101
|
Pleger ST, Brinks H, Ritterhoff J, Raake P, Koch WJ, Katus HA, Most P. Heart failure gene therapy: the path to clinical practice. Circ Res 2013; 113:792-809. [PMID: 23989720 PMCID: PMC11848682 DOI: 10.1161/circresaha.113.300269] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 06/26/2013] [Indexed: 01/08/2023]
Abstract
Gene therapy, aimed at the correction of key pathologies being out of reach for conventional drugs, bears the potential to alter the treatment of cardiovascular diseases radically and thereby of heart failure. Heart failure gene therapy refers to a therapeutic system of targeted drug delivery to the heart that uses formulations of DNA and RNA, whose products determine the therapeutic classification through their biological actions. Among resident cardiac cells, cardiomyocytes have been the therapeutic target of numerous attempts to regenerate systolic and diastolic performance, to reverse remodeling and restore electric stability and metabolism. Although the concept to intervene directly within the genetic and molecular foundation of cardiac cells is simple and elegant, the path to clinical reality has been arduous because of the challenge on delivery technologies and vectors, expression regulation, and complex mechanisms of action of therapeutic gene products. Nonetheless, since the first demonstration of in vivo gene transfer into myocardium, there have been a series of advancements that have driven the evolution of heart failure gene therapy from an experimental tool to the threshold of becoming a viable clinical option. The objective of this review is to discuss the current state of the art in the field and point out inevitable innovations on which the future evolution of heart failure gene therapy into an effective and safe clinical treatment relies.
Collapse
Affiliation(s)
- Sven T. Pleger
- Center for Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Henriette Brinks
- Department of Cardiac and Vascular Surgery, University Hospital Bern, 3010 Bern, Switzerland
| | - Julia Ritterhoff
- Center for Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Philip Raake
- Center for Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Walter J. Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University, Philadelphia, PA 19122, USA
| | - Hugo A. Katus
- Center for Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Patrick Most
- Center for Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, 69120 Heidelberg, Germany
- Center for Translational Medicine, Department of Medicine, Jefferson Medical College, Philadelphia, PA 19107, USA
| |
Collapse
|
102
|
Zouein FA, Booz GW. AAV-mediated gene therapy for heart failure: enhancing contractility and calcium handling. F1000PRIME REPORTS 2013; 5:27. [PMID: 23967378 PMCID: PMC3732072 DOI: 10.12703/p5-27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heart failure is a progressive, debilitating disease that is characterized by inadequate contractility of the heart. With an aging population, the incidence and economic burden of managing heart failure are anticipated to increase substantially. Drugs for heart failure only slow its progression and offer no cure. However, results of recent clinical trials using recombinant adeno-associated virus (AAV) gene delivery offer the promise, for the first time, that heart failure can be reversed. The strategy is to improve contractility of cardiac muscle cells by enhancing their ability to store calcium through increased expression of the sarco(endo)plasmic reticulum Ca(2+)-ATPase pump (SERCA2a). Preclinical trials have also identified other proteins involved in calcium cycling in cardiac muscle that are promising targets for gene therapy in heart failure, including the following: protein phosphatase 1, adenylyl cyclase 6, G-protein-coupled receptor kinase 2, phospholamban, SUMO1, and S100A1. These preclinical and clinical trials represent a "quiet revolution" that may end up being one of the most significant and remarkable breakthroughs in modern medical practice. Of course, a number of uncertainties remain, including the long-term utility and wisdom of improving the contractile performance of "sick" muscle cells. In this regard, gene therapy may turn out to be a way of buying additional time for actual cardiac regeneration to occur using cardiac stem cells or induced pluripotent stem cells.
Collapse
Affiliation(s)
- Fouad A. Zouein
- Department of Pharmacology and Toxicology, School of Medicine and The Jackson Center for Heart ResearchJackson, MississippiUSA
- The Cardiovascular-Renal Research Center, The University of Mississippi Medical CenterJackson, MississippiUSA
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine and The Jackson Center for Heart ResearchJackson, MississippiUSA
- The Cardiovascular-Renal Research Center, The University of Mississippi Medical CenterJackson, MississippiUSA
| |
Collapse
|
103
|
Fan Q, Chen M, Zuo L, Shang X, Huang MZ, Ciccarelli M, Raake P, Brinks H, Chuprun KJ, Dorn GW, Koch WJ, Gao E. Myocardial Ablation of G Protein-Coupled Receptor Kinase 2 (GRK2) Decreases Ischemia/Reperfusion Injury through an Anti-Intrinsic Apoptotic Pathway. PLoS One 2013; 8:e66234. [PMID: 23805205 PMCID: PMC3689757 DOI: 10.1371/journal.pone.0066234] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/02/2013] [Indexed: 01/08/2023] Open
Abstract
Studies from our lab have shown that decreasing myocardial G protein–coupled receptor kinase 2 (GRK2) activity and expression can prevent heart failure progression after myocardial infarction. Since GRK2 appears to also act as a pro-death kinase in myocytes, we investigated the effect of cardiomyocyte-specific GRK2 ablation on the acute response to cardiac ischemia/reperfusion (I/R) injury. To do this we utilized two independent lines of GRK2 knockout (KO) mice where the GRK2 gene was deleted in only cardiomyocytes either constitutively at birth or in an inducible manner that occurred in adult mice prior to I/R. These GRK2 KO mice and appropriate control mice were subjected to a sham procedure or 30 min of myocardial ischemia via coronary artery ligation followed by 24 hrs reperfusion. Echocardiography and hemodynamic measurements showed significantly improved post-I/R cardiac function in both GRK2 KO lines, which correlated with smaller infarct sizes in GRK2 KO mice compared to controls. Moreover, there was significantly less TUNEL positive myocytes, less caspase-3, and -9 but not caspase-8 activities in GRK2 KO mice compared to control mice after I/R injury. Of note, we found that lowering cardiac GRK2 expression was associated with significantly lower cytosolic cytochrome C levels in both lines of GRK2 KO mice after I/R compared to corresponding control animals. Mechanistically, the anti-apoptotic effects of lowering GRK2 expression were accompanied by increased levels of Bcl-2, Bcl-xl, and increased activation of Akt after I/R injury. These findings were reproduced in vitro in cultured cardiomyocytes and GRK2 mRNA silencing. Therefore, lowering GRK2 expression in cardiomyocytes limits I/R-induced injury and improves post-ischemia recovery by decreasing myocyte apoptosis at least partially via Akt/Bcl-2 mediated mitochondrial protection and implicates mitochondrial-dependent actions, solidifying GRK2 as a pro-death kinase in the heart.
Collapse
Affiliation(s)
- Qian Fan
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Mai Chen
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Xijing Hospital, The Fourth Military Medical University, Xian, China
| | - Lin Zuo
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xiying Shang
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Maggie Z. Huang
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michele Ciccarelli
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Philip Raake
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Henriette Brinks
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Kurt J. Chuprun
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Gerald W. Dorn
- The Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Walter J. Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
104
|
Jaba IM, Zhuang ZW, Li N, Jiang Y, Martin KA, Sinusas AJ, Papademetris X, Simons M, Sessa WC, Young LH, Tirziu D. NO triggers RGS4 degradation to coordinate angiogenesis and cardiomyocyte growth. J Clin Invest 2013; 123:1718-31. [PMID: 23454748 DOI: 10.1172/jci65112] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 01/10/2013] [Indexed: 12/11/2022] Open
Abstract
Myocardial hypertrophy is an adaptation to increased hemodynamic demands. An increase in heart tissue must be matched by a corresponding expansion of the coronary vasculature to maintain and adequate supply of oxygen and nutrients for the heart. The physiological mechanisms that underlie the coordination of angiogenesis and cardiomyocyte growth are unknown. We report that induction of myocardial angiogenesis promotes cardiomyocyte growth and cardiac hypertrophy through a novel NO-dependent mechanism. We used transgenic, conditional overexpression of placental growth factor (PlGF) in murine cardiac tissues to stimulate myocardial angiogenesis and increase endothelial-derived NO release. NO production, in turn, induced myocardial hypertrophy by promoting proteasomal degradation of regulator of G protein signaling type 4 (RGS4), thus relieving the repression of the Gβγ/PI3Kγ/AKT/mTORC1 pathway that stimulates cardiomyocyte growth. This hypertrophic response was prevented by concomitant transgenic expression of RGS4 in cardiomyocytes. NOS inhibitor L-NAME also significantly attenuated RGS4 degradation, and reduced activation of AKT/mTORC1 signaling and induction of myocardial hypertrophy in PlGF transgenic mice, while conditional cardiac-specific PlGF expression in eNOS knockout mice did not induce myocardial hypertrophy. These findings describe a novel NO/RGS4/Gβγ/PI3Kγ/AKT mechanism that couples cardiac vessel growth with myocyte growth and heart size.
Collapse
Affiliation(s)
- Irina M Jaba
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Rengo G, Galasso G, Femminella GD, Parisi V, Zincarelli C, Pagano G, De Lucia C, Cannavo A, Liccardo D, Marciano C, Vigorito C, Giallauria F, Ferrara N, Furgi G, Filardi PP, Koch WJ, Leosco D. Reduction of lymphocyte G protein-coupled receptor kinase-2 (GRK2) after exercise training predicts survival in patients with heart failure. Eur J Prev Cardiol 2013; 21:4-11. [PMID: 23689525 DOI: 10.1177/2047487313491656] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Increased cardiac G protein-coupled receptor kinase-2 (GRK2) expression has a pivotal role at inducing heart failure (HF)-related β-adrenergic receptor (βAR) dysfunction. Importantly, abnormalities of βAR signalling in the failing heart, including GRK2 overexpression, are mirrored in circulating lymphocytes and correlate with HF severity. Exercise training has been shown to exert several beneficial effects on the failing heart, including normalization of cardiac βAR function and GRK2 protein levels. In the present study, we evaluated whether lymphocyte GRK2 levels and short-term changes of this kinase after an exercise training programme can predict long-term survival in HF patients. METHODS For this purpose, we prospectively studied 193 HF patients who underwent a 3-month exercise training programme. Lymphocyte GRK2 protein levels, plasma N-terminal pro-brain natriuretic peptide, and norepinephrine were measured at baseline and after training along with clinical and functional parameters (left ventricular ejection fraction, NYHA class, and peak-VO2). Cardiac-related mortality was evaluated during a mean follow-up period of 37 ± 20 months. RESULTS Exercise was associated with a significant reduction of lymphocyte GRK2 protein levels (from 1.29 ± 0.52 to 1.16 ± 0.65 densitometric units, p < 0.0001). Importantly, exercise related changes of GRK2 (delta values) robustly predicted survival in our study population. Interestingly, HF patients who did not show reduced lymphocyte GRK2 protein levels after training presented the poorest outcome. CONCLUSIONS Our data offer the first demonstration that changes of lymphocyte GRK2 after exercise training can strongly predict outcome in advanced HF.
Collapse
Affiliation(s)
- Giuseppe Rengo
- Fondazione S. Maugeri, Istituto di Telese, Benevento, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Tang T, Hammond HK. Gene transfer for congestive heart failure: update 2013. Transl Res 2013; 161:313-20. [PMID: 23261978 PMCID: PMC3602385 DOI: 10.1016/j.trsl.2012.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/19/2012] [Accepted: 11/27/2012] [Indexed: 01/08/2023]
Abstract
Congestive heart failure is a major cause of morbidity and mortality with increasing social and economic costs. There have been no new high impact therapeutic agents for this devastating disease for more than a decade. However, many pivotal regulators of cardiac function have been identified using cardiac-directed transgene expression and gene deletion in preclinical studies. Some of these increase function of the failing heart. Altering the expression of these pivotal regulators using gene transfer is now either being tested in clinical gene transfer trials, or soon will be. In this review, we summarize recent progress in cardiac gene transfer for clinical congestive heart failure.
Collapse
Affiliation(s)
- Tong Tang
- Department of Medicine, University of California San Diego, and VA San Diego Healthcare System, San Diego, Calif., USA
| | | |
Collapse
|
107
|
Swain JD, Fargnoli AS, Katz MG, Tomasulo CE, Sumaroka M, Richardville KC, Koch WJ, Rabinowitz JE, Bridges CR. MCARD-mediated gene transfer of GRK2 inhibitor in ovine model of acute myocardial infarction. J Cardiovasc Transl Res 2013; 6:253-62. [PMID: 23208013 PMCID: PMC3695486 DOI: 10.1007/s12265-012-9418-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 10/15/2012] [Indexed: 01/08/2023]
Abstract
β-Adrenergic receptor (βAR) dysfunction in acute myocardial infarction (MI) is associated with elevated levels of the G-protein-coupled receptor kinase-2 (GRK2), which plays a key role in heart failure progression. Inhibition of GRK2 via expression of a peptide βARKct transferred by molecular cardiac surgery with recirculating delivery (MCARD) may be a promising intervention. Five sheep underwent scAAV6-mediated MCARD delivery of βARKct, and five received no treatment (control). After a 3-week period, the branch of the circumflex artery (OM1) was ligated. Quantitative PCR data showed intense βARKct expression in the left ventricle (LV). Circumferential fractional shortening was 23.4 ± 7.1 % (baseline) vs. -2.9 ± 5.2 % (p < 0.05) in the control at 10 weeks. In the MCARD-βARKct group, this parameter was close to baseline. The same trend was observed with LV wall thickening. Cardiac index fully recovered in the MCARD-βARKct group. LV end-diastolic volume and LV end-diastolic pressure did not differ in both groups. MCARD-mediated βARKct gene expression results in preservation of regional and global systolic function after acute MI without arresting progressive ventricular remodeling.
Collapse
Affiliation(s)
- JaBaris D. Swain
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Anthony S. Fargnoli
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Michael G. Katz
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Catherine E. Tomasulo
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Marina Sumaroka
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Kyle C. Richardville
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Walter J. Koch
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Joseph E. Rabinowitz
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Charles R. Bridges
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| |
Collapse
|
108
|
Katz MG, Fargnoli AS, Bridges CR. Myocardial gene transfer: routes and devices for regulation of transgene expression by modulation of cellular permeability. Hum Gene Ther 2013; 24:375-92. [PMID: 23427834 DOI: 10.1089/hum.2012.241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heart diseases are major causes of morbidity and mortality in Western society. Gene therapy approaches are becoming promising therapeutic modalities to improve underlying molecular processes affecting failing cardiomyocytes. Numerous cardiac clinical gene therapy trials have yet to demonstrate strong positive results and advantages over current pharmacotherapy. The success of gene therapy depends largely on the creation of a reliable and efficient delivery method. The establishment of such a system is determined by its ability to overcome the existing biological barriers, including cellular uptake and intracellular trafficking as well as modulation of cellular permeability. In this article, we describe a variety of physical and mechanical methods, based on the transient disruption of the cell membrane, which are applied in nonviral gene transfer. In addition, we focus on the use of different physiological techniques and devices and pharmacological agents to enhance endothelial permeability. Development of these methods will undoubtedly help solve major problems facing gene therapy.
Collapse
Affiliation(s)
- Michael G Katz
- Thoracic and Cardiovascular Surgery, Sanger Heart & Vascular Institute, Carolinas Healthcare System, Charlotte, NC 28203, USA
| | | | | |
Collapse
|
109
|
Yang L, Xiao X. Creation of a cardiotropic adeno-associated virus: the story of viral directed evolution. Virol J 2013; 10:50. [PMID: 23394344 PMCID: PMC3574030 DOI: 10.1186/1743-422x-10-50] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/16/2013] [Indexed: 12/13/2022] Open
Abstract
Adeno-associated virus (AAV) is an important vector system for human gene therapy. Although use of AAV serotypes can result in efficient myocardial gene transfer, improvements in the transduction efficiency and specificity are still required. As a method for artificial modification and selection of gene function, directed evolution has been used for diverse applications in genetic engineering of enzymes and proteins. Since 2000, pioneering work has been performed on directed evolution of viral vectors. We further attempted to evolve the AAV using DNA shuffling and in vivo biopanning in a mouse model. An AAVM41 mutant was characterized, which was found to have improved transduction efficiency and specificity in myocardium, an attribute unknown for any natural AAV serotypes. This review focuses on the development of AAV vector for cardiac gene transfer, the history of directed evolution of viral vectors, and our creation of a cardiotropic AAV, which might have implications for the future design and application of viral vectors.
Collapse
Affiliation(s)
- Lin Yang
- Wuhan Institute of Virology, Chinese Academy of Sciences, 44 Xiaohongshan, Wuhan 430071, Hubei, China.
| | | |
Collapse
|
110
|
Arrestins in the cardiovascular system. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:297-334. [PMID: 23764059 DOI: 10.1016/b978-0-12-394440-5.00012-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Of the four mammalian arrestins, only the β-arrestins (βarrs; Arrestin2 and -3) are expressed throughout the cardiovascular system, where they regulate, as either desensitizers/internalizers or signal transducers, several G-protein-coupled receptors (GPCRs) critical for cardiovascular homeostasis. The cardiovascular roles of βarrs have been delineated at an accelerated pace via a variety of techniques and tools, such as knockout mice, siRNA knockdown, artificial or naturally occurring polymorphic GPCRs, and availability of new βarr "biased" GPCR ligands. This chapter summarizes the current knowledge of cardiovascular arrestin physiology and pharmacology, addressing the individual cardiovascular receptors affected by βarrs in vivo, as well as the individual cell types, tissues, and organs of the cardiovascular system in which βarr effects are exerted; for example, cardiac myocyte or fibroblast, vascular smooth muscle, adrenal gland and platelet. In the broader scope of cardiovascular βarr pharmacology, a discussion of the βarr "bias" of certain cardiovascular GPCR ligands is also included.
Collapse
|
111
|
Thal DM, Homan KT, Chen J, Wu EK, Hinkle PM, Huang ZM, Chuprun JK, Song J, Gao E, Cheung JY, Sklar LA, Koch WJ, Tesmer JJ. Paroxetine is a direct inhibitor of g protein-coupled receptor kinase 2 and increases myocardial contractility. ACS Chem Biol 2012; 7:1830-9. [PMID: 22882301 DOI: 10.1021/cb3003013] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G protein-coupled receptor kinase 2 (GRK2) is a well-established therapeutic target for the treatment of heart failure. Herein we identify the selective serotonin reuptake inhibitor (SSRI) paroxetine as a selective inhibitor of GRK2 activity both in vitro and in living cells. In the crystal structure of the GRK2·paroxetine-Gβγ complex, paroxetine binds in the active site of GRK2 and stabilizes the kinase domain in a novel conformation in which a unique regulatory loop forms part of the ligand binding site. Isolated cardiomyocytes show increased isoproterenol-induced shortening and contraction amplitude in the presence of paroxetine, and pretreatment of mice with paroxetine before isoproterenol significantly increases left ventricular inotropic reserve in vivo with no significant effect on heart rate. Neither is observed in the presence of the SSRI fluoxetine. Our structural and functional results validate a widely available drug as a selective chemical probe for GRK2 and represent a starting point for the rational design of more potent and specific GRK2 inhibitors.
Collapse
Affiliation(s)
- David M. Thal
- Life Sciences
Institute and
the Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kristoff T. Homan
- Life Sciences
Institute and
the Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jun Chen
- Center for Molecular Discovery, University of New Mexico Health Sciences Center, Albuquerque,
New Mexico 87131, United States
| | - Emily K. Wu
- Life Sciences
Institute and
the Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Patricia M. Hinkle
- Department of Pharmacology and
Physiology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Z. Maggie Huang
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia,
Pennsylvania 19140, United States
| | - J. Kurt Chuprun
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia,
Pennsylvania 19140, United States
| | - Jianliang Song
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia,
Pennsylvania 19140, United States
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia,
Pennsylvania 19140, United States
| | - Joseph Y. Cheung
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia,
Pennsylvania 19140, United States
| | - Larry A. Sklar
- Center for Molecular Discovery, University of New Mexico Health Sciences Center, Albuquerque,
New Mexico 87131, United States
| | - Walter J. Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia,
Pennsylvania 19140, United States
| | - John J.G. Tesmer
- Life Sciences
Institute and
the Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
112
|
Kamal FA, Travers JG, Blaxall BC. G protein-coupled receptor kinases in cardiovascular disease: why "where" matters. Trends Cardiovasc Med 2012; 22:213-9. [PMID: 23062971 DOI: 10.1016/j.tcm.2012.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cardiac function is mainly controlled by β-adrenergic receptors (β-ARs), members of the G protein-coupled receptor (GPCR) family. GPCR signaling and expression are tightly controlled by G protein-coupled receptor kinases (GRKs), which induce GPCR internalization and signal termination through phosphorylation. Reduced β-AR density and activity associated with elevated cardiac GRK expression and activity have been described in various cardiovascular diseases. Moreover, alterations in extracardiac GRKs have been observed in blood vessels, adrenal glands, kidneys, and fat cells. The broad tissue distribution of GPCRs and GRKs suggests that a keen appreciation of integrative physiology may drive future therapeutic development. In this review, we provide a brief summary of GRK isoforms, subcellular localization, and interacting partners that impinge directly or indirectly on the cardiovascular system. We also discuss GRK/GPCR interactions and their implications in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Fadia A Kamal
- The Heart Institute, Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | |
Collapse
|
113
|
Rengo G, Lymperopoulos A, Zincarelli C, Femminella G, Liccardo D, Pagano G, de Lucia C, Cannavo A, Gargiulo P, Ferrara N, Perrone Filardi P, Koch W, Leosco D. Blockade of β-adrenoceptors restores the GRK2-mediated adrenal α(2) -adrenoceptor-catecholamine production axis in heart failure. Br J Pharmacol 2012; 166:2430-2440. [PMID: 22519418 PMCID: PMC3448904 DOI: 10.1111/j.1476-5381.2012.01972.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 02/07/2012] [Accepted: 03/20/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Sympathetic nervous system (SNS) hyperactivity is characteristic of chronic heart failure (HF) and significantly worsens prognosis. The success of β-adrenoceptor antagonist (β-blockers) therapy in HF is primarily attributed to protection of the heart from the noxious effects of augmented catecholamine levels. β-Blockers have been shown to reduce SNS hyperactivity in HF, but the underlying molecular mechanisms are not understood. The GPCR kinase-2 (GRK2)-α(2) adrenoceptor-catecholamine production axis is up-regulated in the adrenal medulla during HF causing α(2) -adrenoceptor dysfunction and elevated catecholamine levels. Here, we sought to investigate if β-blocker treatment in HF could lower SNS activation by directly altering adrenal GRK2 levels. EXPERIMENTAL APPROACH Four weeks after myocardial infarction-induced HF, adult rats were randomized to 10-week treatment with vehicle (HF/C) or bisoprolol (HF/B). Cardiac function and dimensions were measured. In heart and adrenal gland, GRK2 levels were assessed by RT-PCR and Western blotting and adrenoceptors studied with radioligand binding. Catecholamines and α(2) adrenoceptors in adrenal medulla chromaffin cell cultures were also measured. KEY RESULTS Bisoprolol treatment ameliorated HF-related adverse cardiac remodelling and reduced plasma catecholamine levels, compared with HF/C rats. Bisoprolol also attenuated adrenal GRK2 overexpression as observed in HF/C rats and increased α(2) adrenoceptor density. In cultures of adrenal medulla chromaffin cells from all study groups, bisoprolol reversed HF-related α(2) adrenoceptor dysfunction. This effect was reversed by GRK2 overexpression. CONCLUSION AND IMPLICATIONS Blockade of β-adrenoceptors normalized the adrenal α(2) adrenoceptor-catecholamine production axis by reducing GRK2 levels. This effect may contribute significantly to the decrease of HF-related sympathetic overdrive by β-blockers.
Collapse
Affiliation(s)
- G Rengo
- Cardiology Division, Fondazione Salvatore Maugeri, IRCCS, Telese Terme (BN), Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Rengo G, Zincarelli C, Femminella GD, Liccardo D, Pagano G, de Lucia C, Altobelli GG, Cimini V, Ruggiero D, Perrone-Filardi P, Gao E, Ferrara N, Lymperopoulos A, Koch WJ, Leosco D. Myocardial β(2) -adrenoceptor gene delivery promotes coordinated cardiac adaptive remodelling and angiogenesis in heart failure. Br J Pharmacol 2012; 166:2348-2361. [PMID: 22452704 PMCID: PMC3448898 DOI: 10.1111/j.1476-5381.2012.01954.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 02/28/2012] [Accepted: 03/02/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE We investigated whether β(2) -adrenoceptor overexpression could promote angiogenesis and improve blood perfusion and left ventricular (LV) remodeling of the failing heart. EXPERIMENTAL APPROACH We explored the angiogenic effects of β(2) -adrenoceptor overexpression in a rat model of post-myocardial infarction (MI) heart failure (HF). Cardiac adenoviral-mediated β(2) -adrenoceptor overexpression was obtained via direct intramyocardial injection 4-weeks post-MI. Adenovirus(Ad)-GFP and saline injected rats served as controls. Furthermore, we extended our observation to β(2) -adrenoceptor -/- mice undergoing MI. KEY RESULTS Transgenes were robustly expressed in the LV at 2 weeks post-gene therapy, whereas their expression was minimal at 4-weeks post-gene delivery. In HF rats, cardiac β(2) -adrenoceptor overexpression resulted in enhanced basal and isoprenaline-stimulated cardiac contractility at 2-weeks post-gene delivery. At 4 weeks post-gene transfer, Ad-β(2) -adrenoceptor HF rats showed improved LV remodeling and cardiac function. Importantly, β(2) -adrenoceptor overexpression was associated with a markedly increased capillary and arteriolar length density and enhanced in vivo myocardial blood flow and coronary reserve. At the molecular level, cardiac β(2) -adrenoceptor gene transfer induced the activation of the VEGF/PKB/eNOS pro-angiogenic pathway. In β(2) -adrenoceptor-/- mice, we found a ~25% reduction in cardiac capillary density compared with β(2) -adrenoceptor+/+ mice. The lack of β(2) -adrenoceptors was associated with a higher mortality rate at 30 days and LV dilatation, and a worse global cardiac contractility compared with controls. CONCLUSIONS AND IMPLICATION β(2) -Adrenoceptors play an important role in the regulation of the angiogenic response in HF. The activation of VEGF/PKB/eNOS pathway seems to be strongly involved in this mechanism.
Collapse
Affiliation(s)
- G Rengo
- Salvatore Maugeri Foundation, IRCCS, Telese Terme (BN), Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Nagai T, Komuro I. Gene and cytokine therapy for heart failure: molecular mechanisms in the improvement of cardiac function. Am J Physiol Heart Circ Physiol 2012; 303:H501-12. [PMID: 22777420 DOI: 10.1152/ajpheart.00130.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite significant advances in pharmacological and clinical treatment, heart failure (HF) remains a leading cause of morbidity and mortality worldwide. Many new therapeutic strategies, including cell transplantation, gene delivery, and cytokines or other small molecules, have been explored to treat HF. Recent advancement of our understanding of the molecules that regulate cardiac function uncover many of the therapeutic key molecules to treat HF. Furthermore, a theory of paracrine mechanism, which underlies the beneficial effects of cell therapy, leads us to search novel target molecules for genetic or pharmacological strategy. Gene therapy means delivery of genetic materials into cells to achieve therapeutic effects. Recently, gene transfer technology in the cardiovascular system has been improved and several therapeutic target genes have been started to examine in clinical research, and some of the promising results have been emerged. Among the various bioactive reagents, cytokines such as granulocyte colony-stimulating factor and erythropoietin have been well examined, and a number of clinical trials for acute myocardial infarction and chronic HF have been conducted. Although further research is needed in both preclinical and clinical areas in terms of molecular mechanisms, safety, and efficiency, both gene and cytokine therapy have a great possibility to open the new era of the treatment of HF.
Collapse
Affiliation(s)
- Toshio Nagai
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | |
Collapse
|
116
|
Weeks KL, Gao X, Du XJ, Boey EJ, Matsumoto A, Bernardo BC, Kiriazis H, Cemerlang N, Tan JW, Tham YK, Franke TF, Qian H, Bogoyevitch MA, Woodcock EA, Febbraio MA, Gregorevic P, McMullen JR. Phosphoinositide 3-Kinase p110α Is a Master Regulator of Exercise-Induced Cardioprotection and PI3K Gene Therapy Rescues Cardiac Dysfunction. Circ Heart Fail 2012; 5:523-34. [DOI: 10.1161/circheartfailure.112.966622] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background—
Numerous molecular and biochemical changes have been linked with the cardioprotective effects of exercise, including increases in antioxidant enzymes, heat shock proteins, and regulators of cardiac myocyte proliferation. However, a master regulator of exercise-induced protection has yet to be identified. Here, we assess whether phosphoinositide 3-kinase (PI3K) p110α is essential for mediating exercise-induced cardioprotection, and if so, whether its activation independent of exercise can restore function of the failing heart.
Methods and Results—
Cardiac-specific transgenic (Tg) mice with elevated or reduced PI3K(p110α) activity (constitutively active PI3K [caPI3K] and dominant negative PI3K, respectively) and non-Tg controls were subjected to 4 weeks of exercise training followed by 1 week of pressure overload (aortic-banding) to induce pathological remodeling. Aortic-banding in untrained non-Tg controls led to pathological cardiac hypertrophy, depressed systolic function, and lung congestion. This phenotype was attenuated in non-Tg controls that had undergone exercise before aortic-banding. Banded caPI3K mice were protected from pathological remodeling independent of exercise status, whereas exercise provided no protection in banded dominant negative PI3K mice, suggesting that PI3K is necessary for exercise-induced cardioprotection. Tg overexpression of heat shock protein 70 could not rescue the phenotype of banded dominant negative PI3K mice, and deletion of heat shock protein 70 from banded caPI3K mice had no effect. Next, we used a gene therapy approach (recombinant adeno-associated viral vector 6) to deliver caPI3K expression cassettes to hearts of mice with established cardiac dysfunction caused by aortic-banding. Mice treated with recombinant adeno-associated viral 6-caPI3K vectors had improved heart function after 10 weeks.
Conclusions—
PI3K(p110α) is essential for exercise-induced cardioprotection and delivery of caPI3K vector can improve function of the failing heart.
Collapse
Affiliation(s)
- Kate L. Weeks
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Xiaoming Gao
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Xiao-Jun Du
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Esther J.H. Boey
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Aya Matsumoto
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Bianca C. Bernardo
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Helen Kiriazis
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Nelly Cemerlang
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Joon Win Tan
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Yow Keat Tham
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Thomas F. Franke
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Hongwei Qian
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Marie A. Bogoyevitch
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Elizabeth A. Woodcock
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Mark A. Febbraio
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Paul Gregorevic
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| | - Julie R. McMullen
- From the Baker IDI Heart and Diabetes Institute (K.L.W., X.G., X-J.D., E.J.H.B., A.M., B.C.B., H.K., N.C., J.W.T., Y.K.T., H.Q., E.A.W., M.A.F., P.G., J.R.M.); Department of Biochemistry and Molecular Biology, University of Melbourne (K.L.W., M.A.B.), Melbourne, Victoria, Australia; Department of Psychiatry and Department of Pharmacology, New York University, School of Medicine, New York, NY (T.F.F.); Department of Medicine (J.R.M.) and the Department of Physiology (J.R.M.), Monash University,
| |
Collapse
|
117
|
Rengo G, Perrone-Filardi P, Femminella GD, Liccardo D, Zincarelli C, de Lucia C, Pagano G, Marsico F, Lymperopoulos A, Leosco D. Targeting the β-adrenergic receptor system through G-protein-coupled receptor kinase 2: a new paradigm for therapy and prognostic evaluation in heart failure: from bench to bedside. Circ Heart Fail 2012; 5:385-391. [PMID: 22589366 DOI: 10.1161/circheartfailure.112.966895] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/13/2012] [Indexed: 01/14/2023]
Affiliation(s)
- Giuseppe Rengo
- Salvatore Maugeri Foundation, IRCCS, Telese Terme, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Abstract
Congestive heart failure is an inexorable disease associated with unacceptably high morbidity and mortality. Preclinical results indicate that gene transfer using various proteins is a safe and effective approach for increasing function of the failing heart. In the current review, we provide a summary of cardiac gene transfer in general and summarize findings using adenylyl cyclase 6 as therapeutic gene in the failing heart. We also discuss the potential usefulness of a new treatment for congestive heart failure, paracrine-based gene transfer.
Collapse
Affiliation(s)
- T Tang
- Department of Medicine, University of California San Diego, CA, USA
| | | | | |
Collapse
|
119
|
Raake PW, Zhang X, Vinge LE, Brinks H, Gao E, Jaleel N, Li Y, Tang M, Most P, Dorn GW, Houser SR, Katus HA, Chen X, Koch WJ. Cardiac G-protein-coupled receptor kinase 2 ablation induces a novel Ca2+ handling phenotype resistant to adverse alterations and remodeling after myocardial infarction. Circulation 2012; 125:2108-18. [PMID: 22496128 DOI: 10.1161/circulationaha.111.044255] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND G-protein-coupled receptor kinase 2 (GRK2) is a primary regulator of β-adrenergic signaling in the heart. G-protein-coupled receptor kinase 2 ablation impedes heart failure development, but elucidation of the cellular mechanisms has not been achieved, and such elucidation is the aim of this study. METHODS AND RESULTS Myocyte contractility, Ca(2+) handling and excitation-contraction coupling were studied in isolated cardiomyocytes from wild-type and GRK2 knockout (GRK2KO) mice without (sham) or with myocardial infarction (MI). In cardiac myocytes isolated from unstressed wild-type and GRK2KO hearts, myocyte contractions and Ca(2+) transients were similar, but GRK2KO myocytes had lower sarcoplasmic reticulum (SR) Ca(2+) content because of increased sodium-Ca(2+) exchanger activity and inhibited SR Ca(2+) ATPase by local protein kinase A-mediated activation of phosphodiesterase 4 resulting in hypophosphorylated phospholamban. This Ca(2+) handling phenotype is explained by a higher fractional SR Ca(2+) release induced by increased L-type Ca(2+) channel currents. After β-adrenergic stimulation, GRK2KO myocytes revealed significant increases in contractility and Ca(2+) transients, which were not mediated through cardiac L-type Ca(2+) channels but through an increased SR Ca(2+). Interestingly, post-MI GRK2KO mice showed better cardiac function than post-MI control mice, which is explained by an improved Ca(2+) handling phenotype. The SR Ca(2+) content was better maintained in post-MI GRK2KO myocytes than in post-MI control myocytes because of better-maintained L-type Ca(2+) channel current density and no increase in sodium-Ca(2+) exchanger in GRK2KO myocytes. An L-type Ca(2+) channel blocker, verapamil, reversed some beneficial effects of GRK2KO. CONCLUSIONS These data argue for novel differential regulation of L-type Ca(2+) channel currents and SR load by GRK2. G-protein-coupled receptor kinase 2 ablation represents a novel beneficial Ca(2+) handling phenotype resisting adverse remodeling after MI.
Collapse
Affiliation(s)
- Philip W Raake
- Department of Internal Medicine III, Cardiology, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Poller W, Rother M, Skurk C, Scheibenbogen C. Endogenous migration modulators as parent compounds for the development of novel cardiovascular and anti-inflammatory drugs. Br J Pharmacol 2012; 165:2044-58. [PMID: 22035209 PMCID: PMC3413843 DOI: 10.1111/j.1476-5381.2011.01762.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 07/18/2011] [Accepted: 09/16/2011] [Indexed: 01/13/2023] Open
Abstract
Development of novel cell migration modulators for anti-inflammatory and cardiovascular therapy is a complex task since any modulator will necessarily interfere with a balanced system of physiological regulators directing proper positioning of diverse immune cell types within the body. Whereas this shall serve efficient pathogen elimination, lack of proper control over these processes may result in counterproductive chronic inflammation and progressive tissue injury instead of healing. Prediction of the therapeutic potential or side effects of any migration modulator is not possible based on theoretical considerations alone but needs to be experimentally evaluated in preclinical disease models and by clinical studies. Here, we briefly summarize basic mechanism of cell migration, and groups of synthetic drugs currently in use for migration modulation. We then discuss one fundamental problem encountered with single-target approaches that arises from the complexity of any inflammation, with multiple interacting and often redundant factors being involved. This issue is likely to arise for any class of therapeutic agent (small molecules, peptides, antibodies, regulatory RNAs) addressing a single gene or protein. Against this background of studies on synthetic migration modulators addressing single targets, we then discuss the potential of endogenous proteins as therapeutic migration modulators, or as parent compounds for the development of mimetic drugs. Regulatory proteins of this type commonly address multiple receptors and signalling pathways and act upon the immune response in a phase-specific manner. Based on recent evidence, we suggest investigation of such endogenous migration modulators as novel starting points for anti-inflammatory and cardiovascular drug development.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department of Cardiology and Pneumology, Campus Benjamin Franklin CBF, Charite - Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | |
Collapse
|
121
|
Katz MG, Fargnoli AS, Pritchette LA, Bridges CR. Gene delivery technologies for cardiac applications. Gene Ther 2012; 19:659-69. [PMID: 22418063 DOI: 10.1038/gt.2012.11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ischemic heart disease (IHD) and heart failure (HF) are major causes of morbidity and mortality in the Western society. Advances in understanding the molecular pathology of these diseases, the evolution of vector technology, as well as defining the targets for therapeutic interventions has placed these conditions within the reach of gene-based therapy. One of the cornerstones of limiting the effectiveness of gene therapy is the establishment of clinically relevant methods of genetic transfer. Recently there have been advances in direct and transvascular gene delivery methods with the use of new technologies. Current research efforts in IHD are focused primarily on the stimulation of angiogenesis, modify the coronary vascular environment and improve endothelial function with localized gene-eluting catheters and stents. In contrast to standard IHD treatments, gene therapy in HF primarily targets inhibition of apoptosis, reduction in adverse remodeling and increase in contractility through global cardiomyocyte transduction for maximal efficacy. This article will review a variety of gene-transfer strategies in models of coronary artery disease and HF and discuss the relative success of these strategies in improving the efficiency of vector-mediated cardiac gene delivery.
Collapse
Affiliation(s)
- M G Katz
- Department of Thoracic and Cardiovascular Surgery, Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, NC, USA
| | | | | | | |
Collapse
|
122
|
Katz MG, Fargnoli AS, Tomasulo CE, Pritchette LA, Bridges CR. Model-specific selection of molecular targets for heart failure gene therapy. J Gene Med 2012; 13:573-86. [PMID: 21954055 DOI: 10.1002/jgm.1610] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Heart failure (HF) is a complex multifaceted problem of abnormal ventricular function and structure. In recent years, new information has been accumulated allowing for a more detailed understanding of the cellular and molecular alterations that are the underpinnings of diverse causes of HF, including myocardial ischemia, pressure-overload, volume-overload or intrinsic cardiomyopathy. Modern pharmacological approaches to treat HF have had a significant impact on the course of the disease, although they do not reverse the underlying pathological state of the heart. Therefore gene-based therapy holds a great potential as a targeted treatment for cardiovascular diseases. Here, we survey the relative therapeutic efficacy of genetic modulation of β-adrenergic receptor signaling, Ca(2+) handling proteins and angiogenesis in the most common extrinsic models of HF.
Collapse
Affiliation(s)
- Michael G Katz
- Department of Surgery, Division of Cardiovascular Surgery, The University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
123
|
Reinkober J, Tscheschner H, Pleger ST, Most P, Katus HA, Koch WJ, Raake PWJ. Targeting GRK2 by gene therapy for heart failure: benefits above β-blockade. Gene Ther 2012; 19:686-93. [PMID: 22336718 DOI: 10.1038/gt.2012.9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heart failure (HF) is a common pathological end point for several cardiac diseases. Despite reasonable achievements in pharmacological, electrophysiological and surgical treatments, prognosis for chronic HF remains poor. Modern therapies are generally symptom oriented and do not currently address specific intracellular molecular signaling abnormalities. Therefore, new and innovative therapeutic approaches are warranted and, ideally, these could at least complement established therapeutic options if not replace them. Gene therapy has potential to serve in this regard in HF as vectors can be directed toward diseased myocytes and directly target intracellular signaling abnormalities. Within this review, we will dissect the adrenergic system contributing to HF development and progression with special emphasis on G-protein-coupled receptor kinase 2 (GRK2). The levels and activity of GRK2 are increased in HF and we and others have demonstrated that this kinase is a major molecular culprit in HF. We will cover the evidence supporting gene therapy directed against myocardial as well as adrenal GRK2 to improve the function and structure of the failing heart and how these strategies may offer complementary and synergistic effects with the existing HF mainstay therapy of β-adrenergic receptor antagonism.
Collapse
Affiliation(s)
- J Reinkober
- Department of Internal Medicine III, Cardiology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
124
|
Hinkel R, Boekstegers P, Kupatt C. Adjuvant early and late cardioprotective therapy: access to the heart. Cardiovasc Res 2012; 94:226-36. [PMID: 22318936 DOI: 10.1093/cvr/cvs075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Coronary heart disease is still the leading cause of death in industrialized nations, occurring either as acute coronary occlusion and myocardial infarction or as chronic ischaemic cardiomyopathy caused by continuous obstruction of one or more coronary arteries. Even after successful reperfusion, an additional loss of otherwise vital cardiomyocytes may occur in the primary ischaemic area, called lethal reperfusion injury. In experimental settings, delivery of therapeutic agents targeting the reperfusion injury reduces the infarct size by 30%. In addition to the choice of therapeutic agent and time point, the mode of application may be crucial for the therapeutic success. Therefore, this review focuses on the current and future administration techniques for early and late post-myocardial infarction therapies.
Collapse
Affiliation(s)
- Rabea Hinkel
- Medizinische Klinik und Poliklinik I, Klinikum der LMU München, Marchioninistraße 15, Munich, Germany.
| | | | | |
Collapse
|
125
|
Lymperopoulos A, Bathgate A. Pharmacogenomics of the heptahelical receptor regulators G-protein-coupled receptor kinases and arrestins: the known and the unknown. Pharmacogenomics 2012; 13:323-341. [PMID: 22304582 DOI: 10.2217/pgs.11.178] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heptahelical G-protein-coupled receptors are the most diverse and therapeutically important family of receptors, playing major roles in the physiology of various organs and tissues. They couple their ligand binding to G-protein activation, which then transmits intracellular signals. G-protein signaling is terminated by phosphorylation of the receptor by the family of G-protein-coupled receptor kinases (GRKs), followed by arrestin (Arr) binding, which uncouples the phosphorylated receptor from the G-protein and subsequently targets the receptor for internalization. Moreover, Arrs can transmit signals in their own right during receptor internalization. Genetic polymorphisms in receptors, as well as in GRK and Arr family members per se, which affect regulation of receptor signaling and function, have just started being identified and characterized. The present review will discuss what is known so far in this evolving field of GRK/Arr pharmacogenomics, as well as highlight important areas likely to produce invaluable information in the future.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft. Lauderdale, FL 33328, USA.
| | | |
Collapse
|
126
|
Raake PWJ, Schlegel P, Ksienzyk J, Reinkober J, Barthelmes J, Schinkel S, Pleger S, Mier W, Haberkorn U, Koch WJ, Katus HA, Most P, Müller OJ. AAV6.βARKct cardiac gene therapy ameliorates cardiac function and normalizes the catecholaminergic axis in a clinically relevant large animal heart failure model. Eur Heart J 2012; 34:1437-47. [PMID: 22261894 DOI: 10.1093/eurheartj/ehr447] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS G protein-coupled receptor kinase 2 (GRK2), which is markedly upregulated in failing human myocardium, has been implicated as a contributing factor or consequence of heart failure (HF). Importantly, cardiac-specific GRK2 knockout mice have recently proved the pathological nature of GRK2 in HF. Targeted inhibition of GRK2 is possible using a peptide inhibitor known as the βARKct, which has rescued several disparate small animal HF models. This study was designed to evaluate long-term βARKct expression in a clinically relevant large animal HF model, using stable myocardial gene delivery with adeno-associated virus serotype 6 (AAV6). METHODS AND RESULTS A porcine model of HF subsequent to left ventricular (LV) myocardial infarction (MI) was used to study the effects of retrograde injection into the anterior interventricular vein of either AAV6.βARKct or AAV6.luciferase as a control 2 weeks after MI. Echocardiography and LV hemodynamics were performed before and 6 weeks after gene transfer. Robust and long-term βARKct expression was found after AAV6-mediated delivery, leading to significant amelioration of LV haemodynamics and contractile function in HF pigs compared with AAV6.luciferase-treated control animals that showed a continued decline in cardiac function. Interestingly, the neurohormonal axis was virtually normalized in AVV6.βARKct-treated HF animals, represented by reductions in plasma norepinephrine levels, whereas AAV6.luciferase-treated pigs showed further increases in plasma catecholamine levels. As a result, LV remodelling and foetal gene expression was reversed by AVV6.βARKct gene therapy. CONCLUSION These data--showing sustained amelioration of cardiac function in a post-MI pig HF model--demonstrate the therapeutic potential of βARKct gene therapy for HF.
Collapse
Affiliation(s)
- Philip W J Raake
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg 69120, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Schumacher-Bass SM, Traynham CJ, Koch WJ. G protein-coupled Receptor Kinase 2 as a Therapeutic Target for Heart Failure. ACTA ACUST UNITED AC 2012; 9:e155-e162. [PMID: 24839449 DOI: 10.1016/j.ddstr.2014.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An ever-increasing number of people world-wide are developing and suffering from heart failure, and existing therapies, although improved are not ideal. Therefore, innovative treatment strategies are urgently needed. As our understanding of the underlying dysfunction of the myocardium increases, so does our ability to target the mechanisms responsible for heart failure progression. In this review we discuss novel molecular therapies and approaches for the treatment of heart failure. We will focus on the G protein-coupled receptor kinase GRK2, an increasing target for heart failure therapy, based on its important role in disease progression and the therapeutic potential of GRK2 inhibitors.
Collapse
Affiliation(s)
- Sarah M Schumacher-Bass
- Department of Pharmacology and Center for Translational Medicine, Temple University School of Medicine, Medical Education Research Building, 3500 North Broad Street, MERB 940, Philadelphia, PA 19140, USA
| | - Christopher J Traynham
- Department of Pharmacology and Center for Translational Medicine, Temple University School of Medicine, Medical Education Research Building, 3500 North Broad Street, MERB 940, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Department of Pharmacology and Center for Translational Medicine, Temple University School of Medicine, Medical Education Research Building, 3500 North Broad Street, MERB 940, Philadelphia, PA 19140, USA
| |
Collapse
|
128
|
Gurevich EV, Tesmer JJG, Mushegian A, Gurevich VV. G protein-coupled receptor kinases: more than just kinases and not only for GPCRs. Pharmacol Ther 2012; 133:40-69. [PMID: 21903131 PMCID: PMC3241883 DOI: 10.1016/j.pharmthera.2011.08.001] [Citation(s) in RCA: 338] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 08/01/2011] [Indexed: 12/24/2022]
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) are best known for their role in homologous desensitization of GPCRs. GRKs phosphorylate activated receptors and promote high affinity binding of arrestins, which precludes G protein coupling. GRKs have a multidomain structure, with the kinase domain inserted into a loop of a regulator of G protein signaling homology domain. Unlike many other kinases, GRKs do not need to be phosphorylated in their activation loop to achieve an activated state. Instead, they are directly activated by docking with active GPCRs. In this manner they are able to selectively phosphorylate Ser/Thr residues on only the activated form of the receptor, unlike related kinases such as protein kinase A. GRKs also phosphorylate a variety of non-GPCR substrates and regulate several signaling pathways via direct interactions with other proteins in a phosphorylation-independent manner. Multiple GRK subtypes are present in virtually every animal cell, with the highest expression levels found in neurons, with their extensive and complex signal regulation. Insufficient or excessive GRK activity was implicated in a variety of human disorders, ranging from heart failure to depression to Parkinson's disease. As key regulators of GPCR-dependent and -independent signaling pathways, GRKs are emerging drug targets and promising molecular tools for therapy. Targeted modulation of expression and/or of activity of several GRK isoforms for therapeutic purposes was recently validated in cardiac disorders and Parkinson's disease.
Collapse
Affiliation(s)
- Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, Preston Research Building, Rm. 454, Nashville, TN 37232, United States.
| | | | | | | |
Collapse
|
129
|
Bristow MR. Treatment of chronic heart failure with β-adrenergic receptor antagonists: a convergence of receptor pharmacology and clinical cardiology. Circ Res 2011; 109:1176-94. [PMID: 22034480 DOI: 10.1161/circresaha.111.245092] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite the absence of a systematic development plan, β-blockers have reached the top tier of medical therapies for chronic heart failure. The successful outcome was due to the many dedicated investigators who produced, over a 30-year period, increasing evidence that β-blocking agents should or actually did improve the natural history of dilated cardiomyopathies and heart failure. It took 20 years for supportive evidence to become undeniable, at which time in 1993 the formidable drug development resources of large pharmaceutical companies were deployed into Phase 3 trials. Success then came relatively quickly, and within 8 years multiple agents were on the market in the United States and Europe. Importantly, there is ample room to improve antiadrenergic therapy, through novel approaches exploiting the nuances of receptor biology and/or intracellular signaling, as well as through pharmacogenetic targeting.
Collapse
Affiliation(s)
- Michael R Bristow
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
130
|
Abstract
Heart failure (HF) involves changes in cardiac structure, myocardial composition, myocyte deformation, and multiple biochemical and molecular alterations that impact heart function and reserve capacity. Collectively, these changes have been referred to as 'cardiac remodeling'. Understanding the components of this process with the goal of stopping or reversing its progression has become a major objective. This concept is often termed 'reverse remodeling', and is successfully achieved by inhibitors of the renin-angiotensin-aldosterone system, β-blockers, and device therapies such as cardiac resynchronization or ventricular assist devices. Not every method of reverse remodeling has long-lasting clinical efficacy. However, thus far, every successful clinical treatment with long-term benefits on the morbidity and mortality of patients with HF reverses remodeling. Reverse remodeling is defined by lower chamber volumes (particularly end-systolic volume) and is often accompanied by improved β-adrenergic and heart-rate responsiveness. At the cellular level, reverse remodeling impacts on myocyte size, function, excitation-contraction coupling, bioenergetics, and a host of molecular pathways that regulate contraction, cell survival, mitochondrial function, oxidative stress, and other features. Here, we review the current evidence for reverse remodeling by existing therapies, and discuss novel approaches that are rapidly moving from preclinical to clinical trials.
Collapse
|
131
|
Katz MG, Fargnoli AS, Swain JD, Tomasulo CE, Ciccarelli M, Huang ZM, Rabinowitz JE, Bridges CR. AAV6-βARKct gene delivery mediated by molecular cardiac surgery with recirculating delivery (MCARD) in sheep results in robust gene expression and increased adrenergic reserve. J Thorac Cardiovasc Surg 2011; 143:720-726.e3. [PMID: 22143102 DOI: 10.1016/j.jtcvs.2011.08.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 07/12/2011] [Accepted: 08/03/2011] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Genetic modulation of heart function is a novel therapeutic strategy. We investigated the effect of molecular cardiac surgery with recirculating delivery (MCARD)-mediated carboxyl-terminus of the β-adrenergic receptor kinase (βARKct) gene transfer on cardiac mechanoenergetics and β-adrenoreceptor (βAR) signaling. METHODS After baseline measurements, sheep underwent MCARD-mediated delivery of 10(14) genome copies of self-complimentary adeno-associated virus (scAAV6)-βARKct. Four and 8 weeks after MCARD, mechanoenergetic studies using magnetic resonance imaging were performed. Tissues were analyzed with real-time quantitative polymerase chain reaction (RT-qPCR) and Western blotting. βAR density, cyclic adenosine monophosphate levels, and physiologic parameters were evaluated. RESULTS There was a significant increase in dP/dt(max) at 4 weeks: 1384 ± 76 versus 1772 ± 182 mm Hg/s; and the increase persisted at 8 weeks in response to isoproterenol (P < .05). Similarly, the magnitude of dP/dt(min) increased at both 4 weeks and 8 weeks with isoproterenol stimulation (P < .05). At 8 weeks, potential energy was conserved, whereas in controls there was a decrease in potential energy (P < .05) in response to isoproterenol. RT-qPCR confirmed robustness of βARKct expression throughout the left ventricle and undetectable expression in extracardiac tissues. Quantitative Western blot data confirmed higher expression of βARKct in the left ventricle: 0.46 ± 0.05 versus 0.00 in lung and liver (P < .05). Survival was 100% and laboratory parameters of major organ function were within normal limits. CONCLUSIONS MCARD-mediated βARKct delivery is safe, results in robust cardiac-specific gene expression, enhances cardiac contractility and lusitropy, increases adrenergic reserve, and improves energy utilization efficiency in a preclinical large animal model.
Collapse
Affiliation(s)
- Michael G Katz
- Division of Cardiovascular Surgery, Department of Surgery, the University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Agüero J, Almenar L, Montó F, Oliver E, Sánchez-Lázaro I, Vicente D, Martínez-Dolz L, D'Ocon P, Rueda J, Salvador A. Myocardial G protein receptor-coupled kinase expression correlates with functional parameters and clinical severity in advanced heart failure. J Card Fail 2011; 18:53-61. [PMID: 22196842 DOI: 10.1016/j.cardfail.2011.10.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 09/04/2011] [Accepted: 10/07/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND In heart failure (HF), sympathetic hyperactivation induces deleterious effects in myocardial β-adrenergic signaling, with receptor down-regulation and desensitization mediated by G protein receptor-coupled kinases (GRKs). We hypothesised that changes in GRK isoforms may be associated with clinical status in advanced HF, using the Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS) scale. METHODS We included 31 patients with advanced HF undergoing transplantation. According to INTERMACS profiles, mRNA and protein levels of GRK isoforms in left ventricular (LV) myocardium were analyzed and compared with nonfailing LV samples. RESULTS In failing LV myocardium, GRK2 and GRK5 (but not GRK3) protein was up-regulated compared with control samples. Among HF patients, an increase in GRK2 and GRK5 mRNA and protein abundance was observed in β-agonist-treated patients (vs β-blockers: P < .05) and in higher-risk INTERMACS status (profiles 2 and 3 vs 4 and 5: P < .05). A significant negative correlation of GRK2 expression with LV stroke volume supported these findings. CONCLUSIONS Increased GRK2 correlates with clinical severity using the INTERMACS scale and LV stroke volume, supporting it as a potential target in advanced HF. These changes are paralleled by GRK5 expression in the failing myocardium, suggesting a relevant role in human HF.
Collapse
Affiliation(s)
- Jaime Agüero
- Heart Failure and Transplant Unit, Department of Cardiology, La Fe University Hospital, Valencia, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Abstract
G protein-coupled receptors (GPCRs) represent the largest family of membrane receptors and are responsible for regulating a wide variety of physiological processes. This is accomplished via ligand binding to GPCRs, activating associated heterotrimeric G proteins and intracellular signaling pathways. G protein-coupled receptor kinases (GRKs), in concert with β-arrestins, classically desensitize receptor signal transduction, thus preventing hyperactivation of GPCR second-messenger cascades. As changes in GRK expression have featured prominently in many cardiovascular pathologies, including heart failure, myocardial infarction, hypertension, and cardiac hypertrophy, GRKs have been intensively studied as potential diagnostic or therapeutic targets. Herein, we review our evolving understanding of the role of GRKs in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Stephen L Belmonte
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | |
Collapse
|
134
|
Raake PWJ, Tscheschner H, Reinkober J, Ritterhoff J, Katus HA, Koch WJ, Most P. Gene therapy targets in heart failure: the path to translation. Clin Pharmacol Ther 2011; 90:542-53. [PMID: 21866097 DOI: 10.1038/clpt.2011.148] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heart failure (HF) is the common end point of cardiac diseases. Despite the optimization of therapeutic strategies and the consequent overall reduction in HF-related mortality, the key underlying intracellular signal transduction abnormalities have not been addressed directly. In this regard, the gaps in modern HF therapy include derangement of β-adrenergic receptor (β-AR) signaling, Ca(2+) disbalances, cardiac myocyte death, diastolic dysfunction, and monogenetic cardiomyopathies. In this review we discuss the potential of gene therapy to fill these gaps and rectify abnormalities in intracellular signaling. We also examine current vector technology and currently available vector-delivery strategies, and we delineate promising gene therapy structures. Finally, we analyze potential limitations related to the transfer of successful preclinical gene therapy approaches to HF treatment in the clinic, as well as impending strategies aimed at overcoming these limitations.
Collapse
Affiliation(s)
- P W J Raake
- Division of Cardiology, Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
135
|
AAV vectors for cardiac gene transfer: experimental tools and clinical opportunities. Mol Ther 2011; 19:1582-90. [PMID: 21792180 DOI: 10.1038/mt.2011.124] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Since the first demonstration of in vivo gene transfer into myocardium there have been a series of advancements that have driven the evolution of cardiac gene delivery from an experimental tool into a therapy currently at the threshold of becoming a viable clinical option. Innovative methods have been established to address practical challenges related to tissue-type specificity, choice of delivery vehicle, potency of the delivered material, and delivery route. Most importantly for therapeutic purposes, these strategies are being thoroughly tested to ensure safety of the delivery system and the delivered genetic material. This review focuses on the development of recombinant adeno-associated virus (rAAV) as one of the most valuable cardiac gene transfer agents available today. Various forms of rAAV have been used to deliver "pre-event" cardiac protection and to temper the severity of hypertrophy, cardiac ischemia, or infarct size. Adeno-associated virus (AAV) vectors have also been functional delivery tools for cardiac gene expression knockdown studies and successfully improving the cardiac aspects of several metabolic and neuromuscular diseases. Viral capsid manipulations along with the development of tissue-specific and regulated promoters have greatly increased the utility of rAAV-mediated gene transfer. Important clinical studies are currently underway to evaluate AAV-based cardiac gene delivery in humans.
Collapse
|
136
|
Brinks H, Das A, Koch WJ. A role for GRK2 in myocardial ischemic injury: indicators of a potential future therapy and diagnostic. Future Cardiol 2011; 7:547-56. [DOI: 10.2217/fca.11.36] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Morbidity and mortality of myocardial infarction remains significant with resulting left ventricular function presenting as a major determinant of clinical outcome. Protecting the myocardium against ischemia reperfusion injury has become a major therapeutic goal and the identification of key signaling pathways has paved the way for various interventions, but until now with disappointing results. This article describes the recently discovered new role of G-protein-coupled receptor kinase-2 (GRK2), which is known to critically influence the development and progression of heart failure, in acute myocardial injury. This article focuses on potential applications of the GRK2 peptide inhibitor βARKct in ischemic myocardial injury, the use of GRK2 as a biomarker in acute myocardial infarction and discusses the challenges of translating GRK2 inhibition as a cardioprotective strategy to a possible future clinical application.
Collapse
Affiliation(s)
- Henriette Brinks
- Department of Cardiovascular Surgery, Inselspital University Hospital, Berne, Switzerland
| | - Amrit Das
- Center for Translational Medicine, Department of Internal Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Walter J Koch
- Center for Translational Medicine, Department of Internal Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
137
|
Lymperopoulos A. GRK2 and β-arrestins in cardiovascular disease: Something old, something new. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2011; 1:126-137. [PMID: 22254193 PMCID: PMC3253495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 06/23/2011] [Indexed: 05/31/2023]
Abstract
Heptahelical G protein-coupled receptors are the most diverse and therapeutically important family of receptors in the human genome, playing major roles in the physiology of various organs/tissues including the heart and blood vessels. Ligand binding activates heterotrimeric G proteins that transmit intracellular signals by regulating effector enzymes or ion channels. G protein signaling is terminated, in large part, by phosphorylation of the agonist-bound receptor by the family of G-protein coupled receptor kinases (GRKs) followed by βarrestin binding, which uncouples the phosphorylated receptor and G protein and subsequently targets the receptor for internalization. As the receptor-βarrestin complex enters the cell, βarrestins serve as ligand-regulated scaffolds that recruit a host of intracellular proteins and signal transducers, thus promoting their own wave of signal transduction independently of G-proteins. A large number of preclinical studies in small and large animals over the past several years have pinpointed specific pathophysiologic roles played by these two families of receptor-regulating proteins in various cardiovascular diseases, directly implicating them in disease pathology and suggesting them as potential therapeutic targets. The present review gives an account of what is currently known about the benefits of cardiac GRK2 inhibition for cardiovascular disease treatment, and also discusses the exciting new therapeutic possibilities emerging from uncovering the physiological roles of adrenal GRK2 and of βarrestin-mediated signaling in vivo in the cardiovascular system.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy Ft. Lauderdale, FL 33328, USA
| |
Collapse
|
138
|
Fargnoli AS, Katz MG, Yarnall C, Sumaroka MV, Stedman H, Rabinowitz JJ, Koch WJ, Bridges CR. A pharmacokinetic analysis of molecular cardiac surgery with recirculation mediated delivery of βARKct gene therapy: developing a quantitative definition of the therapeutic window. J Card Fail 2011; 17:691-9. [PMID: 21807332 DOI: 10.1016/j.cardfail.2011.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/03/2011] [Accepted: 03/25/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Two major problems for translating gene therapy for heart failure therapy are: safe and efficient delivery and the inability to establish a relationship between vector exposure and in vivo effects. We present a pharmacokinetics (PK) analysis of molecular cardiac surgery with recirculating delivery (MCARD) of scAAV6-βARKct. MCARD's stable cardiac specific delivery profile was exploited to determine vector exposure, half-life, and systemic clearance. METHODS AND RESULTS Five naive sheep underwent MCARD with 10(14) genome copies of scAAV6-βARKct. Blood samples were collected over the recirculation interval time of 20 minutes and evaluated with quantitative polymerase chain reaction (qPCR). C(t) curves were generated and expressed on a log scale. The exposure, half-life, and clearance curves were generated for analysis. qPCR and Western blots were used to determine biodistribution. Finally, all in vivo transduction data was plotted against MCARD's PK to determine if a relationship existed. Vector concentrations at each time point were (cardiac and systemic, respectively): 5 minutes: 9.16 ± 0.15 and 3.21 ± 0.38; 10 minutes: 8.81 ± 0.19 and 3.62 ± 0.37; 15 minutes: 8.75 ± 0.12 and 3.69 ± 0.31; and 20 minutes: 8.66 ± 0.22 and 3.95 ± 0.26; P < .00001. The half life of the vector was 2.66 ± 0.24 minutes. PK model data revealed that only 0.61 ± 0.43% of the original dose remained in the blood after delivery, and complete clearance from the system was achieved at 1 week. A PK transfer function revealed a positive correlation between exposure and in vivo transduction. Robust βARKct expression was found in all cardiac regions with none in the liver. CONCLUSION MCARD may offer a viable method to establish a relationship between vector exposure and in vivo transduction. Using this methodology, it may be possible to address a critical need for establishing an effective therapeutic window.
Collapse
Affiliation(s)
- Anthony S Fargnoli
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Huang ZM, Gold JI, Koch WJ. G protein-coupled receptor kinases in normal and failing myocardium. Front Biosci (Landmark Ed) 2011; 16:3047-60. [PMID: 21622221 DOI: 10.2741/3898] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heart failure (HF) is the end stage of many underlying cardiovascular diseases and is among the leading causes of morbidity and mortality in industrialized countries. One of the striking characteristics of HF is the desensitization of G protein-coupled receptor (GPCR) signaling, particularly the beta-adrenergic receptor (betaAR) system. GPCR desensitization is initiated by phosphorylation by GPCR kinases (GRKs), followed by downregulation and functional uncoupling from their G proteins. In the heart, the major GRK isoforms, GRK2 and GRK5, undergo upregulation due to the heightened sympathetic nervous system activity that is characteristic of HF as catecholamine levels increase in an effort to drive the failing pump. This desensitization leads to the distinctive loss of inotropic reserve and functional capacity of the failing heart. Moreover, GRK2 and GRK5 have an increasing non-GPCR interactome, which may play critical roles in cardiac physiology. In the current review, the canonical GPCR kinase function of GRKs and the novel non-GPCR kinase activity of GRKs, their contribution to the pathogenesis of cardiac hypertrophy and HF, and the possibility of GRKs serving as future drug targets will be discussed.
Collapse
Affiliation(s)
- Zheng Maggie Huang
- Center for Translational Medicine and George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | |
Collapse
|
140
|
Ciccarelli M, Chuprun JK, Rengo G, Gao E, Wei Z, Peroutka RJ, Gold J, Gumpert A, Chen M, Otis NJ, Dorn GW, Trimarco B, Iaccarino G, Koch WJ. G protein-coupled receptor kinase 2 activity impairs cardiac glucose uptake and promotes insulin resistance after myocardial ischemia. Circulation 2011; 123:1953-62. [PMID: 21518983 PMCID: PMC3113597 DOI: 10.1161/circulationaha.110.988642] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 03/01/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND Alterations in cardiac energy metabolism downstream of neurohormonal stimulation play a crucial role in the pathogenesis of heart failure. The chronic adrenergic stimulation that accompanies heart failure is a signaling abnormality that leads to the upregulation of G protein-coupled receptor kinase 2 (GRK2), which is pathological in the myocyte during disease progression in part owing to uncoupling of the β-adrenergic receptor system. In this study, we explored the possibility that enhanced GRK2 expression and activity, as seen during heart failure, can negatively affect cardiac metabolism as part of its pathogenic profile. METHODS AND RESULTS Positron emission tomography studies revealed in transgenic mice that cardiac-specific overexpression of GRK2 negatively affected cardiac metabolism by inhibiting glucose uptake and desensitization of insulin signaling, which increases after ischemic injury and precedes heart failure development. Mechanistically, GRK2 interacts with and directly phosphorylates insulin receptor substrate-1 in cardiomyocytes, causing insulin-dependent negative signaling feedback, including inhibition of membrane translocation of the glucose transporter GLUT4. This identifies insulin receptor substrate-1 as a novel nonreceptor target for GRK2 and represents a new pathological mechanism for this kinase in the failing heart. Importantly, inhibition of GRK2 activity prevents postischemic defects in myocardial insulin signaling and improves cardiac metabolism via normalized glucose uptake, which appears to participate in GRK2-targeted prevention of heart failure. CONCLUSIONS Our data provide novel insights into how GRK2 is pathological in the injured heart. Moreover, it appears to be a critical mechanistic link within neurohormonal crosstalk governing cardiac contractile signaling/function through β-adrenergic receptors and metabolism through the insulin receptor.
Collapse
Affiliation(s)
- Michele Ciccarelli
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Clinical Medicine and Cardiovascular Science, “Federico II” University of Naples, Italy
| | - J. Kurt Chuprun
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Giuseppe Rengo
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
- Division of Cardiology, Fondazione “Salvatore Maugeri” – IRCCS – Istituto di Telese Terme – Italy
| | - Erhe Gao
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhengyu Wei
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Raymond J. Peroutka
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessica Gold
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anna Gumpert
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mai Chen
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nicholas J. Otis
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gerald W. Dorn
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruno Trimarco
- Department of Clinical Medicine and Cardiovascular Science, “Federico II” University of Naples, Italy
| | - Guido Iaccarino
- University of Salerno, Department of Medicine, Salerno, Italy Ciccarelli, GRK2 and Cardiac Metabolism
| | - Walter J. Koch
- George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
141
|
Rengo G, Lymperopoulos A, Leosco D, Koch WJ. GRK2 as a novel gene therapy target in heart failure. J Mol Cell Cardiol 2011; 50:785-92. [PMID: 20800067 PMCID: PMC3005526 DOI: 10.1016/j.yjmcc.2010.08.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 08/16/2010] [Accepted: 08/17/2010] [Indexed: 12/13/2022]
Abstract
Despite significant advances in pharmacological and clinical treatment, heart failure (HF) remains a leading cause of morbidity and mortality worldwide. HF is a chronic and progressive clinical syndrome characterized by a reduction in left ventricular (LV) ejection fraction and adverse remodeling of the myocardium. The past several years have seen remarkable progress using animal models in unraveling the cellular and molecular mechanisms underlying HF pathogenesis and progression. These studies have revealed potentially novel therapeutic targets/strategies. The application of cardiac gene transfer, which allows for the manipulation of targets in cardiomyocytes, appears to be a promising therapeutic tool in HF. β-adrenergic receptor (βAR) dysfunction represents a hallmark abnormality of chronic HF, and increased G protein-coupled receptor kinase 2 (GRK2) levels/activity in failing myocardium is among these alterations. In the past 15years, several animal studies have shown that expression of a peptide inhibitor of GRK2 (βARKct) can improve the contractile function of failing myocardium including promoting reverse remodeling of the LV. Therefore, data support the use of the βARKct as a promising candidate for therapeutic application in human HF. Importantly, recent studies in cardiac-specific GRK2 knockout mice have corroborated GRK2 being pathological in failing myocytes. The purpose of this review is to discuss: 1) the alterations of βAR signaling that occur in HF, 2) the evidence from transgenic mouse studies investigating the impact of GRK2 manipulation in failing myocardium, 3) the therapeutic efficacy of in vivo βARKct gene therapy in HF, and 4) the intriguing possibility of lowering HF-related sympathetic nervous system hyperactivity by inhibiting GRK2 activity in the adrenal gland. This article is part of a Special Section entitled "Special Section: Cardiovascular Gene Therapy".
Collapse
Affiliation(s)
- Giuseppe Rengo
- Center for Translational Medicine and George Zallie and Family Laboratory for Cardiovascular Gene Therapy, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
142
|
Jasmin JF, Rengo G, Lymperopoulos A, Gupta R, Eaton GJ, Quann K, Gonzales DM, Mercier I, Koch WJ, Lisanti MP. Caveolin-1 deficiency exacerbates cardiac dysfunction and reduces survival in mice with myocardial infarction. Am J Physiol Heart Circ Physiol 2011; 300:H1274-H1281. [PMID: 21297026 PMCID: PMC3075024 DOI: 10.1152/ajpheart.01173.2010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 01/28/2011] [Indexed: 11/22/2022]
Abstract
Caveolin (Cav)-1 has been involved in the pathogenesis of ischemic injuries. For instance, modulations of Cav-1 expression have been reported in animal models of myocardial infarction and cerebral ischemia-reperfusion. Furthermore, ablation of the Cav-1 gene in mice has been shown to increase the extent of ischemic injury in models of cerebral and hindlimb ischemia. Cav-1 has also been suggested to play a role in myocardial ischemic preconditioning. However, the role of Cav-1 in myocardial ischemia (MI)-induced cardiac dysfunction still remains to be determined. We determined the outcome of a permanent left anterior descending coronary artery (LAD) ligation in Cav-1 knockout (KO) mice. Wild-type (WT) and Cav-1 KO mice were subjected to permanent LAD ligation for 24 h. The progression of ischemic injury was monitored by echocardiography, hemodynamic measurements, 2,3,5-triphenyltetrazolium chloride staining, β-binding analysis, cAMP level measurements, and Western blot analyses. Cav-1 KO mice subjected to LAD ligation display reduced survival compared with WT mice. Despite similar infarct sizes, Cav-1 KO mice subjected to MI showed reduced left ventricular (LV) ejection fraction and fractional shortening as well as increased LV end-diastolic pressures compared with their WT counterparts. Mechanistically, Cav-1 KO mice subjected to MI exhibit reduced β-adrenergic receptor density at the plasma membrane as well as decreased cAMP levels and PKA phosphorylation. In conclusion, ablation of the Cav-1 gene exacerbates cardiac dysfunction and reduces survival in mice subjected to MI. Mechanistically, Cav-1 KO mice subjected to LAD ligation display abnormalities in β-adrenergic signaling.
Collapse
Affiliation(s)
- Jean-François Jasmin
- Department of Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Briston SJ, Caldwell JL, Horn MA, Clarke JD, Richards MA, Greensmith DJ, Graham HK, Hall MCS, Eisner DA, Dibb KM, Trafford AW. Impaired β-adrenergic responsiveness accentuates dysfunctional excitation-contraction coupling in an ovine model of tachypacing-induced heart failure. J Physiol 2011; 589:1367-82. [PMID: 21242250 PMCID: PMC3082097 DOI: 10.1113/jphysiol.2010.203984] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 01/10/2011] [Indexed: 01/08/2023] Open
Abstract
Reduced inotropic responsiveness is characteristic of heart failure (HF). This study determined the cellular Ca2+ homeostatic and molecular mechanisms causing the blunted β-adrenergic (β-AR) response in HF.We induced HF by tachypacing in sheep; intracellular Ca2+ concentration was measured in voltage-clamped ventricular myocytes. In HF, Ca2+ transient amplitude and peak L-type Ca2+ current (ICa-L) were reduced (to 70 ± 11% and 50 ± 3.7% of control, respectively, P <0.05) whereas sarcoplasmic reticulum (SR) Ca2+ content was unchanged. β-AR stimulation with isoprenaline (ISO) increased Ca2+ transient amplitude, ICa-L and SRCa2+ content in both cell types; however, the response of HF cells was markedly diminished (P <0.05).Western blotting revealed an increase in protein phosphatase levels (PP1, 158 ± 17% and PP2A, 188 ± 34% of control, P <0.05) and reduced phosphorylation of phospholamban in HF (Ser16, 30 ± 10% and Thr17, 41 ± 15% of control, P <0.05). The β-AR receptor kinase GRK-2 was also increased in HF (173 ± 38% of control, P <0.05). In HF, activation of adenylyl cyclase with forskolin rescued the Ca2+ transient, SR Ca2+ content and SR Ca2+ uptake rate to the same levels as control cells in ISO. In conclusion, the reduced responsiveness of the myocardium to β-AR agonists in HF probably arises as a consequence of impaired phosphorylation of key intracellular proteins responsible for regulating the SR Ca2+ content and therefore failure of the systolic Ca2+ transient to increase appropriately during β-AR stimulation.
Collapse
Affiliation(s)
- Sarah J Briston
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9NT, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
D'Souza KM, Malhotra R, Philip JL, Staron ML, Theccanat T, Jeevanandam V, Akhter SA. G protein-coupled receptor kinase-2 is a novel regulator of collagen synthesis in adult human cardiac fibroblasts. J Biol Chem 2011; 286:15507-16. [PMID: 21357420 DOI: 10.1074/jbc.m111.218263] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cardiac fibroblasts (CF) make up 60-70% of the total cell number in the heart and play a critical role in regulating normal myocardial function and in adverse remodeling following myocardial infarction and the transition to heart failure. Recent studies have shown that increased intracellular cAMP can inhibit CF transformation and collagen synthesis in adult rat CF; however, mechanisms by which cAMP production is regulated in CF have not been elucidated. We investigated the potential role of G protein-coupled receptor kinase-2 (GRK2) in modulating collagen synthesis by adult human CF isolated from normal and failing left ventricles. Baseline collagen synthesis was elevated in failing CF and was not inhibited by β-agonist stimulation in contrast to normal controls. β-adrenergic receptor (β-AR) signaling was markedly uncoupled in the failing CF, and expression and activity of GRK2 were increased 3-fold. Overexpression of GRK2 in normal CF recapitulated a heart failure phenotype with minimal inhibition of collagen synthesis following β-agonist stimulation. In contrast, knockdown of GRK2 expression in normal CF enhanced cAMP production and led to greater β-agonist-mediated inhibition of basal and TGFβ-stimulated collagen synthesis versus control. Inhibition of GRK2 activity in failing CF by expression of the GRK2 inhibitor, GRK2ct, or siRNA-mediated knockdown restored β-agonist-stimulated inhibition of collagen synthesis and decreased collagen synthesis in response to TGFβ stimulation. GRK2 appears to play a significant role in regulating collagen synthesis in adult human CF, and increased activity of this kinase may be an important mechanism of maladaptive ventricular remodeling as mediated by cardiac fibroblasts.
Collapse
Affiliation(s)
- Karen M D'Souza
- Department of Surgery, Section of Cardiac and Thoracic Surgery, University of Chicago Medical Center, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
Heart failure is an important cause of morbidity and mortality in individuals of all ages. The many-faceted nature of the clinical heart failure syndrome has historically frustrated attempts to develop an overarching explanative theory. However, much useful information has been gained by basic and clinical investigation, even though a comprehensive understanding of heart failure has been elusive. Heart failure is a growing problem, in both adult and pediatric populations, for which standard medical therapy, as of 2010, can have positive effects, but these are usually limited and progressively diminish with time in most patients. If we want curative or near-curative therapy that will return patients to a normal state of health at a feasible cost, much better diagnostic and therapeutic technologies need to be developed. This review addresses the vexing group of heart failure etiologies that include cardiomyopathies and other ventricular dysfunctions of various types, for which current therapy is only modestly effective. Although there are many unique aspects to heart failure in patients with pediatric and congenital heart disease, many of the innovative approaches that are being developed for the care of adults with heart failure will be applicable to heart failure in childhood.
Collapse
Affiliation(s)
- Daniel J Penny
- Section of Pediatric Cardiology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, 6621 Fannin Street, Houston, TX 77030, USA
| | | |
Collapse
|
146
|
Lymperopoulos A, Rengo G, Zincarelli C, Kim J, Koch WJ. Adrenal beta-arrestin 1 inhibition in vivo attenuates post-myocardial infarction progression to heart failure and adverse remodeling via reduction of circulating aldosterone levels. J Am Coll Cardiol 2011; 57:356-65. [PMID: 21232674 PMCID: PMC3087631 DOI: 10.1016/j.jacc.2010.08.635] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 08/13/2010] [Accepted: 08/17/2010] [Indexed: 12/31/2022]
Abstract
OBJECTIVES We investigated whether adrenal beta-arrestin 1 (βarr1)-mediated aldosterone production plays any role in post-myocardial infarction (MI) heart failure (HF) progression. BACKGROUND Heart failure represents 1 of the most significant health problems worldwide, and new and innovative treatments are needed. Aldosterone contributes significantly to HF progression after MI by accelerating adverse cardiac remodeling and ventricular dysfunction. It is produced by the adrenal cortex after angiotensin II activation of angiotensin II type 1 receptors (AT₁Rs), G protein-coupled receptors that also signal independently of G proteins. The G protein-independent signaling is mediated by βarr1 and βarr2. We recently reported that adrenal βarr1 promotes AT₁R-dependent aldosterone production leading to elevated circulating aldosterone levels in vivo. METHODS Adrenal-targeted, adenoviral-mediated gene delivery in vivo in 2-week post-MI rats, a time point around which circulating aldosterone significantly increases to accelerate HF progression, was performed to either increase the expression of adrenal βarr1 or inhibit its function via expression of a βarr1 C-terminal-derived peptide fragment. RESULTS We found that adrenal βarr1 overexpression promotes aldosterone elevation after MI, resulting in accelerated cardiac adverse remodeling and deterioration of ventricular function. Importantly, these detrimental effects of aldosterone are prevented when adrenal βarr1 is inhibited in vivo, which markedly decreases circulating aldosterone after MI. Finally, the prototypic AT₁R antagonist losartan seems unable to lower this adrenal βarr1-driven aldosterone elevation. CONCLUSIONS Adrenal βarr1 inhibition, either directly or with AT₁R "biased" antagonists that prevent receptor-βarr1 coupling, might be of therapeutic value for curbing HF-exacerbating hyperaldosteronism.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida 33328, USA.
| | | | | | | | | |
Collapse
|
147
|
Völkers M, Weidenhammer C, Herzog N, Qiu G, Spaich K, Wegner FV, Peppel K, Müller OJ, Schinkel S, Rabinowitz JE, Hippe HJ, Brinks H, Katus HA, Koch WJ, Eckhart AD, Friedrich O, Most P. The inotropic peptide βARKct improves βAR responsiveness in normal and failing cardiomyocytes through G(βγ)-mediated L-type calcium current disinhibition. Circ Res 2011; 108:27-39. [PMID: 21106943 PMCID: PMC4013502 DOI: 10.1161/circresaha.110.225201] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 11/15/2010] [Indexed: 12/20/2022]
Abstract
RATIONALE The G(βγ)-sequestering peptide β-adrenergic receptor kinase (βARK)ct derived from the G-protein-coupled receptor kinase (GRK)2 carboxyl terminus has emerged as a promising target for gene-based heart failure therapy. Enhanced downstream cAMP signaling has been proposed as the underlying mechanism for increased β-adrenergic receptor (βAR) responsiveness. However, molecular targets mediating improved cardiac contractile performance by βARKct and its impact on G(βγ)-mediated signaling have yet to be fully elucidated. OBJECTIVE We sought to identify G(βγ)-regulated targets and signaling mechanisms conveying βARKct-mediated enhanced βAR responsiveness in normal (NC) and failing (FC) adult rat ventricular cardiomyocytes. METHODS AND RESULTS Assessing viral-based βARKct gene delivery with electrophysiological techniques, analysis of contractile performance, subcellular Ca²(+) handling, and site-specific protein phosphorylation, we demonstrate that βARKct enhances the cardiac L-type Ca²(+) channel (LCC) current (I(Ca)) both in NCs and FCs on βAR stimulation. Mechanistically, βARKct augments I(Ca) by preventing enhanced inhibitory interaction between the α1-LCC subunit (Cav1.2α) and liberated G(βγ) subunits downstream of activated βARs. Despite improved βAR contractile responsiveness, βARKct neither increased nor restored cAMP-dependent protein kinase (PKA) and calmodulin-dependent kinase II signaling including unchanged protein kinase (PK)Cε, extracellular signal-regulated kinase (ERK)1/2, Akt, ERK5, and p38 activation both in NCs and FCs. Accordingly, although βARKct significantly increases I(Ca) and Ca²(+) transients, being susceptible to suppression by recombinant G(βγ) protein and use-dependent LCC blocker, βARKct-expressing cardiomyocytes exhibit equal basal and βAR-stimulated sarcoplasmic reticulum Ca²(+) load, spontaneous diastolic Ca²(+) leakage, and survival rates and were less susceptible to field-stimulated Ca²(+) waves compared with controls. CONCLUSION Our study identifies a G(βγ)-dependent signaling pathway attenuating cardiomyocyte I(Ca) on βAR as molecular target for the G(βγ)-sequestering peptide βARKct. Targeted interruption of this inhibitory signaling pathway by βARKct confers improved βAR contractile responsiveness through increased I(Ca) without enhancing regular or restoring abnormal cAMP-signaling. βARKct-mediated improvement of I(Ca) rendered cardiomyocytes neither susceptible to βAR-induced damage nor arrhythmogenic sarcoplasmic reticulum Ca²(+) leakage.
Collapse
Affiliation(s)
- Mirko Völkers
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Christian Weidenhammer
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Nicole Herzog
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Gang Qiu
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Kristin Spaich
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Frederic V Wegner
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Karsten Peppel
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Oliver J Müller
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Stefanie Schinkel
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Joseph E Rabinowitz
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Hans-Jorg Hippe
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Henriette Brinks
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Hugo A Katus
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Walter J Koch
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Andrea D Eckhart
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Oliver Friedrich
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Patrick Most
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| |
Collapse
|
148
|
Penela P, Murga C, Ribas C, Lafarga V, Mayor F. The complex G protein-coupled receptor kinase 2 (GRK2) interactome unveils new physiopathological targets. Br J Pharmacol 2010; 160:821-32. [PMID: 20590581 DOI: 10.1111/j.1476-5381.2010.00727.x] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
GRK2 is a ubiquitous member of the G protein-coupled receptor kinase (GRK) family that appears to play a central, integrative role in signal transduction cascades. GRKs participate together with arrestins in the regulation of G protein-coupled receptors (GPCR), a family of hundreds of membrane proteins of key physiological and pharmacological importance, by triggering receptor desensitization from G proteins and GPCR internalization, and also by helping assemble macromolecular signalosomes in the receptor environment acting as agonist-regulated adaptor scaffolds, thus contributing to signal propagation. In addition, emerging evidence indicates that GRK2 can phosphorylate a growing number of non-GPCR substrates and associate with a variety of proteins related to signal transduction, thus suggesting that this kinase could also have diverse 'effector' functions. We discuss herein the increasing complexity of such GRK2 'interactome', with emphasis on the recently reported roles of this kinase in cell migration and cell cycle progression and on the functional impact of the altered GRK2 levels observed in several relevant cardiovascular, inflammatory or tumour pathologies. Deciphering how the different networks of potential GRK2 functional interactions are orchestrated in a stimulus, cell type or context-specific way is critical to unveil the contribution of GRK2 to basic cellular processes, to understand how alterations in GRK2 levels or functionality may participate in the onset or development of several cardiovascular, tumour or inflammatory diseases, and to assess the feasibility of new therapeutic strategies based on the modulation of the activity, levels or specific interactions of GRK2.
Collapse
Affiliation(s)
- Petronila Penela
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Universidad Autónoma, Madrid, Spain
| | | | | | | | | |
Collapse
|
149
|
Zincarelli C, Soltys S, Rengo G, Koch WJ, Rabinowitz JE. Comparative cardiac gene delivery of adeno-associated virus serotypes 1-9 reveals that AAV6 mediates the most efficient transduction in mouse heart. Clin Transl Sci 2010; 3:81-9. [PMID: 20590676 DOI: 10.1111/j.1752-8062.2010.00190.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Cardiac gene transfer is an attractive tool for developing novel heart disease treatments. Adeno-associated viral (AAV) vectors are widely used to mediate transgene expression in animal models and are being evaluated for human gene therapy. However, it is not clear which serotype displays the best cardiac tropism. Therefore, we curried out this study to directly compare AAV serotypes 1-9 heart transduction efficiency after indirect intracoronary injection. AAV-cytomegalovirus immediate early enhancer promoter (CMV)-luciferase serotypes 1-9 were injected in the left ventricular cavity of adult mice, after cross-clamping the ascending aorta and pulmonary artery. An imaging system was used to visualize luciferase expression at 3, 7, 21, 70, and 140 days postinjection. Echocardiography was performed to evaluate cardiac function on day 140. At the end of the study, luciferase enzyme activity and genome copies of the different AAV serotypes were assessed in several tissues and potential AAV immunogenicity was evaluated on heart sections by staining for macrophage and lymphocyte antigens. Among AAV serotypes 1-9, AAV6 showed the best capability of achieving high transduction levels in the myocardium in a tissue-specific manner, whereas the other serotypes had less cardiac transduction and more extracardiac expression, especially in the liver. Importantly, none of the serotypes tested with this marker gene affected cardiac function nor was associated with inflammation.
Collapse
Affiliation(s)
- Carmela Zincarelli
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
150
|
Katz MG, Swain JD, Tomasulo CE, Sumaroka M, Fargnoli A, Bridges CR. Current strategies for myocardial gene delivery. J Mol Cell Cardiol 2010; 50:766-76. [PMID: 20837022 DOI: 10.1016/j.yjmcc.2010.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 08/30/2010] [Accepted: 09/01/2010] [Indexed: 10/19/2022]
Abstract
Existing methods of cardiac gene delivery can be classified by the site of injection, interventional approach and type of cardiac circulation at the time of transfer. General criteria to assess the efficacy of a given delivery method include: global versus regional myocardial transduction, technical complexity and the pathophysiological effects associated with its use, delivery-related collateral expression and the delivery-associated inflammatory and immune response. Direct gene delivery (intramyocardial, endocardial, epicardial) may be useful for therapeutic angiogenesis and for focal arrhythmia therapy but with gene expression which is primarily limited to regions in close proximity to the injection site. An often unappreciated limitation of these techniques is that they are frequently associated with substantial systemic vector delivery. Percutaneous infusion of vector into the coronary arteries is minimally invasive and allows for transgene delivery to the whole myocardium. Unfortunately, efficiency of intracoronary delivery is highly variable and the short residence time of vector within the coronary circulation and significant collateral organ expression limit its clinical potential. Surgical techniques, including the incorporation of cardiopulmonary bypass with isolated cardiac recirculation, represent novel delivery strategies that may potentially overcome these limitations; yet, these techniques are complex with inherent morbidity that must be thoroughly evaluated before safe translation into clinical practice. Characteristics of the optimal technique for gene delivery include low morbidity, increased myocardial transcapillary gradient, extended vector residence time in the coronary circulation and exclusion of residual vector from the systemic circulation after delivery to minimize extracardiac expression and to mitigate a cellular immune response. This article is part of a Special Section entitled "Special Section: Cardiovascular Gene Therapy".
Collapse
Affiliation(s)
- Michael G Katz
- Department of Surgery, Division of Cardiovascular Surgery, The University of Pennsylvania Medical Center, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|