Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Blood Adv ; 6(15): 4524-4536, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35793439

ABSTRACT

The critical role of G protein-coupled receptor kinase 2 (GRK2) in regulating cardiac function has been well documented for >3 decades. Targeting GRK2 has therefore been extensively studied as a novel approach to treating cardiovascular disease. However, little is known about its role in hemostasis and thrombosis. We provide here the first evidence that GRK2 limits platelet activation and regulates the hemostatic response to injury. Deletion of GRK2 in mouse platelets causes increased platelet accumulation after laser-induced injury in the cremaster muscle arterioles, shortens tail bleeding time, and enhances thrombosis in adenosine 5'-diphosphate (ADP)-induced pulmonary thromboembolism and in FeCl3-induced carotid injury. GRK2-/- platelets have increased integrin activation, P-selectin exposure, and platelet aggregation in response to ADP stimulation. Furthermore, GRK2-/- platelets retain the ability to aggregate in response to ADP restimulation, indicating that GRK2 contributes to ADP receptor desensitization. Underlying these changes in GRK2-/- platelets is an increase in Ca2+ mobilization, RAS-related protein 1 activation, and Akt phosphorylation stimulated by ADP, as well as an attenuated rise of cyclic adenosine monophosphate levels in response to ADP in the presence of prostaglandin I2. P2Y12 antagonist treatment eliminates the phenotypic difference in platelet accumulation between wild-type and GRK2-/- mice at the site of injury. Pharmacologic inhibition of GRK2 activity in human platelets increases platelet activation in response to ADP. Finally, we show that GRK2 binds to endogenous Gßγ subunits during platelet activation. Collectively, these results show that GRK2 regulates ADP signaling via P2Y1 and P2Y12, interacts with Gßγ, and functions as a signaling hub in platelets for modulating the hemostatic response to injury.


Subject(s)
Hemostatics , Thrombosis , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/metabolism , Humans , Mice , Platelet Aggregation , Thrombosis/metabolism
2.
Cells ; 10(5)2021 05 15.
Article in English | MEDLINE | ID: mdl-34063503

ABSTRACT

When myocardial function is compromised as in heart failure (HF), there is activation of the sympathetic nervous system with elevated circulating catecholamine levels. These catecholamines activate cardiac and extra-cardiac adrenergic receptors (ARs). Interest in secreted extracellular vesicles (EVs) from the heart is growing and in HF, it is not known whether excessive activation of α- or ß-adrenergic receptors (ARs) could induce specific changes in EV content. In this study, we have evaluated, by next generation sequencing, the small RNA content, including micro-RNAs (miRs), of circulating EVs of mice exposed to chronic selective α- or ß- AR stimulation. EVs from mouse blood were purified by differential ultracentrifugation resulting in EVs with an average size of 116.6 ± 4.8 nm that by immunoblotting included protein markers of EVs. We identified the presence of miRs in blood EVs using miR-21-5p and -16-5p real-time PCR as known constituents of blood exosomes that make up a portion of EVs. We next performed next generation sequencing (NGS) of small non-coding RNAs found in blood EVs from mice following 7 days of chronic treatment with isoproterenol (ISO) or phenylephrine (PE) to stimulate α- or ß-ARs, respectively. PE increased the percent of genomic repeat region reads and decreased the percent of miR reads. In miR expression analysis, PE and ISO displayed specific patterns of miR expression that suggests differential pathway regulation. The top 20 KEGG pathways predicted by differential expressed miRs show that PE and ISO share 11 of 20 pathways analyzed and reveal also key differences including three synapse relative pathways induced by ISO relative to PE treatment. Both α-and ß-AR agonists can alter small RNA content of circulating blood EVs/exosomes including differential expression and loading of miRs that indicate regulation of distinct pathways. This study provides novel insight into chronic sympathetic nervous system activation in HF where excessive catecholamines may not only participate in pathological remodeling of the heart but alter other organs due to secretion of EVs with altered miR content.


Subject(s)
Cardiovascular Diseases/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/blood , Receptors, Adrenergic, alpha/blood , Receptors, Adrenergic, beta/blood , Animals , Biomarkers/blood , Biomarkers/metabolism , Mice
3.
Sci Signal ; 14(676)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785612

ABSTRACT

Aberrant changes in gene expression underlie the pathogenesis and progression of pressure-overload heart failure, leading to maladaptive cardiac hypertrophy, ventricular remodeling, and contractile dysfunction. Signaling through the G protein Gq triggers maladaptation and heart failure, in part through the activation of G protein-coupled receptor kinase 5 (GRK5). Hypertrophic stimuli induce the accumulation of GRK5 in the nuclei of cardiomyocytes, where it regulates pathological gene expression through multiple transcription factors including NFAT. The nuclear targeting of GRK5 is mediated by an amino-terminal (NT) domain that binds to calmodulin (CaM). Here, we sought to prevent GRK5-mediated pathology in pressure-overload maladaptation and heart failure by expressing in cardiomyocytes a peptide encoding the GRK5 NT (GRK5nt) that encompasses the CaM binding domain. In cultured cardiomyocytes, GRK5nt expression abrogated Gq-coupled receptor-mediated hypertrophy, including attenuation of pathological gene expression and the transcriptional activity of NFAT and NF-κB. We confirmed that GRK5nt bound to and blocked Ca2+-CaM from associating with endogenous GRK5, thereby preventing GRK5 nuclear accumulation after pressure overload. We generated mice that expressed GRKnt in a cardiac-specific fashion (TgGRK5nt mice), which exhibited reduced cardiac hypertrophy, ventricular dysfunction, pulmonary congestion, and cardiac fibrosis after chronic transverse aortic constriction. Together, our data support a role for GRK5nt as an inhibitor of pathological GRK5 signaling that prevents heart failure.


Subject(s)
Cardiomegaly , G-Protein-Coupled Receptor Kinase 5/genetics , Heart Failure , Animals , Calmodulin/metabolism , Cardiomegaly/genetics , Cell Nucleus/metabolism , Heart Failure/genetics , Mice , Myocytes, Cardiac/metabolism
4.
J Mol Cell Cardiol ; 154: 137-153, 2021 05.
Article in English | MEDLINE | ID: mdl-33548241

ABSTRACT

G protein-coupled receptor (GPCR) kinase 2 (GRK2) expression and activity are elevated early on in response to several forms of cardiovascular stress and are a hallmark of heart failure. Interestingly, though, in addition to its well-characterized role in regulating GPCRs, mounting evidence suggests a GRK2 "interactome" that underlies a great diversity in its functional roles. Several such GRK2 interacting partners are important for adaptive and maladaptive myocyte growth; therefore, an understanding of domain-specific interactions with signaling and regulatory molecules could lead to novel targets for heart failure therapy. Herein, we subjected transgenic mice with cardiac restricted expression of a short, amino terminal fragment of GRK2 (ßARKnt) to pressure overload and found that unlike their littermate controls or previous GRK2 fragments, they exhibited an increased left ventricular wall thickness and mass prior to cardiac stress that underwent proportional hypertrophic growth to controls after acute pressure overload. Importantly, despite this enlarged heart, ßARKnt mice did not undergo the expected transition to heart failure observed in controls. Further, ßARKnt expression limited adverse left ventricular remodeling and increased cell survival signaling. Proteomic analysis to identify ßARKnt binding partners that may underlie the improved cardiovascular phenotype uncovered a selective functional interaction of both endogenous GRK2 and ßARKnt with AKT substrate of 160 kDa (AS160). AS160 has emerged as a key downstream regulator of insulin signaling, integrating physiological and metabolic cues to couple energy demand to membrane recruitment of Glut4. Our preliminary data indicate that in ßARKnt mice, cardiomyocyte insulin signaling is improved during stress, with a coordinate increase in spare respiratory activity and ATP production without metabolite switching. Surprisingly, these studies also revealed a significant decrease in gonadal fat weight, equivalent to human abdominal fat, in male ßARKnt mice at baseline and following cardiac stress. These data suggest that the enhanced AS160-mediated signaling in the ßARKnt mice may ameliorate pathological cardiac remodeling through direct modulation of insulin signaling within cardiomyocytes, and translate these to beneficial effects on systemic metabolism.


Subject(s)
Cardiomegaly/etiology , Cardiomegaly/physiopathology , G-Protein-Coupled Receptor Kinase 2/chemistry , Peptides/genetics , Protein Interaction Domains and Motifs , Animals , Biomarkers , Cardiomegaly/diagnosis , Disease Models, Animal , Disease Susceptibility , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Expression , Mice , Mice, Transgenic , Peptides/metabolism , Phenotype , Protein Binding , Signal Transduction , Ventricular Remodeling
5.
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
6.
Am J Physiol Heart Circ Physiol ; 320(4): H1276-H1289, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33513081

ABSTRACT

Recent data supporting any benefit of stem cell therapy for ischemic heart disease have suggested paracrine-based mechanisms via extracellular vesicles (EVs) including exosomes. We have previously engineered cardiac-derived progenitor cells (CDCs) to express a peptide inhibitor, ßARKct, of G protein-coupled receptor kinase 2, leading to improvements in cell proliferation, survival, and metabolism. In this study, we tested whether ßARKct-CDC EVs would be efficacious when applied to stressed myocytes in vitro and in vivo. When isolated EVs from ßARKct-CDCs and control GFP-CDCs were added to cardiomyocytes in culture, they both protected against hypoxia-induced apoptosis. We tested whether these EVs could protect the mouse heart in vivo, following exposure either to myocardial infarction (MI) or acute catecholamine toxicity. Both types of EVs significantly protected against ischemic injury and improved cardiac function after MI compared with mice treated with EVs from mouse embryonic fibroblasts; however, ßARKct EVs treated mice did display some unique beneficial properties including significantly altered pro- and anti-inflammatory cytokines. Importantly, in a catecholamine toxicity model of heart failure (HF), myocardial injections of ßARKct-containing EVs were superior at preventing HF compared with control EVs, and this catecholamine toxicity protection was recapitulated in vitro. Therefore, introduction of the ßARKct into cellular EVs can have improved reparative properties in the heart especially against catecholamine damage, which is significant as sympathetic nervous system activity is increased in HF.NEW & NOTEWORTHY ßARKct, the peptide inhibitor of GRK2, improves survival and metabolic functions of cardiac-derived progenitor cells. As any benefit of stem cells in the ischemic and injured heart suggests paracrine mechanisms via secreted EVs, we investigated whether CDC-ßARKct engineered EVs would show any benefit over control CDC-EVs. Compared with control EVs, ßARKct-containing EVs displayed some unique beneficial properties that may be due to altered pro- and anti-inflammatory cytokines within the vesicles.


Subject(s)
Extracellular Vesicles/transplantation , Heart Failure/prevention & control , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , Peptides/metabolism , Recombinant Proteins/metabolism , Stem Cell Transplantation , Animals , Apoptosis , Cell Hypoxia , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Paracrine Communication , Peptides/genetics , Rats , Recombinant Proteins/genetics , Recovery of Function , Signal Transduction , Stem Cells/metabolism
7.
JCI Insight ; 52019 04 04.
Article in English | MEDLINE | ID: mdl-30946029

ABSTRACT

A vast body of literature has established GRK2 as a key player in the development and progression of heart failure. Inhibition of GRK2 improves cardiac function post injury in numerous animal models. In recent years, discovery of several non-canonical GRK2 targets has expanded our view of this kinase. Here, we describe the novel and exciting finding that cardiac GRK2 activity can regulate whole body metabolism. Transgenic mice with cardiac-specific expression of a peptide inhibitor of GRK2 (TgßARKct) display an enhanced obesogenic phenotype when fed a high fat diet (HFD). In contrast, mice with cardiac-specific overexpression of GRK2 (TgGRK2) show resistance to HFD induced obesity. White adipose tissue (WAT) mass was significantly enhanced in HFD fed TgßARKct mice. Furthermore, regulators of adipose differentiation were differentially regulated in WAT from mice with gain or loss of GRK2 function. Using complex metabolomics we found that cardiac GRK2 signaling altered myocardial BCAA and endocannabinoid metabolism and modulated circulating BCAA and endocannabinoid metabolite profiles on a HFD, and one of the BCAA metabolites identified here enhances adipocyte differentiation in vitro. Taken together, these results suggest that metabolic changes in the heart due to GRK2 signaling on a HFD control whole body metabolism.


Subject(s)
Adipose Tissue, White/metabolism , Adiposity/physiology , G-Protein-Coupled Receptor Kinase 2/metabolism , Myocardium/metabolism , Obesity/metabolism , Adipocytes/physiology , Adipose Tissue, White/cytology , Amino Acids, Branched-Chain/metabolism , Animals , Cell Differentiation/physiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Endocannabinoids/metabolism , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/genetics , Humans , Male , Metabolomics , Mice , Mice, Transgenic , Obesity/etiology , Signal Transduction/physiology , Weight Gain/physiology
8.
Sci Signal ; 11(560)2018 12 11.
Article in English | MEDLINE | ID: mdl-30538174

ABSTRACT

Increased abundance of GRK2 [G protein-coupled receptor (GPCR) kinase 2] is associated with poor cardiac function in heart failure patients. In animal models, GRK2 contributes to the pathogenesis of heart failure after ischemia-reperfusion (IR) injury. In addition to its role in down-regulating activated GPCRs, GRK2 also localizes to mitochondria both basally and post-IR injury, where it regulates cellular metabolism. We previously showed that phosphorylation of GRK2 at Ser670 is essential for the translocation of GRK2 to the mitochondria of cardiomyocytes post-IR injury in vitro and that this localization promotes cell death. Here, we showed that mice with a S670A knock-in mutation in endogenous GRK2 showed reduced cardiomyocyte death and better cardiac function post-IR injury. Cultured GRK2-S670A knock-in cardiomyocytes subjected to IR in vitro showed enhanced glucose-mediated mitochondrial respiratory function that was partially due to maintenance of pyruvate dehydrogenase activity and improved glucose oxidation. Thus, we propose that mitochondrial GRK2 plays a detrimental role in cardiac glucose oxidation post-injury.


Subject(s)
Apoptosis , G-Protein-Coupled Receptor Kinase 2/metabolism , Glucose/chemistry , Heart Failure/prevention & control , Ischemia/physiopathology , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Animals , G-Protein-Coupled Receptor Kinase 2/genetics , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Mitochondria/pathology , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxygen Consumption , Phosphorylation , Point Mutation , Serine/chemistry , Serine/genetics , Serine/metabolism , Signal Transduction
9.
Proc Natl Acad Sci U S A ; 114(5): E859-E868, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096338

ABSTRACT

Mitochondrial fusion is thought to be important for supporting cardiac contractility, but is hardly detectable in cultured cardiomyocytes and is difficult to directly evaluate in the heart. We overcame this obstacle through in vivo adenoviral transduction with matrix-targeted photoactivatable GFP and confocal microscopy. Imaging in whole rat hearts indicated mitochondrial network formation and fusion activity in ventricular cardiomyocytes. Promptly after isolation, cardiomyocytes showed extensive mitochondrial connectivity and fusion, which decayed in culture (at 24-48 h). Fusion manifested both as rapid content mixing events between adjacent organelles and slower events between both neighboring and distant mitochondria. Loss of fusion in culture likely results from the decline in calcium oscillations/contractile activity and mitofusin 1 (Mfn1), because (i) verapamil suppressed both contraction and mitochondrial fusion, (ii) after spontaneous contraction or short-term field stimulation fusion activity increased in cardiomyocytes, and (iii) ryanodine receptor-2-mediated calcium oscillations increased fusion activity in HEK293 cells and complementing changes occurred in Mfn1. Weakened cardiac contractility in vivo in alcoholic animals is also associated with depressed mitochondrial fusion. Thus, attenuated mitochondrial fusion might contribute to the pathogenesis of cardiomyopathy.


Subject(s)
Calcium Signaling/physiology , Mitochondria, Heart/physiology , Mitochondrial Dynamics/physiology , Myocardial Contraction/physiology , Animals , Cell Line , Genes, Reporter , Genetic Vectors , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Male , Microscopy, Confocal , Mitochondria, Heart/ultrastructure , Rats , Rats, Sprague-Dawley , Transduction, Genetic
10.
Sci Signal ; 9(420): ra30, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-27016525

ABSTRACT

G protein-coupled receptor (GPCR) kinases (GRKs) play a critical role in cardiac function by regulating GPCR activity. GRK2 suppresses GPCR signaling by phosphorylating and desensitizing active GPCRs, and through protein-protein interactions that uncouple GPCRs from their downstream effectors. Several GRK2 interacting partners, including Gα(q), promote maladaptive cardiac hypertrophy, which leads to heart failure, a leading cause of mortality worldwide. The regulator of G protein signaling (RGS) domain of GRK2 interacts with and inhibits Gα(q) in vitro. We generated TgßARKrgs mice with cardiac-specific expression of the RGS domain of GRK2 and subjected these mice to pressure overload to trigger adaptive changes that lead to heart failure. Unlike their nontransgenic littermate controls, the TgßARKrgs mice exhibited less hypertrophy as indicated by reduced left ventricular wall thickness, decreased expression of genes linked to cardiac hypertrophy, and less adverse structural remodeling. The ßARKrgs peptide, but not endogenous GRK2, interacted with Gα(q) and interfered with signaling through this G protein. These data support the development of GRK2-based therapeutic approaches to prevent hypertrophy and heart failure.


Subject(s)
Cardiomegaly/enzymology , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Heart Failure/enzymology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , G-Protein-Coupled Receptor Kinase 2/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Mice , Mice, Transgenic , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Domains
11.
J Mol Cell Cardiol ; 89(Pt B): 360-4, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26506135

ABSTRACT

The G protein-coupled receptor kinase-2 (GRK2) is upregulated in the injured heart and contributes to heart failure pathogenesis. GRK2 was recently shown to associate with mitochondria but its functional impact in myocytes due to this localization is unclear. This study was undertaken to determine the effect of elevated GRK2 on mitochondrial respiration in cardiomyocytes. Sub-fractionation of purified cardiac mitochondria revealed that basally GRK2 is found in multiple compartments. Overexpression of GRK2 in mouse cardiomyocytes resulted in an increased amount of mitochondrial-based superoxide. Inhibition of GRK2 increased oxygen consumption rates and ATP production. Moreover, fatty acid oxidation was found to be significantly impaired when GRK2 was elevated and was dependent on the catalytic activity and mitochondrial localization of this kinase. Our study shows that independent of cardiac injury, GRK2 is localized in the mitochondria and its kinase activity negatively impacts the function of this organelle by increasing superoxide levels and altering substrate utilization for energy production.


Subject(s)
Fatty Acids/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Oxygen Consumption , Superoxides/metabolism , Animals , Cell Respiration , Mice, Transgenic , Stress, Physiological
12.
Circ Res ; 117(12): 1001-12, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26515328

ABSTRACT

RATIONALE: G protein-coupled receptor kinases (GRKs) are dynamic regulators of cellular signaling. GRK5 is highly expressed within myocardium and is upregulated in heart failure. Although GRK5 is a critical regulator of cardiac G protein-coupled receptor signaling, recent data has uncovered noncanonical activity of GRK5 within nuclei that plays a key role in pathological hypertrophy. Targeted cardiac elevation of GRK5 in mice leads to exaggerated hypertrophy and early heart failure after transverse aortic constriction (TAC) because of GRK5 nuclear accumulation. OBJECTIVE: In this study, we investigated the role of GRK5 in physiological, swimming-induced hypertrophy (SIH). METHODS AND RESULTS: Cardiac-specific GRK5 transgenic mice and nontransgenic littermate control mice were subjected to a 21-day high-intensity swim protocol (or no swim sham controls). SIH and specific molecular and genetic indices of physiological hypertrophy were assessed, including nuclear localization of GRK5, and compared with TAC. Unlike after TAC, swim-trained transgenic GRK5 and nontransgenic littermate control mice exhibited similar increases in cardiac growth. Mechanistically, SIH did not lead to GRK5 nuclear accumulation, which was confirmed in vitro as insulin-like growth factor-1, a known mediator of physiological hypertrophy, was unable to induce GRK5 nuclear translocation in myocytes. We found specific patterns of altered gene expression between TAC and SIH with GRK5 overexpression. Further, SIH in post-TAC transgenic GRK5 mice was able to preserve cardiac function. CONCLUSIONS: These data suggest that although nuclear-localized GRK5 is a pathological mediator after stress, this noncanonical nuclear activity of GRK5 is not induced during physiological hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , G-Protein-Coupled Receptor Kinase 5/physiology , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cardiomegaly/genetics , Cells, Cultured , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Rats
13.
Mol Cell ; 60(1): 47-62, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26387735

ABSTRACT

Mitochondrial permeability transition is a phenomenon in which the mitochondrial permeability transition pore (PTP) abruptly opens, resulting in mitochondrial membrane potential (ΔΨm) dissipation, loss of ATP production, and cell death. Several genetic candidates have been proposed to form the PTP complex, however, the core component is unknown. We identified a necessary and conserved role for spastic paraplegia 7 (SPG7) in Ca(2+)- and ROS-induced PTP opening using RNAi-based screening. Loss of SPG7 resulted in higher mitochondrial Ca(2+) retention, similar to cyclophilin D (CypD, PPIF) knockdown with sustained ΔΨm during both Ca(2+) and ROS stress. Biochemical analyses revealed that the PTP is a heterooligomeric complex composed of VDAC, SPG7, and CypD. Silencing or disruption of SPG7-CypD binding prevented Ca(2+)- and ROS-induced ΔΨm depolarization and cell death. This study identifies an ubiquitously expressed IMM integral protein, SPG7, as a core component of the PTP at the OMM and IMM contact site.


Subject(s)
Cyclophilins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mitochondria/metabolism , Voltage-Dependent Anion Channel 1/metabolism , ATPases Associated with Diverse Cellular Activities , Binding Sites , Calcium/metabolism , Cell Death , Cyclophilins/chemistry , HEK293 Cells , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Metalloendopeptidases/chemistry , Mitochondrial Membranes/metabolism , RNA Interference , Reactive Oxygen Species/metabolism
14.
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
15.
Sci Transl Med ; 7(277): 277ra31, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25739765

ABSTRACT

Heart failure (HF) is a disease of epidemic proportion and is associated with exceedingly high health care costs. G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor (GPCR) kinase 2 (GRK2), which is up-regulated in the failing human heart, appears to play a critical role in HF progression in part because enhanced GRK2 activity promotes dysfunctional adrenergic signaling and myocyte death. Recently, we found that the selective serotonin reuptake inhibitor (SSRI) paroxetine could inhibit GRK2 with selectivity over other GRKs. Wild-type mice were treated for 4 weeks with paroxetine starting at 2 weeks after myocardial infarction (MI). These mice were compared with mice treated with fluoxetine, which does not inhibit GRK2, to control for the SSRI effects of paroxetine. All mice exhibited similar left ventricular (LV) dysfunction before treatment; however, although the control and fluoxetine groups had continued degradation of function, the paroxetine group had considerably improved LV function and structure, and several hallmarks of HF were either inhibited or reversed. Use of genetically engineered mice indicated that paroxetine was working through GRK2 inhibition. The beneficial effects of paroxetine were markedly greater than those of ß-blocker therapy, a current standard of care in human HF. These data demonstrate that paroxetine-mediated inhibition of GRK2 improves cardiac function after MI and represents a potential repurposing of this drug, as well as a starting point for innovative small-molecule GRK2 inhibitor development.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Myocardial Infarction/physiopathology , Paroxetine/pharmacology , Ventricular Remodeling/drug effects , Adrenergic beta-Antagonists/pharmacology , Animals , Fibrosis , G-Protein-Coupled Receptor Kinase 2/metabolism , Heart Failure/complications , Heart Failure/physiopathology , Hemodynamics/drug effects , Male , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardium/pathology , Receptors, Adrenergic, beta/metabolism , Translational Research, Biomedical
16.
Circ Res ; 115(12): 976-85, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25332207

ABSTRACT

RATIONALE: G protein-coupled receptor kinases (GRKs) acting in the cardiomyocyte regulate important signaling events that control cardiac function. Both GRK2 and GRK5, the predominant GRKs expressed in the heart, have been shown to be upregulated in failing human myocardium. Although the canonical role of GRKs is to desensitize G protein-coupled receptors via phosphorylation, it has been demonstrated that GRK5, unlike GRK2, can reside in the nucleus of myocytes and exert G protein-coupled receptor-independent effects that promote maladaptive cardiac hypertrophy and heart failure. OBJECTIVE: To explore novel mechanisms by which GRK5 acting in the nucleus of cardiomyocytes participates in pathological cardiac hypertrophy. METHODS AND RESULTS: In this study, we have found that GRK5-mediated pathological cardiac hypertrophy involves the activation of the nuclear factor of activated T cells (NFAT) because GRK5 causes enhancement of NFAT-mediated hypertrophic gene transcription. Transgenic mice with cardiomyocyte-specific GRK5 overexpression activate an NFAT-reporter in mice basally and after hypertrophic stimulation, including transverse aortic constriction and phenylephrine treatment. Complimentary to this, GRK5 null mice exhibit less NFAT transcriptional activity after transverse aortic constriction. Furthermore, the loss of NFATc3 expression in the heart protected GRK5 overexpressing transgenic mice from the exaggerated hypertrophy and early progression to heart failure seen after transverse aortic constriction. Molecular studies suggest that GRK5 acts in concert with NFAT to increase hypertrophic gene transcription in the nucleus via GRK5's ability to bind DNA directly without a phosphorylation event. CONCLUSIONS: GRK5, acting in a kinase independent manner, is a facilitator of NFAT activity and part of a DNA-binding complex responsible for pathological hypertrophic gene transcription.


Subject(s)
Cardiomegaly/enzymology , G-Protein-Coupled Receptor Kinase 5/metabolism , Myocytes, Cardiac/enzymology , NFATC Transcription Factors/metabolism , Animals , Binding Sites , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Line , Cell Nucleus/enzymology , Disease Models, Animal , Disease Progression , Female , G-Protein-Coupled Receptor Kinase 5/genetics , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myocytes, Cardiac/pathology , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Rats , Time Factors , Transcription, Genetic , Transfection
17.
Antioxid Redox Signal ; 21(14): 2032-43, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-24702056

ABSTRACT

SIGNIFICANCE: Heart failure (HF) is a common end point for many underlying cardiovascular diseases. Down-regulation and desensitization of ß-adrenergic receptors (ß-AR) caused by G-protein-coupled receptor (GPCR) kinase 2 (GRK2) are prominent features of HF. Recent Advances and Critical Issues: Significant progress has been made to understand the pathological role of GRK2 in the heart both as a GPCR kinase and as a molecule that can exert GPCR-independent effects. Inhibition of cardiac GRK2 has proved to be therapeutic in the failing heart and may offer synergistic and additional benefits to ß-blocker therapy. However, the mechanisms of how GRK2 directly contributes to the pathogenesis of HF need further investigation, and additional verification of the mechanistic details are needed before GRK2 inhibition can be used for the treatment of HF. FUTURE DIRECTIONS: The newly identified characteristics of GRK2, including the S-nitrosylation of GRK2 and the localization of GRK2 on mitochondria, merit further investigation. They may contribute to it being a pro-death kinase and result in HF under stressed conditions through regulation of intracellular signaling, including cardiac reduction-oxidation (redox) balance. A thorough understanding of the functions of GRK2 in the heart is necessary in order to finalize it as a candidate for drug development.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Genetic Therapy , Heart Failure/enzymology , Myocardium/enzymology , Adrenergic beta-Antagonists/therapeutic use , Drug Design , Heart Failure/metabolism , Heart Failure/therapy , Humans , Myocardium/pathology , Oxidation-Reduction , Receptors, Adrenergic, beta/biosynthesis , Receptors, Adrenergic, beta/genetics , Signal Transduction/genetics
18.
J Mol Cell Cardiol ; 67: 86-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24361238

ABSTRACT

cAMP-dependent protein kinase (PKA) regulates the L-type calcium channel, the ryanodine receptor, and phospholamban (PLB) thereby increasing inotropy. Cardiac contractility is also regulated by p38 MAPK, which is a negative regulator of cardiac contractile function. The aim of this study was to identify the mechanism mediating the positive inotropic effect of p38 inhibition. Isolated adult and neonatal cardiomyocytes and perfused rat hearts were utilized to investigate the molecular mechanisms regulated by p38. PLB phosphorylation was enhanced in cardiomyocytes by chemical p38 inhibition, by overexpression of dominant negative p38α and by p38α RNAi, but not with dominant negative p38ß. Treatment of cardiomyocytes with dominant negative p38α significantly decreased Ca(2+)-transient decay time indicating enhanced sarco/endoplasmic reticulum Ca(2+)-ATPase function and increased cardiomyocyte contractility. Analysis of signaling mechanisms involved showed that inhibition of p38 decreased the activity of protein phosphatase 2A, which renders protein phosphatase inhibitor-1 phosphorylated and thereby inhibits PP1. In conclusion, inhibition of p38α enhances PLB phosphorylation and diastolic Ca(2+) uptake. Our findings provide evidence for novel mechanism regulating cardiac contractility upon p38 inhibition.


Subject(s)
Muscle Contraction/physiology , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Enzyme Activation/drug effects , Myocytes, Cardiac/drug effects , Phosphorylation , RNA Interference , Rats , p38 Mitogen-Activated Protein Kinases/pharmacology
19.
Sci Signal ; 6(299): ra95, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24170934

ABSTRACT

Heart failure caused by ischemic heart disease is a leading cause of death in the developed world. Treatment is currently centered on regimens involving G protein-coupled receptors (GPCRs) or nitric oxide (NO). These regimens are thought to target distinct molecular pathways. We showed that these pathways were interdependent and converged on the effector GRK2 (GPCR kinase 2) to regulate myocyte survival and function. Ischemic injury coupled to GPCR activation, including GPCR desensitization and myocyte loss, required GRK2 activation, and we found that cardioprotection mediated by inhibition of GRK2 depended on endothelial nitric oxide synthase (eNOS) and was associated with S-nitrosylation of GRK2. Conversely, the cardioprotective effects of NO bioactivity were absent in a knock-in mouse with a form of GRK2 that cannot be S-nitrosylated. Because GRK2 and eNOS inhibit each other, the balance of the activities of these enzymes in the myocardium determined the outcome to ischemic injury. Our findings suggest new insights into the mechanism of action of classic drugs used to treat heart failure and new therapeutic approaches to ischemic heart disease.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Blotting, Western , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Heart/drug effects , Heart/physiopathology , Isoproterenol/pharmacology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Rats , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , S-Nitrosoglutathione/pharmacology , S-Nitrosothiols/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
20.
PLoS One ; 8(9): e73537, 2013.
Article in English | MEDLINE | ID: mdl-24058479

ABSTRACT

AIM: As technological interventions treating acute myocardial infarction (MI) improve, post-ischemic heart failure increasingly threatens patient health. The aim of the current study was to test whether FADD could be a potential target of gene therapy in the treatment of heart failure. METHODS: Cardiomyocyte-specific FADD knockout mice along with non-transgenic littermates (NLC) were subjected to 30 minutes myocardial ischemia followed by 7 days of reperfusion or 6 weeks of permanent myocardial ischemia via the ligation of left main descending coronary artery. Cardiac function were evaluated by echocardiography and left ventricular (LV) catheterization and cardiomyocyte death was measured by Evans blue-TTC staining, TUNEL staining, and caspase-3, -8, and -9 activities. In vitro, H9C2 cells transfected with ether scramble siRNA or FADD siRNA were stressed with chelerythrin for 30 min and cleaved caspase-3 was assessed. RESULTS: FADD expression was significantly decreased in FADD knockout mice compared to NLC. Ischemia/reperfusion (I/R) upregulated FADD expression in NLC mice, but not in FADD knockout mice at the early time. FADD deletion significantly attenuated I/R-induced cardiac dysfunction, decreased myocardial necrosis, and inhibited cardiomyocyte apoptosis. Furthermore, in 6 weeks long term permanent ischemia model, FADD deletion significantly reduced the infarct size (from 41.20 ± 3.90% in NLC to 26.83 ± 4.17% in FADD deletion), attenuated myocardial remodeling, improved cardiac function and improved survival. In vitro, FADD knockdown significantly reduced chelerythrin-induced the level of cleaved caspase-3. CONCLUSION: Taken together, our results suggest FADD plays a critical role in post-ischemic heart failure. Inhibition of FADD retards heart failure progression. Our data supports the further investigation of FADD as a potential target for genetic manipulation in the treatment of heart failure.


Subject(s)
Fas-Associated Death Domain Protein/genetics , Heart Failure/genetics , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/metabolism , Ventricular Dysfunction, Left/genetics , Animals , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cells, Cultured , Coronary Vessels/surgery , Disease Models, Animal , Fas-Associated Death Domain Protein/antagonists & inhibitors , Fas-Associated Death Domain Protein/deficiency , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Severity of Illness Index , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...