1
|
Li J, Hu C, Zhao B, Li J, Chen L. Proteomic and cardiac dysregulation by representative perfluoroalkyl acids of different chemical speciation during early embryogenesis of zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 926:172000. [PMID: 38552965 DOI: 10.1016/j.scitotenv.2024.172000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024]
Abstract
Perfluoroalkyl acids (PFAAs) of different chemical speciation were previously found to cause diverse toxicity. However, the toxicological mechanisms depending on chemical speciation are still largely unknown. In this follow-up study, zebrafish embryos were acutely exposed to only one concentration at 4.67 μM of the acid and salt of representative PFAAs, including perfluorooctanoic acid (PFOA), perfluorobutane carboxylic acid (PFBA), and perfluorobutanesulfonic acid (PFBS), till 96 h post-fertilization (hpf), aiming to gain more mechanistic insights. High-throughput proteomics found that PFAA acid and salt exerted discriminative effects on protein expression pattern. Bioinformatic analyses based on differentially expressed proteins underlined the developmental cardiotoxicity of PFOA acid with regard to cardiac muscle contraction, vascular smooth muscle contraction, adrenergic signaling in cardiomyocytes, and multiple terms related to myocardial contraction. PFOA salt and PFBS acid merely disrupted the cardiac muscle contraction pathway, while cardiac muscle cell differentiation was significantly enriched in PFBA acid-exposed zebrafish larvae. Consistently, under PFAA exposure, especially PFOA and PFBS acid forms, transcriptional levels of key genes for cardiogenesis and the concentrations of troponin and epinephrine associated with myocardial contraction were significantly dysregulated. Moreover, a transgenic line Tg (my17: GFP) expressing green fluorescent protein in myocardial cells was employed to visualize the histopathology of developing heart. PFOA acid concurrently caused multiple deficits in heart morphogenesis and function, which were characterized by the significant increase in sinus venosus and bulbus arteriosus distance (SV-BA distance), the induction of pericardial edema, and the decrease in heart rate, further confirming the stronger toxicity of PFOA acid than the salt counterpart on heart development. Overall, this study highlighted the developmental cardiotoxicity of PFAAs, with potency ranking PFOA > PFBS > PFBA. The acid forms of PFAAs induced stronger cardiac toxicity than their salt counterparts, providing an additional insight into the structure-toxicity relationship.
Collapse
Affiliation(s)
- Jing Li
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430072, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyan Hu
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430072, China
| | - Bin Zhao
- Center for Water and Ecology, State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China.
| | - Jiali Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lianguo Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| |
Collapse
|
2
|
Kise R, Inoue A. GPCR signaling bias: an emerging framework for opioid drug development. J Biochem 2024; 175:367-376. [PMID: 38308136 DOI: 10.1093/jb/mvae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/04/2024] Open
Abstract
Biased signaling, also known as functional selectivity, has emerged as an important concept in drug development targeting G-protein-coupled receptors (GPCRs). Drugs that provoke biased signaling are expected to offer an opportunity for enhanced therapeutic effectiveness with minimized side effects. Opioid analgesics, whilst exerting potent pain-relieving effects, have become a social problem owing to their serious side effects. For the development of safer pain medications, there has been extensive exploration of agonists with a distinct balance of G-protein and β-arrestin (βarr) signaling. Recently, several approaches based on protein-protein interactions have been developed to precisely evaluate individual signal pathways, paving the way for the comprehensive analysis of biased signals. In this review, we describe an overview of bias signaling in opioid receptors, especially the μ-opioid receptor (MOR), and how to evaluate signaling bias in the GPCR field. We also discuss future directions for rational drug development through the integration of diverse signal datasets.
Collapse
Affiliation(s)
- Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
3
|
Yano H, Onoue K, Tokinaga S, Ioka T, Ishihara S, Hashimoto Y, Nakada Y, Nakagawa H, Ueda T, Seno A, Nishida T, Watanabe M, Saito Y. Overexpression of GRK2 in vascular smooth muscle leads to inappropriate hypertension and acute heart failure as in clinical scenario 1. Sci Rep 2023; 13:7707. [PMID: 37173348 PMCID: PMC10182096 DOI: 10.1038/s41598-023-34209-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Clinical scenario 1 (CS1) is acute heart failure (HF) characterized by transient systolic blood pressure (SBP) elevation and pulmonary congestion. Although it is managed by vasodilators, the molecular mechanism remains unclear. The sympathetic nervous system plays a key role in HF, and desensitization of cardiac β-adrenergic receptor (AR) signaling due to G protein-coupled receptor kinase 2 (GRK2) upregulation is known. However, vascular β-AR signaling that regulates cardiac afterload remains unelucidated in HF. We hypothesized that upregulation of vascular GRK2 leads to pathological conditions similar to CS1. GRK2 was overexpressed in vascular smooth muscle (VSM) of normal adult male mice by peritoneally injected adeno-associated viral vectors driven by the myosin heavy chain 11 promoter. Upregulation of GRK2 in VSM of GRK2 overexpressing mice augmented the absolute increase in SBP (+ 22.5 ± 4.3 mmHg vs. + 36.0 ± 4.0 mmHg, P < 0.01) and lung wet weight (4.28 ± 0.05 mg/g vs. 4.76 ± 0.15 mg/g, P < 0.01) by epinephrine as compared to those in control mice. Additionally, the expression of brain natriuretic peptide mRNA was doubled in GRK2 overexpressing mice as compared to that in control mice (P < 0.05). These findings were similar to CS1. GRK2 overexpression in VSM may cause inappropriate hypertension and HF, as in CS1.
Collapse
Affiliation(s)
- Hiroki Yano
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Shiho Tokinaga
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Tomoko Ioka
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Satomi Ishihara
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Yukihiro Hashimoto
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Yasuki Nakada
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Hitoshi Nakagawa
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Tomoya Ueda
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Ayako Seno
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Taku Nishida
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Makoto Watanabe
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan.
| |
Collapse
|
4
|
MiR-150 blunts cardiac dysfunction in mice with cardiomyocyte loss of β 1-adrenergic receptor/β-arrestin signaling and controls a unique transcriptome. Cell Death Dis 2022; 8:504. [PMID: 36585403 PMCID: PMC9803679 DOI: 10.1038/s41420-022-01295-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
The β1-adrenergic receptor (β1AR) is found primarily in hearts (mainly in cardiomyocytes [CMs]) and β-arrestin-mediated β1AR signaling elicits cardioprotection through CM survival. We showed that microRNA-150 (miR-150) is upregulated by β-arrestin-mediated β1AR signaling and that CM miR-150 inhibits maladaptive remodeling post-myocardial infarction. Here, we investigate whether miR-150 rescues cardiac dysfunction in mice bearing CM-specific abrogation of β-arrestin-mediated β1AR signaling. Using CM-specific transgenic (TG) mice expressing a mutant β1AR (G protein-coupled receptor kinase [GRK]-β1AR that exhibits impairment in β-arrestin-mediated β1AR signaling), we first generate a novel double TG mouse line overexpressing miR-150. We demonstrate that miR-150 is sufficient to improve cardiac dysfunction in CM-specific GRK-β1AR TG mice following chronic catecholamine stimulation. Our genome-wide circular RNA, long noncoding RNA (lncRNA), and mRNA profiling analyses unveil a subset of cardiac ncRNAs and genes as heretofore unrecognized mechanisms for beneficial actions of β1AR/β-arrestin signaling or miR-150. We further show that lncRNA Gm41664 and GDAP1L1 are direct novel upstream and downstream regulators of miR-150. Lastly, CM protective actions of miR-150 are attributed to repressing pro-apoptotic GDAP1L1 and are mitigated by pro-apoptotic Gm41664. Our findings support the idea that miR-150 contributes significantly to β1AR/β-arrestin-mediated cardioprotection by regulating unique ncRNA and gene signatures in CMs.
Collapse
|
5
|
Li W, Guo S, Miao N. Transcriptional responses of fluxapyroxad-induced dysfunctional heart in zebrafish (Danio rerio) embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:90034-90045. [PMID: 35864390 DOI: 10.1007/s11356-022-21981-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
Fluxapyroxad (FLU) is a succinate dehydrogenase inhibitor (SDHI) fungicide used in controlling crop diseases. Potential toxicity to aquatic organisms is not known. We exposed zebrafish to 1, 2, and 4 μM FLU for 3 days. The embryonic zebrafish showed developmental cardiac defects, including heart malformation, pericardial edema, and heart rate reduction. Compared with the controls, cardiac-specific transcription factors (nkx2.5, myh7, myl7, and myh6) exhibited dysregulated expression patterns after FLU treatment. We next used transcriptome and qRT-PCR analyses to explore the molecular mechanism of FLU cardiotoxicity. The transcriptome analysis and interaction network showed that the downregulated genes were enriched in calcium signaling pathways, adrenergic signaling in cardiomyocytes, and cardiac muscle contraction. FLU exposure repressed the cardio-related calcium signaling pathway, associated with apoptosis in the heart and other manifestations of cardiotoxicity. Thus, the findings provide valuable evidence that FLU exposure causes disruption of cardiac development in zebrafish embryos. Our findings will help to promote a better understanding of the toxicity mechanisms of FLU and act as a reference to explore the rational use and safety of FLU in agriculture.
Collapse
Affiliation(s)
- Wenhua Li
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Shanshan Guo
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Nan Miao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, People's Republic of China.
| |
Collapse
|
6
|
Joyce W, Scholman KT, Jensen B, Wang T, Boukens BJ. α 1-adrenergic stimulation increases ventricular action potential duration in the intact mouse heart. Facets (Ott) 2021. [DOI: 10.1139/facets-2020-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of α1-adrenergic receptors (α-ARs) in the regulation of myocardial function is less well-understood than that of β-ARs. Previous reports in the mouse heart have described that α1-adrenergic stimulation shortens action potential duration in isolated cells or tissues, in contrast to prolongation of the action potential reported in most other mammalian hearts. It has since become appreciated, however, that the mouse heart exhibits marked variation in inotropic response to α1-adrenergic stimulation between ventricles and even individual cardiomyocytes. We investigated the effects of α1-adrenergic stimulation on action potential duration at 80% of repolarization in the right and left ventricles of Langendorff-perfused mouse hearts using optical mapping. In hearts under β-adrenergic blockade (propranolol), phenylephrine or noradrenaline perfusion both increased action potential duration in both ventricles. The increased action potential duration was partially reversed by subsequent perfusion with the α-adrenergic antagonist phentolamine (1 μmol L−1). These data show that α1-receptor stimulation may lead to a prolonging of action potential in the mouse heart and thereby refine our understanding of how action potential duration adjusts during sympathetic stimulation.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON K1N 6N5, Canada
| | - Koen T. Scholman
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Tobias Wang
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Bastiaan J. Boukens
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1100 DD Amsterdam, the Netherlands
| |
Collapse
|
7
|
Moen JM, Matt MG, Ramirez C, Tarasov KV, Chakir K, Tarasova YS, Lukyanenko Y, Tsutsui K, Monfredi O, Morrell CH, Tagirova S, Yaniv Y, Huynh T, Pacak K, Ahmet I, Lakatta EG. Overexpression of a Neuronal Type Adenylyl Cyclase (Type 8) in Sinoatrial Node Markedly Impacts Heart Rate and Rhythm. Front Neurosci 2019; 13:615. [PMID: 31275103 PMCID: PMC6591434 DOI: 10.3389/fnins.2019.00615] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022] Open
Abstract
Heart rate (HR) and HR variability (HRV), predictors of over-all organism health, are widely believed to be driven by autonomic input to the sinoatrial node (SAN), with sympathetic input increasing HR and reducing HRV. However, variability in spontaneous beating intervals in isolated SAN tissue and single SAN cells, devoid of autonomic neural input, suggests that clocks intrinsic to SAN cells may also contribute to HR and HRV in vivo. We assessed contributions of both intrinsic and autonomic neuronal input mechanisms of SAN cell function on HR and HRV via in vivo, telemetric EKG recordings. This was done in both wild type (WT) mice, and those in which adenylyl cyclase type 8 (ADCY8), a main driver of intrinsic cAMP-PKA-Ca2+ mediated pacemaker function, was overexpressed exclusively in the heart (TGAC8). We hypothesized that TGAC8 mice would: (1) manifest a more coherent pattern of HRV in vivo, i.e., a reduced HRV driven by mechanisms intrinsic to SAN cells, and less so to modulation by autonomic input and (2) utilize unique adaptations to limit sympathetic input to a heart with high levels of intrinsic cAMP-Ca2+ signaling. Increased adenylyl cyclase (AC) activity in TGAC8 SAN tissue was accompanied by a marked increase in HR and a concurrent marked reduction in HRV, both in the absence or presence of dual autonomic blockade. The marked increase in intrinsic HR and coherence of HRV in TGAC8 mice occurred in the context of: (1) reduced HR and HRV responses to β-adrenergic receptor (β-AR) stimulation; (2) increased transcription of genes and expression of proteins [β-Arrestin, G Protein-Coupled Receptor Kinase 5 (GRK5) and Clathrin Adaptor Protein (Dab2)] that desensitize β-AR signaling within SAN tissue, (3) reduced transcripts or protein levels of enzymes [dopamine beta-hydorxylase (DBH) and phenylethanolamine N-methyltransferase (PNMT)] required for catecholamine production in intrinsic cardiac adrenergic cells, and (4) substantially reduced plasma catecholamine levels. Thus, mechanisms driven by cAMP-PKA-Ca2+ signaling intrinsic to SAN cells underlie the marked coherence of TGAC8 mice HRV. Adaptations to limit additional activation of AC signaling, via decreased neuronal sympathetic input, are utilized to ensure the hearts survival and prevent Ca2+ overload.
Collapse
Affiliation(s)
- Jack M Moen
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.,Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
| | - Michael G Matt
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.,School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christopher Ramirez
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Kirill V Tarasov
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Khalid Chakir
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yelena S Tarasova
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yevgeniya Lukyanenko
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Kenta Tsutsui
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Oliver Monfredi
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.,Department of Cardiovascular and Electrophysiology, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Christopher H Morrell
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.,Department of Mathematics, Loyola University Maryland, Baltimore, MD, United States
| | - Syevda Tagirova
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yael Yaniv
- Faculty of Biomedical Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Thanh Huynh
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Ismayil Ahmet
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Edward G Lakatta
- Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
8
|
Abstract
PURPOSE OF THE REVIEW Proinflammatory cytokines are consistently elevated in congestive heart failure. In the current review, we provide an overview on the current understanding of how tumor necrosis factor-α (TNFα), a key proinflammatory cytokine, potentiates heart failure by overwhelming the anti-inflammatory responses disrupting the homeostasis. RECENT FINDINGS Studies have shown co-relationship between severity of heart failure and levels of the proinflammatory cytokine TNFα and one of its secondary mediators interleukin-6 (IL-6), suggesting their potential as biomarkers. Recent efforts have focused on understanding the mechanisms of how proinflammatory cytokines contribute towards cardiac dysfunction and failure. In addition, how unchecked proinflammatory cytokines and their cross-talk with sympathetic system overrides the anti-inflammatory response underlying failure. The review offers insights on how TNFα and IL-6 contribute to cardiac dysfunction and failure. Furthermore, this provides a forum to begin the discussion on the cross-talk between sympathetic drive and proinflammatory cytokines and its determinant role in deleterious outcomes.
Collapse
Affiliation(s)
- Sarah M Schumacher
- NB50, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Sathyamangla V Naga Prasad
- NB50, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
9
|
Abstract
Proinflammatory reaction by the body occurs acutely in response to injury that is considered primarily beneficial. However, sustained proinflammatory cytokines observed with chronic pathologies such as metabolic syndrome, cancer, and arthritis are detrimental and in many cases is a major cardiovascular risk factor. Proinflammatory cytokines such as interleukin-1, interleukin-6, and tumor necrosis factor α (TNFα) have long been implicated in cardiovascular risk and considered to be a major underlying cause for heart failure (HF). The failure of the anti-TNFα therapy for HF indicates our elusive understanding on the dichotomous role of proinflammatory cytokines on acutely beneficial effects versus long-term deleterious effects. Despite these well-described observations, less is known about the mechanistic underpinnings of proinflammatory cytokines especially TNFα in pathogenesis of HF. Increasing evidence suggests the existence of an active cross-talk between the TNFα receptor signaling and G-protein-coupled receptors such as β-adrenergic receptor (βAR). Given that βARs are the key regulators of cardiac function, the review will discuss the current state of understanding on the role of proinflammatory cytokine TNFα in regulating βAR function.
Collapse
Affiliation(s)
- Maradumane L Mohan
- *Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and †Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH
| | | | | |
Collapse
|
10
|
Unique Roles of β-Arrestin in GPCR Trafficking Revealed by Photoinducible Dimerizers. Sci Rep 2018; 8:677. [PMID: 29330504 PMCID: PMC5766490 DOI: 10.1038/s41598-017-19130-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/21/2017] [Indexed: 12/21/2022] Open
Abstract
Intracellular trafficking of G protein-coupled receptors (GPCRs) controls their localization and degradation, which affects a cell's ability to adapt to extracellular stimuli. Although the perturbation of trafficking induces important diseases, these trafficking mechanisms are poorly understood. Herein, we demonstrate an optogenetic method using an optical dimerizer, cryptochrome (CRY) and its partner protein (CIB), to analyze the trafficking mechanisms of GPCRs and their regulatory proteins. Temporally controlling the interaction between β-arrestin and β2-adrenergic receptor (ADRB2) reveals that the duration of the β-arrestin-ADRB2 interaction determines the trafficking pathway of ADRB2. Remarkably, the phosphorylation of ADRB2 by G protein-coupled receptor kinases is unnecessary to trigger clathrin-mediated endocytosis, and β-arrestin interacting with unphosphorylated ADRB2 fails to activate mitogen-activated protein kinase (MAPK) signaling, in contrast to the ADRB2 agonist isoproterenol. Temporal control of β-arrestin-GPCR interactions will enable the investigation of the unique roles of β-arrestin and the mechanism by which it regulates β-arrestin-specific trafficking pathways of different GPCRs.
Collapse
|
11
|
Targeting GPCR-Gβγ-GRK2 signaling as a novel strategy for treating cardiorenal pathologies. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1883-1892. [PMID: 28130200 DOI: 10.1016/j.bbadis.2017.01.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/13/2017] [Accepted: 01/18/2017] [Indexed: 02/06/2023]
Abstract
The pathologic crosstalk between the heart and kidney is known as cardiorenal syndrome (CRS). While the specific mechanisms underlying this crosstalk remain poorly understood, CRS is associated with exacerbated dysfunction of either or both organs and reduced survival. Maladaptive fibrotic remodeling is a key component of both heart and kidney failure pathogenesis and progression. G-protein coupled receptor (GPCR) signaling is a crucial regulator of cardiovascular and renal function. Chronic/pathologic GPCR signaling elicits the interaction of the G-protein Gβγ subunit with GPCR kinase 2 (GRK2), targeting the receptor for internalization, scaffolding to pathologic signals, and receptor degradation. Targeting this pathologic Gβγ-GRK2 interaction has been suggested as a possible strategy for the treatment of HF. In the current review, we discuss recent updates in understanding the role of GPCR-Gβγ-GRK2 signaling as a crucial mediator of maladaptive organ remodeling detected in HF and kidney dysfunction, with specific attention to small molecule-mediated inhibition of pathologic Gβγ-GRK2 interactions. Further, we explore the potential of GPCR-Gβγ-GRK2 signaling as a possible therapeutic target for cardiorenal pathologies.
Collapse
|
12
|
Abstract
G protein-coupled receptors are the largest family of targets for current therapeutics. The classic model of their activation was binary, where agonist binding induced an active conformation and subsequent downstream signaling. Subsequently, the revised concept of biased agonism emerged, where different ligands at the same G protein-coupled receptor selectively activate one downstream pathway versus another. Advances in understanding the mechanism of biased agonism have led to the development of novel ligands, which have the potential for improved therapeutic and safety profiles. In this review, we summarize the theory and most recent breakthroughs in understanding biased signaling, examine recent laboratory investigations concerning biased ligands across different organ systems, and discuss the promising clinical applications of biased agonism.
Collapse
|
13
|
Abstract
Sympathetic nervous system overactivity has been linked to ventricular tachyarrhythmias and sudden death. It has been hypothesized that the extent and nature of the arrhythmogenic effect of sympathetic stimulation depends on the underlying myocardial substrate, the mechanism of the arrhythmia, and the integrated effects of sympathetic stimulation in the particular individual circumstance. Multiple direct and indirect mechanisms of adrenergic action on the heart may benefit from the known antiarrhythmic actions of β-blocker therapy and other interventions that decrease sympathetic tone. The antiarrhythmic mechanism of β-blockade (and possibly α-blockade) will depend on the specific mechanism of the individual arrhythmia and will differ for those arrhythmias caused by tachycardia and ischemia, those caused by reentry and promoted by decreased conduction velocity and shortened refractoriness, and those caused by early or delayed afterdepolarizations, usually in the context of prolonged action potential duration. Antagonism of cardiac adrenergic activity by β-blockade in particular is the best-established drug therapy to prevent ventricular arrhythmias.
Collapse
Affiliation(s)
- Paul Dorian
- Division of Cardiology, St. Michael's Hospital, University of Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Abstract
In the classical two-state model, G protein-coupled receptors (GPCRs) are considered to exist in equilibrium between an active and an inactive conformation. Thus, even at the resting state, some subpopulation of GPCRs is in the active state, which underlies the basal activity of the GPCRs. In this review, we discuss inverse agonists, which are defined as GPCR ligands that shift the equilibrium toward the inactive state and thereby suppress the basal activity. Theoretically, if constitutive activation plays an essential role in the pathogenesis of a disease, only inverse agonists, and not neutral antagonists, can reverse this pathophysiological activation. Although many pharmacological examples of inverse agonism have been identified, its clinical importance is still unclear and debated. Thus, even though inverse agonism of angiotensin receptor blockers (ARBs) has been discussed for more than 10 years, its clinical relevance remains to be completely clarified.
Collapse
Affiliation(s)
- Junichiro Sato
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo 113-8655, Japan
| | | | | |
Collapse
|
15
|
Watson LJ, Alexander KM, Mohan ML, Bowman AL, Mangmool S, Xiao K, Naga Prasad SV, Rockman HA. Phosphorylation of Src by phosphoinositide 3-kinase regulates beta-adrenergic receptor-mediated EGFR transactivation. Cell Signal 2016; 28:1580-92. [PMID: 27169346 DOI: 10.1016/j.cellsig.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 01/08/2023]
Abstract
β2-Adrenergic receptors (β2AR) transactivate epidermal growth factor receptors (EGFR) through formation of a β2AR-EGFR complex that requires activation of Src to mediate signaling. Here, we show that both lipid and protein kinase activities of the bifunctional phosphoinositide 3-kinase (PI3K) enzyme are required for β2AR-stimulated EGFR transactivation. Mechanistically, the generation of phosphatidylinositol (3,4,5)-tris-phosphate (PIP3) by the lipid kinase function stabilizes β2AR-EGFR complexes while the protein kinase activity of PI3K regulates Src activation by direct phosphorylation. The protein kinase activity of PI3K phosphorylates serine residue 70 on Src to enhance its activity and induce EGFR transactivation following βAR stimulation. This newly identified function for PI3K, whereby Src is a substrate for the protein kinase activity of PI3K, is of importance since Src plays a key role in pathological and physiological signaling.
Collapse
Affiliation(s)
- Lewis J Watson
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Kevin M Alexander
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, United States
| | - Amber L Bowman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand
| | - Kunhong Xiao
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States; Department of Pharmacology and Chemical Biology, University of Pittsburg School of Medicine, Pittsburgh, PA 15261, United States
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, United States.
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
16
|
Zhang W, Qu X, Chen B, Snyder M, Wang M, Li B, Tang Y, Chen H, Zhu W, Zhan L, Yin N, Li D, Xie L, Liu Y, Zhang JJ, Fu XY, Rubart M, Song LS, Huang XY, Shou W. Critical Roles of STAT3 in β-Adrenergic Functions in the Heart. Circulation 2016; 133:48-61. [PMID: 26628621 PMCID: PMC4698100 DOI: 10.1161/circulationaha.115.017472] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/02/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND β-Adrenergic receptors (βARs) play paradoxical roles in the heart. On one hand, βARs augment cardiac performance to fulfill the physiological demands, but on the other hand, prolonged activations of βARs exert deleterious effects that result in heart failure. The signal transducer and activator of transcription 3 (STAT3) plays a dynamic role in integrating multiple cytokine signaling pathways in a number of tissues. Altered activation of STAT3 has been observed in failing hearts in both human patients and animal models. Our objective is to determine the potential regulatory roles of STAT3 in cardiac βAR-mediated signaling and function. METHODS AND RESULTS We observed that STAT3 can be directly activated in cardiomyocytes by β-adrenergic agonists. To follow up this finding, we analyzed βAR function in cardiomyocyte-restricted STAT3 knockouts and discovered that the conditional loss of STAT3 in cardiomyocytes markedly reduced the cardiac contractile response to acute βAR stimulation, and caused disengagement of calcium coupling and muscle contraction. Under chronic β-adrenergic stimulation, Stat3cKO hearts exhibited pronounced cardiomyocyte hypertrophy, cell death, and subsequent cardiac fibrosis. Biochemical and genetic data supported that Gαs and Src kinases are required for βAR-mediated activation of STAT3. Finally, we demonstrated that STAT3 transcriptionally regulates several key components of βAR pathway, including β1AR, protein kinase A, and T-type Ca(2+) channels. CONCLUSIONS Our data demonstrate for the first time that STAT3 has a fundamental role in βAR signaling and functions in the heart. STAT3 serves as a critical transcriptional regulator for βAR-mediated cardiac stress adaption, pathological remodeling, and heart failure.
Collapse
Affiliation(s)
- Wenjun Zhang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.).
| | - Xiuxia Qu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Biyi Chen
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Marylynn Snyder
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Meijing Wang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Baiyan Li
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Yue Tang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Hanying Chen
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Wuqiang Zhu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Li Zhan
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Ni Yin
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Deqiang Li
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Li Xie
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Ying Liu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - J Jillian Zhang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Xin-Yuan Fu
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Michael Rubart
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Long-Sheng Song
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Xin-Yun Huang
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.)
| | - Weinian Shou
- From State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (W. Zhang, X.Q., Y.T., W.S.); Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indianapolis, IN (W. Zhang, B.L., H.C., W. Zhu, L.Z., N.Y., D.L., L.X., Y.L., M.R., W.S.); Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City (B.C., L.-S.S.); Department of Physiology and Biophysics, Cornell University Weill Medical College, New York, NY (M.S., J.J.Z., X.-Y.H.); Department of Surgery, Indiana University School of Medicine, Indianapolis (M.W.); Department of Pharmacology, Harbin Medical University, Harbin, China (B.L.); Department of Heart Surgery, Xiangya 2nd Hospital, Central South University, Changsha, China (N.Y., L.X.); and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis (X.-Y.F.).
| |
Collapse
|
17
|
Castaldi A, Zaglia T, Di Mauro V, Carullo P, Viggiani G, Borile G, Di Stefano B, Schiattarella GG, Gualazzi MG, Elia L, Stirparo GG, Colorito ML, Pironti G, Kunderfranco P, Esposito G, Bang ML, Mongillo M, Condorelli G, Catalucci D. MicroRNA-133 modulates the β1-adrenergic receptor transduction cascade. Circ Res 2014; 115:273-83. [PMID: 24807785 DOI: 10.1161/circresaha.115.303252] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RATIONALE The sympathetic nervous system plays a fundamental role in the regulation of myocardial function. During chronic pressure overload, overactivation of the sympathetic nervous system induces the release of catecholamines, which activate β-adrenergic receptors in cardiomyocytes and lead to increased heart rate and cardiac contractility. However, chronic stimulation of β-adrenergic receptors leads to impaired cardiac function, and β-blockers are widely used as therapeutic agents for the treatment of cardiac disease. MicroRNA-133 (miR-133) is highly expressed in the myocardium and is involved in controlling cardiac function through regulation of messenger RNA translation/stability. OBJECTIVE To determine whether miR-133 affects β-adrenergic receptor signaling during progression to heart failure. METHODS AND RESULTS Based on bioinformatic analysis, β1-adrenergic receptor (β1AR) and other components of the β1AR signal transduction cascade, including adenylate cyclase VI and the catalytic subunit of the cAMP-dependent protein kinase A, were predicted as direct targets of miR-133 and subsequently validated by experimental studies. Consistently, cAMP accumulation and activation of downstream targets were repressed by miR-133 overexpression in both neonatal and adult cardiomyocytes following selective β1AR stimulation. Furthermore, gain-of-function and loss-of-function studies of miR-133 revealed its role in counteracting the deleterious apoptotic effects caused by chronic β1AR stimulation. This was confirmed in vivo using a novel cardiac-specific TetON-miR-133 inducible transgenic mouse model. When subjected to transaortic constriction, TetON-miR-133 inducible transgenic mice maintained cardiac performance and showed attenuated apoptosis and reduced fibrosis compared with control mice. CONCLUSIONS miR-133 controls multiple components of the β1AR transduction cascade and is cardioprotective during heart failure.
Collapse
Affiliation(s)
- Alessandra Castaldi
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Tania Zaglia
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Vittoria Di Mauro
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Pierluigi Carullo
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Giacomo Viggiani
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Giulia Borile
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Barbara Di Stefano
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Gabriele Giacomo Schiattarella
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Maria Giovanna Gualazzi
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Leonardo Elia
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Giuliano Giuseppe Stirparo
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Maria Luisa Colorito
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Gianluigi Pironti
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Paolo Kunderfranco
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Giovanni Esposito
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Marie-Louise Bang
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Marco Mongillo
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Gianluigi Condorelli
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.)
| | - Daniele Catalucci
- From the Humanitas Clinical and Research Center, Rozzano, Milan, Italy (A.C., V.D.M., P.C., G.V., M.G.G., G.G.S., P.K., M.-L.B., G.C., D.C.); Multimedica, Milan, Italy (L.E.); University of Milan Bicocca, Milan, Italy (A.C.); Venetian Institute of Molecular Medicine, Padova, Italy (T.Z., G.B., M.M.); University of Padova, Padova, Italy (T.Z., G.B., M.M.); Institute of Genetic and Biomedical Research-Milan Unit, Milan, Italy (P.C., M.-L.B., G.C., D.C.); University "Federico II," Naples, Italy (G.G.S., G.E.); University of Milan, Milan, Italy (G.G.S., G.C.); Duke University Medical Center, Durham, NC (G.P.); and University of Palermo, Palermo, Italy (B.D.S., M.L.C.).
| |
Collapse
|
18
|
Kamal FA, Mickelsen DM, Wegman KM, Travers JG, Moalem J, Hammes SR, Smrcka AV, Blaxall BC. Simultaneous adrenal and cardiac g-protein-coupled receptor-gβγ inhibition halts heart failure progression. J Am Coll Cardiol 2014; 63:2549-2557. [PMID: 24703913 DOI: 10.1016/j.jacc.2014.02.587] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/10/2014] [Accepted: 02/25/2014] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The authors propose simultaneous inhibition of Gβγ signaling in the heart and the adrenal gland as a novel therapeutic approach for heart failure (HF). BACKGROUND Elevated sympathetic nervous system activity is a salient characteristic of HF progression. It causes pathologic desensitization of β-adrenergic receptors (β-AR), facilitated predominantly through Gβγ-mediated signaling. The adrenal glands are key contributors to the chronically elevated plasma catecholamine levels observed in HF, where adrenal α2-AR feedback inhibitory function is impaired also through Gβγ-mediated signaling. METHODS We investigated the efficacy of a small molecule Gβγ inhibitor, gallein, in a clinically relevant, pressure-overload model of HF. RESULTS Daily gallein treatment (10 mg/kg/day), initiated 4 weeks after transverse aortic constriction, improved survival and cardiac function and attenuated cardiac remodeling. Mechanistically, gallein restored β-AR membrane density in cardiomyocytes, attenuated Gβγ-mediated G-protein-coupled receptor kinase 2-phosphoinositide 3-kinase γ membrane recruitment, and reduced Akt (protein kinase B) and glycogen synthase kinase 3β phosphorylation. Gallein also reduced circulating plasma catecholamine levels and catecholamine production in isolated mouse adrenal glands by restoring adrenal α2-AR feedback inhibition. In human adrenal endocrine tumors (pheochromocytoma), gallein attenuated catecholamine secretion, as well as G-protein-coupled receptor kinase 2 expression and membrane translocation. CONCLUSIONS These data suggest small molecule Gβγ inhibition as a systemic pharmacologic therapy for HF by simultaneously normalizing pathologic adrenergic/Gβγ signaling in both the heart and the adrenal gland. Our data also suggest important endocrine/cardiovascular interactions and a possible role for small molecule Gβγ inhibition in treating endocrine tumors such as pheochromocytoma, in addition to HF.
Collapse
Affiliation(s)
- Fadia A Kamal
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Deanne M Mickelsen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Katherine M Wegman
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joshua G Travers
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jacob Moalem
- Department of Surgery, University of Rochester Medical Center, Rochester, New York; Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Stephen R Hammes
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Burns C Blaxall
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
19
|
Heyen JR, Hu W, Jamieson J, Thibault S, Batugo M, Loi CM, Burns-Naas LA, McHarg AD, Jessen B. Cardiovascular differentiation of imatinib and bosutinib in the rat. Int J Hematol 2013; 98:597-607. [DOI: 10.1007/s12185-013-1453-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/13/2013] [Accepted: 10/06/2013] [Indexed: 12/23/2022]
|
20
|
The Arf GAP AGAP2 interacts with β-arrestin2 and regulates β2-adrenergic receptor recycling and ERK activation. Biochem J 2013; 452:411-21. [PMID: 23527545 DOI: 10.1042/bj20121004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
AGAP2 [Arf (ADP-ribosylation factor) GAP (GTPase-activating protein) with GTP-binding-protein-like, ankyrin repeat and PH (pleckstrin homology) domains] is a multidomain Arf GAP that was shown to promote the fast recycling of transferrin receptors. In the present study we tested the hypothesis that AGAP2 regulates the trafficking of β2-adrenergic receptors. We found that AGAP2 formed a complex with β-arrestin1 and β-arrestin2, proteins that are known to regulate β2-adrenergic receptor signalling and trafficking. AGAP2 co-localized with β-arrestin2 on the plasma membrane, and knockdown of AGAP2 expression reduced plasma membrane association of β-arrestin2 upon β2-adrenergic receptor activation. AGAP2 also co-localized with internalized β2-adrenergic receptors on endosomes, and overexpression of AGAP2 slowed accumulation of β2-adrenergic receptor in the perinuclear recycling endosomes. In contrast, knockdown of AGAP2 expression prevented the recycling of the β2-adrenergic receptor back to the plasma membrane. In addition, AGAP2 formed a complex with endogenous ERK (extracellular-signal-regulated kinase) and overexpression of AGAP2 potentiated ERK phosphorylation induced by β2-adrenergic receptors. Taken together, these results support the hypothesis that AGAP2 plays a role in the signalling and recycling of β2-adrenergic receptors.
Collapse
|
21
|
Swain JD, Fargnoli AS, Katz MG, Tomasulo CE, Sumaroka M, Richardville KC, Koch WJ, Rabinowitz JE, Bridges CR. MCARD-mediated gene transfer of GRK2 inhibitor in ovine model of acute myocardial infarction. J Cardiovasc Transl Res 2013; 6:253-62. [PMID: 23208013 PMCID: PMC3695486 DOI: 10.1007/s12265-012-9418-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 10/15/2012] [Indexed: 01/08/2023]
Abstract
β-Adrenergic receptor (βAR) dysfunction in acute myocardial infarction (MI) is associated with elevated levels of the G-protein-coupled receptor kinase-2 (GRK2), which plays a key role in heart failure progression. Inhibition of GRK2 via expression of a peptide βARKct transferred by molecular cardiac surgery with recirculating delivery (MCARD) may be a promising intervention. Five sheep underwent scAAV6-mediated MCARD delivery of βARKct, and five received no treatment (control). After a 3-week period, the branch of the circumflex artery (OM1) was ligated. Quantitative PCR data showed intense βARKct expression in the left ventricle (LV). Circumferential fractional shortening was 23.4 ± 7.1 % (baseline) vs. -2.9 ± 5.2 % (p < 0.05) in the control at 10 weeks. In the MCARD-βARKct group, this parameter was close to baseline. The same trend was observed with LV wall thickening. Cardiac index fully recovered in the MCARD-βARKct group. LV end-diastolic volume and LV end-diastolic pressure did not differ in both groups. MCARD-mediated βARKct gene expression results in preservation of regional and global systolic function after acute MI without arresting progressive ventricular remodeling.
Collapse
Affiliation(s)
- JaBaris D. Swain
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Anthony S. Fargnoli
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Michael G. Katz
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Catherine E. Tomasulo
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Marina Sumaroka
- Department of Surgery, Division of Cardiovascular Surgery, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Kyle C. Richardville
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| | - Walter J. Koch
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Joseph E. Rabinowitz
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Charles R. Bridges
- Sanger Heart and Vascular Institute, Cannon Research Center, Carolinas HealthCare System, Charlotte, North Carolina
| |
Collapse
|
22
|
Vélez JM, Chamorro GA, Calzada CC, Zuñiga CA, Vélez JJ, Ocharán E. A study of prevention and regression of cardiac hypertrophy with a prolactin inhibitor in a biological model of ventricular hypertrophy caused by aorto caval fistulae in rat. Cardiovasc Pathol 2013; 22:357-67. [PMID: 23478013 DOI: 10.1016/j.carpath.2013.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The possibility of decreasing or reverting left ventricular hypertrophy and, therefore, cardiac hypertrophy (CH) is an important medical issue. The aim of the present study was to evaluate these two possibilities with a 3-week daily dose of captopril, losartan, or bromocriptine in a preventive or corrective model. METHODS After aorto caval fistulae (ACF) surgery on adult male Wistar rats to induce CH, animals were assigned to the preventive protocol (drug treatment began immediately after surgery) or corrective protocol (hypertrophy was allowed to develop before drug treatment). After treatments, isoproterenol was administered to half of the animals to further induce CH. The groups included the passive control, the sham-operated animals, those with ACF surgery but without drug treatment, and the 3-week treatments with captopril, losartan, or the low or high dose of bromocriptine. RESULTS Three treatments, with captopril, losartan, or the high dose of bromocriptine, significantly impeded/reverted an increase in CH-related parameters in the preventive/corrective model compared to the surgically treated group without drug treatment. The same effect was found after isoproterenol administration. The present results show an avoidance/reversion of CH with these three treatments. Better results were found with the angiotensin converting enzyme inhibitor (captopril) than with the prolactin inhibitor (bromocriptine). CONCLUSIONS Treatments with captopril, losartan, and the high dose of bromocriptine were effective in preventing/reversing the manifestation of CH in the preventive/corrective rat models. Further studies are needed to identify the initial mediator, the key component, and the molecular events involved in the pathogenesis of CH.
Collapse
Affiliation(s)
- Juan M Vélez
- Intracellular Signaling Laboratory, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México
| | | | | | | | | | | |
Collapse
|
23
|
Goldhaber JI, Philipson KD. Cardiac sodium-calcium exchange and efficient excitation-contraction coupling: implications for heart disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 961:355-64. [PMID: 23224894 DOI: 10.1007/978-1-4614-4756-6_30] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide, with ischemic heart disease alone accounting for >12% of all deaths, more than HIV/AIDS, tuberculosis, lung, and breast cancer combined. Heart disease has been the leading cause of death in the United States for the past 85 years and is a major cause of disability and health-care expenditures. The cardiac conditions most likely to result in death include heart failure and arrhythmias, both a consequence of ischemic coronary disease and myocardial infarction, though chronic hypertension and valvular diseases are also important causes of heart failure. Sodium-calcium exchange (NCX) is the dominant calcium (Ca2+) efflux mechanism in cardiac cells. Using ventricular-specific NCX knockout mice, we have found that NCX is also an essential regulator of cardiac contractility independent of sarcoplasmic reticulum Ca2+ load. During the upstroke of the action potential, sodium (Na+) ions enter the diadic cleft space between the sarcolemma and the sarcoplasmic reticulum. The rise in cleft Na+, in conjunction with depolarization, causes NCX to transiently reverse. Ca2+ entry by this mechanism then "primes" the diadic cleft so that subsequent Ca2+ entry through Ca2+ channels can more efficiently trigger Ca2+ release from the sarcoplasmic reticulum. In NCX knockout mice, this mechanism is inoperative (Na+ current has no effect on the Ca2+ transient), and excitation-contraction coupling relies upon the elevated diadic cleft Ca2+ that arises from the slow extrusion of cytoplasmic Ca2+ by the ATP-dependent sarcolemmal Ca2+ pump. Thus, our data support the conclusion that NCX is an important regulator of cardiac contractility. These findings suggest that manipulation of NCX may be beneficial in the treatment of heart failure.
Collapse
|
24
|
Pleger ST, Shan C, Ksienzyk J, Bekeredjian R, Boekstegers P, Hinkel R, Schinkel S, Leuchs B, Ludwig J, Qiu G, Weber C, Raake P, Koch WJ, Katus HA, Müller OJ, Most P. Cardiac AAV9-S100A1 gene therapy rescues post-ischemic heart failure in a preclinical large animal model. Sci Transl Med 2012; 3:92ra64. [PMID: 21775667 DOI: 10.1126/scitranslmed.3002097] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As a prerequisite for clinical application, we determined the long-term therapeutic effectiveness and safety of adeno-associated virus (AAV)-S100A1 gene therapy in a preclinical large animal model of heart failure. S100A1, a positive inotropic regulator of myocardial contractility, becomes depleted in failing cardiomyocytes in humans and animals, and myocardial-targeted S100A1 gene transfer rescues cardiac contractile function by restoring sarcoplasmic reticulum calcium (Ca(2+)) handling in acutely and chronically failing hearts in small animal models. We induced heart failure in domestic pigs by balloon occlusion of the left circumflex coronary artery, resulting in myocardial infarction. After 2 weeks, when the pigs displayed significant left ventricular contractile dysfunction, we administered, by retrograde coronary venous delivery, AAV serotype 9 (AAV9)-S100A1 to the left ventricular, non-infarcted myocardium. AAV9-luciferase and saline treatment served as control. At 14 weeks, both control groups showed significantly decreased myocardial S100A1 protein expression along with progressive deterioration of cardiac performance and left ventricular remodeling. AAV9-S100A1 treatment prevented and reversed these functional and structural changes by restoring cardiac S100A1 protein levels. S100A1 treatment normalized cardiomyocyte Ca(2+) cycling, sarcoplasmic reticulum calcium handling, and energy homeostasis. Transgene expression was restricted to cardiac tissue, and extracardiac organ function was uncompromised. This translational study shows the preclinical feasibility of long-term therapeutic effectiveness of and a favorable safety profile for cardiac AAV9-S100A1 gene therapy in a preclinical model of heart failure. Our results present a strong rationale for a clinical trial of S100A1 gene therapy for human heart failure that could potentially complement current strategies to treat end-stage heart failure.
Collapse
Affiliation(s)
- Sven T Pleger
- Center for Molecular and Translational Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Multifaceted cardiac signal transduction mediated by G protein-coupled receptors: Potential target sites where an unambiguous attention is needed for exploring new drugs for cardiovascular disorders. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/j.biomag.2011.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
26
|
Abstract
G protein-coupled receptors (GPCRs) represent the largest family of membrane receptors and are responsible for regulating a wide variety of physiological processes. This is accomplished via ligand binding to GPCRs, activating associated heterotrimeric G proteins and intracellular signaling pathways. G protein-coupled receptor kinases (GRKs), in concert with β-arrestins, classically desensitize receptor signal transduction, thus preventing hyperactivation of GPCR second-messenger cascades. As changes in GRK expression have featured prominently in many cardiovascular pathologies, including heart failure, myocardial infarction, hypertension, and cardiac hypertrophy, GRKs have been intensively studied as potential diagnostic or therapeutic targets. Herein, we review our evolving understanding of the role of GRKs in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Stephen L Belmonte
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | |
Collapse
|
27
|
Abstract
Increasing numbers of compounds, previously classified as antagonists, were shown to inhibit this spontaneous or constitutive receptor activity, instead of leave it unaffected as expected for a formal antagonist. In addition, some other antagonists did not have any effect by themselves, but prevented the inhibition of constitutive activity induced by thought-to-be antagonists. These thought-to-be antagonists with negative efficacy are now known as "inverse agonists." Inverse agonism at βAR has been evidenced for both subtypes in wild-type GPCRs systems and in engineered systems with high constitutive activity. It is important to mention that native systems are of particular importance for analyzing the in vivo relevance of constitutive activity because these systems have physiological expression levels of target receptors. Studies of inverse agonism of β blockers in physiological setting have also evidenced that pathophysiological conditions can affect pharmacodynamic properties of these ligands. To date, hundreds of clinically well-known drugs have been tested and classified for this property. Prominent examples include the beta-blockers propranolol, alprenolol, pindolol, and timolol used for treating hypertension, angina pectoris, and arrhythmia that act on the β₂ARs, metoprolol, and bisoprolol used for treating hypertension, coronary heart disease, and arrhythmias by acting on β₁ARs. Inverse agonists seem to be useful in the treatment of chronic disease characterized by harmful effects resulting from β₁AR and β₂AR overactivation, such as heart failure and asthma, respectively.
Collapse
Affiliation(s)
- Carlos A Taira
- Cátedra de Farmacología, Instituto de Fisiopatología y Bioquímica Clínica, Universidad de Buenos Aires, CONICET, Junín 956, Buenos Aires, Argentina
| | | | | |
Collapse
|
28
|
Kamal FA, Smrcka AV, Blaxall BC. Taking the heart failure battle inside the cell: small molecule targeting of Gβγ subunits. J Mol Cell Cardiol 2011; 51:462-7. [PMID: 21256851 DOI: 10.1016/j.yjmcc.2011.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2010] [Revised: 01/09/2011] [Accepted: 01/11/2011] [Indexed: 10/18/2022]
Abstract
Heart failure (HF) is devastating disease with poor prognosis. Elevated sympathetic nervous system activity and outflow, leading to pathologic attenuation and desensitization of β-adrenergic receptors (β-ARs) signaling and responsiveness, are salient characteristic of HF progression. These pathologic effects on β-AR signaling and HF progression occur in part due to Gβγ-mediated signaling, including recruitment of receptor desensitizing kinases such as G-protein coupled receptor (GPCR) kinase 2 (GRK2) and phosphoinositide 3-kinase (PI3K), which subsequently phosphorylate agonist occupied GPCRs. Additionally, chronic GPCR signaling signals chronically dissociated Gβγ subunits to interact with multiple effector molecules that activate various signaling cascades involved in HF pathophysiology. Importantly, targeting Gβγ signaling with large peptide inhibitors has proven a promising therapeutic paradigm in the treatment of HF. We recently described an approach to identify small molecule Gβγ inhibitors that selectively block particular Gβγ functions by specifically targeting a Gβγ protein-protein interaction "hot spot." Here we describe their effects on Gβγ downstream signaling pathways, including their role in HF pathophysiology. We suggest a promising therapeutic role for small molecule inhibition of pathologic Gβγ signaling in the treatment of HF. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."
Collapse
Affiliation(s)
- Fadia A Kamal
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | | |
Collapse
|
29
|
Völkers M, Weidenhammer C, Herzog N, Qiu G, Spaich K, Wegner FV, Peppel K, Müller OJ, Schinkel S, Rabinowitz JE, Hippe HJ, Brinks H, Katus HA, Koch WJ, Eckhart AD, Friedrich O, Most P. The inotropic peptide βARKct improves βAR responsiveness in normal and failing cardiomyocytes through G(βγ)-mediated L-type calcium current disinhibition. Circ Res 2011; 108:27-39. [PMID: 21106943 PMCID: PMC4013502 DOI: 10.1161/circresaha.110.225201] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 11/15/2010] [Indexed: 12/20/2022]
Abstract
RATIONALE The G(βγ)-sequestering peptide β-adrenergic receptor kinase (βARK)ct derived from the G-protein-coupled receptor kinase (GRK)2 carboxyl terminus has emerged as a promising target for gene-based heart failure therapy. Enhanced downstream cAMP signaling has been proposed as the underlying mechanism for increased β-adrenergic receptor (βAR) responsiveness. However, molecular targets mediating improved cardiac contractile performance by βARKct and its impact on G(βγ)-mediated signaling have yet to be fully elucidated. OBJECTIVE We sought to identify G(βγ)-regulated targets and signaling mechanisms conveying βARKct-mediated enhanced βAR responsiveness in normal (NC) and failing (FC) adult rat ventricular cardiomyocytes. METHODS AND RESULTS Assessing viral-based βARKct gene delivery with electrophysiological techniques, analysis of contractile performance, subcellular Ca²(+) handling, and site-specific protein phosphorylation, we demonstrate that βARKct enhances the cardiac L-type Ca²(+) channel (LCC) current (I(Ca)) both in NCs and FCs on βAR stimulation. Mechanistically, βARKct augments I(Ca) by preventing enhanced inhibitory interaction between the α1-LCC subunit (Cav1.2α) and liberated G(βγ) subunits downstream of activated βARs. Despite improved βAR contractile responsiveness, βARKct neither increased nor restored cAMP-dependent protein kinase (PKA) and calmodulin-dependent kinase II signaling including unchanged protein kinase (PK)Cε, extracellular signal-regulated kinase (ERK)1/2, Akt, ERK5, and p38 activation both in NCs and FCs. Accordingly, although βARKct significantly increases I(Ca) and Ca²(+) transients, being susceptible to suppression by recombinant G(βγ) protein and use-dependent LCC blocker, βARKct-expressing cardiomyocytes exhibit equal basal and βAR-stimulated sarcoplasmic reticulum Ca²(+) load, spontaneous diastolic Ca²(+) leakage, and survival rates and were less susceptible to field-stimulated Ca²(+) waves compared with controls. CONCLUSION Our study identifies a G(βγ)-dependent signaling pathway attenuating cardiomyocyte I(Ca) on βAR as molecular target for the G(βγ)-sequestering peptide βARKct. Targeted interruption of this inhibitory signaling pathway by βARKct confers improved βAR contractile responsiveness through increased I(Ca) without enhancing regular or restoring abnormal cAMP-signaling. βARKct-mediated improvement of I(Ca) rendered cardiomyocytes neither susceptible to βAR-induced damage nor arrhythmogenic sarcoplasmic reticulum Ca²(+) leakage.
Collapse
Affiliation(s)
- Mirko Völkers
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Christian Weidenhammer
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Nicole Herzog
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Gang Qiu
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Kristin Spaich
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Frederic V Wegner
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Karsten Peppel
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Oliver J Müller
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Stefanie Schinkel
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Joseph E Rabinowitz
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Hans-Jorg Hippe
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Henriette Brinks
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Hugo A Katus
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Walter J Koch
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Andrea D Eckhart
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Oliver Friedrich
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Patrick Most
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| |
Collapse
|
30
|
Grönefeld GC, Bänsch D. [Antiarrhythmic therapy with β-receptor antagonists]. Herzschrittmacherther Elektrophysiol 2010; 21:222-227. [PMID: 21104261 DOI: 10.1007/s00399-010-0089-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 10/13/2010] [Indexed: 05/30/2023]
Abstract
β-Blockers are an essential component of medical therapy in patients with ischemic heart disease or cardiac dysfunction of any genesis. They have an effect at the level of the sinus and the atrioventricular node, as well as on the atrial and ventricular refractory period of the myocardium. Overall, there are complicated antifibrillatory effects which are involved in the reduction of morbidity and mortality of this the therapy. According to the guidelines, it is important to uptitrate to highest tolerated dose. In patients with atrial fibrillation, antiadrenergic therapy should be the first line treatment; if well tolerated, then β-blockers alone or as a combination with an antiarrhythmic drug is preferable. Future prospective studies on the antiarrhythmic effects in this therapeutic area should include comparisons of different α - and β-selective active substances. Increasing knowledge of the differential therapy with the available active substances including intravenously applicable short-acting β-blockers, e.g., in intensive care therapy - should distinguish the different therapeutic effects.
Collapse
Affiliation(s)
- G C Grönefeld
- I. Medizinische Abteilung, Asklepios Klinik Barmbek, Rübenkamp 220, 22291, Hamburg, Deutschland.
| | | |
Collapse
|
31
|
Mangmool S, Shukla AK, Rockman HA. beta-Arrestin-dependent activation of Ca(2+)/calmodulin kinase II after beta(1)-adrenergic receptor stimulation. ACTA ACUST UNITED AC 2010; 189:573-87. [PMID: 20421423 PMCID: PMC2867304 DOI: 10.1083/jcb.200911047] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
β-Arrestin functions as a scaffold for CaMKII and the Rap guanine nucleotide exchange factor Epac to regulate signaling from β1-ARs. Ca2+/calmodulin kinase II (CaMKII) plays an important role in cardiac contractility and the development of heart failure. Although stimulation of β1–adrenergic receptors (ARs) leads to an increase in CaMKII activity, the molecular mechanism by which β1-ARs activate CaMKII is not completely understood. In this study, we show the requirement for the β1-AR regulatory protein β-arrestin as a scaffold for both CaMKII and Epac (exchange protein directly activated by cAMP). Stimulation of β1-ARs induces the formation of a β-arrestin–CaMKII–Epac1 complex, allowing its recruitment to the plasma membrane, whereby interaction with cAMP leads to CaMKII activation. β-Arrestin binding to the carboxyl-terminal tail of β1-ARs promotes a conformational change within β-arrestin that allows CaMKII and Epac to remain in a stable complex with the receptor. The essential role for β-arrestin and identification of the molecular mechanism by which only β1-ARs and not β2-ARs activate CaMKII significantly advances our understanding of this important cellular pathway.
Collapse
Affiliation(s)
- Supachoke Mangmool
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
32
|
Lompré AM, Hajjar RJ, Harding SE, Kranias EG, Lohse MJ, Marks AR. Ca2+ cycling and new therapeutic approaches for heart failure. Circulation 2010; 121:822-30. [PMID: 20124124 DOI: 10.1161/circulationaha.109.890954] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anne-Marie Lompré
- INSERM UMRS956/Université Pierre et Marie Curie, Faculté de Médecine, 91 Boulevard de l'Hôpital, 75013 Paris, France.
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
The cellular response to a reduced oxygen state (or hypoxia) includes de novo alterations in gene expression patterns, many of which are controlled by hypoxia-inducible factor (HIF) transcription factors. HIF signaling is predominantly regulated by the dioxygenase family of prolyl hydroxylases (PHDs), also known as EGL nine homologs (EGLNs). The PHD family in higher eukaryotes, like the HIF alpha family, is composed of multiple members that have some shared biochemical properties yet have unique biological roles. Although HIF members are the major substrates identified to date for the PHD members, a reasonable expectation is that other proteins whose activities are altered by hypoxia may also serve as PHD substrates. Indeed, the beta(2)-adrenergic receptor, a major adrenergic heterotrimeric guanine nucleotide-binding protein-coupled receptor in the heart, has been identified as a substrate for PHD3.
Collapse
Affiliation(s)
- Joseph A Garcia
- Internal Medicine Department, Cardiology Division, VA North Texas Health Care System, Dallas, TX 75216, USA.
| |
Collapse
|
34
|
Li Z, Liu N, Zhang LS, Gong K, Cai Y, Gao W, Liu Z, Liu S, Han Q, Zhang Y. Proteomic profiling reveals comprehensive insights into adrenergic receptor-mediated hypertrophy in neonatal rat cardiomyocytes. Proteomics Clin Appl 2009; 3:1407-21. [PMID: 21136960 DOI: 10.1002/prca.200900029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 07/27/2009] [Accepted: 08/24/2009] [Indexed: 11/11/2022]
Abstract
Myocardial adrenergic receptors (ARs) play important roles in cardiac hypertrophy. However, the detailed molecular mechanism of AR-mediated cardiac hypertrophy remains elusive to date. To gain full insight into how ARs are involved in the regulation of cardiac hypertrophy, protein expression profiling was performed with comparative proteomics approach on neonatal rat cardiomyocytes. Forty-six proteins were identified as differentially expressed in hypertrophic cardiomyocytes induced by AR stimulation. To better understand the biological significance of the obtained proteomic data, we utilized the ingenuity pathway analysis tool to construct biological networks and analyze function and pathways that might associate with AR-mediated cardiac hypertrophy. Pathway analysis strongly suggested that ROS may be involved in the development of AR-mediated cardiac hypertrophy, which was then confirmed by further experimentation. The results showed that a marked increase in ROS production was detected in AR-mediated cardiac hypertrophy and blocking of ROS production significantly inhibited AR-mediated cardiac hypertrophy. We further proved that the ROS production was through NADPH oxidase or the mitochondrial electron transport chain and this ROS accumulation resulted in activation of extracellular signal-regulated kinase 1/2 leading to AR-mediated cardiac hypertrophy. These experimental results support the hypothesis, from the ingenuity pathway analysis, that AR-mediated cardiac hypertrophy is associated with the dysregulation of a complicated oxidative stress-regulatory network. In conclusion, our results provide a basis for understanding the detailed molecular mechanisms of AR-mediated cardiac hypertrophy.
Collapse
Affiliation(s)
- Zijian Li
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, P. R China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Cohn HI, Xi Y, Pesant S, Harris DM, Hyslop T, Falkner B, Eckhart AD. G protein-coupled receptor kinase 2 expression and activity are associated with blood pressure in black Americans. Hypertension 2009; 54:71-6. [PMID: 19487588 DOI: 10.1161/hypertensionaha.108.125955] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypertension occurs with higher prevalence and morbidity in black Americans compared with other groups. Alterations in the signal transduction pathways of 7-transmembrane spanning receptors are found in hypertensive patients. G protein-coupled receptor kinases (GRKs) play an important role in regulating this receptor signaling. The 2 most abundantly expressed GRKs in the cardiovascular system are GRK2 and GRK5, and each has unique substrates. Understanding changes in expression may give us insight into activated receptors in the pathophysiological progression of hypertension. In heart failure and white hypertensives, increased GRK2 expression arises because of neurohormonal stimulation of particular receptors. GRK2 subsequently desensitizes specific receptors, including beta-adrenergic receptors. In blood pressure control, beta-adrenergic receptor desensitization could lead to increased blood pressure. GRK2 and GRK5 mRNA were evaluated in lymphocytes of black Americans via quantitative real-time PCR. GRK2 mRNA expression directly correlated with systolic blood pressure and norepinephrine levels. GRK2 was elevated >30% among those with systolic blood pressure > or =130 mm Hg. No significant correlation between GRK5 mRNA expression and blood pressure or catecholamines was observed. Diabetic status, age, sex, and body mass index were also compared with GRK2 expression using univariate and multivariate analyses. GRK2 protein expression was elevated 2-fold in subjects with higher blood pressure, and GRK activity was increased >40%. Our data suggest that GRK2, but not GRK5, is correlated with increasing blood pressure in black Americans. Understanding the receptors stimulated by increased neurohormonal activation may give insight into the pathophysiology of hypertension in this at-risk population.
Collapse
Affiliation(s)
- Heather I Cohn
- Center for Translational Medicine, Thomas Jefferson Hospital, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Uncoupling of myocardial beta-adrenergic receptor signaling during coronary artery bypass grafting: the role of GRK2. Ann Thorac Surg 2008; 86:1189-94. [PMID: 18805158 DOI: 10.1016/j.athoracsur.2008.05.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 05/08/2008] [Accepted: 05/15/2008] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cardiopulmonary bypass (CPB) and cardioplegic arrest during cardiac surgery leads to desensitization of myocardial beta-adrenergic receptors (beta-ARs). Impaired signaling through this pathway can have a detrimental effect on ventricular function and increased need for inotropic support. The mechanism of myocardial beta-AR desensitization during cardiac surgery has not been defined. This study investigates the role of G protein-coupled receptor kinase-2 (GRK2), a serine-threonine kinase which phosphorylates and desensitizes agonist-occupied beta-ARs, as a primary mechanism of beta-AR uncoupling during coronary artery bypass grafting (CABG) with CPB and cardioplegic arrest. METHODS Forty-eight patients undergoing elective CABG were enrolled in this study. Myocardial beta-AR signaling was assessed by measuring total beta-AR density and adenylyl cyclase activity in right atrial biopsies obtained before CPB and just before weaning from CPB. Myocardial GRK2 expression and activity were also measured before CPB and just before weaning from CPB. RESULTS Myocardial beta-AR signaling was significantly impaired after CPB and cardioplegic arrest during CABG. Cardiac GRK2 expression was not altered; however, there was a twofold increase in GRK2 activity during CABG. There was an even greater elevation in cardiac GRK2 activity in patients with severely depressed ventricular function. CONCLUSIONS Increased myocardial GRK2 activity appears to be the primary mechanism of impaired beta-AR signaling during CABG with CPB and cardioplegic arrest. This may contribute to the greater need for inotropic support in patients with severe ventricular dysfunction. Strategies to inhibit activation of GRK2 during CABG may decrease morbidity in this patient population.
Collapse
|
37
|
Dobson JG, Shea LG, Fenton RA. Adenosine A2A and beta-adrenergic calcium transient and contractile responses in rat ventricular myocytes. Am J Physiol Heart Circ Physiol 2008; 295:H2364-72. [PMID: 18849328 DOI: 10.1152/ajpheart.00927.2008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The adenosine A2A receptor (A2AR) enhances cardiac contractility, and the adenosine A1R receptor (A1R) is antiadrenergic by reducing the adrenergic beta1 receptor (beta1R)-elicited increase in contractility. In this study we compared the A2AR-, A1R-, and beta1R-elicited actions on isolated rat ventricular myocytes in terms of Ca transient and contractile responses involving PKA and PKC. Stimulation of A2AR with 2 microM (approximately EC50) CGS-21680 (CGS) produced a 17-28% increase in the Ca transient ratio (CTR) and maximum velocities (Vmax) of transient ratio increase (+MVT) and recovery (-MVT) but no change in the time-to-50% recovery (TTR). CGS increased myocyte sarcomere shortening (MSS) and the maximum velocities of shortening (+MVS) and relaxation (-MVS) by 31-34% with no change in time-to-50% relengthening (TTL). beta1R stimulation using 2 nM (approximately EC50) isoproterenol (Iso) increased CTR, +MVT, and -MVT by 67-162% and decreased TTR by 43%. Iso increased MSS, +MVS, and -MVS by 153-174% and decreased TTL by 31%. The A2AR and beta1R Ca transient and contractile responses were not additive. The PKA inhibitor Rp-adenosine 3',5'-cyclic monophosphorothioate triethylamonium salt prevented both the CGS- and Iso-elicited contractile responses. The PKC inhibitors chelerythrine and KIE1-1 peptide (PKCepsilon specific) prevented the antiadrenergic action of A1R but did not influence A2AR-mediated increases in contractile variables. The findings suggest that cardiac A2AR utilize cAMP/PKA like beta1R, but the Ca transient and contractile responses are less in magnitude and not equally affected. Although PKC is important in the A1R antiadrenergic action, it does not seem to play a role in A2AR-elicited Ca transient and contractile events.
Collapse
Affiliation(s)
- James G Dobson
- Department of Physiology, University of Massachusetts Medical School, 55 Lake Ave., North, Worcester, MA 01655, USA.
| | | | | |
Collapse
|
38
|
Graham S, Hammond-Jones D, Gamie Z, Polyzois I, Tsiridis E, Tsiridis E. The effect of β-blockers on bone metabolism as potential drugs under investigation for osteoporosis and fracture healing. Expert Opin Investig Drugs 2008; 17:1281-99. [DOI: 10.1517/13543784.17.9.1281] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Acute beta-blockade prevents myocardial beta-adrenergic receptor desensitization and preserves early ventricular function after brain death. J Thorac Cardiovasc Surg 2008; 135:792-8. [PMID: 18374758 DOI: 10.1016/j.jtcvs.2007.09.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Accepted: 09/06/2007] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Beta-adrenergic receptor desensitization through activation of the G protein-coupled receptor kinase 2 is an important mechanism of early cardiac dysfunction after brain death. We hypothesized that acute beta-blockade can prevent myocardial beta-adrenergic receptor desensitization after brain death through attenuation of G protein-coupled receptor kinase 2 activity, resulting in improved cardiac function. METHODS Adult pigs underwent either sham operation, induction of brain death, or treatment with esmolol (beta-blockade) for 30 minutes before and 45 minutes after brain death (n = 8 per group). Cardiac function was assessed at baseline and for 6 hours after the operation. Myocardial beta-adrenergic receptor signaling was assessed 6 hours after operation by measuring sarcolemmal membrane adenylate cyclase activity, beta-adrenergic receptor density, and G protein-coupled receptor kinase 2 expression and activity. RESULTS Baseline left ventricular preload recruitable stroke work was similar among sham, brain death, and beta-blockade groups. Preload recruitable stroke work was significantly decreased 6 hours after brain death versus sham, and beta-blockade resulted in maintenance of baseline preload recruitable stroke work relative to brain death and not different from sham. Basal and isoproterenol-stimulated adenylate cyclase activities were preserved in the beta-blockade group relative to the brain death group and were not different from the sham group. Left ventricular G protein-coupled receptor kinase 2 expression and activity in the beta-blockade group were markedly decreased relative to the brain death group and similar to the sham group. Beta-adrenergic receptor density was not different among groups. CONCLUSION Acute beta-blockade before brain death attenuates beta-adrenergic receptor desensitization mediated by G protein-coupled receptor kinase 2 and preserves early cardiac function after brain death. These data support the hypothesis that acute beta-adrenergic receptor desensitization is an important mechanism in early ventricular dysfunction after brain death. Future studies with beta-blocker therapy immediately after brain death appear warranted.
Collapse
|
40
|
Patil PN, Li C, Kumari V, Hieble JP. Analysis of efficacy of chiral adrenergic agonists. Chirality 2008; 20:529-43. [DOI: 10.1002/chir.20506] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
41
|
Poller W, Suckau L, Pinkert S, Fechner H. RNA Interference and MicroRNA Modulation for the Treatment of Cardiac Disorders. RNA TECHNOLOGIES IN CARDIOVASCULAR MEDICINE AND RESEARCH 2008. [PMCID: PMC7121055 DOI: 10.1007/978-3-540-78709-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The current status and challenges of RNA interference (RNAi) and microRNA modulation strategies for the treatment of myocardial disorders are discussed and related to the classical gene therapeutic approaches of the past decade. Section 2 summarizes the key issues of current vector technologies which determine if they may be suitable for clinical translation of experimental RNAi or microRNA therapeutic protocols. We then present and discuss examples dealing with the potential of cardiac RNAi therapy. First, an approach to block a key early step in the pathogenesis of a virus-induced cardiomyopathy by RNAi targeting of a cellular receptor for cardiopathogenic viruses (Section 3). Second, an approach to improve cardiac function by RNAi targeting of late pathway of heart failure pathogenesis common to myocardial disorders of multiple etiologies. This strategy is directed at myocardial Ca2+ homeostasis which is disturbed in heart failure due to coronary heart disease, heart valve dysfunction, cardiac inflammation, or genetic defects (Section 4). Whereas the first type of strategies (directed at early pathogenesis) need to be tailor-made for each different type of pathomechanism, the second type (targeting late common pathways) has a much broader range of application. This advantage of the second type of approaches is of key importance since enormous efforts need to be undertaken before any regulatory RNA therapy enters the stage of possible clinical translation. If then the number of patients eligible for this protocol is large, the actual transformation of the experimental therapy into a new therapeutic option of clinical importance is far more likely to occur.
Collapse
|
42
|
Abstract
Protein transport between intracellular organelles is coordinated by Rab GTPases. As an initial approach to defining the function of Rab GTPases in cardiomyocytes, our laboratory focused on Rab1, which regulates protein transport specifically from the endoplasmic reticulum (ER) to the Golgi apparatus. Our studies have demonstrated that adenovirus-driven expression of Rab1 promotes cell growth of primary cultures of neonatal cardiomyocytes in vitro and that transgenic expression of Rab1 in the myocardium induces cardiac hypertrophy in mouse hearts in vivo. These data provide strong evidence implicating that ER-to-Golgi protein transport functions as a regulatory site for control of cardiomyocyte growth. Here we describe a sets of methods used in our laboratory to characterize the function of Rab1 GTPase in modulating cardiac myocyte growth.
Collapse
Affiliation(s)
- Catalin M Filipeanu
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | | | | |
Collapse
|
43
|
Noma T, Lemaire A, Naga Prasad SV, Barki-Harrington L, Tilley DG, Chen J, Le Corvoisier P, Violin JD, Wei H, Lefkowitz RJ, Rockman HA. Beta-arrestin-mediated beta1-adrenergic receptor transactivation of the EGFR confers cardioprotection. J Clin Invest 2007; 117:2445-58. [PMID: 17786238 PMCID: PMC1952636 DOI: 10.1172/jci31901] [Citation(s) in RCA: 353] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 06/12/2007] [Indexed: 12/16/2022] Open
Abstract
Deleterious effects on the heart from chronic stimulation of beta-adrenergic receptors (betaARs), members of the 7 transmembrane receptor family, have classically been shown to result from Gs-dependent adenylyl cyclase activation. Here, we identify a new signaling mechanism using both in vitro and in vivo systems whereby beta-arrestins mediate beta1AR signaling to the EGFR. This beta-arrestin-dependent transactivation of the EGFR, which is independent of G protein activation, requires the G protein-coupled receptor kinases 5 and 6. In mice undergoing chronic sympathetic stimulation, this novel signaling pathway is shown to promote activation of cardioprotective pathways that counteract the effects of catecholamine toxicity. These findings suggest that drugs that act as classical antagonists for G protein signaling, but also stimulate signaling via beta-arrestin-mediated cytoprotective pathways, would represent a novel class of agents that could be developed for multiple members of the 7 transmembrane receptor family.
Collapse
Affiliation(s)
- Takahisa Noma
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Anthony Lemaire
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Sathyamangla V. Naga Prasad
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Liza Barki-Harrington
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Douglas G. Tilley
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Juhsien Chen
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Philippe Le Corvoisier
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Jonathan D. Violin
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Huijun Wei
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert J. Lefkowitz
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Howard A. Rockman
- Department of Medicine,
Howard Hughes Medical Institute,
Department of Cell Biology, and
Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
44
|
Abstract
Despite advances in treatment, atrial fibrillation (AF) remains the most common arrhythmia in humans. Antiarrhythmic drug therapy continues to be a cornerstone of AF treatment, even in light of emerging non-pharmacologic therapies. Conventional antiarrhythmic drugs target cardiac ion channels and are often associated with modest AF suppression and the risk of ventricular proarrhythmia. Ongoing drug development has focused on targeting atrial-specific ion channels as well as novel non-ionic targets. Targeting non-ionic mechanisms may also provide new drugs directed towards the underlying mechanisms responsible for AF and possibly greater antiarrhythmic potency. Agents that act against these new targets may offer improved safety and efficacy in AF treatment.
Collapse
Affiliation(s)
- Deepak Bhakta
- Indiana University School of Medicine, Krannert Institute of Cardiology, 1800 N. Capitol Avenue, Indianapolis, IN 46202, USA.
| | | |
Collapse
|
45
|
Quaile MP, Rossman EI, Berretta RM, Bratinov G, Kubo H, Houser SR, Margulies KB. Reduced sarcoplasmic reticulum Ca(2+) load mediates impaired contractile reserve in right ventricular pressure overload. J Mol Cell Cardiol 2007; 43:552-63. [PMID: 17931654 DOI: 10.1016/j.yjmcc.2007.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 08/12/2007] [Accepted: 08/16/2007] [Indexed: 11/25/2022]
Abstract
Myocardial contractile reserve is significantly attenuated in patients with advanced heart failure. The aim of this study was to identify mechanisms of impaired contractile reserve in a large animal model that closely mimics human myocardial failure. Progressive right ventricular hypertrophy and failure were induced by banding the pulmonary artery in kittens. Isometric contractile force was measured in right ventricular trabeculae (n=115) from age-matched Control and Banded feline hearts. Rapid cooling contractures (RCC) were used to determine sarcoplasmic reticulum (SR) Ca(2+) load while assessing the ability of changes in rate, adrenergic stimulation and bath Ca(2+) to augment contractility. The positive force-frequency relationship and robust pre- and post-receptor adrenergic responses observed in Control trabeculae were closely paralleled by increases in RCC amplitude and the RCC2/RCC1 ratio. Conversely, the severely blunted force-frequency and adrenergic responses in Banded trabeculae were paralleled by an unchanged RCC amplitude and RCC2/RCC1 ratio. Likewise, supraphysiologic levels of bath Ca(2+) were associated with severely reduced contractility and RCC amplitude in Banded trabeculae compared to Controls. There were no differences in myofilament Ca(2+) sensitivity or length-dependent increases in contractility between Control and Banded trabeculae. There was a significant decrease in SR Ca(2+)-ATPase pump abundance and phosphorylation of phospholamban and ryanodine receptor in Banded trabeculae compared with Controls. A reduced ability to increase SR Ca(2+) load is the primary mechanism of reduced contractile reserve in failing feline myocardium. The similarity of impaired contractile reserve phenomenology in this feline model and transplanted hearts suggests mechanistic relevance to human myocardial failure.
Collapse
Affiliation(s)
- Michael P Quaile
- Department of Physiology and the Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Cardiac hypertrophy and heart failure are major causes of morbidity and mortality in Western societies. Many factors have been implicated in cardiac remodeling, including alterations in gene expression in myocytes, cardiomyocytes apoptosis, cytokines and growth factors that influence cardiac dynamics, and deficits in energy metabolism as well as alterations in cardiac extracellular matrix composition. Many therapeutic means have been shown to prevent or reverse cardiac hypertrophy. New concepts for characterizing the pathophysiology of cardiac hypertrophy have been drawn from various aspects, including medical therapy and gene therapy, or use of stem cells for tissue regeneration. In this review, we focus on various types of cardiac hypertrophy, defining the causes of hypertrophy, describing available animal models of hypertrophy, discussing the mechanisms for development of hypertrophy and its transition to heart failure, and presenting the potential use of novel promising therapeutic strategies derived from new advances in basic scientific research.
Collapse
Affiliation(s)
- Sudhiranjan Gupta
- Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | |
Collapse
|
47
|
Oikawa R, Nasa Y, Ishii R, Kuwaki T, Tanoue A, Tsujimoto G, Takeo S. Vasopressin V1A receptor enhances baroreflex via the central component of the reflex arc. Eur J Pharmacol 2007; 558:144-50. [PMID: 17224142 DOI: 10.1016/j.ejphar.2006.11.063] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 11/24/2006] [Accepted: 11/29/2006] [Indexed: 11/19/2022]
Abstract
The neurohypophyseal peptide [Arg(8)]-vasopressin (AVP) exerts its physiological actions via 3 distinct receptor isoforms designated V1A, V1B, and V2. We recently showed that V1A receptor was involved in the baroreflex control of heart rate using V1A receptor knockout mice. The present study was undertaken to further clarify this finding. In conscious mice, resting blood pressure of the knockout group was lower than that of the wild-type group (wild-type, 108+/-2.0 mm Hg; knockout, 98+/-3.8 mm Hg; n=6-7) without notable change in heart rate. Although phenylephrine and nitroprusside-induced changes in blood pressure did not differ in these strains, the subsequent bradycardia and tachycardia were markedly blunted in the knockout mice (mean slopes for baroreflex curve after phenylephrine treatment; wild-type, -5.65+/-0.30 bpm/mm Hg; knockout, -3.97+/-0.52 bpm/mm Hg; those after nitroprusside treatment; wild-type, -0.51+/-0.10 bpm/mm Hg; knockout, -0.18+/-0.05 bpm/mm Hg; n=6-7). Under urethane anesthesia (1.0-1.2 g/kg, i.p.), electrical stimulation of the vagal afferent nerve evoked frequency-dependent hypotension and bradycardia in the wild-type mice. In contrast, in the knockout mice such stimulation induced a pressor, not a depressor, response and diminished bradycardia. Moreover, electrical stimulation-induced hemodynamic changes through the vagal afferent nerve in the wild-type mice were significantly attenuated by pretreatment with intravenously administered V1A receptor antagonist d(CH(2))(5)Tyr(Me)AVP. Electrical stimulation of the vagal efferent nerve-induced hemodynamic changes (depressor and bradycardia) and chronotropic responses to adrenergic and cholinergic stimuli were not different between the 2 strains. These results suggest that the V1A receptor in the central nervous system is involved in the regulation of the heart rate via the baroreflex arc.
Collapse
Affiliation(s)
- Ryo Oikawa
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Science, 1432-1, Horinouchi, Hachioji, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Koshimizu TA, Tanoue A, Tsujimoto G. Clinical implications from studies of alpha1 adrenergic receptor knockout mice. Biochem Pharmacol 2006; 73:1107-12. [PMID: 17141736 DOI: 10.1016/j.bcp.2006.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 10/27/2006] [Accepted: 11/02/2006] [Indexed: 11/18/2022]
Abstract
alpha1-Adrenergic receptors (alpha1-ARs) modulate a large number of physiological functions in cardiovascular and noncardiovascular tissues. Because individual members of the alpha1-AR family (alpha1A-, alpha1B-, and alpha1D-ARs) have overlapping expression profiles in most tissues, elucidation of the precise physiological roles of individual alpha1-AR subtypes remains a challenging task. To alleviate this constraint, a gene targeting approach has been employed to generate mutant mice lacking one or two alpha1-AR genes. Recent studies on these mutant mouse strains are discussed in this article, with an emphasis on the role of alpha1-AR in the central nervous system and lower urinary tracts. These are two major tissues of particular interest for the development of new therapeutic strategies targeted to the alpha1-ARs. By combining gene targeting techniques with pharmacological tools, the specific roles of alpha1-AR subtypes could be delineated.
Collapse
Affiliation(s)
- Taka-aki Koshimizu
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | |
Collapse
|
49
|
Hansen JL, Theilade J, Aplin M, Sheikh SP. Role of G-protein-coupled receptor kinase 2 in the heart--do regulatory mechanisms open novel therapeutic perspectives? Trends Cardiovasc Med 2006; 16:169-77. [PMID: 16781951 DOI: 10.1016/j.tcm.2006.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Revised: 03/03/2006] [Accepted: 03/09/2006] [Indexed: 01/08/2023]
Abstract
G-protein-coupled receptor kinase (GRK) 2 regulates a plethora of cellular processes, including cardiac expression and function of key seven-transmembrane receptors (7TM receptors) such as the beta-adrenergic and angiotensin receptors (Penela P, Murga C, Ribas C, et al.: 2006. Mechanisms of regulation of G-protein-coupled receptor kinases [GRKs] and cardiovascular disease. Cardiovasc Res 69:46-56, Rockman HA, Koch WJ, Lefkowitz RJ: 2002. Seven-transmembrane-spanning receptors and heart function. Nature 415:206-212). Interestingly, these two G-protein-coupled receptor systems are targeted by modern heart failure treatment including beta-adrenergic blockers, angiotensin-converting enzyme inhibitors, and angiotensin receptor blockers. Although GRK2 is ubiquitously expressed, its particular importance in the heart has been demonstrated by interesting phenotypes of genetically altered mice that suggest GRK2 inhibition can ameliorate heart failure. In essence, this work suggests GRK2 could be an endogenous receptor blocker targeting both the beta-adrenergic and angiotensin receptors in the heart. This notion immediately suggests it is important to understand the molecular mechanisms that regulate GRK2 activity in the heart. In this review, we provide a detailed presentation of the tight regulation of GRK2 expression levels and protein activity, and we discuss the cardiovascular GRK2 functions and possible therapeutic perspectives.
Collapse
Affiliation(s)
- Jakob Lerche Hansen
- Laboratory of Molecular and Cellular Cardiology, H:S Rigshospitalet 9312, Department of Medicine B, University of Copenhagen, Juliane Mariesvej 20, DK-2100 Copenhagen, Denmark
| | | | | | | |
Collapse
|
50
|
Mills GD, Kubo H, Harris DM, Berretta RM, Piacentino V, Houser SR. Phosphorylation of phospholamban at threonine-17 reduces cardiac adrenergic contractile responsiveness in chronic pressure overload-induced hypertrophy. Am J Physiol Heart Circ Physiol 2006; 291:H61-70. [PMID: 16772527 DOI: 10.1152/ajpheart.01353.2005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Physiological hemodynamic stress, such as aerobic exercise, is intermittent and requires an increase in Ca2+ -dependent contractility through sympathetic nervous system activation. Pathological hemodynamic stress, such as hypertension, is persistent and requires sustained increases in cardiac function. Over time, this causes left ventricular hypertrophy (LVH)-reduced responsiveness to sympathetic stimulation. In this study, we examined the hypothesis that blunted in vivo adrenergic contractile responsiveness in pressure overload (PO)-induced cardiac hypertrophy is caused by abnormalities in the abundance and/or basal phosphorylation state of Ca2+ regulatory proteins. PO, induced by aortic constriction, caused concentric LVH or dilated LVH. Only animals with dilation exhibited a decrease in baseline left ventricle function [fractional area change (FAC); measured with echocardiography]. All PO animals had a reduced contractile response to adrenergic agonists (increase in FAC with 40 microg.kg(-1).min(-1) dobutamine, control 0.30 +/- 0.04, n = 5 vs. banded 0.10 +/- 0.03, n = 10; P < 0.01). PO animals had reduced phospholamban (PLB) protein abundance (P = 0.07, not significant) and increased PLB phosphorylation at the calmodulin-dependent kinase II (CaMKII)-specific site (PLB-Thr17, P < 0.05) but not at the protein kinase A-specific site (PLB-Ser16). PLB-Thr17 phosphorylation was inversely correlated with dobutamine-induced increases in contractility in PO animals (r2 = 0.81, P < 0.05). Continuous induction of Ca2+ transients in isolated ventricular myocytes for 24 h increased phosphorylation at PLB-Thr17 and diminished inotropic responsiveness and PLB-Ser16 phosphorylation after exposure to isoproterenol (P < 0.05). These data show that reduced adrenergic responsiveness in feline PO hypertrophy and failure involves increases in basal PLB-Thr17 phosphorylation, suggesting that activation of CaMKII in PO hypertrophy contributes to defective adrenergic reserve in compensated LVH and early heart failure.
Collapse
Affiliation(s)
- Geoffrey D Mills
- Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|