1
|
Kehr D, Ritterhoff J, Glaser M, Jarosch L, Salazar RE, Spaich K, Varadi K, Birkenstock J, Egger M, Gao E, Koch WJ, Sauter M, Freichel M, Katus HA, Frey N, Jungmann A, Busch C, Mather PJ, Ruhparwar A, Busch M, Völkers M, Wade RC, Most P. S100A1ct: A Synthetic Peptide Derived From S100A1 Protein Improves Cardiac Performance and Survival in Preclinical Heart Failure Models. Circulation 2025; 151:548-565. [PMID: 39569500 PMCID: PMC11850016 DOI: 10.1161/circulationaha.123.066961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND The EF-hand Ca2+ sensor protein S100A1 has been identified as a molecular regulator and enhancer of cardiac performance. The ability of S100A1 to recognize and modulate the activity of targets such as SERCA2a (sarcoplasmic reticulum Ca2+ ATPase) and RyR2 (ryanodine receptor 2) in cardiomyocytes has mostly been ascribed to its hydrophobic C-terminal α-helix (residues 75-94). We hypothesized that a synthetic peptide consisting of residues 75 through 94 of S100A1 and an N-terminal solubilization tag (S100A1ct) could mimic the performance-enhancing effects of S100A1 and may be suitable as a peptide therapeutic to improve the function of diseased hearts. METHODS We applied an integrative translational research pipeline ranging from in silico computational molecular modeling and in vitro biochemical molecular assays as well as isolated rodent and human cardiomyocyte performance assessments to in vivo safety and efficacy studies in small and large animal cardiac disease models. RESULTS We characterize S100A1ct as a cell-penetrating peptide with positive inotropic and antiarrhythmic properties in normal and failing myocardium in vitro and in vivo. This activity translates into improved contractile performance and survival in preclinical heart failure models with reduced ejection fraction after S100A1ct systemic administration. S100A1ct exerts a fast and sustained dose-dependent enhancement of cardiomyocyte Ca2+ cycling and prevents β-adrenergic receptor-triggered Ca2+ imbalances by targeting SERCA2a and RyR2 activity. In line with the S100A1ct-mediated enhancement of SERCA2a activity, modeling suggests an interaction of the peptide with the transmembrane segments of the sarcoplasmic Ca2+ pump. Incorporation of a cardiomyocyte-targeting peptide tag into S100A1ct (cor-S100A1ct) further enhanced its biological and therapeutic potency in vitro and in vivo. CONCLUSIONS S100A1ct is a promising lead for the development of novel peptide-based therapeutics against heart failure with reduced ejection fraction.
Collapse
Affiliation(s)
- Dorothea Kehr
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Julia Ritterhoff
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Manuel Glaser
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany (M.G., R.C.W.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Lukas Jarosch
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
| | - Rafael E. Salazar
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
| | - Kristin Spaich
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
| | - Karl Varadi
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Jennifer Birkenstock
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
| | - Michael Egger
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G.)
| | - Walter J. Koch
- Division of Cardiovascular and Thoracic Surgery, Duke University, Durham, NC (W.J.K.)
| | - Max Sauter
- Department of Clinical Pharmacology and Pharmacoepidemiology (M.S.), Heidelberg University Hospital (UKHD), Germany
| | - Marc Freichel
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Department of Pharmacology, Heidelberg Medical Faculty, Germany (M.F.)
| | - Hugo A. Katus
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Andreas Jungmann
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Cornelius Busch
- Department of Anesthesiology (C.B.), Heidelberg University Hospital (UKHD), Germany
| | - Paul J. Mather
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (P.J.M.)
| | - Arjang Ruhparwar
- Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany (A.R.)
| | - Martin Busch
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Mirko Völkers
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Rebecca C. Wade
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany (M.G., R.C.W.)
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg, Germany (R.C.W.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Patrick Most
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
- Center for Translational Medicine, Jefferson University, Philadelphia, PA (P.M.)
| |
Collapse
|
2
|
Alhajri N, Rustom M, Adegbile A, Ahmed W, Kilidar S, Afify N. Deciphering the Basis of Molecular Biology of Selected Cardiovascular Diseases: A View on Network Medicine. Int J Mol Sci 2022; 23:ijms231911421. [PMID: 36232723 PMCID: PMC9569471 DOI: 10.3390/ijms231911421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death across the world. For decades, researchers have been studying the causes of cardiovascular disease, yet many of them remain undiscovered or poorly understood. Network medicine is a recently expanding, integrative field that attempts to elucidate this issue by conceiving of disease as the result of disruptive links between multiple interconnected biological components. Still in its nascent stages, this revolutionary application of network science facilitated a number of important discoveries in complex disease mechanisms. As methodologies become more advanced, network medicine harbors the potential to expound on the molecular and genetic complexities of disease to differentiate how these intricacies govern disease manifestations, prognosis, and therapy. This is of paramount importance for confronting the incredible challenges of current and future cardiovascular disease research. In this review, we summarize the principal molecular and genetic mechanisms of common cardiac pathophysiologies as well as discuss the existing knowledge on therapeutic strategies to prevent, halt, or reverse these pathologies.
Collapse
Affiliation(s)
- Noora Alhajri
- Department of Internal Medicine, Cleveland Clinic Abu Dhabi (CCAD), Abu Dhabi P.O. Box 112412, United Arab Emirates
- Correspondence:
| | - Mohammad Rustom
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Adedayo Adegbile
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Weshah Ahmed
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Salsabeel Kilidar
- Department of Emergency Medicine, Sheikh Shakhbout Medical City SSMC, Abu Dhabi P.O. Box 11001, United Arab Emirates
| | - Nariman Afify
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
3
|
Wang J, Shi Q, Wang Y, Dawson LW, Ciampa G, Zhao W, Zhang G, Chen B, Weiss RM, Grueter CE, Hall DD, Song LS. Gene Therapy With the N-Terminus of Junctophilin-2 Improves Heart Failure in Mice. Circ Res 2022; 130:1306-1317. [PMID: 35317607 PMCID: PMC9050933 DOI: 10.1161/circresaha.121.320680] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/11/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Transcriptional remodeling is known to contribute to heart failure (HF). Targeting stress-dependent gene expression mechanisms may represent a clinically relevant gene therapy option. We recently uncovered a salutary mechanism in the heart whereby JP2 (junctophilin-2), an essential component of the excitation-contraction coupling apparatus, is site-specifically cleaved and releases an N-terminal fragment (JP2NT [N-terminal fragment of JP2]) that translocates into the nucleus and functions as a transcriptional repressor of HF-related genes. This study aims to determine whether JP2NT can be leveraged by gene therapy techniques for attenuating HF progression in a preclinical pressure overload model. METHODS We intraventricularly injected adeno-associated virus (AAV) (2/9) vectors expressing eGFP (enhanced green fluorescent protein), JP2NT, or DNA-binding deficient JP2NT (JP2NTΔbNLS/ARR) into neonatal mice and induced cardiac stress by transaortic constriction (TAC) 9 weeks later. We also treated mice with established moderate HF from TAC stress with either AAV-JP2NT or AAV-eGFP. RNA-sequencing analysis was used to reveal changes in hypertrophic and HF-related gene transcription by JP2NT gene therapy after TAC. Echocardiography, confocal imaging, and histology were performed to evaluate heart function and pathological myocardial remodeling following stress. RESULTS Mice preinjected with AAV-JP2NT exhibited ameliorated cardiac remodeling following TAC. The JP2NT DNA-binding domain is required for cardioprotection as its deletion within the AAV-JP2NT vector prevented improvement in TAC-induced cardiac dysfunction. Functional and histological data suggest that JP2NT gene therapy after the onset of cardiac dysfunction is effective at slowing the progression of HF. RNA-sequencing analysis further revealed a broad reversal of hypertrophic and HF-related gene transcription by JP2NT overexpression after TAC. CONCLUSIONS Our prevention- and intervention-based approaches here demonstrated that AAV-mediated delivery of JP2NT into the myocardium can attenuate stress-induced transcriptional remodeling and the development of HF when administered either before or after cardiac stress initiation. Our data indicate that JP2NT gene therapy holds great potential as a novel therapeutic for treating hypertrophy and HF.
Collapse
Affiliation(s)
- Jinxi Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Qian Shi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Yihui Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Logan W. Dawson
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
| | - Grace Ciampa
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
| | - Weiyang Zhao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Guangqin Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Robert M. Weiss
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Chad E. Grueter
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Duane D. Hall
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
- Department of Veterans Affairs, Iowa City Medical Center, IA 52242
| |
Collapse
|
4
|
Sakata T, Mazurek R, Mavropoulos SA, Romeo FJ, Ravichandran AJ, Ishikawa K. Assessing the Effect of Cardiac Gene Therapy Using Catheter-Based Pressure-Volume Measurement in Large Animals. Methods Mol Biol 2022; 2573:313-321. [PMID: 36040605 DOI: 10.1007/978-1-0716-2707-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Gene therapy for heart failure targets various pathways that modulate cardiac function. Its detailed evaluation is crucial for proving the efficacy of cardiac gene therapies. Parameters that can be obtained by noninvasive approaches are generally influenced by loading conditions of the heart. In contrast, catheter-based left ventricular pressure-volume assessment provides a unique option to minimally invasively assess intrinsic myocardial function in a load-insensitive manner. In this chapter, we describe procedural steps for performing pressure-volume measurements and analysis in a preclinical large animal model.
Collapse
Affiliation(s)
- Tomoki Sakata
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Renata Mazurek
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Spyros A Mavropoulos
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francisco J Romeo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anjali J Ravichandran
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyotake Ishikawa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Bilal AS, Thuerauf DJ, Blackwood EA, Glembotski CC. Design and Production of Heart Chamber-Specific AAV9 Vectors. Methods Mol Biol 2022; 2573:89-113. [PMID: 36040589 DOI: 10.1007/978-1-0716-2707-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Adeno-associated virus serotype 9 (AAV9) is often used in heart research involving gene delivery due to its cardiotropism, high transduction efficiency, and little to no pathogenicity, making it highly applicable for gene manipulation, in vivo. However, current AAV9 technology is limited by the lack of strains that can selectively express and elucidate gene function in an atrial- and ventricular-specific manner. In fact, study of gene function in cardiac atria has been limited due to the lack of an appropriate tool to study atrial gene expression in vivo, hindering progress in the study of atrial-specific diseases such as atrial fibrillation, the most common cardiac arrhythmia in the USA.This chapter describes the method for the design and production of such chamber-specific AAV9 vectors, with the use of Nppa and Myl2 promoters to enhance atrial- and ventricular-specific expression. While several gene promoter candidates were considered and tested, Nppa and Myl2 were selected for use here because of their clearly defined regulatory elements that confer cardiac chamber-specific expression. Accordingly, Nppa (-425/+25) and Myl2 (-226/+36) promoter fragments are inserted into AAV9 vectors. The atrial- and ventricular-specific expression conferred by these new recombinant AAV9 was confirmed in a double-fluorescent Cre-dependent reporter mouse model. At only 450 and 262 base pairs of Nppa and Myl2 promoters, respectively, these AAV9 that drive chamber-specific AAV9 transgene expression address two major limitations of AAV9 technology, i.e., achieving chamber-specificity while maximizing space in the AAV genome for insertion of larger transgenes.
Collapse
Affiliation(s)
- Alina S Bilal
- Translational Cardiovascular Research Center and Department of Internal Medicine, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Donna J Thuerauf
- Department of Cellular and Molecular Biology, San Diego State University, San Diego, CA, USA
| | - Erik A Blackwood
- Translational Cardiovascular Research Center and Department of Internal Medicine, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Christopher C Glembotski
- Translational Cardiovascular Research Center and Department of Internal Medicine, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| |
Collapse
|
6
|
Isomi M, Sadahiro T, Fujita R, Abe Y, Yamada Y, Akiyama T, Mizukami H, Shu T, Fukuda K, Ieda M. Direct reprogramming with Sendai virus vectors repaired infarct hearts at the chronic stage. Biochem Biophys Res Commun 2021; 560:87-92. [PMID: 33984769 DOI: 10.1016/j.bbrc.2021.04.121] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
Adult hearts have limited regenerative capacity. Hence, after acute myocardial infarction (MI), dead myocardial tissues are digested by immune cells and replaced by fibrosis, leading to ventricular remodeling and heart failure at the chronic stage. Direct reprogramming of the cardiac fibroblasts (CFs) into induced cardiomyocytes (iCMs) with cardiac transcription factors, including Gata4, Mef2c, and Tbx5 (GMT), may have significant potential for cardiac repair. Sendai virus (SeV) vectors expressing GMT have been reported to reprogram the mouse cardiac fibroblasts into iCMs without any risk of insertional mutagenesis. In vivo reprogramming improved the cardiac function after acute MI in immunodeficient mice. However, it is unknown whether the newly generated iCMs could exist in infarct hearts for a prolonged period and SeV-GMT can improve cardiac function after MI at the chronic stage in immunocompetent mice. Here, we show that SeV vectors efficiently infect CFs in vivo and reprogram them into iCMs, which existed for at least four weeks after MI, in fibroblast-linage tracing mice. Moreover, SeV-GMT improved cardiac function and reduced fibrosis and collagen I expression at 12 weeks after MI in immunocompetent mice. Thus, direct cardiac reprogramming with SeV vectors could be a promising therapy for MI.
Collapse
Affiliation(s)
- Mari Isomi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| | - Taketaro Sadahiro
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| | - Ryo Fujita
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan; Division of Regenerative Medicine, Transborder Medical Research Center, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| | - Yuto Abe
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| | - Yu Yamada
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| | - Tatsuya Akiyama
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan; Department of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Tsugumine Shu
- ID Pharma Co., Ltd., R&D Center, Techno Park Oho, 6 Ohkubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan.
| |
Collapse
|
7
|
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
|
8
|
Development of an AAV9-RNAi-mediated silencing strategy to abrogate TRPM4 expression in the adult heart. Pflugers Arch 2021; 473:533-546. [PMID: 33580817 PMCID: PMC7940300 DOI: 10.1007/s00424-021-02521-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
The cation channel transient receptor potential melastatin 4 (TRPM4) is a calcium-activated non-selective cation channel and acts in cardiomyocytes as a negative modulator of the L-type Ca2+ influx. Global deletion of TRPM4 in the mouse led to increased cardiac contractility under β-adrenergic stimulation. Consequently, cardiomyocyte-specific inactivation of the TRPM4 function appears to be a promising strategy to improve cardiac contractility in heart failure patients. The aim of this study was to develop a gene therapy approach in mice that specifically silences the expression of TRPM4 in cardiomyocytes. First, short hairpin RNAmiR30 (shRNAmiR30) sequences against the TRPM4 mRNA were screened in vitro using lentiviral transduction for a stable expression of the shRNA cassettes. Western blot analysis identified three efficient shRNAmiR30 sequences out of six, which reduced the endogenous TRPM4 protein level by up to 90 ± 6%. Subsequently, the most efficient shRNAmiR30 sequences were delivered into cardiomyocytes of adult mice using adeno-associated virus serotype 9 (AAV9)-mediated gene transfer. Initially, the AAV9 vector particles were administered via the lateral tail vein, which resulted in a downregulation of TRPM4 by 46 ± 2%. Next, various optimization steps were carried out to improve knockdown efficiency in vivo. First, the design of the expression cassette was streamlined for integration in a self-complementary AAV vector backbone for a faster expression. Compared to the application via the lateral tail vein, intravenous application via the retro-orbital sinus has the advantage that the vector solution reaches the heart directly and in a high concentration, and eventually a TRPM4 knockdown efficiency of 90 ± 7% in the heart was accomplished by this approach. By optimization of the shRNAmiR30 constructs and expression cassette as well as the route of AAV9 vector application, a 90% reduction of TRPM4 expression was achieved in the adult mouse heart. In the future, AAV9-RNAi-mediated inactivation of TRPM4 could be a promising strategy to increase cardiac contractility in preclinical animal models of acute and chronic forms of cardiac contractile failure.
Collapse
|
9
|
Hu C, Zhang X, Zhang N, Wei WY, Li LL, Ma ZG, Tang QZ. Osteocrin attenuates inflammation, oxidative stress, apoptosis, and cardiac dysfunction in doxorubicin-induced cardiotoxicity. Clin Transl Med 2020; 10:e124. [PMID: 32618439 PMCID: PMC7418805 DOI: 10.1002/ctm2.124] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Inflammation, oxidative stress, and apoptosis contribute to the evolution of doxorubicin (DOX)‐induced cardiotoxicity. Osteocrin (OSTN) is a novel secretory peptide mainly derived from the bone and skeletal muscle, and plays critical roles in regulating bone growth and physical endurance. Inspiringly, OSTN was also reported to be abundant in the myocardium that functioned as a therapeutic agent against cardiac rupture and congestive heart failure in mice after myocardial infarction. Herein, we investigated the role and potential mechanism of OSTN in DOX‐induced cardiotoxicity. Methods Cardiac‐restrict OSTN overexpression was performed by the intravenous injection of a cardiotropic AAV9 vector, and subsequently the mice received 15 mg/kg DOX injection (i.p., once) to induce acute cardiac injury. Besides, H9C2 cell lines were used to assess the possible role of OSTN in vitro by incubating with recombinant human OSTN or small interfering RNA against Ostn (siOstn). To clarify the involvement of protein kinase G (PKG), KT5823 and siPkg were used in vivo and in vitro. Mice were also administrated intraperitoneally with 5 mg/kg DOX weekly for consecutive 3 weeks at a cumulative dose of 15 mg/kg to mimic the cardiotoxic effects upon chronic DOX exposure. Results OSTN treatment notably attenuated, whereas OSTN silence exacerbated inflammation, oxidative stress, and cardiomyocyte apoptosis in DOX‐treated H9C2 cells. Besides, cardiac‐restrict OSTN‐overexpressed mice showed an alleviated cardiac injury and malfunction upon DOX injection. Mechanistically, we found that OSTN activated PKG, while PKG inhibition abrogated the beneficial effect of OSTN in vivo and in vitro. As expected, OSTN overexpression also improved cardiac function and survival rate in mice after chronic DOX treatment. Conclusions OSTN protects against DOX‐elicited inflammation, oxidative stress, apoptosis, and cardiac dysfunction via activating PKG, and cardiac gene therapy with OSTN provides a novel therapeutic strategy against DOX‐induced cardiotoxicity.
Collapse
Affiliation(s)
- Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| | - Ning Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| | - Wen-Ying Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| | - Ling-Li Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P. R. China
| |
Collapse
|
10
|
Reddy YNV, Borlaug BA, O’Connor CM, Gersh BJ. Novel approaches to the management of chronic systolic heart failure: future directions and unanswered questions. Eur Heart J 2019; 41:1764-1774. [DOI: 10.1093/eurheartj/ehz364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/25/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
Abstract
Despite improvements in outcomes in the last few decades for heart failure (HF) with reduced ejection fraction (HFrEF), there still remains a need for novel therapies as many patients incompletely recover with existing therapies and progress to advanced HF. In this review, we will discuss recent advances in the management of HFrEF with a focus on upcoming therapies that hold the greatest promise for clinical use. We will discuss novel pharmacological therapies and areas of uncertainty with existing therapies. We will also discuss the potential utility and controversy surrounding novel interventions for HF such as percutaneous mitral valve repair, atrial fibrillation ablation, and other emerging interventions with positive signals for benefit in HFrEF. Finally, we will summarize the current state of stem cell and gene therapy for HFrEF and future directions.
Collapse
Affiliation(s)
- Yogesh N V Reddy
- The Department of Cardiovascular Medicine, Mayo Clinic Rochester, 200 First Street SW, MN 55906, USA
| | - Barry A Borlaug
- The Department of Cardiovascular Medicine, Mayo Clinic Rochester, 200 First Street SW, MN 55906, USA
| | | | - Bernard J Gersh
- The Department of Cardiovascular Medicine, Mayo Clinic Rochester, 200 First Street SW, MN 55906, USA
| |
Collapse
|
11
|
Guenther CM, Brun MJ, Bennett AD, Ho ML, Chen W, Zhu B, Lam M, Yamagami M, Kwon S, Bhattacharya N, Sousa D, Evans AC, Voss J, Sevick-Muraca EM, Agbandje-McKenna M, Suh J. Protease-Activatable Adeno-Associated Virus Vector for Gene Delivery to Damaged Heart Tissue. Mol Ther 2019; 27:611-622. [PMID: 30772143 DOI: 10.1016/j.ymthe.2019.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/08/2019] [Accepted: 01/23/2019] [Indexed: 01/08/2023] Open
Abstract
Adeno-associated virus (AAV) has emerged as a promising gene delivery vector because of its non-pathogenicity, simple structure and genome, and low immunogenicity compared to other viruses. However, its adoption as a safe and effective delivery vector for certain diseases relies on altering its tropism to deliver transgenes to desired cell populations. To this end, we have developed a protease-activatable AAV vector, named provector, that responds to elevated extracellular protease activity commonly found in diseased tissue microenvironments. The AAV9-based provector is initially inactive, but then it can be switched on by matrix metalloproteinases (MMP)-2 and -9. Cryo-electron microscopy and image reconstruction reveal that the provector capsid is structurally similar to that of AAV9, with a flexible peptide insertion at the top of the 3-fold protrusions. In an in vivo model of myocardial infarction (MI), the provector is able to deliver transgenes site specifically to high-MMP-activity regions of the damaged heart, with concomitant decreased delivery to many off-target organs, including the liver. The AAV provector may be useful in the future for enhanced delivery of transgenes to sites of cardiac damage.
Collapse
Affiliation(s)
- Caitlin M Guenther
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Mitchell J Brun
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Antonette D Bennett
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Michelle L Ho
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Weitong Chen
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Banghe Zhu
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 6767 Bertner Avenue, Houston, TX 77225, USA
| | - Michael Lam
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Momona Yamagami
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Sunkuk Kwon
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 6767 Bertner Avenue, Houston, TX 77225, USA
| | - Nilakshee Bhattacharya
- Biological Science Imaging facility (BSIR), Department of Biology, 89 Chieftan Way, Florida State University, Tallahassee, FL 32306, USA
| | - Duncan Sousa
- Biological Science Imaging facility (BSIR), Department of Biology, 89 Chieftan Way, Florida State University, Tallahassee, FL 32306, USA
| | - Annicka C Evans
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Julie Voss
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 6767 Bertner Avenue, Houston, TX 77225, USA
| | - Eva M Sevick-Muraca
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 6767 Bertner Avenue, Houston, TX 77225, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Junghae Suh
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA.
| |
Collapse
|
12
|
Makarewich CA, Munir AZ, Schiattarella GG, Bezprozvannaya S, Raguimova ON, Cho EE, Vidal AH, Robia SL, Bassel-Duby R, Olson EN. The DWORF micropeptide enhances contractility and prevents heart failure in a mouse model of dilated cardiomyopathy. eLife 2018; 7:e38319. [PMID: 30299255 PMCID: PMC6202051 DOI: 10.7554/elife.38319] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/26/2018] [Indexed: 01/01/2023] Open
Abstract
Calcium (Ca2+) dysregulation is a hallmark of heart failure and is characterized by impaired Ca2+ sequestration into the sarcoplasmic reticulum (SR) by the SR-Ca2+-ATPase (SERCA). We recently discovered a micropeptide named DWORF (DWarf Open Reading Frame) that enhances SERCA activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Here we show that DWORF has a higher apparent binding affinity for SERCA than PLN and that DWORF overexpression mitigates the contractile dysfunction associated with PLN overexpression, substantiating its role as a potent activator of SERCA. Additionally, using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to genetic deletion of the muscle-specific LIM domain protein (MLP), we show that DWORF overexpression restores cardiac function and prevents the pathological remodeling and Ca2+ dysregulation classically exhibited by MLP knockout mice. Our results establish DWORF as a potent activator of SERCA within the heart and as an attractive candidate for a heart failure therapeutic.
Collapse
Affiliation(s)
- Catherine A Makarewich
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Amir Z Munir
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Gabriele G Schiattarella
- Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Svetlana Bezprozvannaya
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Olga N Raguimova
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Ellen E Cho
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Alexander H Vidal
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Seth L Robia
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Eric N Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
13
|
Non-invasive detection of adeno-associated viral gene transfer using a genetically encoded CEST-MRI reporter gene in the murine heart. Sci Rep 2018; 8:4638. [PMID: 29545551 PMCID: PMC5854573 DOI: 10.1038/s41598-018-22993-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/05/2018] [Indexed: 01/02/2023] Open
Abstract
Research into gene therapy for heart failure has gained renewed interest as a result of improved safety and availability of adeno-associated viral vectors (AAV). While magnetic resonance imaging (MRI) is standard for functional assessment of gene therapy outcomes, quantitation of gene transfer/expression relies upon tissue biopsy, fluorescence or nuclear imaging. Imaging of gene expression through the use of genetically encoded chemical exchange saturation transfer (CEST)-MRI reporter genes could be combined with clinical cardiac MRI methods to comprehensively probe therapeutic gene expression and subsequent outcomes. The CEST-MRI reporter gene Lysine Rich Protein (LRP) was cloned into an AAV9 vector and either administered systemically via tail vein injection or directly injected into the left ventricular free wall of mice. Longitudinal in vivo CEST-MRI performed at days 15 and 45 after direct injection or at 1, 60 and 90 days after systemic injection revealed robust CEST contrast in myocardium that was later confirmed to express LRP by immunostaining. Ventricular structure and function were not impacted by expression of LRP in either study arm. The ability to quantify and link therapeutic gene expression to functional outcomes can provide rich data for further development of gene therapy for heart failure.
Collapse
|
14
|
Jungi S, Fu X, Segiser A, Busch M, Most P, Fiedler M, Carrel T, Tevaearai Stahel H, Longnus SL, Most H. Enhanced Cardiac S100A1 Expression Improves Recovery from Global Ischemia-Reperfusion Injury. J Cardiovasc Transl Res 2018; 11:236-245. [PMID: 29392537 DOI: 10.1007/s12265-018-9788-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022]
Abstract
Gene-targeted therapy with the inotropic Ca2 + -sensor protein S100A1 rescues contractile function in post-ischemic heart failure and is being developed towards clinical trials. Its proven beneficial effect on cardiac metabolism and mitochondrial function suggests a cardioprotective effect of S100A1 in myocardial ischemia-reperfusion injury (IRI). Fivefold cardiomyocyte-specific S100A1 overexpressing, isolated rat hearts perfused in working mode were subjected to 28 min ischemia (37 °C) followed by 60 min reperfusion. S100A1 overexpressing hearts showed superior hemodynamic recover: Left ventricular pressure recovered to 57 ± 7.3% of baseline compared to 51 ± 4.6% in control (p = 0.025), this effect mirrored in LV work and dP/dt(max). Troponin T and lactate dehydrogenase was decreased in the S100A1 group, as well as FoxO pro-apoptotic transcription factor, indicating less tissue necrosis, whereas phosphocreatine content was higher after reperfusion. This is the first report of a cardioprotective effect of S100A1 overexpression in a global IRI model.
Collapse
Affiliation(s)
- S Jungi
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - X Fu
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - A Segiser
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - M Busch
- Section for Molecular and Translational Cardiology, Department of Cardiology, Pneumology and Angiology, Karl-Ruprechts University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - P Most
- Section for Molecular and Translational Cardiology, Department of Cardiology, Pneumology and Angiology, Karl-Ruprechts University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - M Fiedler
- Center for Laboratory Medicine, Inselspital University Hospital, University of Bern, Bern, Switzerland
| | - T Carrel
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - H Tevaearai Stahel
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - S L Longnus
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Henriette Most
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
15
|
Gyöngyösi M, Winkler J, Ramos I, Do QT, Firat H, McDonald K, González A, Thum T, Díez J, Jaisser F, Pizard A, Zannad F. Myocardial fibrosis: biomedical research from bench to bedside. Eur J Heart Fail 2017; 19:177-191. [PMID: 28157267 PMCID: PMC5299507 DOI: 10.1002/ejhf.696] [Citation(s) in RCA: 284] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/07/2016] [Accepted: 10/01/2016] [Indexed: 01/05/2023] Open
Abstract
Myocardial fibrosis refers to a variety of quantitative and qualitative changes in the interstitial myocardial collagen network that occur in response to cardiac ischaemic insults, systemic diseases, drugs, or any other harmful stimulus affecting the circulatory system or the heart itself. Myocardial fibrosis alters the architecture of the myocardium, facilitating the development of cardiac dysfunction, also inducing arrhythmias, influencing the clinical course and outcome of heart failure patients. Focusing on myocardial fibrosis may potentially improve patient care through the targeted diagnosis and treatment of emerging fibrotic pathways. The European Commission funded the FIBROTARGETS consortium as a multinational academic and industrial consortium with the primary aim of performing a systematic and collaborative search of targets of myocardial fibrosis, and then translating these mechanisms into individualized diagnostic tools and specific therapeutic pharmacological options for heart failure. This review focuses on those methodological and technological aspects considered and developed by the consortium to facilitate the transfer of the new mechanistic knowledge on myocardial fibrosis into potential biomedical applications.
Collapse
Affiliation(s)
| | | | - Isbaal Ramos
- Innovative Technologies in Biological Systems SL (INNOPROT), Bizkaia, Spain
| | | | | | | | - Arantxa González
- Program of Cardiovascular Diseases, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Germany.,National Heart and Lung Institute, Imperial College London, UK
| | - Javier Díez
- Program of Cardiovascular Diseases, Center for Applied Medical Research, University of Navarra, Pamplona, Spain.,Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, University of Navarra, Pamplona, Spain
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, Inserm U1138, Université Pierre et Marie Curie, Paris, France
| | - Anne Pizard
- UMRS U1116 Inserm, CIC 1433, Pierre Drouin, CHU, Université de Lorraine, Nancy, France
| | - Faiez Zannad
- UMRS U1116 Inserm, CIC 1433, Pierre Drouin, CHU, Université de Lorraine, Nancy, France
| |
Collapse
|
16
|
Teichman SL, Thomson KS, Regnier M. Cardiac Myosin Activation with Gene Therapy Produces Sustained Inotropic Effects and May Treat Heart Failure with Reduced Ejection Fraction. Handb Exp Pharmacol 2017; 243:447-464. [PMID: 27590227 DOI: 10.1007/164_2016_31] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic inotropic therapy is effective for the treatment of heart failure with reduced ejection fraction, but has been limited by adverse long-term safety profiles, development of tolerance, and the need for chronic parenteral administration. A safe and convenient therapeutic agent that produces sustained inotropic effects could improve symptoms, functional capacity, and quality of life. Small amounts of 2-deoxy-adenosine triphosphate (dATP) activate cardiac myosin leading to enhanced contractility in normal and failing heart muscle. Cardiac myosin activation triggers faster myosin crossbridge cycling with greater force generation during each contraction. This paper describes the rationale and results of a translational medicine effort to increase dATP levels using a gene therapy strategy to deliver and upregulate ribonucleotide reductase (R1R2), the enzyme responsible for dATP synthesis, selectively in cardiomyocytes. In small and large animal models of heart failure, a single dose of this gene therapy has led to sustained inotropic effects with a benign safety profile. Further animal studies are appropriate with the goal of testing this agent in patients with heart failure.
Collapse
Affiliation(s)
- Sam L Teichman
- BEAT Biotherapeutics Corp, 1380 112th Ave., NE, Suite 200, Seattle, WA, 98004, USA.
| | | | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
The evolution of heart failure with reduced ejection fraction pharmacotherapy: What do we have and where are we going? Pharmacol Ther 2017; 178:67-82. [DOI: 10.1016/j.pharmthera.2017.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
18
|
Krummen DE, Ho G. Left Atrial Venous Anatomy. JACC Clin Electrophysiol 2017; 3:1033-1036. [DOI: 10.1016/j.jacep.2017.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 11/25/2022]
|
19
|
Status of Therapeutic Gene Transfer to Treat Cardiovascular Disease in Dogs and Cats. Vet Clin North Am Small Anim Pract 2017. [PMID: 28647114 DOI: 10.1016/j.cvsm.2017.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Gene therapy is a procedure resulting in the transfer of a gene into an individual's cells to treat a disease. One goal of gene transfer is to express a functional gene when the endogenous gene is inactive. However, because heart failure is a complex disease characterized by multiple abnormalities at the cellular level, an alternate gene delivery approach is to alter myocardial protein levels to improve function. This article discusses background information on gene delivery, including packaging, administration, and a brief discussion of some of the candidate transgenes likely to alter the progression of naturally occurring heart disease in dogs and cats.
Collapse
|
20
|
Peana D, Domeier TL. Cardiomyocyte Ca 2+ homeostasis as a therapeutic target in heart failure with reduced and preserved ejection fraction. Curr Opin Pharmacol 2017; 33:17-26. [PMID: 28437711 DOI: 10.1016/j.coph.2017.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/22/2017] [Indexed: 12/28/2022]
Abstract
Heart failure is a highly prevalent syndrome of multiple etiologies and associated comorbidities, and aberrant intracellular Ca2+ homeostasis is a hallmark finding in heart failure patients. The cyclical changes in Ca2+ concentration within cardiomyocytes control cycles of cardiac contraction and relaxation, and dysregulation of Ca2+ handling processes leads to systolic dysfunction, diastolic dysfunction, and adverse remodeling. For this reason, greater understanding of Ca2+ handling mechanisms in heart failure is critical for selection of appropriate treatment strategies. In this review, we summarize the mechanisms of altered Ca2+ handling in two subsets of heart failure, heart failure with reduced ejection fraction and heart failure with preserved ejection fraction, and outline current and experimental treatments that target cardiomyocyte Ca2+ handling processes.
Collapse
Affiliation(s)
- Deborah Peana
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
21
|
Gu X, Matsumura Y, Tang Y, Roy S, Hoff R, Wang B, Wagner WR. Sustained viral gene delivery from a micro-fibrous, elastomeric cardiac patch to the ischemic rat heart. Biomaterials 2017; 133:132-143. [PMID: 28433936 DOI: 10.1016/j.biomaterials.2017.04.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/06/2017] [Accepted: 04/12/2017] [Indexed: 01/14/2023]
Abstract
Biodegradable and elastomeric patches have been applied to the surface of infarcted hearts as temporary mechanical supports to effectively alter adverse left ventricular remodeling processes. In this report, recombinant adeno-associated virus (AAV), known for its persistent transgene expression and low pathogenicity, was incorporated into elastomeric polyester urethane urea (PEUU) and polyester ether urethane urea (PEEUU) and processed by electrospinning into two formats (solid fibers and core-sheath fibers) designed to influence the controlled release behavior. The extended release of AAV encoding green fluorescent protein (GFP) was assessed in vitro. Sustained and localized viral particle delivery was achieved over 2 months in vitro. The biodegradable cardiac patches with or without AAV-GFP were implanted over rat left ventricular lesions three days following myocardial infarction to evaluate the transduction effect of released viral vectors. AAV particles were directly injected into the infarcted hearts as a control. Cardiac function and remodeling were significantly improved for 12 weeks after patch implantation compared to AAV injection. More GFP genes was expressed in the AAV patch group than AAV injection group, with both α-SMA positive cells and cardiac troponin T positive cells transduced in the patch group. Overall, the extended release behavior, prolonged transgene expression, and elastomeric mechanical properties make the AAV-loaded scaffold an attractive option for cardiac tissue engineering where both gene delivery and appropriate mechanical support are desired.
Collapse
Affiliation(s)
- Xinzhu Gu
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Yasumoto Matsumura
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Souvik Roy
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Richard Hoff
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Bing Wang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
22
|
Shirani J, Singh A, Agrawal S, Dilsizian V. Cardiac molecular imaging to track left ventricular remodeling in heart failure. J Nucl Cardiol 2017; 24:574-590. [PMID: 27480973 DOI: 10.1007/s12350-016-0620-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/13/2016] [Indexed: 12/11/2022]
Abstract
Cardiac left ventricular (LV) remodeling is the final common pathway of most primary cardiovascular diseases that manifest clinically as heart failure (HF). The more advanced the systolic HF and LV dysfunction, the worse the prognosis. The knowledge of the molecular, cellular, and neurohormonal mechanisms that lead to myocardial dysfunction and symptomatic HF has expanded rapidly and has allowed sophisticated approaches to understanding and management of the disease. New therapeutic targets for pharmacologic intervention in HF have also been identified through discovery of novel cellular and molecular components of membrane-bound receptor-mediated intracellular signal transduction cascades. Despite all advances, however, the prognosis of systolic HF has remained poor in general. This is, at least in part, related to the (1) relatively late institution of treatment due to reliance on gross functional and structural abnormalities that define the "heart failure phenotype" clinically; (2) remarkable genetic-based interindividual variations in the contribution of each of the many molecular components of cardiac remodeling; and (3) inability to monitor the activity of individual pathways to cardiac remodeling in order to estimate the potential benefits of pharmacologic agents, monitor the need for dose titration, and minimize side effects. Imaging of the recognized ultrastructural components of cardiac remodeling can allow redefinition of heart failure based on its "molecular phenotype," and provide a guide to implementation of "personalized" and "evidence-based" evaluation, treatment, and longitudinal monitoring of the disease beyond what is currently available through randomized controlled clinical trials.
Collapse
Affiliation(s)
- Jamshid Shirani
- Department of Cardiology, St. Luke's University Health Network, 801 Ostrum Street, Bethlehem, PA, USA.
| | - Amitoj Singh
- Department of Cardiology, St. Luke's University Health Network, 801 Ostrum Street, Bethlehem, PA, USA
| | - Sahil Agrawal
- Department of Cardiology, St. Luke's University Health Network, 801 Ostrum Street, Bethlehem, PA, USA
| | - Vasken Dilsizian
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Abu-Taha IH, Heijman J, Hippe HJ, Wolf NM, El-Armouche A, Nikolaev VO, Schäfer M, Würtz CM, Neef S, Voigt N, Baczkó I, Varró A, Müller M, Meder B, Katus HA, Spiger K, Vettel C, Lehmann LH, Backs J, Skolnik EY, Lutz S, Dobrev D, Wieland T. Nucleoside Diphosphate Kinase-C Suppresses cAMP Formation in Human Heart Failure. Circulation 2016; 135:881-897. [PMID: 27927712 DOI: 10.1161/circulationaha.116.022852] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/23/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND Chronic heart failure (HF) is associated with altered signal transduction via β-adrenoceptors and G proteins and with reduced cAMP formation. Nucleoside diphosphate kinases (NDPKs) are enriched at the plasma membrane of patients with end-stage HF, but the functional consequences of this are largely unknown, particularly for NDPK-C. Here, we investigated the potential role of NDPK-C in cardiac cAMP formation and contractility. METHODS Real-time polymerase chain reaction, (far) Western blot, immunoprecipitation, and immunocytochemistry were used to study the expression, interaction with G proteins, and localization of NDPKs. cAMP levels were determined with immunoassays or fluorescent resonance energy transfer, and contractility was determined in cardiomyocytes (cell shortening) and in vivo (fractional shortening). RESULTS NDPK-C was essential for the formation of an NDPK-B/G protein complex. Protein and mRNA levels of NDPK-C were upregulated in end-stage human HF, in rats after long-term isoprenaline stimulation through osmotic minipumps, and after incubation of rat neonatal cardiomyocytes with isoprenaline. Isoprenaline also promoted translocation of NDPK-C to the plasma membrane. Overexpression of NDPK-C in cardiomyocytes increased cAMP levels and sensitized cardiomyocytes to isoprenaline-induced augmentation of contractility, whereas NDPK-C knockdown decreased cAMP levels. In vivo, depletion of NDPK-C in zebrafish embryos caused cardiac edema and ventricular dysfunction. NDPK-B knockout mice had unaltered NDPK-C expression but showed contractile dysfunction and exacerbated cardiac remodeling during long-term isoprenaline stimulation. In human end-stage HF, the complex formation between NDPK-C and Gαi2 was increased whereas the NDPK-C/Gαs interaction was decreased, producing a switch that may contribute to an NDPK-C-dependent cAMP reduction in HF. CONCLUSIONS Our findings identify NDPK-C as an essential requirement for both the interaction between NDPK isoforms and between NDPK isoforms and G proteins. NDPK-C is a novel critical regulator of β-adrenoceptor/cAMP signaling and cardiac contractility. By switching from Gαs to Gαi2 activation, NDPK-C may contribute to lower cAMP levels and the related contractile dysfunction in HF.
Collapse
Affiliation(s)
- Issam H Abu-Taha
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jordi Heijman
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hans-Jörg Hippe
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nadine M Wolf
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ali El-Armouche
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Viacheslav O Nikolaev
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marina Schäfer
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christina M Würtz
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Stefan Neef
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Niels Voigt
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - István Baczkó
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - András Varró
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marion Müller
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Benjamin Meder
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hugo A Katus
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katharina Spiger
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christiane Vettel
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lorenz H Lehmann
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Johannes Backs
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Edward Y Skolnik
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Susanne Lutz
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Dobromir Dobrev
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Thomas Wieland
- From Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty (I.H.A.-T., N.M.W., K.S., C.V., S.L., T.W.), and Department of Internal Medicine III (H.-J.H., N.M.W., M.M., B.M., H.-A.K., L.H.L., J.B.), Heidelberg University, Heidelberg-Mannheim, Germany; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (I.H.A.-T., J.H., M.S., N.V., D.D.); Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Germany (A.E.-A., C.M.W., S.L.); Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Germany (A.E.-A.); Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.); Department of Internal Medicine II, University of Regensburg, Germany (S.N.); Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Hungary (I.B., A.V.); Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.); and DZHK (German Center for Cardiovascular Research), Partner Site HD/MA, Heidelberg-Mannheim, Germany (B.M., H.A.K., C.V., J.B., T.W.). The current affiliation for H.-J.H. is the Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
24
|
Sorriento D, Ciccarelli M, Cipolletta E, Trimarco B, Iaccarino G. "Freeze, Don't Move": How to Arrest a Suspect in Heart Failure - A Review on Available GRK2 Inhibitors. Front Cardiovasc Med 2016; 3:48. [PMID: 27999776 PMCID: PMC5138235 DOI: 10.3389/fcvm.2016.00048] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/21/2016] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease and heart failure (HF) still collect the largest toll of death in western societies and all over the world. A growing number of molecular mechanisms represent possible targets for new therapeutic strategies, which can counteract the metabolic and structural changes observed in the failing heart. G protein-coupled receptor kinase 2 (GRK2) is one of such targets for which experimental and clinical evidence are established. Indeed, several strategies have been carried out in place to interface with the known GRK2 mechanisms of action in the failing heart. This review deals with results from basic and preclinical studies. It shows different strategies to inhibit GRK2 in HF in vivo (βARK-ct gene therapy, treatment with gallein, and treatment with paroxetine) and in vitro (RNA aptamer, RKIP, and peptide-based inhibitors). These strategies are based either on the inhibition of the catalytic activity of the kinase (“Freeze!”) or the prevention of its shuttling within the cell (“Don’t Move!”). Here, we review the peculiarity of each strategy with regard to the ability to interact with the multiple tasks of GRK2 and the perspective development of eventual clinical use.
Collapse
Affiliation(s)
- Daniela Sorriento
- Department of Advanced Biomedical Sciences, University of Naples Federico II , Naples , Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno , Baronissi, SA , Italy
| | - Ersilia Cipolletta
- Department of Advanced Biomedical Sciences, University of Naples Federico II , Naples , Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, University of Naples Federico II , Naples , Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno , Baronissi, SA , Italy
| |
Collapse
|
25
|
Translation of Cardiac Myosin Activation with 2-deoxy-ATP to Treat Heart Failure via an Experimental Ribonucleotide Reductase-Based Gene Therapy. JACC Basic Transl Sci 2016; 1:666-679. [PMID: 28553667 PMCID: PMC5444879 DOI: 10.1016/j.jacbts.2016.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Despite recent advances, chronic heart failure remains a significant and growing unmet medical need, reaching epidemic proportions carrying substantial morbidity, mortality, and costs. A safe and convenient therapeutic agent that produces sustained inotropic effects could ameliorate symptoms and improve functional capacity and quality of life. The authors discovered that small amounts of 2-deoxy-ATP (dATP) activate cardiac myosin leading to enhanced contractility in normal and failing heart muscle. Cardiac myosin activation triggers faster myosin cross-bridge cycling with greater force generation during each contraction. They describe the rationale and results of a translational medicine effort to increase dATP levels using a gene therapy strategy that up-regulates ribonucleotide reductase, the rate-limiting enzyme for dATP synthesis, selectively in cardiomyocytes. In small and large animal models of heart failure, a single dose of this gene therapy has led to sustained inotropic effects with no toxicity or safety concerns identified to date. Further animal studies are being conducted with the goal of testing this agent in patients with heart failure.
Collapse
|
26
|
Greenberg B. Gene therapy for heart failure. Trends Cardiovasc Med 2016; 27:216-222. [PMID: 28063800 DOI: 10.1016/j.tcm.2016.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 10/29/2016] [Accepted: 11/01/2016] [Indexed: 12/22/2022]
Abstract
Novel strategies are needed to treat the growing population of heart failure patients. While new drug and device based therapies have improved outcomes over the past several decades, heart failure patients continue to experience amongst the lowest quality of life of any chronic disease, high likelihood of being hospitalized and marked reduction in survival. Better understanding of many of the basic mechanisms involved in the development of heart failure has helped identify abnormalities that could potentially be targeted by gene transfer. Despite success in experimental animal models, translating gene transfer strategies from the laboratory to the clinic remains at an early stage. This review provides an introduction to gene transfer as a therapy for treating heart failure, describes some of the many factors that need to be addressed in order for it to be successful and discusses some of the recent studies that have been carried out in heart failure patients. Insights from these studies highlight both the enormous promise of gene transfer and the obstacles that still need to be overcome for this treatment approach to be successful.
Collapse
|
27
|
Abstract
Despite advances in therapy, patients with heart failure (HF) continue to experience unacceptably high rates of hospitalization and death, as well as poor quality of life. As a consequence, there is an urgent need for new treatments that can improve the clinical course of the growing worldwide population of HF patients. Serelaxin and ularatide, both based on naturally occurring peptides, have potent vasodilatory as well as other effects on the heart and kidneys. For both agents, phase 3 studies that are designed to determine whether they improve outcomes in patients with acute HF have completed enrollment. TRV027, a biased ligand for the type 1 angiotensin receptor with effects that extend beyond traditional angiotensin-receptor blockers is also being studied in the acute HF population. Omecamtiv mecarbil, an inotropic agent that improves myocardial contractility by a novel mechanism, and vericiguat, a drug that stimulates soluble guanylate cyclase, are both being developed to treat patients with chronic HF. Finally, despite the negative results of the CUPID study, gene transfer therapy continues to be explored as a means of improving the function of the failing heart. The basis for the use of these drugs and their current status in clinical trials are discussed. (Circ J 2016; 80: 1882-1891).
Collapse
|
28
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
29
|
Cadeddu C, Mercurio V, Spallarossa P, Nodari S, Triggiani M, Monte I, Piras R, Madonna R, Pagliaro P, Tocchetti CG, Mercuro G. Preventing antiblastic drug-related cardiomyopathy: old and new therapeutic strategies. J Cardiovasc Med (Hagerstown) 2016; 17 Suppl 1:e64-e75. [PMID: 27183527 DOI: 10.2459/jcm.0000000000000382] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Because of the recent advances in chemotherapeutic protocols, cancer survival has improved significantly, although cardiovascular disease has become a major cause of morbidity and mortality among cancer survivors: in addition to the well-known cardiotoxicity (CTX) from anthracyclines, biologic drugs that target molecules that are active in cancer biology also interfere with cardiovascular homeostasis.Pharmacological and non-pharmacological strategies to protect the cardiovascular structure and function are the best approaches to reducing the prevalence of cardiomyopathy linked to anticancer drugs. Extensive efforts have been devoted to identifying and testing strategies to achieve this end, but little consensus has been reached on a common and shared operability.Timing, dose and mode of chemotherapy administration play a crucial role in the development of acute or late myocardial dysfunction. Primary prevention initiatives cover a wide area that ranges from conventional heart failure drugs, such as β-blockers and renin-angiotensin-aldosterone system antagonists to nutritional supplementation and physical training. Additional studies on the pathophysiology and cellular mechanisms of anticancer-drug-related CTX will enable the introduction of novel therapies.We present various typologies of prevention strategies, describing the approaches that have already been used and those that could be effective on the basis of a better understanding of pharmacokinetic and pharmacodynamic CTX mechanisms.
Collapse
Affiliation(s)
- Christian Cadeddu
- aDepartment of Medical Sciences 'Mario Aresu', University of Cagliari, Cagliari bDepartment of Translational Medical Sciences, Division of Internal Medicine, Federico II University, Naples cClinic of Cardiovascular Diseases, IRCCS San Martino IST, Genoa dDepartment of Clinical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia eDepartment of General Surgery and Medical-Surgery Specialities, University of Catania, Catania fInstitute of Cardiology, Center of Excellence on Aging, 'G. d'Annunzio' University, Chieti gDepartment of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mercurio V, Pirozzi F, Lazzarini E, Marone G, Rizzo P, Agnetti G, Tocchetti CG, Ghigo A, Ameri P. Models of Heart Failure Based on the Cardiotoxicity of Anticancer Drugs. J Card Fail 2016; 22:449-58. [PMID: 27103426 DOI: 10.1016/j.cardfail.2016.04.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Abstract
Heart failure (HF) is a complication of oncological treatments that may have dramatic clinical impact. It may acutely worsen a patient's condition or it may present with delayed onset, even years after treatment, when cancer has been cured or is in stable remission. Several studies have addressed the mechanisms of cancer therapy-related HF and some have led to the definition of disease models that hold valid for other and more common types of HF. Here, we review these models of HF based on the cardiotoxicity of antineoplastic drugs and classify them in cardiomyocyte-intrinsic, paracrine, or potentially secondary to effects on cardiac progenitor cells. The first group includes HF resulting from the combination of oxidative stress, mitochondrial dysfunction, and activation of the DNA damage response, which is typically caused by anthracyclines, and HF resulting from deranged myocardial energetics, such as that triggered by anthracyclines and sunitinib. Blockade of the neuregulin-1/ErbB4/ErbB2, vascular endothelial growth factor/vascular endothelial growth factor receptor and platelet-derived growth factor /platelet-derived growth factor receptor pathways by trastuzumab, sorafenib and sunitinib is proposed as paradigm of cancer therapy-related HF associated with alterations of myocardial paracrine pathways. Finally, anthracyclines and trastuzumab are also presented as examples of antitumor agents that induce HF by affecting the cardiac progenitor cell population.
Collapse
Affiliation(s)
- Valentina Mercurio
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Flora Pirozzi
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Giulio Agnetti
- Johns Hopkins University, Cardiology, Baltimore, Maryland; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carlo G Tocchetti
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| |
Collapse
|
31
|
Schlegel P, Huditz R, Meinhardt E, Rapti K, Geis N, Most P, Katus HA, Müller OJ, Bekeredjian R, Raake PW. Locally Targeted Cardiac Gene Delivery by AAV Microbubble Destruction in a Large Animal Model. Hum Gene Ther Methods 2016; 27:71-8. [DOI: 10.1089/hgtb.2015.120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Philipp Schlegel
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Regina Huditz
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Eric Meinhardt
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Kleopatra Rapti
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Nicolas Geis
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Patrick Most
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hugo A. Katus
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Raffi Bekeredjian
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Philip W. Raake
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
32
|
Prevalence of AAV1 neutralizing antibodies and consequences for a clinical trial of gene transfer for advanced heart failure. Gene Ther 2015; 23:313-9. [PMID: 26699914 DOI: 10.1038/gt.2015.109] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/16/2015] [Accepted: 10/23/2015] [Indexed: 01/16/2023]
Abstract
Adeno-associated virus serotype 1 (AAV1) has many advantages as a gene therapy vector, but the presence of pre-existing neutralizing antibodies (NAbs) is an important limitation. This study was designed to determine: (1) characteristics of AAV NAbs in human subjects, (2) prevalence of AAV1 NAbs in heart failure patients and (3) utility of aggressive immunosuppressive therapy in reducing NAb seroconversion in an animal model. NAb titers were assessed in a cohort of heart failure patients and in patients screened for a clinical trial of gene therapy with AAV1 carrying the sarcoplasmic reticulum calcium ATPase gene (AAV1/SERCA2a). AAV1 NAbs were found in 59.5% of 1552 heart failure patients. NAb prevalence increased with age (P=0.001) and varied geographically. The pattern of NAb titers suggested that exposure is against AAV2, with AAV1 NAb seropositivity due to crossreactivity. The effects of immunosuppression on NAb formation were tested in mini-pigs treated with immunosuppressant therapy before, during and after a single AAV1/SERCA2a infusion. Aggressive immunosuppression did not prevent formation of AAV1 NAbs. We conclude that immunosuppression is unlikely to be a viable solution for repeat AAV1 dosing. Strategies to reduce NAbs in heart failure patients are needed to increase eligibility for gene transfer using AAV vectors.
Collapse
|
33
|
Lobenwein D, Tepeköylü C, Kozaryn R, Pechriggl EJ, Bitsche M, Graber M, Fritsch H, Semsroth S, Stefanova N, Paulus P, Czerny M, Grimm M, Holfeld J. Shock Wave Treatment Protects From Neuronal Degeneration via a Toll-Like Receptor 3 Dependent Mechanism: Implications of a First-Ever Causal Treatment for Ischemic Spinal Cord Injury. J Am Heart Assoc 2015; 4:e002440. [PMID: 26508745 PMCID: PMC4845137 DOI: 10.1161/jaha.115.002440] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Paraplegia following spinal cord ischemia represents a devastating complication of both aortic surgery and endovascular aortic repair. Shock wave treatment was shown to induce angiogenesis and regeneration in ischemic tissue by modulation of early inflammatory response via Toll‐like receptor (TLR) 3 signaling. In preclinical and clinical studies, shock wave treatment had a favorable effect on ischemic myocardium. We hypothesized that shock wave treatment also may have a beneficial effect on spinal cord ischemia. Methods and Results A spinal cord ischemia model in mice and spinal slice cultures ex vivo were performed. Treatment groups received immediate shock wave therapy, which resulted in decreased neuronal degeneration and improved motor function. In spinal slice cultures, the activation of TLR3 could be observed. Shock wave effects were abolished in spinal slice cultures from TLR3−/− mice, whereas the effect was still present in TLR4−/− mice. TLR4 protein was found to be downregulated parallel to TLR3 signaling. Shock wave–treated animals showed significantly better functional outcome and survival. The protective effect on neurons could be reproduced in human spinal slices. Conclusions Shock wave treatment protects from neuronal degeneration via TLR3 signaling and subsequent TLR4 downregulation. Consequently, it represents a promising treatment option for the devastating complication of spinal cord ischemia after aortic repair.
Collapse
Affiliation(s)
- Daniela Lobenwein
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| | - Can Tepeköylü
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| | - Radoslaw Kozaryn
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| | - Elisabeth J Pechriggl
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.) Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology, Innsbruck Medical University, Innsbruck, Austria (E.J.P., M.B., H.F.)
| | - Mario Bitsche
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology, Innsbruck Medical University, Innsbruck, Austria (E.J.P., M.B., H.F.)
| | - Michael Graber
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| | - Helga Fritsch
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology, Innsbruck Medical University, Innsbruck, Austria (E.J.P., M.B., H.F.)
| | - Severin Semsroth
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (N.S.)
| | - Patrick Paulus
- Department of Anesthesiology and Operative Intensive Care Medicine, Kepler University Hospital Linz, Linz, Austria (P.P.)
| | - Martin Czerny
- Department for Cardiovascular Surgery, University Hospital Freiburg, Freiburg, Germany (M.C.)
| | - Michael Grimm
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| | - Johannes Holfeld
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.L., C.T., R.K., E.J.P., M.G., S.S., M.G., J.H.)
| |
Collapse
|
34
|
Molinaro M, Ameri P, Marone G, Petretta M, Abete P, Di Lisa F, De Placido S, Bonaduce D, Tocchetti CG. Recent Advances on Pathophysiology, Diagnostic and Therapeutic Insights in Cardiac Dysfunction Induced by Antineoplastic Drugs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:138148. [PMID: 26583088 PMCID: PMC4637019 DOI: 10.1155/2015/138148] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/01/2015] [Indexed: 12/28/2022]
Abstract
Along with the improvement of survival after cancer, cardiotoxicity due to antineoplastic treatments has emerged as a clinically relevant problem. Potential cardiovascular toxicities due to anticancer agents include QT prolongation and arrhythmias, myocardial ischemia and infarction, hypertension and/or thromboembolism, left ventricular (LV) dysfunction, and heart failure (HF). The latter is variable in severity, may be reversible or irreversible, and can occur soon after or as a delayed consequence of anticancer treatments. In the last decade recent advances have emerged in clinical and pathophysiological aspects of LV dysfunction induced by the most widely used anticancer drugs. In particular, early, sensitive markers of cardiac dysfunction that can predict this form of cardiomyopathy before ejection fraction (EF) is reduced are becoming increasingly important, along with novel therapeutic and cardioprotective strategies, in the attempt of protecting cardiooncologic patients from the development of congestive heart failure.
Collapse
Affiliation(s)
- Marilisa Molinaro
- Department of Medicine and Health Sciences, University of Molise, 86100 Campobasso, Italy
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Mario Petretta
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
- National Researches Council, Neuroscience Institute, University of Padova, 35121 Padova, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| |
Collapse
|
35
|
Woitek F, Zentilin L, Hoffman NE, Powers JC, Ottiger I, Parikh S, Kulczycki AM, Hurst M, Ring N, Wang T, Shaikh F, Gross P, Singh H, Kolpakov MA, Linke A, Houser SR, Rizzo V, Sabri A, Madesh M, Giacca M, Recchia FA. Intracoronary Cytoprotective Gene Therapy: A Study of VEGF-B167 in a Pre-Clinical Animal Model of Dilated Cardiomyopathy. J Am Coll Cardiol 2015; 66:139-53. [PMID: 26160630 DOI: 10.1016/j.jacc.2015.04.071] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/24/2015] [Accepted: 04/28/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF)-B activates cytoprotective/antiapoptotic and minimally angiogenic mechanisms via VEGF receptors. Therefore, VEGF-B might be an ideal candidate for the treatment of dilated cardiomyopathy, which displays modest microvascular rarefaction and increased rate of apoptosis. OBJECTIVES This study evaluated VEGF-B gene therapy in a canine model of tachypacing-induced dilated cardiomyopathy. METHODS Chronically instrumented dogs underwent cardiac tachypacing for 28 days. Adeno-associated virus serotype 9 viral vectors carrying VEGF-B167 genes were infused intracoronarily at the beginning of the pacing protocol or during compensated heart failure. Moreover, we tested a novel VEGF-B167 transgene controlled by the atrial natriuretic factor promoter. RESULTS Compared with control subjects, VEGF-B167 markedly preserved diastolic and contractile function and attenuated ventricular chamber remodeling, halting the progression from compensated to decompensated heart failure. Atrial natriuretic factor-VEGF-B167 expression was low in normally functioning hearts and stimulated by cardiac pacing; it thus functioned as an ideal therapeutic transgene, active only under pathological conditions. CONCLUSIONS Our results, obtained with a standard technique of interventional cardiology in a clinically relevant animal model, support VEGF-B167 gene transfer as an affordable and effective new therapy for nonischemic heart failure.
Collapse
Affiliation(s)
- Felix Woitek
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Nicholas E Hoffman
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Jeffery C Powers
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Isabel Ottiger
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Suraj Parikh
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Anna M Kulczycki
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Marykathryn Hurst
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Nadja Ring
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Tao Wang
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Farah Shaikh
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Polina Gross
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Harinder Singh
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mikhail A Kolpakov
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Axel Linke
- University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Steven R Houser
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Victor Rizzo
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Abdelkarim Sabri
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Muniswamy Madesh
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Fabio A Recchia
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
36
|
|
37
|
S100A1 DNA-based Inotropic Therapy Protects Against Proarrhythmogenic Ryanodine Receptor 2 Dysfunction. Mol Ther 2015; 23:1320-1330. [PMID: 26005840 DOI: 10.1038/mt.2015.93] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 04/28/2015] [Indexed: 12/21/2022] Open
Abstract
Restoring expression levels of the EF-hand calcium (Ca(2+)) sensor protein S100A1 has emerged as a key factor in reconstituting normal Ca(2+) handling in failing myocardium. Improved sarcoplasmic reticulum (SR) function with enhanced Ca(2+) resequestration appears critical for S100A1's cyclic adenosine monophosphate-independent inotropic effects but raises concerns about potential diastolic SR Ca(2+) leakage that might trigger fatal arrhythmias. This study shows for the first time a diminished interaction between S100A1 and ryanodine receptors (RyR2s) in experimental HF. Restoring this link in failing cardiomyocytes, engineered heart tissue and mouse hearts, respectively, by means of adenoviral and adeno-associated viral S100A1 cDNA delivery normalizes diastolic RyR2 function and protects against Ca(2+)- and β-adrenergic receptor-triggered proarrhythmogenic SR Ca(2+) leakage in vitro and in vivo. S100A1 inhibits diastolic SR Ca(2+) leakage despite aberrant RyR2 phosphorylation via protein kinase A and calmodulin-dependent kinase II and stoichiometry with accessory modulators such as calmodulin, FKBP12.6 or sorcin. Our findings demonstrate that S100A1 is a regulator of diastolic RyR2 activity and beneficially modulates diastolic RyR2 dysfunction. S100A1 interaction with the RyR2 is sufficient to protect against basal and catecholamine-triggered arrhythmic SR Ca(2+) leak in HF, combining antiarrhythmic potency with chronic inotropic actions.
Collapse
|
38
|
Rohde D, Busch M, Volkert A, Ritterhoff J, Katus HA, Peppel K, Most P. Cardiomyocytes, endothelial cells and cardiac fibroblasts: S100A1's triple action in cardiovascular pathophysiology. Future Cardiol 2015; 11:309-21. [PMID: 26021637 PMCID: PMC11544369 DOI: 10.2217/fca.15.18] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Over the past decade, basic and translational research delivered comprehensive evidence for the relevance of the Ca(2+)-binding protein S100A1 in cardiovascular diseases. Aberrant expression levels of S100A1 surfaced as molecular key defects, driving the pathogenesis of chronic heart failure, arterial and pulmonary hypertension, peripheral artery disease and disturbed myocardial infarction healing. Loss of intracellular S100A1 renders entire Ca(2+)-controlled networks dysfunctional, thereby leading to cardiomyocyte failure and endothelial dysfunction. Lack of S100A1 release in ischemic myocardium compromises cardiac fibroblast function, entailing impaired damage healing. This review focuses on molecular pathways and signaling cascades regulated by S100A1 in cardiomyocytes, endothelial cells and cardiac fibroblasts in order to provide an overview of our current mechanistic understanding of S100A1's action in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- David Rohde
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Martin Busch
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Anne Volkert
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Julia Ritterhoff
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Hugo A. Katus
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Karsten Peppel
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
- uniQure GmbH, INF 410, 69120 Heidelberg, Germany
| | - Patrick Most
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
- uniQure GmbH, INF 410, 69120 Heidelberg, Germany
| |
Collapse
|
39
|
Abstract
Heart failure is a major public health problem throughout the world and it is likely that its prevalence will continue to grow over the next several decades. Despite advances in the treatment of heart failure, morbidity and mortality remain unacceptably high. Gene transfer therapy provides a novel strategy for targeting abnormalities in cardiac cells that adversely affect cardiac function. New vectors for gene delivery, mainly adeno-associated viruses (AAVs) that are preferentially taken up by cardiomyocytes, can result in sustained transgene expression. The cardiac isoform of sarco(endo)plasmic reticulum Ca(2+)ATPase (SERCA2a) plays a major role in regulating calcium levels in cardiomyocytes. Abnormal calcium handling by the failing heart caused by a reduction in SERCA2a activity adversely affects both systolic and diastolic function. The Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) study was a Phase 2a double-blind, randomized, placebo-controlled, dose-finding study that was performed in patients with advanced heart failure due to systolic dysfunction. Eligible patients received AAV/SERCA2a or placebo by direct antegrade infusion into the coronary circulation. At the end of 12 months, patients receiving high-dose therapy (i.e. 1×10(13) DNase Resistant Particles) had evidence of favorable changes in several clinically relevant domains compared to patients treated with placebo. There were no safety concerns at any dose of AAV/SERCA2a. Patients treated with AAV/SERCA2a exhibited a striking reduction in cardiovascular events that persisted through 36 months of follow-up compared to patients who received placebo. Transgene expression was detected in the myocardium of patients receiving AAV/SERCA2a gene therapy as long as 31 months after delivery. A second Phase 2b study, CUPID 2, designed to confirm this favorable effect on heart failure events, is currently underway with the results expected to be presented later in 2015. Additional studies using other vectors and targets are in planning or underway making gene transfer therapy one of the most exciting new approaches under development for treating heart failure.
Collapse
|
40
|
Abstract
Heart failure is a global problem with an estimated prevalence of 38 million patients worldwide, a number that is increasing with the ageing of the population. It is the most common diagnosis in patients aged 65 years or older admitted to hospital and in high-income nations. Despite some progress, the prognosis of heart failure is worse than that of most cancers. Because of the seriousness of the condition, a declaration of war on five fronts has been proposed for heart failure. Efforts are underway to treat heart failure by enhancing myofilament sensitivity to Ca(2+); transfer of the gene for SERCA2a, the protein that pumps calcium into the sarcoplasmic reticulum of the cardiomyocyte, seems promising in a phase 2 trial. Several other abnormal calcium-handling proteins in the failing heart are candidates for gene therapy; many short, non-coding RNAs--ie, microRNAs (miRNAs)--block gene expression and protein translation. These molecules are crucial to calcium cycling and ventricular hypertrophy. The actions of miRNAs can be blocked by a new class of drugs, antagomirs, some of which have been shown to improve cardiac function in animal models of heart failure; cell therapy, with autologous bone marrow derived mononuclear cells, or autogenous mesenchymal cells, which can be administered as cryopreserved off the shelf products, seem to be promising in both preclinical and early clinical heart failure trials; and long-term ventricular assistance devices are now used increasingly as a destination therapy in patients with advanced heart failure. In selected patients, left ventricular assistance can lead to myocardial recovery and explantation of the device. The approaches to the treatment of heart failure described, when used alone or in combination, could become important weapons in the war against heart failure.
Collapse
Affiliation(s)
- Eugene Braunwald
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Affiliation(s)
- Ali J Marian
- From the Institute of Molecular Medicine, Center for Cardiovascular Genetic Research, University of Texas Health Science Center, Houston.
| |
Collapse
|
42
|
Holfeld J, Tepeköylü C, Blunder S, Lobenwein D, Kirchmair E, Dietl M, Kozaryn R, Lener D, Theurl M, Paulus P, Kirchmair R, Grimm M. Low energy shock wave therapy induces angiogenesis in acute hind-limb ischemia via VEGF receptor 2 phosphorylation. PLoS One 2014; 9:e103982. [PMID: 25093816 PMCID: PMC4122398 DOI: 10.1371/journal.pone.0103982] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 07/04/2014] [Indexed: 12/13/2022] Open
Abstract
Objectives Low energy shock waves have been shown to induce angiogenesis, improve left ventricular ejection fraction and decrease angina symptoms in patients suffering from chronic ischemic heart disease. Whether there is as well an effect in acute ischemia was not yet investigated. Methods Hind-limb ischemia was induced in 10–12 weeks old male C57/Bl6 wild-type mice by excision of the left femoral artery. Animals were randomly divided in a treatment group (SWT, 300 shock waves at 0.1 mJ/mm2, 5 Hz) and untreated controls (CTR), n = 10 per group. The treatment group received shock wave therapy immediately after surgery. Results Higher gene expression and protein levels of angiogenic factors VEGF-A and PlGF, as well as their receptors Flt-1 and KDR have been found. This resulted in significantly more vessels per high-power field in SWT compared to controls. Improvement of blood perfusion in treatment animals was confirmed by laser Doppler perfusion imaging. Receptor tyrosine kinase profiler revealed significant phosphorylation of VEGF receptor 2 as an underlying mechanism of action. The effect of VEGF signaling was abolished upon incubation with a VEGFR2 inhibitor indicating that the effect is indeed VEGFR 2 dependent. Conclusions Low energy shock wave treatment induces angiogenesis in acute ischemia via VEGF receptor 2 stimulation and shows the same promising effects as known from chronic myocardial ischemia. It may therefore develop as an adjunct to the treatment armentarium of acute muscle ischemia in limbs and myocardium.
Collapse
Affiliation(s)
- Johannes Holfeld
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
- * E-mail:
| | - Can Tepeköylü
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology, Innsbruck Medical University, Innsbruck, Austria
| | - Stefan Blunder
- University Hospital for Dermatology and Venerology, Innsbruck Medical University, Innsbruck, Austria
| | - Daniela Lobenwein
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Elke Kirchmair
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Marion Dietl
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Radoslaw Kozaryn
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Daniela Lener
- University Hospital for Internal Medicine III, Department of Cardiology and Angiology, Innsbruck Medical University, Innsbruck, Austria
| | - Markus Theurl
- University Hospital for Internal Medicine III, Department of Cardiology and Angiology, Innsbruck Medical University, Innsbruck, Austria
| | - Patrick Paulus
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Rudolf Kirchmair
- University Hospital for Internal Medicine III, Department of Cardiology and Angiology, Innsbruck Medical University, Innsbruck, Austria
| | - Michael Grimm
- University Hospital for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
43
|
Pellicena P, Schulman H. CaMKII inhibitors: from research tools to therapeutic agents. Front Pharmacol 2014; 5:21. [PMID: 24600394 PMCID: PMC3929941 DOI: 10.3389/fphar.2014.00021] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 02/03/2014] [Indexed: 11/23/2022] Open
Abstract
The cardiac field has benefited from the availability of several CaMKII inhibitors serving as research tools to test putative CaMKII pathways associated with cardiovascular physiology and pathophysiology. Successful demonstrations of its critical pathophysiological roles have elevated CaMKII as a key target in heart failure, arrhythmia, and other forms of heart disease. This has caught the attention of the pharmaceutical industry, which is now racing to develop CaMKII inhibitors as safe and effective therapeutic agents. While the first generation of CaMKII inhibitor development is focused on blocking its activity based on ATP binding to its catalytic site, future inhibitors can also target sites affecting its regulation by Ca2+/CaM or translocation to some of its protein substrates. The recent availability of crystal structures of the kinase in the autoinhibited and activated state, and of the dodecameric holoenzyme, provides insights into the mechanism of action of existing inhibitors. It is also accelerating the design and development of better pharmacological inhibitors. This review examines the structure of the kinase and suggests possible sites for its inhibition. It also analyzes the uses and limitations of current research tools. Development of new inhibitors will enable preclinical proof of concept tests and clinical development of successful lead compounds, as well as improved research tools to more accurately examine and extend knowledge of the role of CaMKII in cardiac health and disease.
Collapse
|
44
|
The road ahead: working towards effective clinical translation of myocardial gene therapies. Ther Deliv 2014; 5:39-51. [PMID: 24341816 DOI: 10.4155/tde.13.134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
During the last two decades the fields of molecular and cellular cardiology, and more recently molecular cardiac surgery, have developed rapidly. The concept of delivering cDNA encoding a therapeutic gene to cardiomyocytes using a vector system with substantial cardiac tropism, allowing for long-term expression of a therapeutic protein, has moved from hypothesis to bench to clinical application. However, the clinical results to date are still disappointing. The ideal gene transfer method should be explored in clinically relevant animal models of heart disease to evaluate the relative roles of specific molecular pathways in disease pathogenesis, helping to validate the potential targets for therapeutic intervention. Successful clinical cardiovascular gene therapy also requires the use of nonimmunogenic cardiotropic vectors capable of expressing the requisite amount of therapeutic protein in vivo and in situ. Depending on the desired application either regional or global myocardial gene delivery is required. Cardiac-specific delivery techniques incorporating mapping technologies for regional delivery and highly efficient methodologies for global delivery should improve the precision and specificity of gene transfer to the areas of interest and minimize collateral organ gene expression.
Collapse
|
45
|
Affiliation(s)
- Sven T. Pleger
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, Germany
| | - Philip Raake
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, Germany
| | - Hugo A. Katus
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, Germany
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Patrick Most
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, Germany
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
- DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, Germany
| |
Collapse
|
46
|
Weber C, Neacsu I, Krautz B, Schlegel P, Sauer S, Raake P, Ritterhoff J, Jungmann A, Remppis AB, Stangassinger M, Koch WJ, Katus HA, Müller OJ, Most P, Pleger ST. Therapeutic safety of high myocardial expression levels of the molecular inotrope S100A1 in a preclinical heart failure model. Gene Ther 2013; 21:131-8. [PMID: 24305416 DOI: 10.1038/gt.2013.63] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/29/2013] [Accepted: 09/30/2013] [Indexed: 12/11/2022]
Abstract
Low levels of the molecular inotrope S100A1 are sufficient to rescue post-ischemic heart failure (HF). As a prerequisite to clinical application and to determine the safety of myocardial S100A1 DNA-based therapy, we investigated the effects of high myocardial S100A1 expression levels on the cardiac contractile function and occurrence of arrhythmia in a preclinical large animal HF model. At 2 weeks after myocardial infarction domestic pigs presented significant left ventricular (LV) contractile dysfunction. Retrograde application of AAV6-S100A1 (1.5 × 10(13) tvp) via the anterior cardiac vein (ACV) resulted in high-level myocardial S100A1 protein peak expression of up to 95-fold above control. At 14 weeks, pigs with high-level myocardial S100A1 protein overexpression did not show abnormalities in the electrocardiogram. Electrophysiological right ventricular stimulation ruled out an increased susceptibility to monomorphic ventricular arrhythmia. High-level S100A1 protein overexpression in the LV myocardium resulted in a significant increase in LV ejection fraction (LVEF), albeit to a lesser extent than previously reported with low S100A1 protein overexpression. Cardiac remodeling was, however, equally reversed. High myocardial S100A1 protein overexpression neither increases the occurrence of cardiac arrhythmia nor causes detrimental effects on myocardial contractile function in vivo. In contrast, this study demonstrates a broad therapeutic range of S100A1 gene therapy in post-ischemic HF using a preclinical large animal model.
Collapse
Affiliation(s)
- C Weber
- 1] Center for Molecular and Translational Cardiology, Heidelberg University Hospital, Heidelberg, Germany [2] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - I Neacsu
- 1] Center for Molecular and Translational Cardiology, Heidelberg University Hospital, Heidelberg, Germany [2] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - B Krautz
- 1] Center for Molecular and Translational Cardiology, Heidelberg University Hospital, Heidelberg, Germany [2] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - P Schlegel
- Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - S Sauer
- Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
| | - P Raake
- Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - J Ritterhoff
- 1] Center for Molecular and Translational Cardiology, Heidelberg University Hospital, Heidelberg, Germany [2] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - A Jungmann
- Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - A B Remppis
- Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - M Stangassinger
- Institute for Animal Physiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - W J Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - H A Katus
- 1] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany [2] Deutsches Zentrum für Herz-/Kreislaufforschung, University Hospital Heidelberg, Heidelberg, Germany
| | - O J Müller
- Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - P Most
- 1] Center for Molecular and Translational Cardiology, Heidelberg University Hospital, Heidelberg, Germany [2] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany [3] Deutsches Zentrum für Herz-/Kreislaufforschung, University Hospital Heidelberg, Heidelberg, Germany [4] Laboratory for Cardiac Stem Cell and Gene Therapy, Temple University School of Medicine, Philadelphia, PA, USA
| | - S T Pleger
- 1] Center for Molecular and Translational Cardiology, Heidelberg University Hospital, Heidelberg, Germany [2] Department of Internal Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
47
|
Affiliation(s)
- Eugene Braunwald
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|