Safety assessments including ELISPOT were serially performed. Although designed as a 24 subject trial, recruitment was stopped after five subjects had been randomised and received infusion due to the neutral result from the CUPID 2 trial. Here we describe the results from the 5 patients at 3 years follow up, which confirmed that viral DNA was delivered to the failing human heart in 2 patients receiving gene therapy with vector detectable at follow up endomyocardial biopsy or cardiac transplantation. Absolute levels of detectable transgene DNA were low, and no functional benefit was observed. There were no safety concerns in this small cohort. This trial identified some of the challenges of performing gene therapy trials in this LVAD patient cohort which may help guide future trial design.
Collapse
Affiliation(s)
- A R Lyon
- National Heart and Lung Institute, Imperial College London, London, UK. .,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK.
| | - D Babalis
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - A C Morley-Smith
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - M Hedger
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - A Suarez Barrientos
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - G Foldes
- National Heart and Lung Institute, Imperial College London, London, UK
| | - L S Couch
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R A Chowdhury
- National Heart and Lung Institute, Imperial College London, London, UK
| | - K N Tzortzis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - N S Peters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - E A Rog-Zielinska
- National Heart and Lung Institute, Imperial College London, London, UK.,Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
| | - H-Y Yang
- National Heart and Lung Institute, Imperial College London, London, UK
| | - S Welch
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - C T Bowles
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - S Rahman Haley
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - A R Bell
- Department of Histopathology, Royal Brompton and Harefield Hospitals NHS Trust, Freiburg, Germany
| | - A Rice
- Department of Histopathology, Royal Brompton and Harefield Hospitals NHS Trust, Freiburg, Germany
| | - T Sasikaran
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - N A Johnson
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | - E Falaschetti
- Imperial Clinical Trials Unit (ICTU), School of Public Health, Imperial College London, London, UK
| | | | - C Lewis
- Royal Papworth Hospital NHS Trust, Cambridge, UK
| | - S Tsui
- Royal Papworth Hospital NHS Trust, Cambridge, UK
| | - A Simon
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - J Pepper
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - J J Rudy
- Celladon Corporation, San Diego, CA, USA
| | - K M Zsebo
- Celladon Corporation, San Diego, CA, USA
| | - K T Macleod
- National Heart and Lung Institute, Imperial College London, London, UK
| | - C M Terracciano
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R J Hajjar
- Phospholamban Foundation, Amsterdam, Netherlands
| | - N Banner
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield Hospitals NHS Trust, London, UK
| | - S E Harding
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
123
|
Vakrou S, Nana MA, Nanas IA, Nana-Leventaki E, Bonios M, Kapelios C, Nanas J. Safety and efficacy of global intracoronary administration of cardiosphere-derived cells or conditioned medium immediately after coronary reperfusion in rats. Hellenic J Cardiol 2020; 61:256-261. [PMID: 30904729 DOI: 10.1016/j.hjc.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 03/01/2019] [Accepted: 03/15/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Cardiosphere-derived cells (CDCs) have been shown to reduce infarct size after myocardial infarction (MI). In the present study we investigated the safety and efficacy of global intracoronary administration (GIA) of CDCs or CDC-conditioned medium (CM) immediately after reperfusion in a rat model of ischemia-reperfusion. METHODS CDCs were grown from myocardial biopsies obtained from male Wistar Kyoto rats (WKY). Female WKY rats underwent MI for 45minutes, followed by reperfusion for 1hour. Infarcted rats were randomized to receive GIA of CDCs (CDC group), CM (CM group) or vehicle (control group) immediately after the onset of reperfusion. Cell retention was quantified by PCR for the male specific SRY gene; area at risk (AR) and no reflow area (NR) were measured by histopathology. Cardiac function was evaluated by echocardiography at 1 and 2 months post-MI. RESULTS Cell retention at 1hour after GIA was 25.1% ±5.1. The myocardial AR and NR (measured at 1 hour post-reperfusion) were similar between groups [AR: 28.8% ±7.4 of LV mass in control vs 27.2% ±8 in CM vs 27% ±7 in CDCs group. NR: 7.0% ±3.3 in control vs 7.3% ±3.8 in CM vs 7.1% ±3.6 in CDCs]. One and 2 months post-MI, systolic function and LV volumes did not differ between control and CM groups. CONCLUSION Intracoronary administration of CDCs during the acute phase of MI, at the beginning of reperfusion, does not aggravate microvascular obstruction and results in high cell retention. Delivery of CM in the acute phase of MI did not confer long-term cardiac functional benefits.
Collapse
Affiliation(s)
- Styliani Vakrou
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria A Nana
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ioannis A Nanas
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Emmeleia Nana-Leventaki
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Michael Bonios
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Chris Kapelios
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - John Nanas
- 3(rd) Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
124
|
Tang X, Ma S, Li Y, Sun Y, Zhang K, Zhou Q, Yu R. Evaluating the Activity of Sodium Butyrate to Prevent Osteoporosis in Rats by Promoting Osteal GSK-3β/Nrf2 Signaling and Mitochondrial Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6588-6603. [PMID: 32459091 DOI: 10.1021/acs.jafc.0c01820] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Oxidative stress (OS) and mitochondrial dysfunction are key pathophysiological features of osteoporosis and obesity. Sodium butyrate (NaB), produced by fermentation by the gut microbiota of the large intestine, has been demonstrated to protect against OS by improving specific antioxidant enzymes and to regulate mitochondria redox homeostasis in vivo. Here, in an unblinded study, we identified femur mitochondria as the main target of the beneficial effects of NaB, consisting of reversion of bone loss and body-weight gain in obesity-prone rats. In particular, NaB promoted the activity of mitochondrial antioxidant enzymes and energy metabolism, preserved the bone microstructure and calcium homeostasis, and activated bone metabolism, as shown by increased Nrf2/GSK-3β signaling and the upregulation of PGC-1α and TFAM. In vitro experiments showed that moderate NaB treatment prevented H2O2-induced oxidative damage in MC3T3-E1 cells, improved osteoblast mineralization and differentiation, and maintained the balance in bone metabolism by enhancing intracellular antioxidant enzyme activity and ATP production and decreasing the ROS level. In conclusion, NaB promoted the Nrf2/GSK-3β signaling pathway and mitochondrial function and is a potential new therapeutic strategy for obesity and osteoporosis.
Collapse
Affiliation(s)
- Xue Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shuhua Ma
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yingrui Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yongjuan Sun
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Kai Zhang
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qin Zhou
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
| |
Collapse
|
125
|
Aguayo-Ortiz R, Espinoza-Fonseca LM. Linking Biochemical and Structural States of SERCA: Achievements, Challenges, and New Opportunities. Int J Mol Sci 2020; 21:ijms21114146. [PMID: 32532023 PMCID: PMC7313052 DOI: 10.3390/ijms21114146] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Sarcoendoplasmic reticulum calcium ATPase (SERCA), a member of the P-type ATPase family of ion and lipid pumps, is responsible for the active transport of Ca2+ from the cytoplasm into the sarcoplasmic reticulum lumen of muscle cells, into the endoplasmic reticulum (ER) of non-muscle cells. X-ray crystallography has proven to be an invaluable tool in understanding the structural changes of SERCA, and more than 70 SERCA crystal structures representing major biochemical states (defined by bound ligand) have been deposited in the Protein Data Bank. Consequently, SERCA is one of the best characterized components of the calcium transport machinery in the cell. Emerging approaches in the field, including spectroscopy and molecular simulation, now help integrate and interpret this rich structural information to understand the conformational transitions of SERCA that occur during activation, inhibition, and regulation. In this review, we provide an overview of the crystal structures of SERCA, focusing on identifying metrics that facilitate structure-based categorization of major steps along the catalytic cycle. We examine the integration of crystallographic data with different biophysical approaches and computational methods to link biochemical and structural states of SERCA that are populated in the cell. Finally, we discuss the challenges and new opportunities in the field, including structural elucidation of functionally important and novel regulatory complexes of SERCA, understanding the structural basis of functional divergence among homologous SERCA regulators, and bridging the gap between basic and translational research directed toward therapeutic modulation of SERCA.
Collapse
|
126
|
Baehr A, Umansky KB, Bassat E, Jurisch V, Klett K, Bozoglu T, Hornaschewitz N, Solyanik O, Kain D, Ferraro B, Cohen-Rabi R, Krane M, Cyran C, Soehnlein O, Laugwitz KL, Hinkel R, Kupatt C, Tzahor E. Agrin Promotes Coordinated Therapeutic Processes Leading to Improved Cardiac Repair in Pigs. Circulation 2020; 142:868-881. [PMID: 32508131 DOI: 10.1161/circulationaha.119.045116] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Ischemic heart diseases are leading causes of death and reduced life quality worldwide. Although revascularization strategies significantly reduce mortality after acute myocardial infarction (MI), a large number of patients with MI develop chronic heart failure over time. We previously reported that a fragment of the extracellular matrix protein agrin promotes cardiac regeneration after MI in adult mice. METHODS To test the therapeutic potential of agrin in a preclinical porcine model, we performed ischemia-reperfusion injuries using balloon occlusion for 60 minutes followed by a 3-, 7-, or 28-day reperfusion period. RESULTS We demonstrated that local (antegrade) delivery of recombinant human agrin to the infarcted pig heart can target the affected regions in an efficient and clinically relevant manner. A single dose of recombinant human agrin improved heart function, infarct size, fibrosis, and adverse remodeling parameters 28 days after MI. Short-term MI experiments along with complementary murine studies revealed myocardial protection, improved angiogenesis, inflammatory suppression, and cell cycle reentry as agrin's mechanisms of action. CONCLUSIONS A single dose of agrin is capable of reducing ischemia-reperfusion injury and improving heart function, demonstrating that agrin could serve as a therapy for patients with acute MI and potentially heart failure.
Collapse
Affiliation(s)
- Andrea Baehr
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Kfir Baruch Umansky
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel (K.B.U., E.B., D.K., R.C.-R., E.T.)
| | - Elad Bassat
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel (K.B.U., E.B., D.K., R.C.-R., E.T.)
| | - Victoria Jurisch
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Katharina Klett
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Tarik Bozoglu
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Nadja Hornaschewitz
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Olga Solyanik
- Department of Radiology, Klinikum Großhadern (O.S., C.C.), LMU Munich, Germany
| | - David Kain
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel (K.B.U., E.B., D.K., R.C.-R., E.T.)
| | - Bartolo Ferraro
- Institute for Cardiovascular Prevention (B.F., O.S.), LMU Munich, Germany
| | - Renee Cohen-Rabi
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel (K.B.U., E.B., D.K., R.C.-R., E.T.)
| | - Markus Krane
- Department of Surgery, German Heart Center Munich, Germany (M.K.)
| | - Clemens Cyran
- Department of Radiology, Klinikum Großhadern (O.S., C.C.), LMU Munich, Germany
| | - Oliver Soehnlein
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.).,Institute for Cardiovascular Prevention (B.F., O.S.), LMU Munich, Germany
| | - Karl Ludwig Laugwitz
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Rabea Hinkel
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.).,Department of Laboratory Animal Science, Deutsches Primatenzentrum GmbH, Leibniz-Institut für Primatenforschung, Göttingen, Germany (R.H.)
| | - Christian Kupatt
- I Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar, Technical University Munich, Germany (A.B., V.J., K.K., T.B., N.H., K.L.L., R.H., C.K.).,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (A.B., V.J., K.K., T.B., N.H., B.F., O.S., K.L.L., R.H., C.K.)
| | - Eldad Tzahor
- The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel (K.B.U., E.B., D.K., R.C.-R., E.T.)
| |
Collapse
|
127
|
Two-variable nullcline analysis of ionic general equilibrium predicts calcium homeostasis in ventricular myocytes. PLoS Comput Biol 2020; 16:e1007572. [PMID: 32502205 PMCID: PMC7316341 DOI: 10.1371/journal.pcbi.1007572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/25/2020] [Accepted: 05/05/2020] [Indexed: 01/16/2023] Open
Abstract
Ventricular contraction is roughly proportional to the amount of calcium released from the Sarcoplasmic Reticulum (SR) during systole. While it is rather straightforward to measure calcium levels and contractibility under different physiological conditions, the complexity of calcium handling during systole and diastole has made the prediction of its release at steady state impossible. Here we approach the problem analyzing the evolution of intracellular and extracellular calcium fluxes during a single beat which is away from homeostatic balance. Using an in-silico subcellular model of rabbit ventricular myocyte, we show that the high dimensional nonlinear problem of finding the steady state can be reduced to a two-variable general equilibrium condition where pre-systolic calcium level in the cytosol and in the SR must fulfill simultaneously two different equalities. This renders calcium homeostasis as a problem that can be studied in terms of its equilibrium structure, leading to precise predictions of steady state from single-beat measurements. We show how changes in ion channels modify the general equilibrium, as shocks would do in general equilibrium macroeconomic models. This allows us to predict when an enhanced entrance of calcium in the cell reduces its contractibility and explain why SERCA gene therapy, a change in calcium handling to treat heart failure, might fail to improve contraction even when it successfully increases SERCA expression. Cardiomyocytes, upon voltage excitation, release calcium, which leads to cell contraction. However, under some pathological conditions, calcium handling is impaired. Recently, SERCA gene therapy, whose aim is to improve Ca2+ sequestration by the Sarcoplasmic Reticulum (SR), has failed to improve the prognosis of patients with Heart Failure. This, together with recent counterintuitive results in calcium handling, has highlighted the need for a framework to understand calcium homeostasis across species and pathologies. We show here that the proper framework is a general equilibrium approach of two independent variables. The development of this framework allows us to find a possible mechanism for the failure of SERCA gene therapy even when it manages to increase Ca SERCA expression.
Collapse
|
128
|
Jin G, Li W, Song F, Zhao J, Wang M, Liu Q, Li A, Huang G, Xu F. Fluorescent conjugated polymer nanovector for in vivo tracking and regulating the fate of stem cells for restoring infarcted myocardium. Acta Biomater 2020; 109:195-207. [PMID: 32294553 DOI: 10.1016/j.actbio.2020.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/24/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
Stem cell therapy holds great promise for cardiac regeneration. However, the lack of ability to control stem cell fate after in vivo transplantation greatly restricts its therapeutic outcomes. MicroRNA delivery has emerged as a powerful tool to control stem cell fate for enhanced cardiac regeneration. However, the clinical translation of therapy based on gene-transfected stem cells remains challenging, due to the unknown in vivo behaviors of stem cells. Here, we developed a nano-platform (i.e., PFBT@miR-1-Tat NPs) that can achieve triggered release of microRNA-1 to promote cardiac differentiation of mesenchymal stem cells (MSCs), and long-term tracking of transplanted MSCs through bright and ultra-stable fluorescence of conjugated polymer poly(9,9-dioctylfluorene-alt-benzothiadiazole) (PFBT). We found that PFBT@miR-1-Tat NP-treated MSCs significantly restored the infarcted myocardium by promoting stem cell cardiac differentiation and integration with the in situ cardiac tissues. Meanwhile, MSCs without gene delivery improved the infarcted heart functions mainly through a paracrine effect and blood vessel formation. The developed conjugated polymer nanovector should be a powerful tool for manipulating as well as revealing the fate of therapeutic cells in vivo, which is critical for optimizing the therapeutic route of gene and cell combined therapy and therefore for accelerating clinical translation. STATEMENT OF SIGNIFICANCE: The lack of controllability in stem cell fate and the unclear in vivo cellular behaviors restrict the therapeutic outcomes of stem cell therapy. Herein, we engineered fluorescent conjugated polymer nanoparticles as gene delivery nanovectors with controlled release and high intracellular delivery capability to harness the fate of mesenchymal stem cells (MSCs) in vivo, meanwhile to reveal the cellular mechanism of gene-treated stem cell therapy. As compared with only MSC treatment that improves infarcted myocardium functions through paracrine effect, treatment with conjugated polymer nanovector-treated MSCs significantly restored infarcted myocardium through enhancing MSC cardiac differentiation and integration with the in-situ cardiac tissues. These findings demonstrate that the conjugated polymer nanovector would be a powerful tool in optimizing gene and cell combined therapy.
Collapse
Affiliation(s)
- Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, PR China
| | - Wenfang Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, PR China
| | - Fan Song
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an 710032, PR China
| | - Jing Zhao
- Shaanxi Key Lab Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai North Road, Xi'an 710069, P. R. China
| | - Mengqi Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Qian Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, PR China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
129
|
Ge Z, Li A, McNamara J, Dos Remedios C, Lal S. Pathogenesis and pathophysiology of heart failure with reduced ejection fraction: translation to human studies. Heart Fail Rev 2020; 24:743-758. [PMID: 31209771 DOI: 10.1007/s10741-019-09806-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Heart failure represents the end result of different pathophysiologic processes, which culminate in functional impairment. Regardless of its aetiology, the presentation of heart failure usually involves symptoms of pump failure and congestion, which forms the basis for clinical diagnosis. Pathophysiologic descriptions of heart failure with reduced ejection fraction (HFrEF) are being established. Most commonly, HFrEF is centred on a reactive model where a significant initial insult leads to reduced cardiac output, further triggering a cascade of maladaptive processes. Predisposing factors include myocardial injury of any cause, chronically abnormal loading due to hypertension, valvular disease, or tachyarrhythmias. The pathophysiologic processes behind remodelling in heart failure are complex and reflect systemic neurohormonal activation, peripheral vascular effects and localised changes affecting the cardiac substrate. These abnormalities have been the subject of intense research. Much of the translational successes in HFrEF have come from targeting neurohormonal responses to reduced cardiac output, with blockade of the renin-angiotensin-aldosterone system (RAAS) and beta-adrenergic blockade being particularly fruitful. However, mortality and morbidity associated with heart failure remains high. Although systemic neurohormonal blockade slows disease progression, localised ventricular remodelling still adversely affects contractile function. Novel therapy targeted at improving cardiac contractile mechanics in HFrEF hold the promise of alleviating heart failure at its source, yet so far none has found success. Nevertheless, there are increasing calls for a proximal, 'cardiocentric' approach to therapy. In this review, we examine HFrEF therapy aimed at improving cardiac function with a focus on recent trials and emerging targets.
Collapse
Affiliation(s)
- Zijun Ge
- Sydney Medical School, University of Sydney, Camperdown, Australia
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Amy Li
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
- Department of Pharmacy and Biomedical Science, La Trobe University, Melbourne, Australia
| | - James McNamara
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Cris Dos Remedios
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Sean Lal
- Sydney Medical School, University of Sydney, Camperdown, Australia.
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
- Cardiac Research Laboratory, Discipline of Anatomy and Histology, University of Sydney, Anderson Stuart Building (F13), Camperdown, NSW, 2006, Australia.
| |
Collapse
|
130
|
Kaul U, Ray S, Prabhakar D, Kochar A, Sharma K, Hazra PK, Chandra S, Solanki DRB, Dutta AL, Kumar V, Rao MS, Oomman A, Dani S, Pinto B, Raghu TR. Consensus document: management of heart failure in type 2 diabetes mellitus. Heart Fail Rev 2020; 26:1037-1062. [PMID: 32447488 DOI: 10.1007/s10741-020-09955-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a known predisposing factor for heart failure (HF). The growing burden of these two conditions and their impact on health of the individual and on society in general needs urgent attention from the health care professionals. Availability of multiple treatment choices for managing T2DM and HF may make therapeutic decisions more complex for clinicians. Recent cardiovascular outcome trials of antidiabetic drugs have added very robust evidence to effectively manage subjects with this dual condition. This consensus statement provides the prevalence trends and the impact of this dual burden on patients. In addition, it concisely narrates the types of HF, the different treatment algorithms, and recommendations for physicians to comprehensively manage such patients.
Collapse
Affiliation(s)
- Upendra Kaul
- Batra Heart Centre and Dean Academics and Research of BHMRC, Batra Hospital & Medical Research Centre, 1, Tughlakabad Institutional Area, Mehrauli Badarpur Road, New Delhi, 110 062, India.
| | - Saumitra Ray
- Heart Clinic, Kolkata, West Bengal, 700019, India
| | - D Prabhakar
- Apollo First Med Hospitals, Chennai, 600 010, India
| | - Arun Kochar
- Fortis Hospital, Mohali, Punjab, 160062, India
| | - Kamal Sharma
- SAL Hospital & Medical Institute, Ahmedabad, Gujarat, 380054, India
| | | | - Subhash Chandra
- BLK Super Speciality Hospital, Pusa Road, Karol Bagh, New Delhi, 110005, India
| | | | - Anjan Lal Dutta
- Peerless Hospital, Pancha Sayar Rd, Sahid Smirity Colony, Pancha Sayar, 700094, Kolkata, West Bengal, India
| | - Viveka Kumar
- Cath Labs MSSH (East) Saket, Max Super Speciality Hospital, New Delhi, 110017, India
| | - M Srinivas Rao
- Care Hospitals, Road No 1, Banjara Hills, Hyderabad, 500034, India
| | - Abraham Oomman
- Apollo Hospitals Greams Road Chennai, Apollo Hospitals 21, Greams Lane, Off Greams Road, Chennai, 600 006, India
| | - Sameer Dani
- Apollo Hospitals, Plot No.1 A, Bhat GIDC Estate, Gandhinagar, Gujarat, 382428, India
| | - Brian Pinto
- Holy Family Hospital, Mumbai, 400 050, India
| | - T R Raghu
- Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore, 560 069, India
| |
Collapse
|
131
|
Abstract
While clinical gene therapy celebrates its first successes, with several products already approved for clinical use and several hundreds in the final stages of the clinical approval pipeline, there is not a single gene therapy approach that has worked for the heart. Here, we review the past experience gained in the several cardiac gene therapy clinical trials that had the goal of inducing therapeutic angiogenesis in the ischemic heart and in the attempts at modulating cardiac function in heart failure. Critical assessment of the results so far achieved indicates that the efficiency of cardiac gene delivery remains a major hurdle preventing success but also that improvements need to be sought in establishing more reliable large animal models, choosing more effective therapeutic genes, better designing clinical trials, and more deeply understanding cardiac biology. We also emphasize a few areas of cardiac gene therapy development that hold great promise for the future. In particular, the transition from gene addition studies using protein-coding cDNAs to the modulation of gene expression using small RNA therapeutics and the improvement of precise gene editing now pave the way to applications such as cardiac regeneration after myocardial infarction and gene correction for inherited cardiomyopathies that were unapproachable until a decade ago.
Collapse
Affiliation(s)
- Antonio Cannatà
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Hashim Ali
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| | - Gianfranco Sinagra
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Mauro Giacca
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| |
Collapse
|
132
|
Bachar-Wikstrom E, Curman P, Ahanian T, Leong IUS, Larsson H, Cederlöf M, Wikstrom JD. Darier disease is associated with heart failure: a cross-sectional case-control and population based study. Sci Rep 2020; 10:6886. [PMID: 32327688 PMCID: PMC7181854 DOI: 10.1038/s41598-020-63832-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/03/2020] [Indexed: 01/16/2023] Open
Abstract
Human data supporting a role for endoplasmic reticulum (ER) stress and calcium dyshomeostasis in heart disease is scarce. Darier disease (DD) is a hereditary skin disease caused by mutations in the ATP2A2 gene encoding the sarcoendoplasmic-reticulum Ca2+ ATPase isoform 2 (SERCA2), which causes calcium dyshomeostasis and ER stress. We hypothesized that DD patients would have an increased risk for common heart disease. We performed a cross-sectional case-control clinical study on 25 DD patients and 25 matched controls; and a population-based cohort study on 935 subjects with DD and matched comparison subjects. Main outcomes and measures were N-terminal pro-brain natriuretic peptide, ECG and heart diagnosis (myocardial infarction, heart failure and arrythmia). DD subjects showed normal clinical heart phenotype including heart failure markers and ECG. The risk for heart failure was 1.59 (1,16-2,19) times elevated in DD subjects, while no major differences were found in myocardial infarcation or arrhythmias. Risk for heart failure when corrected for cardivascular risk factors or alcohol misuse was 1.53 (1.11-2.11) and 1.58 (1,15-2,18) respectively. Notably, heart failure occurred several years earlier in DD patients as compared to controls. We conclude that DD patients show a disease specific increased risk of heart failure which should be taken into account in patient management. The observation also strenghtens the clinical evidence on the important role of SERCA2 in heart failure pathophysiology.
Collapse
Affiliation(s)
- Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Philip Curman
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Tara Ahanian
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Ivone U S Leong
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Cederlöf
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Norra Stationsgatan 69, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
133
|
Fransen P, Chen J, Vangheluwe P, Guns PJ. Contractile Behavior of Mouse Aorta Depends on SERCA2 Isoform Distribution: Effects of Replacing SERCA2a by SERCA2b. Front Physiol 2020; 11:282. [PMID: 32296344 PMCID: PMC7136392 DOI: 10.3389/fphys.2020.00282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/12/2020] [Indexed: 01/16/2023] Open
Abstract
The Sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) actively pumps Ca2+ into the sarco/endoplasmic reticulum, thereby regulating intracellular Ca2+ concentrations and associated physiological processes. Different SERCA isoforms have been described (SERCA1, 2, and 3) with SERCA2 playing a pivotal role in Ca2+ homeostasis in cardiovascular tissues. In the heart, SERCA2a is the dominant isoform and has been proposed as therapeutic target in patients with heart failure. In the vasculature, both SERCA2a and SERCA2b are expressed with SERCA2b being the predominant isoform. The physiological role of SERCA2a in the vasculature, however, remains incompletely understood. In the present study, we used gene-modified mice in which the alternative splicing of the SERCA2-encoding gene (Atp2a2), underlying the expression of SERCA2a, is prevented and SERCA2a is replaced by SERCA2b. The resulting SERCA2b/b mice provide a unique opportunity to investigate the specific contribution of SERCA2a versus SERCA2b to vascular physiology. Aortic segments of SERCA2b/b (SERCA2a-deficient) and SERCA2a/b (control) mice were mounted in organ baths to evaluate vascular reactivity. SERCA2b/b aortic rings displayed higher contractions induced by phenylephrine (1 μM). Surprisingly, the initial inositol-3-phosphate mediated phasic contraction showed a faster decay of force in SERCA2b/b mice, while the subsequent tonic contraction was larger in SERCA2b/b segments. Moreover, in the presence of the calcium channel blocker diltiazem (35 μM) SERCA2b/b aortic rings showed higher contractions compared to SERCA2a/b, suggesting that SERCA2a (deficiency) modulates the activity of non-selective cation channels. Additionally, in endothelial cell (EC)-denuded aortic segments, the SERCA-inhibitor cyclopiazonic acid (CPA) caused markedly larger contractions in SERCA2b/b mice, while the increases of cytosolic Ca2+ were similar in both strains. Hence, aortas of SERCA2b/b mice appear to have a stronger coupling of intracellular Ca2+ to contraction, which may be in agreement with the reported difference in intracellular localization of SERCA2a versus SERCA2b. Finally, EC-mediated relaxation by acetylcholine and ATP was assessed. Concentration-response-curves for ATP showed a higher sensitivity of aortic segments of SERCA2b/b mice, while no difference in potency between strains were observed for acetylcholine. In summary, despite the relative low expression of SERCA2a in the murine aorta, our results point toward a distinct role in vascular physiology.
Collapse
Affiliation(s)
- Paul Fransen
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical Sciences and Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Jialin Chen
- Laboratory of Cellular Transport Systems, KU Leuven, Leuven, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, KU Leuven, Leuven, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical Sciences and Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
134
|
Dong M, Yang Z, Fang H, Xiang J, Xu C, Zhou Y, Wu Q, Liu J. Aging Attenuates Cardiac Contractility and Affects Therapeutic Consequences for Myocardial Infarction. Aging Dis 2020; 11:365-376. [PMID: 32257547 PMCID: PMC7069457 DOI: 10.14336/ad.2019.0522] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac function of the human heart changes with age. The age-related change of systolic function is subtle under normal conditions, but abrupt under stress or in a pathogenesis state. Aging decreases the cardiac tolerance to stress and increases susceptibility to ischemia, which caused by aging-induced Ca2+ transient impairment and metabolic dysfunction. The changes of contractility proteins and the relative molecules are in a non-linear fashion. Specifically, the expression and activation of cMLCK increase first then fall during ischemia and reperfusion (I/R). This change is responsible for the nonmonotonic contractility alteration in I/R which the underlying mechanism is still unclear. Contractility recovery in I/R is also attenuated by age. The age-related change in cardiac contractility influences the therapeutic effect and intervention timepoint. For most cardiac ischemia therapies, the therapeutic result in the elderly is not identical to the young. Anti-aging treatment has the potential to prevent the development of ischemic injury and improves cardiac function. In this review we discuss the mechanism underlying the contractility changes in the aged heart and age-induced ischemic injury. The potential mechanism underlying the increased susceptibility to ischemic injury in advanced age is highlighted. Furthermore, we discuss the effect of age and the administration time for intervention in cardiac ischemia therapies.
Collapse
Affiliation(s)
- Ming Dong
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Ziyi Yang
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Hongcheng Fang
- Shenzhen Shajing Hospital, Affiliated of Guangzhou Medical University, Shenzhen, Guangdong, China
| | - Jiaqing Xiang
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Cong Xu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Yanqing Zhou
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Qianying Wu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Jie Liu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| |
Collapse
|
135
|
Gorski PA, Jang SP, Jeong D, Lee A, Lee P, Oh JG, Chepurko V, Yang DK, Kwak TH, Eom SH, Park ZY, Yoo YJ, Kim DH, Kook H, Sunagawa Y, Morimoto T, Hasegawa K, Sadoshima J, Vangheluwe P, Hajjar RJ, Park WJ, Kho C. Role of SIRT1 in Modulating Acetylation of the Sarco-Endoplasmic Reticulum Ca 2+-ATPase in Heart Failure. Circ Res 2020; 124:e63-e80. [PMID: 30786847 DOI: 10.1161/circresaha.118.313865] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE SERCA2a, sarco-endoplasmic reticulum Ca2+-ATPase, is a critical determinant of cardiac function. Reduced level and activity of SERCA2a are major features of heart failure. Accordingly, intensive efforts have been made to develop efficient modalities for SERCA2a activation. We showed that the activity of SERCA2a is enhanced by post-translational modification with SUMO1 (small ubiquitin-like modifier 1). However, the roles of other post-translational modifications on SERCA2a are still unknown. OBJECTIVE In this study, we aim to assess the role of lysine acetylation on SERCA2a function and determine whether inhibition of lysine acetylation can improve cardiac function in the setting of heart failure. METHODS AND RESULTS The acetylation of SERCA2a was significantly increased in failing hearts of humans, mice, and pigs, which is associated with the reduced level of SIRT1 (sirtuin 1), a class III histone deacetylase. Downregulation of SIRT1 increased the SERCA2a acetylation, which in turn led to SERCA2a dysfunction and cardiac defects at baseline. In contrast, pharmacological activation of SIRT1 reduced the SERCA2a acetylation, which was accompanied by recovery of SERCA2a function and cardiac defects in failing hearts. Lysine 492 (K492) was of critical importance for the regulation of SERCA2a activity via acetylation. Acetylation at K492 significantly reduced the SERCA2a activity, presumably through interfering with the binding of ATP to SERCA2a. In failing hearts, acetylation at K492 appeared to be mediated by p300 (histone acetyltransferase p300), a histone acetyltransferase. CONCLUSIONS These results indicate that acetylation/deacetylation at K492, which is regulated by SIRT1 and p300, is critical for the regulation of SERCA2a activity in hearts. Pharmacological activation of SIRT1 can restore SERCA2a activity through deacetylation at K492. These findings might provide a novel strategy for the treatment of heart failure.
Collapse
Affiliation(s)
- Przemek A Gorski
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Seung Pil Jang
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Dongtak Jeong
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Ahyoung Lee
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Philyoung Lee
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Jae Gyun Oh
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Vadim Chepurko
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Dong Kwon Yang
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | | | - Soo Hyun Eom
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Zee-Yong Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Yung Joon Yoo
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Do Han Kim
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Hyun Kook
- Basic Research Laboratory, Chonnam National University Medical School, Hwasun-gun, Jeollanam-do, Korea (H.K.)
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Japan (Y.S., T.M.)
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Japan (Y.S., T.M.)
| | - Koji Hasegawa
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, Japan (K.H.)
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Peter Vangheluwe
- Department of Cellular and Molecular Medicine, KU Leuven, Belgium (P.V.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Korea (S.P.J., D.K.Y., S.H.E., Z.-Y.P., Y.J.Y., D.H.K., W.J.P.)
| | - Changwon Kho
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (P.A.G., D.J., A.L., P.L., J.G.O., V.C., R.J.H., C.K.)
| |
Collapse
|
136
|
Nyns ECA, Poelma RH, Volkers L, Plomp JJ, Bart CI, Kip AM, van Brakel TJ, Zeppenfeld K, Schalij MJ, Zhang GQ, de Vries AAF, Pijnappels DA. An automated hybrid bioelectronic system for autogenous restoration of sinus rhythm in atrial fibrillation. Sci Transl Med 2020; 11:11/481/eaau6447. [PMID: 30814339 DOI: 10.1126/scitranslmed.aau6447] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/05/2018] [Accepted: 01/17/2019] [Indexed: 11/02/2022]
Abstract
Because of suboptimal therapeutic strategies, restoration of sinus rhythm in symptomatic atrial fibrillation (AF) often requires in-hospital delivery of high-voltage shocks, thereby precluding ambulatory AF termination. Continuous, rapid restoration of sinus rhythm is desired given the recurring and progressive nature of AF. Here, we present an automated hybrid bioelectronic system for shock-free termination of AF that enables the heart to act as an electric current generator for autogenous restoration of sinus rhythm. We show that local, right atrial delivery of adenoassociated virus vectors encoding a light-gated depolarizing ion channel results in efficient and spatially confined transgene expression. Activation of an implanted intrathoracic light-emitting diode device allows for termination of AF by illuminating part of the atria. Combining this newly obtained antiarrhythmic effector function of the heart with the arrhythmia detector function of a machine-based cardiac rhythm monitor in the closed chest of adult rats allowed automated and rapid arrhythmia detection and termination in a safe, effective, repetitive, yet shock-free manner. These findings hold translational potential for the development of shock-free antiarrhythmic device therapy for ambulatory treatment of AF.
Collapse
Affiliation(s)
- Emile C A Nyns
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - René H Poelma
- Department of Microelectronics, Delft University of Technology, 2628 CD, Delft, Netherlands
| | - Linda Volkers
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Jaap J Plomp
- Department of Neurology and Neurophysiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Cindy I Bart
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Annemarie M Kip
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Thomas J van Brakel
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Katja Zeppenfeld
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Martin J Schalij
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Guo Qi Zhang
- Department of Microelectronics, Delft University of Technology, 2628 CD, Delft, Netherlands
| | - Antoine A F de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands.
| |
Collapse
|
137
|
Law ML, Cohen H, Martin AA, Angulski ABB, Metzger JM. Dysregulation of Calcium Handling in Duchenne Muscular Dystrophy-Associated Dilated Cardiomyopathy: Mechanisms and Experimental Therapeutic Strategies. J Clin Med 2020; 9:jcm9020520. [PMID: 32075145 PMCID: PMC7074327 DOI: 10.3390/jcm9020520] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is an X-linked recessive disease resulting in the loss of dystrophin, a key cytoskeletal protein in the dystrophin-glycoprotein complex. Dystrophin connects the extracellular matrix with the cytoskeleton and stabilizes the sarcolemma. Cardiomyopathy is prominent in adolescents and young adults with DMD, manifesting as dilated cardiomyopathy (DCM) in the later stages of disease. Sarcolemmal instability, leading to calcium mishandling and overload in the cardiac myocyte, is a key mechanistic contributor to muscle cell death, fibrosis, and diminished cardiac contractile function in DMD patients. Current therapies for DMD cardiomyopathy can slow disease progression, but they do not directly target aberrant calcium handling and calcium overload. Experimental therapeutic targets that address calcium mishandling and overload include membrane stabilization, inhibition of stretch-activated channels, ryanodine receptor stabilization, and augmentation of calcium cycling via modulation of the Serca2a/phospholamban (PLN) complex or cytosolic calcium buffering. This paper addresses what is known about the mechanistic basis of calcium mishandling in DCM, with a focus on DMD cardiomyopathy. Additionally, we discuss currently utilized therapies for DMD cardiomyopathy, and review experimental therapeutic strategies targeting the calcium handling defects in DCM and DMD cardiomyopathy.
Collapse
Affiliation(s)
- Michelle L. Law
- Department of Family and Consumer Sciences, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA;
| | - Houda Cohen
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Ashley A. Martin
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Addeli Bez Batti Angulski
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
- Correspondence: ; Tel.: +1-612-625-5902; Fax: +1-612-625-5149
| |
Collapse
|
138
|
Toya T, Ito K, Kagami K, Osaki A, Sato A, Kimura T, Horii S, Yasuda R, Namba T, Ido Y, Nagatomo Y, Hayashi K, Masaki N, Yada H, Adachi T. Impact of oxidative posttranslational modifications of SERCA2 on heart failure exacerbation in young patients with non-ischemic cardiomyopathy: A pilot study. IJC HEART & VASCULATURE 2020; 26:100437. [PMID: 31763443 PMCID: PMC6864308 DOI: 10.1016/j.ijcha.2019.100437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Oxidative posttranslational modifications (OPTM) impair the function of Sarcoplasmic/endoplasmic reticulum (SR) calcium (Ca2+) ATPase (SERCA) 2 and trigger cytosolic Ca2+ dysregulation. We investigated the extent of OPTM of SERCA2 in patients with non-ischemic cardiomyopathy (NICM). METHODS AND RESULTS Endomyocardial biopsy (EMB) was obtained in 40 consecutive patients with NICM. Total expression and OPTM of SERCA2, including sulfonylation at cysteine-674 (S-SERCA2) and nitration at tyrosine-294/295 (N-SERCA2), were examined by immunohistochemical analysis. S-SERCA2 increased in the presence of late gadolinium enhancement on cardiac magnetic resonance imaging. S-SERCA2/SERCA2 and N-SERCA2/SERCA2 correlated with cardiac fibrosis evaluated by Masson's trichrome staining of EMB. SERCA2 expression modestly increased in parallel with an upward trend in OPTM of SERCA2 with aging. This tendency became prominent only in patients aged >65 years. OPTM of SERCA2 positively correlated with brain natriuretic peptide (BNP) values only in patients aged ≤65 years. Composite major adverse cardiac events (MACE) increased more in the high OPTM group of younger patients; however, MACE-free survival was similar irrespective of the extent of OPTM in older patients. CONCLUSIONS OPTM of SERCA2 correlate with myocardial fibrosis in NICM. In younger patients, OPTM of SERCA2 correlate with elevated BNP and increased composite MACE.
Collapse
Affiliation(s)
- Takumi Toya
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Kei Ito
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Kazuki Kagami
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Ayumu Osaki
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Atsushi Sato
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Toyokazu Kimura
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Shunpei Horii
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Risako Yasuda
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Takayuki Namba
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Yasuo Ido
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Yuji Nagatomo
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Katsumi Hayashi
- Department of Radiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Nobuyuki Masaki
- Department of Intensive Care Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Hirotaka Yada
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| | - Takeshi Adachi
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan1
| |
Collapse
|
139
|
Zhang J, Hu G, Yang S. Intracoronary Sarcoplasmic Reticulum Calcium-ATPase Gene Therapy in Advanced Heart Failure Patients with reduced Ejection Fraction: A Prospective Cohort Study. Clinics (Sao Paulo) 2020; 75:e1530. [PMID: 32187281 PMCID: PMC7061318 DOI: 10.6061/clinics/2020/e1530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 01/07/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Heart failure is a progressive and debilitating disease. Intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy may improve the function of cardiac muscle cells. This study aimed to test the hypothesis that intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy can improve outcomes and reduce the number of recurrent and terminal events in advanced heart failure patients with reduced ejection fraction. METHODS A total of 768 heart failure patients with reduced ejection fraction and New York Heart Association classification II to IV were included in this prospective cohort study. Patients either underwent intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy (CA group, n=384) or received oral placebo (PA group; n=384). Data regarding recurrent and terminal event(s), treatment-emergent adverse effects, and outcome measures were collected and analyzed. RESULTS After a follow-up period of 18 months, intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy reduced the number of hospital admissions (p=0.001), ambulatory treatments (p=0.0004), and deaths (p=0.024). Additionally, intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy improved the left ventricular ejection fraction (p<0.0001) and Kansas City Cardiomyopathy Questionnaire score (p<0.0001). The number of recurrent and terminal events/patients were higher in the PA group than in the CA group after the follow-up period of 18 months (p=0.015). The effect of the intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy was independent of the confounding variables. No new arrhythmias were reported in the CA group. CONCLUSIONS Intracoronary sarcoplasmic reticulum calcium-ATPase gene therapy reduces the number of recurrent and terminal events and improves the clinical course of advanced heart failure patients with reduced ejection fraction.
Collapse
Affiliation(s)
- Jianfeng Zhang
- Cardio-Pulmonary Rehabilitation Inpatient Area, The Second Rehabilitation Hospital of Shanghai, Shanghai 200431, China
- *Corresponding author. E-mail:
| | - Guojin Hu
- Cardio-Pulmonary Rehabilitation Inpatient Area, The Second Rehabilitation Hospital of Shanghai, Shanghai 200431, China
| | - Shengyong Yang
- Cardio-Pulmonary Rehabilitation Inpatient Area, The Second Rehabilitation Hospital of Shanghai, Shanghai 200431, China
| |
Collapse
|
140
|
Bisserier M, Hadri L. Lung-targeted SERCA2a Gene Therapy: From Discovery to Therapeutic Application in Bleomycin-Induced Pulmonary Fibrosis. JOURNAL OF CELLULAR IMMUNOLOGY 2020; 2:149-156. [PMID: 32587955 PMCID: PMC7316402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by an accumulation of scar tissue within the lungs and the common presence of usual interstitial pneumonia. Unfortunately, only a few FDA-approved therapeutic options are currently available for the treatment of IPF and IPF remains associated with poor prognosis. Therefore, the identification of new pharmacological targets and strategies are critical for the treatment of IPF. This commentary aims to further discuss the role of sarcoplasmic reticulum Ca2+-ATPase 2a and its downstream signaling in IPF. Finally, this commentary offers new insights and perspectives regarding the therapeutic potential of AAV-mediated SERCA2A gene therapy as an emerging therapy for respiratory diseases.
Collapse
Affiliation(s)
| | - Lahouaria Hadri
- Correspondence should be addressed to Lahouaria Hadri, PhD, Cardiovascular Research Center, Box 1030, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, New York, NY 10029;
| |
Collapse
|
141
|
Witman N, Zhou C, Grote Beverborg N, Sahara M, Chien KR. Cardiac progenitors and paracrine mediators in cardiogenesis and heart regeneration. Semin Cell Dev Biol 2019; 100:29-51. [PMID: 31862220 DOI: 10.1016/j.semcdb.2019.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/13/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
Abstract
The mammalian hearts have the least regenerative capabilities among tissues and organs. As such, heart regeneration has been and continues to be the ultimate goal in the treatment against acquired and congenital heart diseases. Uncovering such a long-awaited therapy is still extremely challenging in the current settings. On the other hand, this desperate need for effective heart regeneration has developed various forms of modern biotechnologies in recent years. These involve the transplantation of pluripotent stem cell-derived cardiac progenitors or cardiomyocytes generated in vitro and novel biochemical molecules along with tissue engineering platforms. Such newly generated technologies and approaches have been shown to effectively proliferate cardiomyocytes and promote heart repair in the diseased settings, albeit mainly preclinically. These novel tools and medicines give somehow credence to breaking down the barriers associated with re-building heart muscle. However, in order to maximize efficacy and achieve better clinical outcomes through these cell-based and/or cell-free therapies, it is crucial to understand more deeply the developmental cellular hierarchies/paths and molecular mechanisms in normal or pathological cardiogenesis. Indeed, the morphogenetic process of mammalian cardiac development is highly complex and spatiotemporally regulated by various types of cardiac progenitors and their paracrine mediators. Here we discuss the most recent knowledge and findings in cardiac progenitor cell biology and the major cardiogenic paracrine mediators in the settings of cardiogenesis, congenital heart disease, and heart regeneration.
Collapse
Affiliation(s)
- Nevin Witman
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Chikai Zhou
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Niels Grote Beverborg
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Surgery, Yale University School of Medicine, CT, USA.
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
142
|
Abi-Gerges N, Miller PE, Ghetti A. Human Heart Cardiomyocytes in Drug Discovery and Research: New Opportunities in Translational Sciences. Curr Pharm Biotechnol 2019; 21:787-806. [PMID: 31820682 DOI: 10.2174/1389201021666191210142023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
In preclinical drug development, accurate prediction of drug effects on the human heart is critically important, whether in the context of cardiovascular safety or for the purpose of modulating cardiac function to treat heart disease. Current strategies have significant limitations, whereby, cardiotoxic drugs can escape detection or potential life-saving therapies are abandoned due to false positive toxicity signals. Thus, new and more reliable translational approaches are urgently needed to help accelerate the rate of new therapy development. Renewed efforts in the recovery of human donor hearts for research and in cardiomyocyte isolation methods, are providing new opportunities for preclinical studies in adult primary cardiomyocytes. These cells exhibit the native physiological and pharmacological properties, overcoming the limitations presented by artificial cellular models, animal models and have great potential for providing an excellent tool for preclinical drug testing. Adult human primary cardiomyocytes have already shown utility in assessing drug-induced cardiotoxicity risk and helping in the identification of new treatments for cardiac diseases, such as heart failure and atrial fibrillation. Finally, strategies with actionable decision-making trees that rely on data derived from adult human primary cardiomyocytes will provide the holistic insights necessary to accurately predict human heart effects of drugs.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| |
Collapse
|
143
|
Trivedi A, Hoffman J, Arora R. Gene therapy for atrial fibrillation - How close to clinical implementation? Int J Cardiol 2019; 296:177-183. [PMID: 31439427 PMCID: PMC6907402 DOI: 10.1016/j.ijcard.2019.07.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 07/03/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022]
Abstract
In this review we examine the current state of gene therapy for the treatment of cardiac arrhythmias. We describe advances and challenges in successfully creating and incorporating gene vectors into the myocardium. After summarizing the current scientific research in gene transfer technology we then focus on the most promising areas of gene therapy, the treatment of atrial fibrillation and ventricular tachyarrhythmias. We review the scientific literature to determine how gene therapy could potentially be used to treat patients with cardiac arrhythmias.
Collapse
Affiliation(s)
- Amar Trivedi
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University - Feinberg School of Medicine, United States of America
| | - Jacob Hoffman
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University - Feinberg School of Medicine, United States of America
| | - Rishi Arora
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University - Feinberg School of Medicine, United States of America.
| |
Collapse
|
144
|
Dikolayev V, Tuganbekov T, Nikolaev VO. Visualizing Cyclic Adenosine Monophosphate in Cardiac Microdomains Involved in Ion Homeostasis. Front Physiol 2019; 10:1406. [PMID: 31849691 PMCID: PMC6888371 DOI: 10.3389/fphys.2019.01406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022] Open
Abstract
3′,5′-Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates function of proteins involved in ion homeostasis and cardiac excitation-contraction coupling. Over the last decade, it has been increasingly appreciated that cAMP conveys its numerous effects by acting in discrete subcellular compartments or “microdomains.” In this mini review, we describe how such localized signals can be visualized in living cardiomyocytes to better understand cardiac physiology and disease. Special focus is made on targeted biosensors that can be used to resolve second messenger signals within nanometers of cardiac ion channels and transporters. Potential directions for future research and the translational importance of cAMP compartmentalization are discussed.
Collapse
Affiliation(s)
- Vladimir Dikolayev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Surgical Diseases, Astana Medical University, Nur-Sultan, Kazakhstan
| | - Turlybek Tuganbekov
- Department of Surgical Diseases, Astana Medical University, Nur-Sultan, Kazakhstan
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Hamburg, Germany
| |
Collapse
|
145
|
Stroik DR, Ceholski DK, Bidwell PA, Mleczko J, Thanel PF, Kamdar F, Autry JM, Cornea RL, Thomas DD. Viral expression of a SERCA2a-activating PLB mutant improves calcium cycling and synchronicity in dilated cardiomyopathic hiPSC-CMs. J Mol Cell Cardiol 2019; 138:59-65. [PMID: 31751570 DOI: 10.1016/j.yjmcc.2019.11.147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022]
Abstract
There is increasing momentum toward the development of gene therapy for heart failure (HF) that is defined by impaired calcium (Ca2+) transport and reduced contractility. We have used FRET (fluorescence resonance energy transfer) between fluorescently-tagged SERCA2a (the cardiac Ca2+ pump) and PLB (phospholamban, ventricular peptide inhibitor of SERCA) to test directly the effectiveness of loss-of-inhibition/gain-of-binding (LOI/GOB) PLB mutants (PLBM) that were engineered to compete with the binding of inhibitory wild-type PLB (PLBWT). Our therapeutic strategy is to relieve PLBWT inhibition of SERCA2a by using the reserve adrenergic capacity mediated by PLB to enhance cardiac contractility. Using a FRET assay, we determined that the combination of a LOI PLB mutation (L31A) and a GOB PLB mutation (I40A) results in a novel engineered LOI/GOB PLBM (L31A/I40A) that effectively competes with PLBWT binding to cardiac SERCA2a in HEK293-6E cells. We demonstrated that co-expression of PLBM enhances SERCA Ca-ATPase activity by increasing enzyme Ca2+ affinity (1/KCa) in PLBWT-inhibited HEK293 cell homogenates. For an initial assessment of PLBM physiological effectiveness, we used human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) from a healthy individual. In this system, we observed that adeno-associated virus 2 (rAAV2)-driven expression of PLBM enhances the amplitude of SR Ca2+ release and the rate of SR Ca2+ re-uptake. To assess therapeutic potential, we used a hiPSC-CM model of dilated cardiomyopathy (DCM) containing PLB mutation R14del, where we observed that rAAV2-driven expression of PLBM rescues arrhythmic Ca2+ transients and alleviates decreased Ca2+ transport. Thus, we propose that PLBM transgene expression is a promising gene therapy strategy that directly targets the underlying pathophysiology of abnormal Ca2+ transport and thus contractility in underlying systolic heart failure.
Collapse
Affiliation(s)
- Daniel R Stroik
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Delaine K Ceholski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, United States of America
| | - Philip A Bidwell
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America; Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Justyna Mleczko
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, United States of America
| | - Paul F Thanel
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Forum Kamdar
- Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Joseph M Autry
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
146
|
Bistola V, Arfaras-Melainis A, Polyzogopoulou E, Ikonomidis I, Parissis J. Inotropes in Acute Heart Failure: From Guidelines to Practical Use: Therapeutic Options and Clinical Practice. Card Fail Rev 2019; 5:133-139. [PMID: 31768269 PMCID: PMC6848944 DOI: 10.15420/cfr.2019.11.2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 09/11/2019] [Indexed: 01/10/2023] Open
Abstract
Inotropes are pharmacological agents that are indicated for the treatment of patients presenting with acute heart failure (AHF) with concomitant hypoperfusion due to decreased cardiac output. They are usually administered for a short period during the initial management of AHF until haemodynamic stabilisation and restoration of peripheral perfusion occur. They can be used for longer periods to support patients as a bridge to a more definite treatment, such as transplant of left ventricular assist devices, or as part of a palliative care regimen. The currently available inotropic agents in clinical practice fall into three main categories: beta-agonists, phosphodiesterase III inhibitors and calcium sensitisers. However, due to the well-documented potential for adverse events and their association with increased long-term mortality, physicians should be aware of the indications and dosing strategies suitable for different types of patients. Novel inotropes that use alternative intracellular pathways are under investigation, in an effort to minimise the drawbacks that conventional inotropes exhibit.
Collapse
Affiliation(s)
- Vasiliki Bistola
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Athens, Greece
| | - Angelos Arfaras-Melainis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Athens, Greece
| | - Eftihia Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, National and Kapodistrian University of Athens Athens, Greece
| | - Ignatios Ikonomidis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Athens, Greece
| | - John Parissis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Athens, Greece
| |
Collapse
|
147
|
Upadhya B, Haykowsky MJ, Kitzman DW. Therapy for heart failure with preserved ejection fraction: current status, unique challenges, and future directions. Heart Fail Rev 2019; 23:609-629. [PMID: 29876843 DOI: 10.1007/s10741-018-9714-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is the most common form of HF. Among elderly women, HFpEF comprises more than 80% of incident HF cases. Adverse outcomes-exercise intolerance, poor quality of life, frequent hospitalizations, and reduced survival-approach those of classic HF with reduced EF (HFrEF). However, despite its importance, our understanding of the pathophysiology of HFpEF is incomplete, and despite intensive efforts, optimal therapy remains uncertain, as most trials to date have been negative. This is in stark contrast to management of HFrEF, where dozens of positive trials have established a broad array of effective, guidelines-based therapies that definitively improve a range of clinically meaningful outcomes. In addition to providing an overview of current management status, we examine evolving data that may help explain this paradox, overcome past challenges, provide a roadmap for future success, and that underpin a wave of new trials that will test novel approaches based on these insights.
Collapse
Affiliation(s)
- Bharathi Upadhya
- Cardiovascular Medicine Section, Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157-1045, USA
| | - Mark J Haykowsky
- College of Nursing and Health Innovation, University of Texas Arlington, Arlington, TX, USA
| | - Dalane W Kitzman
- Cardiovascular Medicine Section, Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157-1045, USA.
| |
Collapse
|
148
|
Belletti A, Landoni G, Lomivorotov VV, Oriani A, Ajello S. Adrenergic Downregulation in Critical Care: Molecular Mechanisms and Therapeutic Evidence. J Cardiothorac Vasc Anesth 2019; 34:1023-1041. [PMID: 31839459 DOI: 10.1053/j.jvca.2019.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/09/2019] [Accepted: 10/10/2019] [Indexed: 02/08/2023]
Abstract
Catecholamines remain the mainstay of therapy for acute cardiovascular dysfunction. However, adrenergic receptors quickly undergo desensitization and downregulation after prolonged stimulation. Moreover, prolonged exposure to high circulating catecholamines levels is associated with several adverse effects on different organ systems. Unfortunately, in critically ill patients, adrenergic downregulation translates into progressive reduction of cardiovascular response to exogenous catecholamine administration, leading to refractory shock. Accordingly, there has been a growing interest in recent years toward use of noncatecholaminergic inotropes and vasopressors. Several studies investigating a wide variety of catecholamine-sparing strategies (eg, levosimendan, vasopressin, β-blockers, steroids, and use of mechanical circulatory support) have been published recently. Use of these agents was associated with improvement in hemodynamics and decreased catecholamine use but without a clear beneficial effect on major clinical outcomes. Accordingly, additional research is needed to define the optimal management of catecholamine-resistant shock.
Collapse
Affiliation(s)
- Alessandro Belletti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Vladimir V Lomivorotov
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia; Novosibirsk State University, Novosibirsk, Russia
| | - Alessandro Oriani
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Ajello
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
149
|
Oh JG, Watanabe S, Lee A, Gorski PA, Lee P, Jeong D, Liang L, Liang Y, Baccarini A, Sahoo S, Brown BD, Hajjar RJ, Kho C. miR-146a Suppresses SUMO1 Expression and Induces Cardiac Dysfunction in Maladaptive Hypertrophy. Circ Res 2019; 123:673-685. [PMID: 30355233 DOI: 10.1161/circresaha.118.312751] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RATIONALE Abnormal SUMOylation has emerged as a characteristic of heart failure (HF) pathology. Previously, we found reduced SUMO1 (small ubiquitin-like modifier 1) expression and SERCA2a (sarcoplasmic reticulum Ca2+-ATPase) SUMOylation in human and animal HF models. SUMO1 gene delivery or small molecule activation of SUMOylation restored SERCA2a SUMOylation and cardiac function in HF models. Despite the critical role of SUMO1 in HF, the regulatory mechanisms underlying SUMO1 expression are largely unknown. OBJECTIVE To examine miR-146a-mediated SUMO1 regulation and its consequent effects on cardiac morphology and function. METHODS AND RESULTS In this study, miR-146a was identified as a SUMO1-targeting microRNA in the heart. A strong correlation was observed between miR-146a and SUMO1 expression in failing mouse and human hearts. miR-146a was manipulated in cardiomyocytes through AAV9 (adeno-associated virus serotype 9)-mediated gene delivery, and cardiac morphology and function were analyzed by echocardiography and hemodynamics. Overexpression of miR-146a reduced SUMO1 expression, SERCA2a SUMOylation, and cardiac contractility in vitro and in vivo. The effects of miR-146a inhibition on HF pathophysiology were examined by transducing a tough decoy of miR-146a into mice subjected to transverse aortic constriction. miR-146a inhibition improved cardiac contractile function and normalized SUMO1 expression. The regulatory mechanisms of miR-146a upregulation were elucidated by examining the major miR-146a-producing cell types and transfer mechanisms. Notably, transdifferentiation of fibroblasts triggered miR-146a overexpression and secretion through extracellular vesicles, and the extracellular vesicle-associated miR-146a transfer was identified as the causative mechanism of miR-146a upregulation in failing cardiomyocytes. Finally, extracellular vesicles isolated from failing hearts were shown to contain high levels of miR-146a and exerted negative effects on the SUMO1/SERCA2a signaling axis and hence cardiomyocyte contractility. CONCLUSIONS Taken together, our results show that miR-146a is a novel regulator of the SUMOylation machinery in the heart, which can be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Jae Gyun Oh
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Shin Watanabe
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Ahyoung Lee
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Przemek A Gorski
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Philyoung Lee
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Dongtak Jeong
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Lifan Liang
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Yaxuan Liang
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Alessia Baccarini
- Department of Genetics and Genomic Sciences (A.B., B.D.B.), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York
| | - Susmita Sahoo
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Brian D Brown
- Department of Genetics and Genomic Sciences (A.B., B.D.B.), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York
| | - Roger J Hajjar
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Changwon Kho
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| |
Collapse
|
150
|
Abstract
In the past 10 years, there has been tremendous progress made in the field of gene therapy. Effective treatments of Leber congenital amaurosis, hemophilia, and spinal muscular atrophy have been largely based on the efficiency and safety of adeno-associated vectors. Myocardial gene therapy has been tested in patients with heart failure using adeno-associated vectors with no safety concerns but lacking clinical improvements. Cardiac gene therapy is adapting to the new developments in vectors, delivery systems, targets, and clinical end points and is poised for success in the near future.
Collapse
Affiliation(s)
- Kiyotake Ishikawa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thomas Weber
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|