1
|
Snyder J, Jiang CS, Choi RH, Morgan T, Roman J, Underwood L, Lucchese AM, Montgomery S, Grisanti LA, Doliba N, Holland WL, Sato PY. Cardioprotective effect of genetic ablation of the G-protein-coupled receptor kinase GRK2 in adult pancreatic β-cells during high-fat diet. J Biol Chem 2025; 301:108388. [PMID: 40054692 PMCID: PMC12018985 DOI: 10.1016/j.jbc.2025.108388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 04/17/2025] Open
Abstract
Cardiovascular diseases are a major comorbidity factor in patients with type 2 diabetes and a leading cause of death among them. Yet, mechanistically, how impairment in pancreatic islets alters cardiac function under different metabolic states remains largely unknown. Here, we investigate the role of the G-protein-coupled receptor kinase 2 (GRK2) in regulating islet adaptations to an obesogenic diet and its impact on myocardial function. Using a novel inducible β-cell-specific GRK2 knockout mouse model (βGRK2KO), we establish that loss of adult β-cell GRK2 delays metabolic islet maladaptation, protecting the heart against obesity-induced cardiac dysfunction. βGRK2KO are more insulin-sensitive than control mice and have improved cardiac function and myocardial morphology. Thus, genetic ablation of GRK2 in adult β-cells during an obesogenic diet play a cardioprotective role. This study prompts a novel therapeutic window for GRK2 intervention strategies for diabetic patients prone to cardiac dysfunction.
Collapse
Affiliation(s)
- Jonathan Snyder
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Chun-Sun Jiang
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ran Hee Choi
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - Taylor Morgan
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Jeffrey Roman
- Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lilly Underwood
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anna Maria Lucchese
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Sarah Montgomery
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Laurel A Grisanti
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Nicolai Doliba
- Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - Priscila Y Sato
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
2
|
Gatto C, Rusciano MR, Visco V, Ciccarelli M. GRK2 and Mitochondrial Dynamics in Cardiovascular Health and Disease. Int J Mol Sci 2025; 26:2299. [PMID: 40076919 PMCID: PMC11900936 DOI: 10.3390/ijms26052299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
G protein-coupled receptors (GPCRs) represent a family of membrane proteins that regulate several cellular processes. Among the GPCRs, G protein-coupled receptor kinases (GRKs) regulate downstream signaling pathways and receptor desensitization. GRK2 has gained significant interest due to its cardiovascular physiology and pathological involvement. GRK2's presence in cardiac tissue and its influence on cardiac function, β-adrenergic signaling, and myocardial remodeling underlies its involvement in cardiovascular diseases such as heart failure and ischemia. GRK2's canonical role is receptor desensitization, but emerging evidence suggests its involvement in mitochondrial dynamics and bioenergetics, influencing processes such as oxidative phosphorylation, reactive oxygen species production, and apoptosis. Moreover, GRK2's localization within mitochondria suggests a direct role in regulating mitochondrial health and function. Notably, while GRK2 inhibition seems to be a therapeutic approach to heart failure, its precise role in mitochondrial dynamics and pathology needs further investigation. This review explores the complex relationship between mitochondrial function and GRK2 and clarifies the implications for cardiovascular health. Cardiovascular medicine might greatly benefit from future studies that focus on understanding the processes behind GRK2-mitochondrial crosstalk to develop personalized therapies.
Collapse
Affiliation(s)
- Cristina Gatto
- Department of Medicine, Surgery and Dentistry, University of Salerno “Scuola Medica Salernitana”, 84081 Baronissi, Italy; (C.G.); (M.R.R.); (V.V.)
- Scuola di Specializzazione in Patologia Clinica e Biochimica Clinica, University of Salerno “Scuola Medica Salernitana”, 84081 Baronissi, Italy
| | - Maria Rosaria Rusciano
- Department of Medicine, Surgery and Dentistry, University of Salerno “Scuola Medica Salernitana”, 84081 Baronissi, Italy; (C.G.); (M.R.R.); (V.V.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry, University of Salerno “Scuola Medica Salernitana”, 84081 Baronissi, Italy; (C.G.); (M.R.R.); (V.V.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, University of Salerno “Scuola Medica Salernitana”, 84081 Baronissi, Italy; (C.G.); (M.R.R.); (V.V.)
| |
Collapse
|
3
|
Underwood L, Jiang CS, Oh JY, Sato PY. Unheralded Adrenergic Receptor Signaling in Cellular Oxidative Stress and Death. CURRENT OPINION IN PHYSIOLOGY 2024; 40:100766. [PMID: 39070968 PMCID: PMC11271747 DOI: 10.1016/j.cophys.2024.100766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Catecholamines (CAs) bind and activate adrenergic receptors (ARs), thus exuding a key role in cardiac adaptations to global physiological queues. Prolonged exposure to high levels of CAs promotes deleterious effects on the cardiovascular system, leading to organ dysfunction and heart failure (HF). In addition to the prominent role of ARs in inotropic and chronotropic responses, recent studies have delved into elucidating mechanisms contributing to CA toxicity and cell death. Central to this process is understanding the involvement of α1AR and βAR in cardiac remodeling and mechanisms of cellular survival. Here, we highlight the complexity of AR signaling and the fundamental need for a better understanding of its contribution to oxidative stress and cell death. This crucial informational nexus remains a barrier to the development of new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Lilly Underwood
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Chun-Sun Jiang
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Joo-Yeun Oh
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Priscila Y Sato
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
4
|
Sunny S, Jyothidasan A, David CL, Parsawar K, Veerappan A, Jones DP, Pogwizd S, Rajasekaran NS. Tandem Mass Tagging Based Identification of Proteome Signatures for Reductive Stress Cardiomyopathy. Front Cardiovasc Med 2022; 9:848045. [PMID: 35770227 PMCID: PMC9234166 DOI: 10.3389/fcvm.2022.848045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/14/2022] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2), a redox sensor, is vital for cellular redox homeostasis. We reported that transgenic mice expressing constitutively active Nrf2 (CaNrf2-TG) exhibit reductive stress (RS). In this study, we identified novel protein signature for RS-induced cardiomyopathy using Tandem Mass Tag (TMT) proteomic analysis in heart tissues of TG (CaNrf2-TG) mice at 6–7 months of age. A total of 1,105 proteins were extracted from 22,544 spectra. About 560 proteins were differentially expressed in TG vs. NTg hearts, indicating a global impact of RS on the myocardial proteome. Over 32 proteins were significantly altered in response to RS -20 were upregulated and 12 were downregulated in the hearts of TG vs. NTg mice, suggesting that these proteins could be putative signatures of RS. Scaffold analysis revealed a clear distinction between TG vs. NTg hearts. The majority of the differentially expressed proteins (DEPs) that were significantly altered in RS mice were found to be involved in stress related pathways such as antioxidants, NADPH, protein quality control, etc. Interestingly, proteins that were involved in mitochondrial respiration, lipophagy and cardiac rhythm were dramatically decreased in TG hearts. Of note, we identified the glutathione family of proteins as the significantly changed subset of the proteome in TG heart. Surprisingly, our comparative analysis of NGS based transcriptome and TMT-proteome indicated that ~50% of the altered proteins in TG myocardium was found to be negatively correlated with their transcript levels. In association with the altered proteome the TG mice displayed pathological cardiac remodeling.
Collapse
Affiliation(s)
- Sini Sunny
- Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Arun Jyothidasan
- Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Cynthia L David
- Analytical and Biological Mass Spectrometry Core Facility, The University of Arizona, Tuscon, AZ, United States
| | - Krishna Parsawar
- Analytical and Biological Mass Spectrometry Core Facility, The University of Arizona, Tuscon, AZ, United States
| | - Arul Veerappan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, New York University School of Medicine, New York, NY, United States.,Department of Environmental Medicine, New York University School of Medicine, New York, NY, United States
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States
| | - Steven Pogwizd
- Comprehensive Cardiovascular Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Namakkal S Rajasekaran
- Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States.,Division of Cardiovascular Medicine, Department of Medicine, The University of Utah, Salt Lake City, UT, United States.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
5
|
Tadinada SM, Weatherford ET, Collins GV, Bhardwaj G, Cochran J, Kutschke W, Zimmerman K, Bosko A, O'Neill BT, Weiss RM, Abel ED. Functional resilience of C57BL/6J mouse heart to dietary fat overload. Am J Physiol Heart Circ Physiol 2021; 321:H850-H864. [PMID: 34477461 PMCID: PMC8616610 DOI: 10.1152/ajpheart.00419.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 01/22/2023]
Abstract
Molecular mechanisms underlying cardiac dysfunction and subsequent heart failure in diabetic cardiomyopathy are incompletely understood. Initially we intended to test the role of G protein-coupled receptor kinase 2 (GRK2), a potential mediator of cardiac dysfunction in diabetic cardiomyopathy, but found that control animals on HFD did not develop cardiomyopathy. Cardiac function was preserved in both wild-type and GRK2 knockout animals fed high-fat diet as indicated by preserved left ventricular ejection fraction (LVEF) although heart mass was increased. The absence of cardiac dysfunction led us to rigorously evaluate the utility of diet-induced obesity to model diabetic cardiomyopathy in mice. Using pure C57BL/6J animals and various diets formulated with different sources of fat-lard (32% saturated fat, 68% unsaturated fat) or hydrogenated coconut oil (95% saturated fat), we consistently observed left ventricular hypertrophy, preserved LVEF, and preserved contractility measured by invasive hemodynamics in animals fed high-fat diet. Gene expression patterns that characterize pathological hypertrophy were not induced, but a modest induction of various collagen isoforms and matrix metalloproteinases was observed in heart with high-fat diet feeding. PPARα-target genes that enhance lipid utilization such as Pdk4, CD36, AcadL, and Cpt1b were induced, but mitochondrial energetics was not impaired. These results suggest that although long-term fat feeding in mice induces cardiac hypertrophy and increases cardiac fatty acid metabolism, it may not be sufficient to activate pathological hypertrophic mechanisms that impair cardiac function or induce cardiac fibrosis. Thus, additional factors that are currently not understood may contribute to the cardiac abnormalities previously reported by many groups.NEW & NOTEWORTHY Dietary fat overload (DFO) is widely used to model diabetic cardiomyopathy but the utility of this model is controversial. We comprehensively characterized cardiac contractile and mitochondrial function in C57BL6/J mice fed with lard-based or saturated fat-enriched diets initiated at two ages. Despite cardiac hypertrophy, contractile and mitochondrial function is preserved, and molecular adaptations likely limit lipotoxicity. The resilience of these hearts to DFO underscores the need to develop robust alternative models of diabetic cardiomyopathy.
Collapse
MESH Headings
- Age Factors
- Animals
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/physiopathology
- Diet, High-Fat
- Disease Models, Animal
- Energy Metabolism
- Female
- Fibrosis
- G-Protein-Coupled Receptor Kinase 2/genetics
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Myocardium/enzymology
- Myocardium/pathology
- Obesity/complications
- Stroke Volume
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
- Mice
Collapse
Affiliation(s)
- Satya Murthy Tadinada
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Eric T Weatherford
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Greg V Collins
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jesse Cochran
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - William Kutschke
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Kathy Zimmerman
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Alyssa Bosko
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Brian T O'Neill
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| | - Robert M Weiss
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Division of Cardiology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - E Dale Abel
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
6
|
The Metabolic Role of GRK2 in Insulin Resistance and Associated Conditions. Cells 2021; 10:cells10010167. [PMID: 33467677 PMCID: PMC7830135 DOI: 10.3390/cells10010167] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance (IRES) is a pathophysiological condition characterized by the reduced response to insulin of several tissues, including myocardial and skeletal muscle. IRES is associated with obesity, glucose intolerance, dyslipidemia, and hypertension, evolves toward type 2 diabetes, and increases the risk of developing cardiovascular diseases. Several studies designed to explore the mechanisms involved in IRES allowed the identification of a multitude of potential molecular targets. Among the most promising, G Protein Coupled Receptor Kinase type 2 (GRK2) appears to be a suitable one given its functional implications in many cellular processes. In this review, we will discuss the metabolic role of GRK2 in those conditions that are characterized by insulin resistance (diabetes, hypertension, heart failure), and the potentiality of its inhibition as a therapeutic strategy to revert both insulin resistance and its associated phenotypes.
Collapse
|
7
|
Sun X, Zhou M, Wen G, Huang Y, Wu J, Peng L, Jiang W, Yuan H, Lu Y, Cai J. Paroxetine Attenuates Cardiac Hypertrophy Via Blocking GRK2 and ADRB1 Interaction in Hypertension. J Am Heart Assoc 2020; 10:e016364. [PMID: 33372534 PMCID: PMC7955481 DOI: 10.1161/jaha.120.016364] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background ADRB1 (adrenergic receptor beta 1) responds to neuroendocrine stimulations, which have great implications in hypertension. GRK2 (G protein‐coupled receptor kinase 2) is an essential regulator for many G protein‐coupled receptors and subsequent cell signaling cascades, but its role as a regulator of ADRB1 and associated cardiac hypertrophy in hypertension remains to be elucidated. Methods and Results In this study, we found the expressions of GRK2 and ADRB1 in peripheral blood mononuclear cells were positively associated with blood pressure levels in hypertensive patients and with their expression in heart. In vitro evidence showed a direct interaction in ADRB1 and GRK2 and genetic depletion of GRK2 blocks epinephrine‐induced upregulation of hypertrophic and fibrotic genes in cardiomyocytes. Meanwhile, we discovered a selective serotonin reuptake inhibitor paroxetine specifically blockades GRK2 and ADRB1 interaction. In vivo, paroxetine treatment ameliorates hypertension‐induced cardiac hypertrophy, dysfunction, and fibrosis in animal models. We found that paroxetine suppressed sympathetic overdrive and increased the adrenergic receptor sensitivity to catecholamines. Paroxetine treatment also blocks epinephrine‐induced upregulation of hypertrophic and fibrotic genes as well as ADRB1 internalization in cardiomyocytes. Coadministration of paroxetine further potentiates metoprolol‐induced reductions in blood pressure and heart rate, further attenuating cardiac hypertrophy in spontaneously hypertensive rats. Furthermore, in patients with hypertension accompanied with depression, we observed that cardiac remodeling was less severe in those with paroxetine treatment compared with those with other types of anti‐depressive agents. Conclusions Paroxetine promotes ADRB1 sensitivity and attenuates cardiac hypertrophy partially via blocking GRK2‐mediated ADRB1 activation and internalization in the context of hypertension.
Collapse
Affiliation(s)
- Xuejing Sun
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Mengli Zhou
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Gaiyan Wen
- Department of Pharmacy Zhejiang Hospital Hangzhou China
| | - Yun Huang
- Ningbo Medical Center Lihuili Hospital Ningbo China
| | - Junru Wu
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Liping Peng
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Weihong Jiang
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Hong Yuan
- The Center of Clinical Pharmacology The Third Xiangya HospitalCentral South University Changsha China
| | - Yao Lu
- The Center of Clinical Pharmacology The Third Xiangya HospitalCentral South University Changsha China
| | - Jingjing Cai
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China.,The Center of Clinical Pharmacology The Third Xiangya HospitalCentral South University Changsha China
| |
Collapse
|
8
|
Pol CJ, Pollak NM, Jurczak MJ, Zacharia E, Karagiannides I, Kyriazis ID, Ntziachristos P, Scerbo DA, Brown BR, Aifantis I, Shulman GI, Goldberg IJ, Drosatos K. Cardiac myocyte KLF5 regulates body weight via alteration of cardiac FGF21. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2125-2137. [PMID: 31029826 PMCID: PMC6614009 DOI: 10.1016/j.bbadis.2019.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 01/22/2023]
Abstract
Cardiac metabolism affects systemic energetic balance. Previously, we showed that Krüppel-like factor (KLF)-5 regulates cardiomyocyte PPARα and fatty acid oxidation-related gene expression in diabetes. We surprisingly found that cardiomyocyte-specific KLF5 knockout mice (αMHC-KLF5-/-) have accelerated diet-induced obesity, associated with increased white adipose tissue (WAT). Alterations in cardiac expression of the mediator complex subunit 13 (Med13) modulates obesity. αMHC-KLF5-/- mice had reduced cardiac Med13 expression likely because KLF5 upregulates Med13 expression in cardiomyocytes. We then investigated potential mechanisms that mediate cross-talk between cardiomyocytes and WAT. High fat diet-fed αMHC-KLF5-/- mice had increased levels of cardiac and plasma FGF21, while food intake, activity, plasma leptin, and natriuretic peptides expression were unchanged. Consistent with studies reporting that FGF21 signaling in WAT decreases sumoylation-driven PPARγ inactivation, αMHC-KLF5-/- mice had less SUMO-PPARγ in WAT. Increased diet-induced obesity found in αMHC-KLF5-/- mice was absent in αMHC-[KLF5-/-;FGF21-/-] double knockout mice, as well as in αMHC-FGF21-/- mice that we generated. Thus, cardiomyocyte-derived FGF21 is a component of pro-adipogenic crosstalk between heart and WAT.
Collapse
Affiliation(s)
- Christine J Pol
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Nina M Pollak
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael J Jurczak
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Effimia Zacharia
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Iordanes Karagiannides
- Inflammatory Bowel Disease Center and Neuroendocrine Assay Core, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ioannis D Kyriazis
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Panagiotis Ntziachristos
- Howard Hughes Medical Institute, Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Diego A Scerbo
- Division of Preventive Medicine and Nutrition, Columbia University, New York, NY 10032, USA
| | - Brett R Brown
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Iannis Aifantis
- Howard Hughes Medical Institute, Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Gerald I Shulman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ira J Goldberg
- Division of Preventive Medicine and Nutrition, Columbia University, New York, NY 10032, USA
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA.
| |
Collapse
|
9
|
Ahern BM, Levitan BM, Veeranki S, Shah M, Ali N, Sebastian A, Su W, Gong MC, Li J, Stelzer JE, Andres DA, Satin J. Myocardial-restricted ablation of the GTPase RAD results in a pro-adaptive heart response in mice. J Biol Chem 2019; 294:10913-10927. [PMID: 31147441 PMCID: PMC6635439 DOI: 10.1074/jbc.ra119.008782] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/16/2019] [Indexed: 12/25/2022] Open
Abstract
Existing therapies to improve heart function target β-adrenergic receptor (β-AR) signaling and Ca2+ handling and often lead to adverse outcomes. This underscores an unmet need for positive inotropes that improve heart function without any adverse effects. The GTPase Ras associated with diabetes (RAD) regulates L-type Ca2+ channel (LTCC) current (ICa,L). Global RAD-knockout mice (gRAD-/-) have elevated Ca2+ handling and increased cardiac hypertrophy, but RAD is expressed also in noncardiac tissues, suggesting the possibility that pathological remodeling is due also to noncardiac effects. Here, we engineered a myocardial-restricted inducible RAD-knockout mouse (RADΔ/Δ). Using an array of methods and techniques, including single-cell electrophysiological and calcium transient recordings, echocardiography, and radiotelemetry monitoring, we found that RAD deficiency results in a sustained increase of inotropy without structural or functional remodeling of the heart. ICa,L was significantly increased, with RAD loss conferring a β-AR-modulated phenotype on basal ICa,L Cardiomyocytes from RADΔ/Δ hearts exhibited enhanced cytosolic Ca2+ handling, increased contractile function, elevated sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2a) expression, and faster lusitropy. These results argue that myocardial RAD ablation promotes a beneficial elevation in Ca2+ dynamics, which would obviate a need for increased β-AR signaling to improve cardiac function.
Collapse
Affiliation(s)
| | - Bryana M Levitan
- Department of Physiology,; Gill Heart and Vascular Institute, and
| | - Sudhakar Veeranki
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536 and
| | | | - Nemat Ali
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536 and
| | | | | | | | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536 and.
| | | |
Collapse
|
10
|
Sorriento D, Gambardella J, Fiordelisi A, Iaccarino G, Illario M. GRKs and β-Arrestins: "Gatekeepers" of Mitochondrial Function in the Failing Heart. Front Pharmacol 2019; 10:64. [PMID: 30809146 PMCID: PMC6379454 DOI: 10.3389/fphar.2019.00064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/18/2019] [Indexed: 01/14/2023] Open
Abstract
Mitochondrial regulation of energy production, calcium homeostasis, and cell death are critical for cardiac function. Accordingly, the structural and functional abnormalities of these organelles (mitochondrial dysfunction) contribute to developing cardiovascular diseases and heart failure. Therefore the preservation of mitochondrial integrity is essential for cardiac cell survival. Mitochondrial function is regulated by several proteins, including GRK2 and β-arrestins which act in a GPCR independent manner to orchestrate intracellular signaling associated with key mitochondrial processes. It is now ascertained that GRK2 is able to recover mitochondrial function in response to insults. β-arrestins affect several intracellular signaling pathways within the cell which in turn are involved in the regulation of mitochondrial function, but a direct regulation of mitochondria needs further investigations. In this review, we discuss the recent acquisitions on the role of GRK2 and β-arrestins in the regulation of mitochondrial function.
Collapse
Affiliation(s)
- Daniela Sorriento
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Jessica Gambardella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
11
|
Sato PY, Chuprun JK, Grisanti LA, Woodall MC, Brown BR, Roy R, Traynham CJ, Ibetti J, Lucchese AM, Yuan A, Drosatos K, Tilley DG, Gao E, Koch WJ. Restricting mitochondrial GRK2 post-ischemia confers cardioprotection by reducing myocyte death and maintaining glucose oxidation. Sci Signal 2018; 11:11/560/eaau0144. [PMID: 30538174 DOI: 10.1126/scisignal.aau0144] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Increased abundance of GRK2 [G protein-coupled receptor (GPCR) kinase 2] is associated with poor cardiac function in heart failure patients. In animal models, GRK2 contributes to the pathogenesis of heart failure after ischemia-reperfusion (IR) injury. In addition to its role in down-regulating activated GPCRs, GRK2 also localizes to mitochondria both basally and post-IR injury, where it regulates cellular metabolism. We previously showed that phosphorylation of GRK2 at Ser670 is essential for the translocation of GRK2 to the mitochondria of cardiomyocytes post-IR injury in vitro and that this localization promotes cell death. Here, we showed that mice with a S670A knock-in mutation in endogenous GRK2 showed reduced cardiomyocyte death and better cardiac function post-IR injury. Cultured GRK2-S670A knock-in cardiomyocytes subjected to IR in vitro showed enhanced glucose-mediated mitochondrial respiratory function that was partially due to maintenance of pyruvate dehydrogenase activity and improved glucose oxidation. Thus, we propose that mitochondrial GRK2 plays a detrimental role in cardiac glucose oxidation post-injury.
Collapse
Affiliation(s)
- Priscila Y Sato
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - J Kurt Chuprun
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Laurel A Grisanti
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Meryl C Woodall
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Brett R Brown
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rajika Roy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Christopher J Traynham
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jessica Ibetti
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Anna M Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ancai Yuan
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Konstantinos Drosatos
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Doug G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA. .,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
12
|
Manning JR, Chelvarajan L, Levitan BM, Withers CN, Nagareddy PR, Haggerty CM, Fornwalt BK, Gao E, Tripathi H, Abdel-Latif A, Andres DA, Satin J. Rad GTPase deletion attenuates post-ischemic cardiac dysfunction and remodeling. ACTA ACUST UNITED AC 2018; 3:83-96. [PMID: 29732439 PMCID: PMC5931223 DOI: 10.1016/j.jacbts.2017.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Rad-GTPase is an LTCC component that functions to govern calcium current in the myocardium. Deletion of Rad increases myocardial contractility secondary to increased trigger calcium entry. AMI induces heart failure, including reduced calcium homeostasis, but deletion of Rad prevents AMI myocardial calcium alterations. Rad deletion prevents post-MI scar spread by attenuating the inflammatory response. Future studies will explore whether Rad deletion is an effective therapeutic direction for providing combined safe, stable inotropic support to the failing heart in concert with protection against inflammatory signaling.
The protein Rad interacts with the L-type calcium channel complex to modulate trigger Ca2+ and hence to govern contractility. Reducing Rad levels increases cardiac output. Ablation of Rad also attenuated the inflammatory response following acute myocardial infarction. Future studies to target deletion of Rad in the heart could be conducted to establish a novel treatment paradigm whereby pathologically stressed hearts would be given safe, stable positive inotropic support without arrhythmias and without pathological structural remodeling. Future investigations will also focus on establishing inhibitors of Rad and testing the efficacy of Rad deletion in cardioprotection relative to the time of onset of acute myocardial infarction.
Collapse
Affiliation(s)
- Janet R Manning
- Department of Physiology, University of Kentucky, Lexington KY.,Department of Biochemistry, University of Kentucky, Lexington KY
| | - Lakshman Chelvarajan
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY
| | - Bryana M Levitan
- Department of Physiology, University of Kentucky, Lexington KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington KY
| | | | | | - Christopher M Haggerty
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY.,Department of Imaging Science and Innovation, Geisinger, Danville PA
| | - Brandon K Fornwalt
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY.,Department of Imaging Science and Innovation, Geisinger, Danville PA
| | - Erhe Gao
- Department of Physiology, University of Kentucky, Lexington KY.,Center for Translational Medicine, Temple University School of Medicine, Philadelphia PA
| | - Himi Tripathi
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY
| | - Ahmed Abdel-Latif
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington KY
| | - Douglas A Andres
- Department of Biochemistry, University of Kentucky, Lexington KY
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington KY
| |
Collapse
|
13
|
Schreckenberg R, Bencsik P, Weber M, Abdallah Y, Csonka C, Gömöri K, Kiss K, Pálóczi J, Pipis J, Sárközy M, Ferdinandy P, Schulz R, Schlüter KD. Adverse Effects on β-Adrenergic Receptor Coupling: Ischemic Postconditioning Failed to Preserve Long-Term Cardiac Function. J Am Heart Assoc 2017; 6:e006809. [PMID: 29273639 PMCID: PMC5779008 DOI: 10.1161/jaha.117.006809] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) are currently among the most efficient strategies protecting the heart against ischemia/reperfusion injury. However, the effect of IPC and IPoC on functional recovery following ischemia/reperfusion is less clear, particularly with regard to the specific receptor-mediated signaling of the postischemic heart. The current article examines the effect of IPC or IPoC on the regulation and coupling of β-adrenergic receptors and their effects on postischemic left ventricular function. METHODS AND RESULTS The β-adrenergic signal transduction was analyzed in 3-month-old Wistar rats for each of the intervention strategies (Sham, ischemia/reperfusion, IPC, IPoC) immediately and 7 days after myocardial infarction. Directly after the infarction a cardioprotective potential was demonstrated for both IPC and IPoC: the infarct size was reduced, apoptosis and production of reactive oxygen species were lowered, and the myocardial tissue was preserved. Seven days after myocardial ischemia, only IPC hearts showed significant functional improvement. Along with a deterioration in fractional shortening, IPoC hearts no longer responded adequately to β-adrenergic stimulation. The stabilization of β-adrenergic receptor kinase-2 via increased phosphorylation of Mdm2 (an E3-ubiquitin ligase) was responsible for desensitization of β-adrenergic receptors and identified as a characteristic specific to IPoC hearts. CONCLUSIONS Immediately after myocardial infarction, rapid and transient activation of β-adrenergic receptor kinase-2 may be an appropriate means to protect the injured heart from excessive stress. In the long term, however, induction and stabilization of β-adrenergic receptor kinase-2, with the resultant loss of positive inotropic function, leads to the functional picture of heart failure.
Collapse
Affiliation(s)
- Rolf Schreckenberg
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Péter Bencsik
- Pharmahungary Group, Szeged, Hungary
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Martin Weber
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Yaser Abdallah
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Csaba Csonka
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Krisztina Kiss
- Pharmahungary Group, Szeged, Hungary
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - János Pálóczi
- Pharmahungary Group, Szeged, Hungary
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | | | - Márta Sárközy
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | | |
Collapse
|
14
|
Schlegel P, Reinkober J, Meinhardt E, Tscheschner H, Gao E, Schumacher SM, Yuan A, Backs J, Most P, Wieland T, Koch WJ, Katus HA, Raake PW. G protein-coupled receptor kinase 2 promotes cardiac hypertrophy. PLoS One 2017; 12:e0182110. [PMID: 28759639 PMCID: PMC5536362 DOI: 10.1371/journal.pone.0182110] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 07/12/2017] [Indexed: 01/06/2023] Open
Abstract
The increase in protein activity and upregulation of G-protein coupled receptor kinase 2 (GRK2) is a hallmark of cardiac stress and heart failure. Inhibition of GRK2 improved cardiac function and survival and diminished cardiac remodeling in various animal heart failure models. The aim of the present study was to investigate the effects of GRK2 on cardiac hypertrophy and dissect potential molecular mechanisms. In mice we observed increased GRK2 mRNA and protein levels following transverse aortic constriction (TAC). Conditional GRK2 knockout mice showed attenuated hypertrophic response with preserved ventricular geometry 6 weeks after TAC operation compared to wild-type animals. In isolated neonatal rat ventricular cardiac myocytes stimulation with angiotensin II and phenylephrine enhanced GRK2 expression leading to enhanced signaling via protein kinase B (PKB or Akt), consecutively inhibiting glycogen synthase kinase 3 beta (GSK3β), such promoting nuclear accumulation and activation of nuclear factor of activated T-cells (NFAT). Cardiac myocyte hypertrophy induced by in vitro GRK2 overexpression increased the cytosolic interaction of GRK2 and phosphoinositide 3-kinase γ (PI3Kγ). Moreover, inhibition of PI3Kγ as well as GRK2 knock down prevented Akt activation resulting in halted NFAT activity and reduced cardiac myocyte hypertrophy. Our data show that enhanced GRK2 expression triggers cardiac hypertrophy by GRK2-PI3Kγ mediated Akt phosphorylation and subsequent inactivation of GSK3β, resulting in enhanced NFAT activity.
Collapse
Affiliation(s)
- Philipp Schlegel
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Julia Reinkober
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Eric Meinhardt
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Henrike Tscheschner
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Erhe Gao
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Sarah M. Schumacher
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Ancai Yuan
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Johannes Backs
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
- Department of Molecular Cardiology and Epigenetics, University of Heidelberg, Heidelberg, Germany
| | - Patrick Most
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Thomas Wieland
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany
| | - Walter J. Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Hugo A. Katus
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Philip W. Raake
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
15
|
Liu Y, Baumgardt SL, Fang J, Shi Y, Qiao S, Bosnjak ZJ, Vásquez-Vivar J, Xia Z, Warltier DC, Kersten JR, Ge ZD. Transgenic overexpression of GTP cyclohydrolase 1 in cardiomyocytes ameliorates post-infarction cardiac remodeling. Sci Rep 2017; 7:3093. [PMID: 28596578 PMCID: PMC5465102 DOI: 10.1038/s41598-017-03234-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
GTP cyclohydrolase 1 (GCH1) and its product tetrahydrobiopterin play crucial roles in cardiovascular health and disease, yet the exact regulation and role of GCH1 in adverse cardiac remodeling after myocardial infarction are still enigmatic. Here we report that cardiac GCH1 is degraded in remodeled hearts after myocardial infarction, concomitant with increases in the thickness of interventricular septum, interstitial fibrosis, and phosphorylated p38 mitogen-activated protein kinase and decreases in left ventricular anterior wall thickness, cardiac contractility, tetrahydrobiopterin, the dimers of nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and the expression of sarcoplasmic reticulum Ca2+ handling proteins. Intriguingly, transgenic overexpression of GCH1 in cardiomyocytes reduces the thickness of interventricular septum and interstitial fibrosis and increases anterior wall thickness and cardiac contractility after infarction. Moreover, we show that GCH1 overexpression decreases phosphorylated p38 mitogen-activated protein kinase and elevates tetrahydrobiopterin levels, the dimerization and phosphorylation of neuronal nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and sarcoplasmic reticulum Ca2+ handling proteins in post-infarction remodeled hearts. Our results indicate that the pivotal role of GCH1 overexpression in post-infarction cardiac remodeling is attributable to preservation of neuronal nitric oxide synthase and sarcoplasmic reticulum Ca2+ handling proteins, and identify a new therapeutic target for cardiac remodeling after infarction.
Collapse
Affiliation(s)
- Yanan Liu
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Medicine, Columbia University, 630 W. 168th Street, New York, New York, 10032, USA
| | - Shelley L Baumgardt
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, Wisconsin, 53234, USA
| | - Shigang Qiao
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zeljko J Bosnjak
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Physiology, Medical College of Wiscosin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Jeannette Vásquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, People's Republic of China
| | - David C Warltier
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Judy R Kersten
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhi-Dong Ge
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.
| |
Collapse
|
16
|
Chen J, Wang D, Wang F, Shi S, Chen Y, Yang B, Tang Y, Huang C. Exendin-4 inhibits structural remodeling and improves Ca 2+ homeostasis in rats with heart failure via the GLP-1 receptor through the eNOS/cGMP/PKG pathway. Peptides 2017; 90:69-77. [PMID: 28242257 DOI: 10.1016/j.peptides.2017.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) agonist exendin-4 is a long-acting analog of GLP-1, which stimulates insulin secretion and is clinically used in the treatment of type 2 diabetes. Previous studies have demonstrated that GLP-1 agonists and analogs serve as cardioprotective factors in various conditions. Disturbances in calcium cycling are characteristic of heart failure (HF); therefore, the aim of this study was to investigate the effect of exendin-4 (a GLP-1 mimetic) on the regulation of calcium handling and to identify the underlying mechanisms in an HF rat model after myocardial infarction (MI). Rats underwent surgical ligation of the left anterior descending coronary artery or sham surgery prior to infusion with vehicle, exendin-4, or exendin-4 and exendin9-39 for 4 weeks. Exendin-4 treatment decreased MI size, suppressed chamber dilation, myocyte hypertrophy, and fibrosis and improved in vivo heart function in the rats subjected to MI. Exendin-4 resulted in an increase in circulating GLP-1 and GLP-1R in ventricular tissues. Additionally, exendin-4 activated the eNOS/cGMP/PKG signaling pathway and inhibited the Ca2+/calmodulin-dependent kinase II (CaMKII) pathways. Myocytes isolated from exendin-4-treated hearts displayed higher Ca2+ transients, higher sarcoplasmic reticulum Ca2+ content, and higher l-type Ca2+ current densities than MI hearts. Exendin-4 treatment restored the protein expression of sarcoplasmic reticulum Ca2+ uptake ATPase (SERCA2a), phosphorylated phospholamban (PLB) and Cav1.2 and decreased the levels of phosphorylated ryanodine receptor (RyR). Moreover, the favorable effects of exendin-4 were significantly inhibited by exendin9-39 (a GLP-1 receptor antagonist). Exendin-4 treatment of an HF rat model after MI inhibited cardiac and cardiomyocytes progressive remodeling. In addition, Ca2+ handling and its molecular modulation were also improved by exendin-4 treatment. The beneficial effects of exendin-4 on cardiac remodeling may be mediated through activation of the eNOS/cGMP/PKG pathway.
Collapse
Affiliation(s)
- Jingjing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Dandan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Fangai Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Yuting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
17
|
Sorriento D, Ciccarelli M, Cipolletta E, Trimarco B, Iaccarino G. "Freeze, Don't Move": How to Arrest a Suspect in Heart Failure - A Review on Available GRK2 Inhibitors. Front Cardiovasc Med 2016; 3:48. [PMID: 27999776 PMCID: PMC5138235 DOI: 10.3389/fcvm.2016.00048] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/21/2016] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease and heart failure (HF) still collect the largest toll of death in western societies and all over the world. A growing number of molecular mechanisms represent possible targets for new therapeutic strategies, which can counteract the metabolic and structural changes observed in the failing heart. G protein-coupled receptor kinase 2 (GRK2) is one of such targets for which experimental and clinical evidence are established. Indeed, several strategies have been carried out in place to interface with the known GRK2 mechanisms of action in the failing heart. This review deals with results from basic and preclinical studies. It shows different strategies to inhibit GRK2 in HF in vivo (βARK-ct gene therapy, treatment with gallein, and treatment with paroxetine) and in vitro (RNA aptamer, RKIP, and peptide-based inhibitors). These strategies are based either on the inhibition of the catalytic activity of the kinase (“Freeze!”) or the prevention of its shuttling within the cell (“Don’t Move!”). Here, we review the peculiarity of each strategy with regard to the ability to interact with the multiple tasks of GRK2 and the perspective development of eventual clinical use.
Collapse
Affiliation(s)
- Daniela Sorriento
- Department of Advanced Biomedical Sciences, University of Naples Federico II , Naples , Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno , Baronissi, SA , Italy
| | - Ersilia Cipolletta
- Department of Advanced Biomedical Sciences, University of Naples Federico II , Naples , Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, University of Naples Federico II , Naples , Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno , Baronissi, SA , Italy
| |
Collapse
|
18
|
Woodall BP, Woodall MC, Luongo TS, Grisanti LA, Tilley DG, Elrod JW, Koch WJ. Skeletal Muscle-specific G Protein-coupled Receptor Kinase 2 Ablation Alters Isolated Skeletal Muscle Mechanics and Enhances Clenbuterol-stimulated Hypertrophy. J Biol Chem 2016; 291:21913-21924. [PMID: 27566547 DOI: 10.1074/jbc.m116.721282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 02/04/2023] Open
Abstract
GRK2, a G protein-coupled receptor kinase, plays a critical role in cardiac physiology. Adrenergic receptors are the primary target for GRK2 activity in the heart; phosphorylation by GRK2 leads to desensitization of these receptors. As such, levels of GRK2 activity in the heart directly correlate with cardiac contractile function. Furthermore, increased expression of GRK2 after cardiac insult exacerbates injury and speeds progression to heart failure. Despite the importance of this kinase in both the physiology and pathophysiology of the heart, relatively little is known about the role of GRK2 in skeletal muscle function and disease. In this study we generated a novel skeletal muscle-specific GRK2 knock-out (KO) mouse (MLC-Cre:GRK2fl/fl) to gain a better understanding of the role of GRK2 in skeletal muscle physiology. In isolated muscle mechanics testing, GRK2 ablation caused a significant decrease in the specific force of contraction of the fast-twitch extensor digitorum longus muscle yet had no effect on the slow-twitch soleus muscle. Despite these effects in isolated muscle, exercise capacity was not altered in MLC-Cre:GRK2fl/fl mice compared with wild-type controls. Skeletal muscle hypertrophy stimulated by clenbuterol, a β2-adrenergic receptor (β2AR) agonist, was significantly enhanced in MLC-Cre:GRK2fl/fl mice; mechanistically, this seems to be due to increased clenbuterol-stimulated pro-hypertrophic Akt signaling in the GRK2 KO skeletal muscle. In summary, our study provides the first insights into the role of GRK2 in skeletal muscle physiology and points to a role for GRK2 as a modulator of contractile properties in skeletal muscle as well as β2AR-induced hypertrophy.
Collapse
Affiliation(s)
- Benjamin P Woodall
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| | - Meryl C Woodall
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| | - Timothy S Luongo
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| | - Laurel A Grisanti
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| | - Douglas G Tilley
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| | - John W Elrod
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| | - Walter J Koch
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140-4106
| |
Collapse
|
19
|
Gardner RT, Ripplinger CM, Myles RC, Habecker BA. Molecular Mechanisms of Sympathetic Remodeling and Arrhythmias. Circ Arrhythm Electrophysiol 2016; 9:e001359. [PMID: 26810594 DOI: 10.1161/circep.115.001359] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ryan T Gardner
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.)
| | - Crystal M Ripplinger
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.)
| | - Rachel C Myles
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.)
| | - Beth A Habecker
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.).
| |
Collapse
|
20
|
Hullmann J, Traynham CJ, Coleman RC, Koch WJ. The expanding GRK interactome: Implications in cardiovascular disease and potential for therapeutic development. Pharmacol Res 2016; 110:52-64. [PMID: 27180008 DOI: 10.1016/j.phrs.2016.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/05/2016] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is a global epidemic with the highest degree of mortality and morbidity of any disease presently studied. G protein-coupled receptors (GPCRs) are prominent regulators of cardiovascular function. Activated GPCRs are "turned off" by GPCR kinases (GRKs) in a process known as "desensitization". GRKs 2 and 5 are highly expressed in the heart, and known to be upregulated in HF. Over the last 20 years, both GRK2 and GRK5 have been demonstrated to be critical mediators of the molecular alterations that occur in the failing heart. In the present review, we will highlight recent findings that further characterize "non-canonical" GRK signaling observed in HF. Further, we will also present potential therapeutic strategies (i.e. small molecule inhibition, microRNAs, gene therapy) that may have potential in combating the deleterious effects of GRKs in HF.
Collapse
Affiliation(s)
| | - Christopher J Traynham
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Ryan C Coleman
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States.
| |
Collapse
|
21
|
Ripplinger CM, Noujaim SF, Linz D. The nervous heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:199-209. [PMID: 26780507 DOI: 10.1016/j.pbiomolbio.2015.12.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/29/2015] [Accepted: 12/31/2015] [Indexed: 12/23/2022]
Abstract
Many cardiac electrophysiological abnormalities are accompanied by autonomic nervous system dysfunction. Here, we review mechanisms by which the cardiac nervous system controls normal and abnormal excitability and may contribute to atrial and ventricular tachyarrhythmias. Moreover, we explore the potential antiarrhythmic and/or arrhythmogenic effects of modulating the autonomic nervous system by several strategies, including ganglionated plexi ablation, vagal and spinal cord stimulations, and renal sympathetic denervation as therapies for atrial and ventricular arrhythmias.
Collapse
Affiliation(s)
- Crystal M Ripplinger
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA.
| | - Sami F Noujaim
- Molecular Pharmacology and Physiology, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA.
| | - Dominik Linz
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg, Saar, Germany.
| |
Collapse
|
22
|
Schmid E, Neef S, Berlin C, Tomasovic A, Kahlert K, Nordbeck P, Deiss K, Denzinger S, Herrmann S, Wettwer E, Weidendorfer M, Becker D, Schäfer F, Wagner N, Ergün S, Schmitt JP, Katus HA, Weidemann F, Ravens U, Maack C, Hein L, Ertl G, Müller OJ, Maier LS, Lohse MJ, Lorenz K. Cardiac RKIP induces a beneficial β-adrenoceptor-dependent positive inotropy. Nat Med 2015; 21:1298-306. [PMID: 26479924 DOI: 10.1038/nm.3972] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/12/2015] [Indexed: 01/08/2023]
Abstract
In heart failure therapy, it is generally assumed that attempts to produce a long-term increase in cardiac contractile force are almost always accompanied by structural and functional damage. Here we show that modest overexpression of the Raf kinase inhibitor protein (RKIP), encoded by Pebp1 in mice, produces a well-tolerated, persistent increase in cardiac contractility that is mediated by the β1-adrenoceptor (β1AR). This result is unexpected, as β1AR activation, a major driver of cardiac contractility, usually has long-term adverse effects. RKIP overexpression achieves this tolerance via simultaneous activation of the β2AR subtype. Analogously, RKIP deficiency exaggerates pressure overload-induced cardiac failure. We find that RKIP expression is upregulated in mouse and human heart failure, indicative of an adaptive role for RKIP. Pebp1 gene transfer in a mouse model of heart failure has beneficial effects, suggesting a new therapeutic strategy for heart failure therapy.
Collapse
Affiliation(s)
- Evelyn Schmid
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Stefan Neef
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Christopher Berlin
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Angela Tomasovic
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Katrin Kahlert
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Peter Nordbeck
- Comprehensive Heart Failure Center, Würzburg, Germany.,Department of Internal Medicine I, University of Würzburg, Würzburg, Germany
| | - Katharina Deiss
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Sabrina Denzinger
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Sebastian Herrmann
- Comprehensive Heart Failure Center, Würzburg, Germany.,Department of Internal Medicine I, University of Würzburg, Würzburg, Germany
| | - Erich Wettwer
- Department of Pharmacology and Toxicology, Medical Faculty Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Markus Weidendorfer
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Daniel Becker
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Florian Schäfer
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Nicole Wagner
- Institute of Anatomy and Cell Biology, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Würzburg, Germany
| | - Joachim P Schmitt
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.,German Centre for Cardiovascular Research, Heidelberg University Hospital, Heidelberg, Germany
| | - Frank Weidemann
- Comprehensive Heart Failure Center, Würzburg, Germany.,Department of Internal Medicine I, University of Würzburg, Würzburg, Germany
| | - Ursula Ravens
- Department of Pharmacology and Toxicology, Medical Faculty Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Christoph Maack
- Clinic for Internal Medicine III, Saarland University Hospital, Homburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Freiburg, Germany.,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany
| | - Georg Ertl
- Comprehensive Heart Failure Center, Würzburg, Germany.,Department of Internal Medicine I, University of Würzburg, Würzburg, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.,German Centre for Cardiovascular Research, Heidelberg University Hospital, Heidelberg, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Martin J Lohse
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany.,Comprehensive Heart Failure Center, Würzburg, Germany
| | - Kristina Lorenz
- Department of Pharmacology, Institute of Pharmacology and Toxicology, Würzburg, Germany.,Comprehensive Heart Failure Center, Würzburg, Germany
| |
Collapse
|
23
|
Sallam K, Li Y, Sager PT, Houser SR, Wu JC. Finding the rhythm of sudden cardiac death: new opportunities using induced pluripotent stem cell-derived cardiomyocytes. Circ Res 2015; 116:1989-2004. [PMID: 26044252 DOI: 10.1161/circresaha.116.304494] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sudden cardiac death is a common cause of death in patients with structural heart disease, genetic mutations, or acquired disorders affecting cardiac ion channels. A wide range of platforms exist to model and study disorders associated with sudden cardiac death. Human clinical studies are cumbersome and are thwarted by the extent of investigation that can be performed on human subjects. Animal models are limited by their degree of homology to human cardiac electrophysiology, including ion channel expression. Most commonly used cellular models are cellular transfection models, which are able to mimic the expression of a single-ion channel offering incomplete insight into changes of the action potential profile. Induced pluripotent stem cell-derived cardiomyocytes resemble, but are not identical, adult human cardiomyocytes and provide a new platform for studying arrhythmic disorders leading to sudden cardiac death. A variety of platforms exist to phenotype cellular models, including conventional and automated patch clamp, multielectrode array, and computational modeling. Induced pluripotent stem cell-derived cardiomyocytes have been used to study long QT syndrome, catecholaminergic polymorphic ventricular tachycardia, hypertrophic cardiomyopathy, and other hereditary cardiac disorders. Although induced pluripotent stem cell-derived cardiomyocytes are distinct from adult cardiomyocytes, they provide a robust platform to advance the science and clinical care of sudden cardiac death.
Collapse
Affiliation(s)
- Karim Sallam
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Yingxin Li
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Philip T Sager
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Steven R Houser
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| | - Joseph C Wu
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| |
Collapse
|
24
|
Sorriento D, Santulli G, Franco A, Cipolletta E, Napolitano L, Gambardella J, Gomez-Monterrey I, Campiglia P, Trimarco B, Iaccarino G, Ciccarelli M. Integrating GRK2 and NFkappaB in the Pathophysiology of Cardiac Hypertrophy. J Cardiovasc Transl Res 2015. [DOI: 10.1007/s12265-015-9646-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
25
|
Sato PY, Chuprun JK, Schwartz M, Koch WJ. The evolving impact of g protein-coupled receptor kinases in cardiac health and disease. Physiol Rev 2015; 95:377-404. [PMID: 25834229 PMCID: PMC4551214 DOI: 10.1152/physrev.00015.2014] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are important regulators of various cellular functions via activation of intracellular signaling events. Active GPCR signaling is shut down by GPCR kinases (GRKs) and subsequent β-arrestin-mediated mechanisms including phosphorylation, internalization, and either receptor degradation or resensitization. The seven-member GRK family varies in their structural composition, cellular localization, function, and mechanism of action (see sect. II). Here, we focus our attention on GRKs in particular canonical and novel roles of the GRKs found in the cardiovascular system (see sects. III and IV). Paramount to overall cardiac function is GPCR-mediated signaling provided by the adrenergic system. Overstimulation of the adrenergic system has been highly implicated in various etiologies of cardiovascular disease including hypertension and heart failure. GRKs acting downstream of heightened adrenergic signaling appear to be key players in cardiac homeostasis and disease progression, and herein we review the current data on GRKs related to cardiac disease and discuss their potential in the development of novel therapeutic strategies in cardiac diseases including heart failure.
Collapse
Affiliation(s)
- Priscila Y Sato
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - J Kurt Chuprun
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Mathew Schwartz
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Walter J Koch
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| |
Collapse
|
26
|
Li Y, Zhang S, Zhang X, Li J, Ai X, Zhang L, Yu D, Ge S, Peng Y, Chen X. Blunted cardiac beta-adrenergic response as an early indication of cardiac dysfunction in Duchenne muscular dystrophy. Cardiovasc Res 2014; 103:60-71. [PMID: 24812281 DOI: 10.1093/cvr/cvu119] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIMS To determine whether altered beta-adrenergic responses contribute to early cardiac dysfunction in mdx (X-linked muscular dystrophy) mice, an animal model for human Duchenne muscular dystrophy. METHODS AND RESULTS Replacement fibrosis in mdx hearts gradually increased with age, suggesting a gradual loss of cardiomyocytes. Echocardiography and intra-left ventricular haemodynamic measurements detected baseline cardiac dysfunction in mdx mice at ≥8 months. However, a reduction of cardiac beta-adrenergic response to isoproterenol (ISO) was already present in mdx mice at 4 months. Ventricular myocytes (VMs) isolated from 4- and 8-month-old mdx mice had greater baseline contractile function {fractional shortening, [Ca(2+)]i, and sarcoplasmic reticulum (SR) Ca(2+) content} and ICa-L than age-matched control VMs and than myocytes isolated from 2-month-old mdx mice. ISO increased myocyte function in the VMs of 4- and 8-month-old mdx mice to the same level as in age-matched control VMs. In the VMs of 12-month-old mdx mice, ISO failed to increase myocyte function to the level in VMs of 12-month-old control mice and could not further increaseICa-L. No differences were observed in the expression of Cav1.2α1c, Cav1.2β1, Cav1.2β2, sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA), and the Na(+)/Ca(2+) exchanger. In contrast, total ryanodine receptor 2 (RyR2) and basal phosphorylation of RyR2, phospholamban, and Cav1.2α1c were found to be increased in hearts of 4-month-old mdx mice; baseline protein kinase A activity was also increased. After ISO treatment, phosphorylation levels were the same in mdx and control hearts. VMs of 4-month-old mdx mice had reduced beta1-adrenergic receptor (β1-AR) density and beta-adrenergic sensitivity. CONCLUSION In young mdx mice, the myocyte increases its contractile function to compensate for myocyte loss. However, these myocytes with enhanced baseline function have reduced potential for stimulation, decreased β1-AR density/sensitivity, leading to blunted cardiac beta-adrenergic response.
Collapse
Affiliation(s)
- Ying Li
- Institute of Burn Research, Southwest Hospital, The Third Military Medical University, Chongqing, China Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Shuai Zhang
- Institute of Burn Research, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoying Zhang
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jing Li
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA School of Medicine, Nankai University, Tianjin, China
| | - Xiaojie Ai
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA College of Biological Sciences, Shanghai Jiaotong University, Shanghai, China
| | - Li Zhang
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA Drexel University College of Medicine, Philadelphia, PA, USA
| | - Daohai Yu
- Department of Clinical Sciences, Temple University School of Medicine, Philadelphia, PA, USA
| | - Shuping Ge
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Yizhi Peng
- Institute of Burn Research, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Xiongwen Chen
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
Affiliation(s)
- David Eisner
- From the Unit of Cardiac Physiology, University of Manchester, United Kingdom
| | | | | |
Collapse
|
28
|
Gao WQ, Han CG, Lu XC, Liu YX, Hui HP, Wang H. GRK 2 level in peripheral blood lymphocytes of elderly patients with acute myocardial infarction. J Geriatr Cardiol 2013; 10:281-285. [PMID: 24133517 PMCID: PMC3796703 DOI: 10.3969/j.issn.1671-5411.2013.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/22/2013] [Accepted: 06/22/2013] [Indexed: 06/02/2023] Open
Abstract
OBJECTIVE To investigate the G protein-coupled receptor kinase 2 (GRK 2) level in peripheral blood lymphocytes with cardiac function in elderly patients with acute myocardial infarction. METHODS This study enrolled 40 patients with acute ST-segment elevation myocardial infarction (STEMI) and 40 patients with unstable angina. All patients were 65 years or older. Cardiac function was evaluated by echocardiography, and the GRK 2 level in peripheral blood lymphocytes was measured. Patients with STEMI were followed up for 2 years. RESULTS The GRK 2 level in peripheral blood lymphocytes was significantly higher in patients with STEMI than in patients with unstable angina, and was negatively correlated with left ventricular ejection fraction, cardiac output, stroke volume, and left ventricular fractional shortening. The GRK 2 level was significantly elevated in some patients with acute STEMI and poor cardiac function. CONCLUSIONS Increased GRK 2 level in patients with acute STEMI may contribute to poor myocardial systolic function and myocardial remodeling. Measurement of the GRK 2 level in peripheral blood lymphocytes may assist in the evaluation of cardiac function and myocardial remodeling in elderly patients with acute STEMI.
Collapse
Affiliation(s)
- Wen-Qian Gao
- The First Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | | | - Xiao-Chun Lu
- The First Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yong-Xue Liu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hai-Peng Hui
- The First Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Wang
- The First Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
29
|
Pal SN, Kofidis T. Therapeutic potential of genes in cardiac repair. Expert Rev Cardiovasc Ther 2013; 11:1015-28. [PMID: 23945013 DOI: 10.1586/14779072.2013.814867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cardiovascular diseases remain the primary reason of premature death and contribute to a major percentage of global patient morbidity. Recent knowledge in the molecular mechanisms of myocardial complications have identified novel therapeutic targets along with the availability of vectors that offer the chance for designing gene therapy technique for protection and revival of the diseased heart functions. Gene transfer procedure into the myocardium is demonstrated through direct injection of plasmid DNA or through the coronary vasculature using the direct or indirect delivery of viral vectors. Direct DNA injection to the myocardium is reported to be of immense value in research studies that aims at understanding the activities of various elements in myocardium. It is also deemed vital for investigating the effect of the myocardial pathophysiology on expression of the foreign genes that are transferred. Gene therapies have been reported to heal cardiac pathologies such as myocardial ischemia, heart failure and inherited myopathies in several animal models. The results obtained from these animal studies have also encouraged a flurry of early clinical trials. This translational research has been triggered by an enhanced understanding of the biological mechanisms involved in tissue repair after ischemic injury. While safety concerns take utmost priority in these trials, several combinational therapies, various routes and dose of delivery are being tested before concrete optimization and complete potential of gene therapy is convincingly understood.
Collapse
Affiliation(s)
- Shripad N Pal
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
30
|
Garbern JC, Mummery CL, Lee RT. Model systems for cardiovascular regenerative biology. Cold Spring Harb Perspect Med 2013; 3:a014019. [PMID: 23545574 DOI: 10.1101/cshperspect.a014019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
There is an urgent clinical need to develop new therapeutic approaches to treat heart failure, but the biology of cardiovascular regeneration is complex. Model systems are required to advance our understanding of biological mechanisms of cardiac regeneration as well as to test therapeutic approaches to regenerate tissue and restore cardiac function following injury. An ideal model system should be inexpensive, easily manipulated, easily reproducible, physiologically representative of human disease, and ethically sound. In this review, we discuss computational, cell-based, tissue, and animal models that have been used to elucidate mechanisms of cardiovascular regenerative biology or to test proposed therapeutic methods to restore cardiac function following disease or injury.
Collapse
|
31
|
Santulli G, Trimarco B, Iaccarino G. G-protein-coupled receptor kinase 2 and hypertension: molecular insights and pathophysiological mechanisms. High Blood Press Cardiovasc Prev 2013; 20:5-12. [PMID: 23532739 DOI: 10.1007/s40292-013-0001-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 11/06/2012] [Indexed: 12/17/2022] Open
Abstract
Numerous factors partake in the fine-tuning of arterial blood pressure. The heptahelical G-protein-coupled receptors (GPCRs) represent one of the largest classes of cell-surface receptors. Further, ligands directed at GPCRs account for nearly 30 % of current clinical pharmaceutical agents available. Given the wide variety of GPCRs involved in blood pressure control, it is reasonable to speculate for a potential role of established intermediaries involved in the GPCR desensitization process, like the G-protein-coupled receptor kinases (GRKs), in the regulation of vascular tone. Of the seven mammalian GRKs, GRK2 seems to be the most relevant isoform at the cardiovascular level. This review attempts to assemble the currently available information concerning GRK2 and hypertension, opening new potential fields of translational investigation to treat this vexing disease.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131, Naples, Italy.
| | | | | |
Collapse
|
32
|
Fu X, Koller S, Abd Alla J, Quitterer U. Inhibition of G-protein-coupled receptor kinase 2 (GRK2) triggers the growth-promoting mitogen-activated protein kinase (MAPK) pathway. J Biol Chem 2013; 288:7738-7755. [PMID: 23362259 DOI: 10.1074/jbc.m112.428078] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inhibition of G-protein-coupled receptor kinase 2 (GRK2) is an emerging treatment option for heart failure. Because GRK2 is also indispensable for growth and development, we analyzed the impact of GRK2 inhibition on cell growth and proliferation. Inhibition of GRK2 by the dominant-negative GRK2-K220R did not affect the proliferation of cultured cells. In contrast, upon xenograft transplantation of cells into immunodeficient mice, the dominant-negative GRK2-K220R or a GRK2-specific peptide inhibitor increased tumor mass. The enhanced tumor growth upon GRK2 inhibition was attributed to the growth-promoting MAPK pathway because dual inhibition of the GRK2 and RAF-MAPK axis by the Raf kinase inhibitor protein (RKIP) did not increase tumor mass. The MAPK cascade contributed to the cardioprotective profile of GRK2 inhibition by preventing cardiomyocyte death, whereas dual inhibition of RAF/MAPK and GRK2 by RKIP induced cardiomyocyte apoptosis, cardiac dysfunction, and signs of heart failure. Thus, cardioprotective signaling induced by GRK2 inhibition is overlapping with tumor growth promotion.
Collapse
Affiliation(s)
- Xuebin Fu
- Department of Chemistry and Applied Biosciences, Molecular Pharmacology Unit, Swiss Federal Institute of Technology (ETH) Zuerich, CH-8057 Zuerich, Switzerland
| | - Samuel Koller
- Department of Chemistry and Applied Biosciences, Molecular Pharmacology Unit, Swiss Federal Institute of Technology (ETH) Zuerich, CH-8057 Zuerich, Switzerland
| | - Joshua Abd Alla
- Department of Chemistry and Applied Biosciences, Molecular Pharmacology Unit, Swiss Federal Institute of Technology (ETH) Zuerich, CH-8057 Zuerich, Switzerland
| | - Ursula Quitterer
- Department of Chemistry and Applied Biosciences, Molecular Pharmacology Unit, Swiss Federal Institute of Technology (ETH) Zuerich, CH-8057 Zuerich, Switzerland; Department of Medicine, Institute of Pharmacology and Toxicology, University of Zuerich, CH-8057 Zuerich, Switzerland.
| |
Collapse
|
33
|
|