Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 169(3): 407-421.e16, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431242

ABSTRACT

The phosphorylation of agonist-occupied G-protein-coupled receptors (GPCRs) by GPCR kinases (GRKs) functions to turn off G-protein signaling and turn on arrestin-mediated signaling. While a structural understanding of GPCR/G-protein and GPCR/arrestin complexes has emerged in recent years, the molecular architecture of a GPCR/GRK complex remains poorly defined. We used a comprehensive integrated approach of cross-linking, hydrogen-deuterium exchange mass spectrometry (MS), electron microscopy, mutagenesis, molecular dynamics simulations, and computational docking to analyze GRK5 interaction with the ß2-adrenergic receptor (ß2AR). These studies revealed a dynamic mechanism of complex formation that involves large conformational changes in the GRK5 RH/catalytic domain interface upon receptor binding. These changes facilitate contacts between intracellular loops 2 and 3 and the C terminus of the ß2AR with the GRK5 RH bundle subdomain, membrane-binding surface, and kinase catalytic cleft, respectively. These studies significantly contribute to our understanding of the mechanism by which GRKs regulate the function of activated GPCRs. PAPERCLIP.


Subject(s)
G-Protein-Coupled Receptor Kinase 5/chemistry , Mammals/metabolism , Receptors, Adrenergic, beta-2/chemistry , Animals , Camelids, New World , Cattle , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Humans , Mass Spectrometry , Microscopy, Electron , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Rats , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism
2.
Mol Cell ; 81(2): 323-339.e11, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33321095

ABSTRACT

The phosphorylation of G protein-coupled receptors (GPCRs) by GPCR kinases (GRKs) facilitates arrestin binding and receptor desensitization. Although this process can be regulated by Ca2+-binding proteins such as calmodulin (CaM) and recoverin, the molecular mechanisms are poorly understood. Here, we report structural, computational, and biochemical analysis of a CaM complex with GRK5, revealing how CaM shapes GRK5 response to calcium. The CaM N and C domains bind independently to two helical regions at the GRK5 N and C termini to inhibit GPCR phosphorylation, though only the C domain interaction disrupts GRK5 membrane association, thereby facilitating cytoplasmic translocation. The CaM N domain strongly activates GRK5 via ordering of the amphipathic αN-helix of GRK5 and allosteric disruption of kinase-RH domain interaction for phosphorylation of cytoplasmic GRK5 substrates. These results provide a framework for understanding how two functional effects, GRK5 activation and localization, can cooperate under control of CaM for selective substrate targeting by GRK5.


Subject(s)
Calcium/metabolism , Calmodulin/chemistry , G-Protein-Coupled Receptor Kinase 5/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Calmodulin/genetics , Calmodulin/metabolism , Cloning, Molecular , Crystallography, X-Ray , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Kinetics , Molecular Dynamics Simulation , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera , Substrate Specificity , Thermodynamics
3.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article in English | MEDLINE | ID: mdl-33500351

ABSTRACT

Pathological remodeling of the heart is a hallmark of chronic heart failure (HF) and these structural changes further perpetuate the disease. Cardiac fibroblasts are the critical cell type that is responsible for maintaining the structural integrity of the heart. Stress conditions, such as a myocardial infarction (MI), can activate quiescent fibroblasts into synthetic and contractile myofibroblasts. G protein-coupled receptor kinase 5 (GRK5) is an important mediator of cardiovascular homeostasis through dampening of GPCR signaling, and is expressed in the heart and up-regulated in human HF. Of note, GRK5 has been demonstrated to translocate to the nucleus in cardiomyocytes in a calcium-calmodulin (Ca2+-CAM)-dependent manner, promoting hypertrophic gene transcription through activation of nuclear factor of activated T cells (NFAT). Interestingly, NFAT is also involved in fibroblast activation. GRK5 is highly expressed and active in cardiac fibroblasts; however, its pathophysiological role in these crucial cardiac cells is unknown. We demonstrate using adult cardiac fibroblasts that genetic deletion of GRK5 inhibits angiotensin II (AngII)-mediated fibroblast activation. Fibroblast-specific deletion of GRK5 in mice led to decreased fibrosis and cardiac hypertrophy after chronic AngII infusion or after ischemic injury compared to nontransgenic littermate controls (NLCs). Mechanistically, we show that nuclear translocation of GRK5 is involved in fibroblast activation. These data demonstrate that GRK5 is a regulator of fibroblast activation in vitro and cardiac fibrosis in vivo. This adds to previously published data which demonstrate the potential beneficial effects of GRK5 inhibition in the context of cardiac disease.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , G-Protein-Coupled Receptor Kinase 5/metabolism , Myocardium/pathology , Angiotensin II , Animals , Animals, Newborn , Cardiomegaly/complications , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Transdifferentiation , Fibrosis , Mice, Knockout , Models, Biological , Myocardial Ischemia/complications , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myofibroblasts/pathology , Rats
4.
Biochem Soc Trans ; 51(2): 715-724, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37013982

ABSTRACT

G protein-coupled receptors (GPCRs) are key modulators of cell signaling. Multiple GPCRs are present in the heart where they regulate cardiac homeostasis including processes such as myocyte contraction, heart rate and coronary blood flow. GPCRs are pharmacological targets for several cardiovascular disorders including heart failure (HF) such as beta-adrenergic receptor (ßAR) blockers and angiotensin II receptor (AT1R) antagonists. The activity of GPCRs are finely regulated by GPCR kinases (GRKs), which phosphorylate agonist-occupied receptors and start the process of desensitization. Among the seven members of the GRK family, GRK2 and GRK5 are predominantly expressed in the heart, where they exhibit both canonical and non-canonical functions. Both kinases are known to be increased in cardiac pathologies and contribute to pathogenesis through their roles in different cellular compartments. Lowering or inhibiting their actions mediate cardioprotective effects against pathological cardiac growth and failing heart. Therefore, given their importance in cardiac dysfunction, these kinases are drawing attention as promising targets for the treatment of HF, which needs improved therapies. Over the past three decades, broad knowledge on GRK inhibition in HF has been gained by studies using genetically engineered animal models or through gene therapy with peptide inhibitors or using small molecule inhibitors. In this mini review, we summarize the work focusing on GRK2 and GRK5 but also discuss a couple of the non-abundant cardiac subtypes and their multi-functional roles in the normal and diseased heart and the potential and therapeutic targets.


Subject(s)
G-Protein-Coupled Receptor Kinases , Heart Failure , Animals , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , G-Protein-Coupled Receptor Kinases/therapeutic use , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Signal Transduction , Receptors, G-Protein-Coupled
5.
Physiol Rev ; 95(2): 377-404, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25834229

ABSTRACT

G protein-coupled receptors (GPCRs) are important regulators of various cellular functions via activation of intracellular signaling events. Active GPCR signaling is shut down by GPCR kinases (GRKs) and subsequent ß-arrestin-mediated mechanisms including phosphorylation, internalization, and either receptor degradation or resensitization. The seven-member GRK family varies in their structural composition, cellular localization, function, and mechanism of action (see sect. II). Here, we focus our attention on GRKs in particular canonical and novel roles of the GRKs found in the cardiovascular system (see sects. III and IV). Paramount to overall cardiac function is GPCR-mediated signaling provided by the adrenergic system. Overstimulation of the adrenergic system has been highly implicated in various etiologies of cardiovascular disease including hypertension and heart failure. GRKs acting downstream of heightened adrenergic signaling appear to be key players in cardiac homeostasis and disease progression, and herein we review the current data on GRKs related to cardiac disease and discuss their potential in the development of novel therapeutic strategies in cardiac diseases including heart failure.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Heart Diseases/enzymology , Myocardium/enzymology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Diseases/physiopathology , Humans
6.
Circ Res ; 127(6): 796-810, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32507058

ABSTRACT

RATIONALE: Cardiotoxic ß1 adrenergic receptor (ß1AR)-CaMKII (calmodulin-dependent kinase II) signaling is a major and critical feature associated with development of heart failure. SAP97 (synapse-associated protein 97) is a multifunctional scaffold protein that binds directly to the C-terminus of ß1AR and organizes a receptor signalosome. OBJECTIVE: We aim to elucidate the dynamics of ß1AR-SAP97 signalosome and its potential role in chronic cardiotoxic ß1AR-CaMKII signaling that contributes to development of heart failure. METHODS AND RESULTS: The integrity of cardiac ß1AR-SAP97 complex was examined in heart failure. Cardiac-specific deletion of SAP97 was developed to examine ß1AR signaling in aging mice, after chronic adrenergic stimulation, and in pressure overload hypertrophic heart failure. We show that the ß1AR-SAP97 signaling complex is reduced in heart failure. Cardiac-specific deletion of SAP97 yields an aging-dependent cardiomyopathy and exacerbates cardiac dysfunction induced by chronic adrenergic stimulation and pressure overload, which are associated with elevated CaMKII activity. Loss of SAP97 promotes PKA (protein kinase A)-dependent association of ß1AR with arrestin2 and CaMKII and turns on an Epac (exchange protein directly activated by cAMP)-dependent activation of CaMKII, which drives detrimental functional and structural remodeling in myocardium. Moreover, we have identified that GRK5 (G-protein receptor kinase-5) is necessary to promote agonist-induced dissociation of SAP97 from ß1AR. Cardiac deletion of GRK5 prevents adrenergic-induced dissociation of ß1AR-SAP97 complex and increases in CaMKII activity in hearts. CONCLUSIONS: These data reveal a critical role of SAP97 in maintaining the integrity of cardiac ß1AR signaling and a detrimental cardiac GRK5-CaMKII axis that can be potentially targeted in heart failure therapy. Graphical Abstract: A graphical abstract is available for this article.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Discs Large Homolog 1 Protein/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Failure/enzymology , Myocytes, Cardiac/enzymology , Receptors, Adrenergic, beta-1/metabolism , Animals , Apoptosis , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Discs Large Homolog 1 Protein/genetics , Disease Models, Animal , Excitation Contraction Coupling , G-Protein-Coupled Receptor Kinase 5/genetics , Guanine Nucleotide Exchange Factors/metabolism , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/pathology , beta-Arrestin 1/metabolism
7.
Proc Natl Acad Sci U S A ; 116(32): 15895-15900, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31337679

ABSTRACT

G protein-coupled receptor (GPCR) kinases (GRKs) are responsible for initiating desensitization of activated GPCRs. GRK5 is potently inhibited by the calcium-sensing protein calmodulin (CaM), which leads to nuclear translocation of GRK5 and promotion of cardiac hypertrophy. Herein, we report the architecture of the Ca2+·CaM-GRK5 complex determined by small-angle X-ray scattering and negative-stain electron microscopy. Ca2+·CaM binds primarily to the small lobe of the kinase domain of GRK5 near elements critical for receptor interaction and membrane association, thereby inhibiting receptor phosphorylation while activating the kinase for phosphorylation of soluble substrates. To define the role of each lobe of Ca2+·CaM, we utilized the natural product malbrancheamide as a chemical probe to show that the C-terminal lobe of Ca2+·CaM regulates membrane binding while the N-terminal lobe regulates receptor phosphorylation and kinase domain activation. In cells, malbrancheamide attenuated GRK5 nuclear translocation and effectively blocked the hypertrophic response, demonstrating the utility of this natural product and its derivatives in probing Ca2+·CaM-dependent hypertrophy.


Subject(s)
Biological Products/chemistry , Calmodulin/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Calcium/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Enzyme Activation/drug effects , G-Protein-Coupled Receptor Kinase 5/chemistry , Hypertrophy , Indole Alkaloids/chemistry , Indole Alkaloids/pharmacology , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation/drug effects , Protein Domains , Protein Transport/drug effects , Substrate Specificity/drug effects
8.
Biochem Cell Biol ; 99(4): 508-518, 2021 08.
Article in English | MEDLINE | ID: mdl-33507833

ABSTRACT

G protein coupled receptor kinase 5 (GRK5) is localized within the nucleus and moderates functions such as DNA transcription, in addition to its localization at the plasma membrane. In this report, we show that GRK5 modifies the nucleolar stress response activated by the DNA polymerase inhibitor, actinomycin D (ActD). We show an increased sensitivity to the apoptotic effects of ActD on cervical HeLa cells and the breast cancer cell line MDA MB 231 with reduced protein expression of GRK5. We also tested two types of breast cancer cells (MDA MB 231 and MCF7 cells) and found that the rate of response to ActD varied between them because they have innate differences in the protein expression of GRK5. We also found that GRK5 phosphorylates nucleophosmin (NPM1) at T199 before and during the early stages of ActD treatment. Phosphorylation at T199 increases the ability of NPM1 to interact with p14ARF in vitro, which may affect the protein expression levels of p14ARF. We found that the expression levels of p14ARF were lower in the cells transfected with the control shRNA, but higher in cells transfected with GRK5 shRNA. Collectively, this suggests that GRK5 modifies the nucleolar stress response associated with ActD.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Nucleolus/pathology , Dactinomycin/pharmacology , G-Protein-Coupled Receptor Kinase 5/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Apoptosis , Cell Nucleolus/drug effects , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , G-Protein-Coupled Receptor Kinase 5/genetics , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Nucleophosmin , Phosphorylation , Protein Binding , Tumor Cells, Cultured
9.
Mol Cell Biochem ; 476(3): 1505-1516, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33392923

ABSTRACT

G protein-coupled receptor kinases (GRKs), in addition to their role in modulating signal transduction mechanisms associated with activated G protein-coupled receptors (GPCRs), can also interact with many non-GPCR proteins to mediate cellular responses to chemotherapeutics. The rationale for this study is based on the presumption that GRK2 modulates the responses of cancer cells to the chemotherapeutic cisplatin. In this report, we show that GRK2 modulates the responses of cancer cells to cisplatin. Cervical cancer HeLa cells stably transfected with GRK2 shRNA, to decrease GRK2 protein expression, show increased sensitivity to cisplatin. Of interest, these cells also show increased accumulation of NADPH, associating with decreased NADP buildup, at low concentrations of cisplatin tested. These changes in NADPH and NADP levels are also observed in the breast cancer MDA MB 231 cells, which has lower endogenous GRK2 protein expression levels, but not BT549, a breast cancer cell line with higher GRK2 protein expression. This effect of NADPH accumulation may be associated with a decrease in NADPH oxidase 4 (NOX4) protein expression, which is found to correlate with GRK2 protein expression in cancer cells-a relationship which mimics that observed in cardiomyocytes. Furthermore, like in cardiomyocytes, GRK2 and NOX4 interact to form complexes in cancer cells. Collectively, these results suggest that GRK2 interacts with NOX4 to modify cisplatin sensitivity in cancer cells and may also factor into the success of cisplatin-based regimens.


Subject(s)
Cisplatin/pharmacology , G-Protein-Coupled Receptor Kinase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Profiling , HeLa Cells , Humans , Neoplasms/metabolism , Phosphorylation , Protein Binding , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors
10.
Int J Mol Sci ; 22(4)2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33671974

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors and they are responsible for the transduction of extracellular signals, regulating almost all aspects of mammalian physiology. These receptors are specifically regulated by a family of serine/threonine kinases, called GPCR kinases (GRKs). Given the biological role of GPCRs, it is not surprising that GRKs are also involved in several pathophysiological processes. Particular importance is emerging for GRK5, which is a multifunctional protein, expressed in different cell types, and it has been found located in single or multiple subcellular compartments. For instance, when anchored to the plasma membrane, GRK5 exerts its canonical function, regulating GPCRs. However, under certain conditions (e.g., pro-hypertrophic stimuli), GRK5 translocates to the nucleus of cells where it can interact with non-GPCR-related proteins as well as DNA itself to promote "non-canonical" signaling, including gene transcription. Importantly, due to these actions, several studies have demonstrated that GRK5 has a pivotal role in the pathogenesis of chronic-degenerative disorders. This is true in the cardiac cells, tumor cells, and neurons. For this reason, in this review article, we will inform the readers of the most recent evidence that supports the importance of targeting GRK5 to prevent the development or progression of cancer, cardiovascular, and neurological diseases.


Subject(s)
Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , G-Protein-Coupled Receptor Kinase 5/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 5/metabolism , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Animals , G-Protein-Coupled Receptor Kinase 5/chemistry , Humans , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
11.
Protein Expr Purif ; 168: 105547, 2020 04.
Article in English | MEDLINE | ID: mdl-31786308

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors in humans and regulate numerous physiological processes through the activation of heterotrimeric G proteins. GPCR kinases (GRKs) selectively phosphorylate active GPCRs, which promotes arrestin binding, receptor internalization, and initiation of alternative signaling pathways. GRK5 is a representative member of one of three GRK subfamilies that does not need post-translational lipidation or other binding partners to exhibit full activity against GPCRs, rendering it a useful tool for biophysical studies directed at characterizing GRK function. However, recombinant expression of GRK5 has thus far been limited to insect and mammalian systems. Here, we describe the expression of functional GRK5 in E. coli and its purification and biochemical characterization. Bacterially expressed GRK5 is hyperphosphorylated, primarily in regions known to be flexible from prior crystal structures, which slightly decreases its catalytic activity toward receptor substrates. Mutation of a single phosphorylation site, Thr10, restores kinetic parameters to those of GRK5 purified from insect cells. Consequently, bacterial expression will allow for production of GRK5 at a reduced cost and faster pace and would facilitate production of isotopically labeled kinase for NMR studies or for the incorporation of unnatural amino acids.


Subject(s)
Adenosine Triphosphate/chemistry , G-Protein-Coupled Receptor Kinase 5/chemistry , Protein Processing, Post-Translational , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
12.
Proc Natl Acad Sci U S A ; 114(51): 13561-13566, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203659

ABSTRACT

Opioids are powerful analgesics, but also carry significant side effects and abuse potential. Here we describe a modulator of the µ-opioid receptor (MOR1), the transient receptor potential channel subfamily vanilloid member 1 (TRPV1). We show that TRPV1 binds MOR1 and blocks opioid-dependent phosphorylation of MOR1 while leaving G protein signaling intact. Phosphorylation of MOR1 initiates recruitment and activation of the ß-arrestin pathway, which is responsible for numerous opioid-induced adverse effects, including the development of tolerance and respiratory depression. Phosphorylation stands in contrast to G protein signaling, which is responsible for the analgesic effect of opioids. Calcium influx through TRPV1 causes a calcium/calmodulin-dependent translocation of G protein-coupled receptor kinase 5 (GRK5) away from the plasma membrane, thereby blocking its ability to phosphorylate MOR1. Using TRPV1 to block phosphorylation of MOR1 without affecting G protein signaling is a potential strategy to improve the therapeutic profile of opioids.


Subject(s)
Receptors, Opioid, mu/metabolism , TRPV Cation Channels/metabolism , Cell Membrane/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Transport
13.
Int J Mol Sci ; 21(8)2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32326036

ABSTRACT

Aldosterone (Aldo), when overproduced, is a cardiotoxic hormone underlying heart failure and hypertension. Aldo exerts damaging effects via the mineralocorticoid receptor (MR) but also activates the antiapoptotic G protein-coupled estrogen receptor (GPER) in the heart. G protein-coupled receptor (GPCR)-kinase (GRK)-2 and -5 are the most abundant cardiac GRKs and phosphorylate GPCRs as well as non-GPCR substrates. Herein, we investigated whether they phosphorylate and regulate cardiac MR and GPER. To this end, we used the cardiomyocyte cell line H9c2 and adult rat ventricular myocytes (ARVMs), in which we manipulated GRK5 protein levels via clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and GRK2 activity via pharmacological inhibition. We report that GRK5 phosphorylates and inhibits the cardiac MR whereas GRK2 phosphorylates and desensitizes GPER. In H9c2 cardiomyocytes, GRK5 interacts with and phosphorylates the MR upon ß2-adrenergic receptor (AR) activation. In contrast, GRK2 opposes agonist-activated GPER signaling. Importantly, GRK5-dependent MR phosphorylation of the MR inhibits transcriptional activity, since aldosterone-induced gene transcription is markedly suppressed in GRK5-overexpressing cardiomyocytes. Conversely, GRK5 gene deletion augments cardiac MR transcriptional activity. ß2AR-stimulated GRK5 phosphorylates and inhibits the MR also in ARVMs. Additionally, GRK5 is necessary for the protective effects of the MR antagonist drug eplerenone against Aldo-induced apoptosis and oxidative stress in ARVMs. In conclusion, GRK5 blocks the cardiotoxic MR-dependent effects of Aldo in the heart, whereas GRK2 may hinder beneficial effects of Aldo through GPER. Thus, cardiac GRK5 stimulation (e.g., via ß2AR activation) might be of therapeutic value for heart disease treatment via boosting the efficacy of MR antagonists against Aldo-mediated cardiac injury.


Subject(s)
Aldosterone/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Myocytes, Cardiac/metabolism , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 5/genetics , Models, Biological , Oxidative Stress , Phosphorylation , Protein Binding , Rats , Receptors, Adrenergic, beta-2/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Transcriptional Activation
14.
J Biol Chem ; 293(21): 8056-8064, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29628444

ABSTRACT

PH domain leucine-rich repeat protein phosphatase (PHLPP) is a serine/threonine phosphatase that has been shown to regulate cell growth and survival through dephosphorylation of several members of the AGC family of kinases. G-protein-coupled receptor kinase 5 (GRK5) is an AGC kinase that regulates phenylephrine (PE)-induced cardiac hypertrophy through its noncanonical function of directly targeting proteins to the nucleus to regulate transcription. Here we investigated the possibility that the PHLPP2 isoform can regulate GRK5-induced cardiomyocyte hypertrophy in neonatal rat ventricular myocytes (NRVMs). We show that removal of PHLPP2 by siRNA induces hypertrophic growth of NRVMs as measured by cell size changes at baseline, potentiated PE-induced cell size changes, and re-expression of fetal genes atrial natriuretic factor and brain natriuretic peptide. Endogenous GRK5 and PHLPP2 were found to interact in NRVMs, and PE-induced nuclear accumulation of GRK5 was enhanced upon down-regulation of PHLPP2. Conversely, overexpression of PHLPP2 blocked PE-induced hypertrophic growth, re-expression of fetal genes, and nuclear accumulation of GRK5, which depended on its phosphatase activity. Finally, using siRNA against GRK5, we found that GRK5 was necessary for the hypertrophic response induced by PHLPP2 knockdown. Our findings demonstrate for the first time a novel regulation of GRK5 by the phosphatase PHLPP2, which modulates hypertrophic growth. Understanding the signaling pathways affected by PHLPP2 has potential for new therapeutic targets in the treatment of cardiac hypertrophy and failure.


Subject(s)
Cardiomegaly/pathology , G-Protein-Coupled Receptor Kinase 5/metabolism , Gene Expression Regulation , Myocytes, Cardiac/pathology , Phosphoprotein Phosphatases/metabolism , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiotonic Agents/toxicity , Cells, Cultured , G-Protein-Coupled Receptor Kinase 5/genetics , In Vitro Techniques , Myocytes, Cardiac/metabolism , Phenylephrine/toxicity , Phosphoprotein Phosphatases/genetics , Rats , Rats, Sprague-Dawley
15.
J Cell Physiol ; 234(7): 10411-10420, 2019 07.
Article in English | MEDLINE | ID: mdl-30511419

ABSTRACT

G protein-coupled receptor kinase 5 (GRK5) is a serine/threonine kinase whose dysfunction results in cognitive impairment and Alzheimer-like pathology, including tau hyperphosphorylation. However, the mechanisms whereby GRK5 influences tau phosphorylation remain incompletely understood. In the current study, we showed that GRK5 influenced the phosphorylation of tau via glycogen synthase kinase 3ß (GSK3ß). The activity of both tau and GSK3ß in the hippocampus was increased in aged GRK5-knockout mice, which is consistent with what occurs in APP/PS1 transgenic mice. Furthermore, GRK5 regulated the activity of GSK3ß and phosphorylated tau in vitro. Regardless of changes of GRK5 protein levels, tau hyperphosphorylation remained reduced after GSK3ß activity was inhibited, suggesting that GRK5 may specifically influence tau hyperphosphorylation by modulating GSK3ß activity. Taken together, our findings suggest that GRK5 deficiency contributes to the pathogenesis of Alzheimer's disease by influencing the hyperphosphorylation of tau through the activation of GSK3ß.


Subject(s)
Alzheimer Disease/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Phosphorylation/physiology , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cognitive Dysfunction/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/physiology
16.
Biochem Biophys Res Commun ; 520(2): 327-332, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31604529

ABSTRACT

The Raf kinase inhibitor protein (RKIP) activates ß-adrenoceptors (ß-AR) and thereby induces a well-tolerated cardiac contractility and prevents heart failure in mice. Different to RKIP-mediated ß-AR activation, chronic activation of ß-AR by catecholamines was shown to be detrimental for the heart. RKIP is an endogenous inhibitor of G protein coupled receptor kinase 2 (GRK2); it binds GRK2 and thereby inhibits GRK2 mediated ß-AR phosphorylation and desensitization. Here, we evaluate RKIP-mediated effects on ß-AR to explore new strategies for ß-AR modulation. Co-immunoprecipitation assays and pull-down assays revealed subtype specificity of RKIP for the cardiac GRK isoforms GRK2 and GRK3 - not GRK5 - as well as several RKIP binding sites within their N-termini (GRK21-185 and GRK31-185). Overexpression of these N-termini prevented ß2-AR phosphorylation and internalization, subsequently increased receptor signaling in HEK293 cells and cardiomyocyte contractility. Co-immunoprecipitation assays of ß2-AR with these N-terminal GRK fragments revealed a direct interaction suggesting a steric interference of the fragments with the functional GRK-receptor interaction. Altogether, N-termini of GRK2 and GRK3 efficiently simulate RKIP effects on ß-AR signaling in HEK293 cells and in cardiomyocytes by their binding to ß2-AR and, thus, provide important insights for the development of new strategies to modulate ß2-AR signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Binding Sites , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , HEK293 Cells , Humans , Mice, Inbred Strains , Myocytes, Cardiac , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Phosphorylation , Receptors, Adrenergic, beta-2/genetics
17.
Amino Acids ; 51(6): 973-976, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31004228

ABSTRACT

We synthesized a previously identified ß-tubulin-derived G protein-coupled receptor kinase 2 (GKR2) peptide (GR-11-1; DEMEFTEAESNMN) and its amino-terminal extension (GR-11-1-N; GEGMDEMEFTEAESNMN) and carboxyl-terminal extension (GR-11-1-C; DEMEFTEAESNMNDLVSEYQ) peptides with the aim of finding a high-affinity peptide substrate for GRK2. GR-11-1-C showed high affinity for GRK2, but very low affinity for GKR5. Its specificity and sensitivity for GKR2 were greater than those of GR-11-1 and GR-11-1-N. These findings should be useful in designing tools for probing GKR2-mediated intracellular signaling pathways, as well as GRK2-specific drugs.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Insecta , Phosphorylation , Signal Transduction/physiology , Tubulin/metabolism
18.
Mol Cell Biochem ; 461(1-2): 103-118, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31363957

ABSTRACT

G protein-coupled receptor kinases (GRKs) phosphorylate the activated forms of G protein-coupled receptors (GPCRs), leading to receptor desensitization and internalization. In addition, GRKs can modify the activity of many non-GPCR-signaling pathways as well, controlling other cellular functions beyond that directly associated with a GPCR. In this report, we show that cervical cancer HeLa cells and breast cancer MDA MB 231 cells with reduced GRK5 expression display increased sensitivity to the apoptotic effects of paclitaxel (Taxol). This effect in cancer cells with low GRK5 levels could be because of blunted histone deacetylase 6 (HDAC6) activity that leads to an increase in α-tubulin acetylation levels, which augments paclitaxel sensitivity. We demonstrate that GRK5 and HDAC6 form a signaling complex in cells and in vitro. GRK5 phosphorylates HDAC6 at Ser-21 to promote its deacetylase activity. Therefore, the GRK5-HDAC6 interaction may contribute to paclitaxel resistance in cancer cells.


Subject(s)
Drug Resistance, Neoplasm/drug effects , G-Protein-Coupled Receptor Kinase 5/metabolism , Paclitaxel/pharmacology , Acetylation , Apoptosis/drug effects , Biocatalysis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Docetaxel/pharmacology , Female , G-Protein-Coupled Receptor Kinase 3/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , HeLa Cells , Histone Deacetylase 6/metabolism , Histones/metabolism , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Tubulin/metabolism
19.
Am J Physiol Cell Physiol ; 314(3): C310-C322, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29167148

ABSTRACT

Estradiol (E2) prevents cardiac hypertrophy, and these protective actions are mediated by estrogen receptor (ER)α and ERß. The G protein-coupled estrogen receptor (GPER) mediates many estrogenic effects, and its activation in the heart has been observed in ischemia and reperfusion injury or hypertension models; however, the underlying mechanisms need to be fully elucidated. Herein, we investigated whether the protective effect of E2 against cardiomyocyte hypertrophy induced by endothelin-1 (ET-1) is mediated by GPER and the signaling pathways involved. Isolated neonatal female rat cardiomyocytes were treated with ET-1 (100 nmol/l) for 48 h in the presence or absence of E2 (10 nmol/l) or GPER agonist G-1 (10 nmol/l) and GPER antagonist G-15 (10 nmol/l). ET-1 increased the surface area of cardiomyocytes, and this was associated with increased expression of atrial and brain natriuretic peptides. Additionally, ET-1 increased the phosphorylation of extracellular signal-related protein kinases-1/2 (ERK1/2). Notably, E2 or G-1 abolished the hypertrophic actions of ET-1, and that was reversed by G-15. Likewise, E2 reversed the ET-1-mediated increase of ERK1/2 phosphorylation as well as the decrease of phosphorylated Akt and its upstream activator 3-phosphoinositide-dependent protein kinase-1 (PDK1). These effects were inhibited by G-15, indicating that they are GPER dependent. Confirming the participation of GPER, siRNA silencing of GPER inhibited the antihypertrophic effect of E2. In conclusion, E2 plays a key role in antagonizing ET-1-induced hypertrophy in cultured neonatal cardiomyocytes through GPER signaling by a mechanism involving activation of the PDK1 pathway, which would prevent the increase of ERK1/2 activity and consequently the development of hypertrophy.


Subject(s)
Cardiomegaly/prevention & control , Endothelin-1/toxicity , Estradiol/pharmacology , Myocytes, Cardiac/drug effects , Receptors, G-Protein-Coupled/agonists , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Active Transport, Cell Nucleus , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiotoxicity , Cells, Cultured , Cytoprotection , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , G-Protein-Coupled Receptor Kinase 5/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
20.
J Biol Chem ; 292(39): 16032-16043, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28808053

ABSTRACT

G protein-coupled receptors (GPCRs) are essential for transferring extracellular signals into carefully choreographed intracellular responses controlling diverse aspects of cell physiology. The duration of GPCR-mediated signaling is primarily regulated via GPCR kinase (GRK)-mediated phosphorylation of activated receptors. Although many GRK structures have been reported, the mechanisms underlying GRK activation are not well-understood, in part because it is unknown how these structures map to the conformational landscape available to this enzyme family. Unlike most other AGC kinases, GRKs rely on their interaction with GPCRs for activation and not phosphorylation. Here, we used principal component analysis of available GRK and protein kinase A crystal structures to identify their dominant domain motions and to provide a framework that helps evaluate how close each GRK structure is to being a catalytically competent state. Our results indicated that disruption of an interface formed between the large lobe of the kinase domain and the regulator of G protein signaling homology domain (RHD) is highly correlated with establishment of the active conformation. By introducing point mutations in the GRK5 RHD-kinase domain interface, we show with both in silico and in vitro experiments that perturbation of this interface leads to higher phosphorylation activity. Navigation of the conformational landscape defined by this bioinformatics-based study is likely common to all GPCR-activated GRKs.


Subject(s)
G-Protein-Coupled Receptor Kinase 5/metabolism , Models, Molecular , Protein Processing, Post-Translational , Allosteric Regulation , Amino Acid Substitution , Animals , Cell Line , Computational Biology , Crystallography, X-Ray , Databases, Protein , Energy Transfer , Enzyme Activation , Expert Systems , G-Protein-Coupled Receptor Kinase 5/chemistry , G-Protein-Coupled Receptor Kinase 5/genetics , Humans , Insecta , Kinetics , Molecular Dynamics Simulation , Phosphorylation , Point Mutation , Principal Component Analysis , Protein Interaction Domains and Motifs , Protein Refolding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL