Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Determining the Absolute Requirement of G Protein-Coupled Receptor Kinase 5 for Pathological Cardiac Hypertrophy: Short Communication

Circulation Research, 2012
...Read more
1048 Integrative Physiology H eart failure (HF), a leading cause of death in the Western world, often occurs as an end phase of pathological myocardial hypertrophy. 1 Hypertrophy is initially an adaptive response to stresses ranging from hypertension, valve disease, or cardiac injury. 2,3 In an attempt to normalize wall stress, car- diomyocytes enlarge, sarcomeres reorganize, fibroblasts pro- liferate, and hypertrophic genes, including the so-called fetal gene program, are upregulated. If prolonged, these responses lead to chamber dilation, myocardial apoptosis, and HF. 2,3 In pathological cardiac growth, the molecular pathways affect- ing transcription lie downstream of the nodal hypertrophic signal transducer, Gq. 1 Among these complex pathways are the class II histone deacetylases (HDACs). Physiologically opposed to car- diac growth, the HDACs repress expression of key hypertrophic genes, primarily through inhibition of myocyte enhancer factor 2 (MEF2). 3 Genetic deletion of the class II HDAC, HDAC5, sensitizes mice to cardiac stress, whereas murine deletion of MEF2 confers cardioprotection, decreasing pathological hyper- trophy and attenuating upregulation of the fetal gene program. 4 HDAC kinases control nuclear HDAC activity as phosphorylation of HDAC5 induces its nuclear export and MEF2 derepression. 3–5 In This Issue, see p 947 Editorial, see p 957 We recently identified G protein-coupled receptor (GPCR) kinase-5 (GRK5) as a nuclear HDAC kinase, joining protein ki- nase D (PKD) and calmodulin-dependent kinase II (CaMKII) as HDAC-mediated facilitators of cardiac growth after hypertro- phic stimuli. 5–7 This represents a novel, non-GPCR cardiac role for this GRK. We showed that mice with cardiac overexpres- sion of GRK5 demonstrate Gq-dependent nuclear transloca- tion of GRK5, where it can phosphorylate HDAC5 and induce Original received May 8, 2012; revision received July 30, 2012; accepted August 1, 2012. In July 2012, the average time from submission to first decision for all original research papers submitted to Circulation Research was 11.2 days. From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.I.G., W.J.K.); Center for Translational Medicine (E.G., X.S., W.J.K.) and the Department of Pharmacology (W.J.K.), Temple University School of Medicine, Philadelphia, PA; and the Department of Medicine, Duke University Medical Center, Durham, NC (R.T.P.). This manuscript was sent to Elizabeth Murphy, Consulting Editor, for review by expert referees, editorial decision, and final disposition. The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA.112. 273367/-/DC1. Correspondence to Walter J. Koch, MD, Center for Translational Medicine, Temple University School of Medicine, 3500 N Broad St, MERB 941, Philadelphia, PA 19140. E-mail Walter.Koch@temple.edu © 2012 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.112.273367 Determining the Absolute Requirement of G Protein–Coupled Receptor Kinase 5 for Pathological Cardiac Hypertrophy Short Communication Jessica I. Gold, Erhe Gao, Xiying Shang, Richard T. Premont, Walter J. Koch Rationale: Heart failure (HF) is often the end phase of maladaptive cardiac hypertrophy. A contributing factor is activation of a hypertrophic gene expression program controlled by decreased class II histone deacetylase (HDAC) transcriptional repression via HDAC phosphorylation. Cardiac-specific overexpression of G proteinen–coupled receptor kinase-5 (GRK5) has previously been shown to possess nuclear activity as a HDAC5 kinase, promoting an intolerance to in vivo ventricular pressure overload; however, its endogenous requirement in adaptive and maladaptive hypertrophy remains unknown. Objective: We used mouse models with global or cardiomyocyte-specific GRK5 gene deletion to determine the absolute requirement of endogenous GRK5 for cardiac hypertrophy and HF development after chronic hypertrophic stimuli. Methods and Results: Mice with global deletion of GRK5 were subjected to transverse aortic constriction. At 12 weeks, these mice showed attenuated hypertrophy, remodeling, and hypertrophic gene transcription along with preserved cardiac function. Global GRK5 deletion also diminished hypertrophy and related gene expression due to chronic phenylephrine infusion. We then generated mice with conditional, cardiac-specific deletion of GRK5 that also demonstrated similar protection from pathological cardiac hypertrophy and HF after transverse aortic constriction. Conclusions: These results define myocyte GRK5 as a critical regulator of pathological cardiac growth after ventricular pressure overload, supporting its role as an endogenous (patho)-physiological HDAC kinase. Further, these results define GRK5 as a potential therapeutic target to limit HF development after hypertrophic stress. (Circ Res. 2012;111:1048–1053.) Key Words: G protein–coupled receptor kinases cardiac hypertrophy conditional transgenic mouse by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from
Gold et al GRK5 in Pathological Cardiac Hypertrophy 1049 MEF2 activity. 6 Moreover, cardiac GRK5-overexpressing mice displayed an intolerance to ventricular pressure overload with potentiated maladaptive hypertrophy and accelerated HF after transverse aortic constriction (TAC). 6 Importantly, these trans- genic studies did not address whether endogenous GRK5 plays a role in the hypertrophic response. In the present study, we used global and cardiomyocyte- specific GRK5 knockout (KO) mice to directly address the importance of endogenous GRK5 in cardiac hypertrophy. We found that GRK5 in cardiomyocytes is absolutely required for hypertrophic responses after stress. Further, our data indicate that limiting GRK5 expression in the heart can protect against maladaptive cardiac growth and HF development. Methods Generation of GRK5cKO Mice Detailed mouse protocols are described in the Online Data Supplement Methods. Surgical Procedures and Echocardiography TAC methodology, minipump implantation, and determination of in vivo cardiac function and morphology are described in the Online Data Supplement and Methods. RNA Analysis Methods for RNA analysis are described in the Online Data Supplement. Immunoblot Analysis Detailed methods for heart subfractionation and immunoblotting are described in the Online Data Supplement. Results Global GRK5 Deletion Diminishes In Vivo Cardiac Hypertrophy We subjected male global GRK5 gene knockout mice (GRK5gKO) 8 and littermate wild-type (WT) control mice to TAC. Constitutive GRK5 deletion attenuated pressure-overload- induced cardiac growth seen in WT mice at 12 weeks after TAC (Figure 1A). Cardiac dimensions were measured serially by echocardiography to track development of hypertrophy and left ventricular (LV) dilatation over 12 weeks. WT mice showed a quick rise in LV posterior wall thickness (LVPWT), with a peak thickness of 1.99±0.05 mm at 4 weeks after TAC, which then decreased rapidly, indicative of adverse remodeling (Figure 1B). Interestingly, global GRK5 deletion significantly delays the ini- tiation of cardiac hypertrophy after TAC, as GRK5gKO mice do reach a similar LVPWT (1.89±0.09 mm) as WT mice but not until 12 weeks after TAC (Figure 1B). At the end of 12 weeks, GRK5gKO mice had significantly less cardiac hypertrophy as determined by smaller heart weighten-to-enbody weight (HW/ BW) ratios (Figure 1C). Importantly, GRK5gKO mice show no signs of LV dilatation via echocardiographic measurements of systolic LV interior diameter (LVIDs), which were significantly increased in post-TAC WT mice (Figure 1D and Online Figure I). Further, post-TAC GRK5gKO mice showed preserved car- diac function as determined by ejection fraction percent (%EF) compared with post-TAC WT mice that had significant LV dys- function (Figure 1E and Online Figure I). We have previously shown that elevated GRK5 plays a critical role in myocytes as an HDAC5 kinase, derepressing hypertrophy-related transcription. 6 Upregulation of generalized hypertrophy markers, including those of the fetal gene program— atrial natriuretic factor (ANF), β-myosin heavy chain (βMHC), and procollagen, type Iα2 (Col1a2)—were significantly attenu- ated in the GRK5gKO mice compared with WT mice 12 weeks after TAC (Figure 1F). Loss of GRK5 expression globally also prevented the post-TAC upregulation of hypertrophic genes direct- ly regulated by MEF2 9 : brain natriuretic peptide (BNP), actin-α1 (Acta-1), and connective tissue growth factor (CTGF) (Figure 1G). Thus, deletion of GRK5 is protective against hypertrophy at the molecular level, consistent with the in vivo phenotype. Grk5gKO Mice Are Resistant to Phenylephrine-Dependent Hypertrophy Phenylephrine (PE), acting through α-adrenergic receptors (αARs), induces cardiomyocyte hypertrophy in vitro and in vivo. Previously, we have found that PE causes GRK5 nuclear trans- location and increased MEF2 activity in myocytes. 6 Therefore, we tested whether endogenous GRK5 was necessary for devel- opment of PE-induced cardiac hypertrophy. Male mice (WT and GRK5gKO) were treated with a subpressor dose of PE (35 mg/kg per day) or phospho-buffered saline (PBS) for 14 days via osmotic minipumps (Figure 2A). This period covers only an initial hypertrophy stage without decompensation, thereby testing the requirement of endogenous GRK5 in myocardial αAR-mediated hypertrophy. PE treatment caused cardiac growth in WT mice, with a significant 26.9±8% increase in HW/BW ratio at 2 weeks, whereas GRK5gKO mice subjected to PE had only an 11.2±7% increase in HW/BW, insignificant compared with PBS-treated GRK5gKO mice (Figure 2B). Two weeks of PE treatment does not change cardiac function, which is what we found for %EF (Figure 2C), although PE significantly altered morphology in WT mice (Figure 2D and 2E). Importantly, PE-treated GRK5gKO mice showed no changes in cardiac dimensions (Figure 2D, 2E). Non-standard Abbreviations and Acronyms AR adrenergic receptor CaMKII calmodulin-dependent kinase II %EF ejection fraction percent GPCR G protein–coupled receptor GRK G protein–coupled receptor kinase HDAC histone deacetylase HF heart failure HW/BW heart weight–to–body weight ratio KO knockout LV left ventricular LVIDs systolic left ventricular internal diameter LVPWT left ventricular posterior wall thickness MEF2 myocyte enhancer factor 2 PBS phospho-buffered saline PE phenylephrine PKD protein kinase D TAC transverse aortic constriction WT wild-type by guest on June 13, 2016 http://circres.ahajournals.org/ Downloaded from
Integrative Physiology Determining the Absolute Requirement of G Protein–Coupled Receptor Kinase 5 for Pathological Cardiac Hypertrophy Short Communication Jessica I. Gold, Erhe Gao, Xiying Shang, Richard T. Premont, Walter J. Koch Rationale: Heart failure (HF) is often the end phase of maladaptive cardiac hypertrophy. A contributing factor is activation of a hypertrophic gene expression program controlled by decreased class II histone deacetylase (HDAC) transcriptional repression via HDAC phosphorylation. Cardiac-specific overexpression of G proteinen–coupled receptor kinase-5 (GRK5) has previously been shown to possess nuclear activity as a HDAC5 kinase, promoting an intolerance to in vivo ventricular pressure overload; however, its endogenous requirement in adaptive and maladaptive hypertrophy remains unknown. Objective: We used mouse models with global or cardiomyocyte-specific GRK5 gene deletion to determine the absolute requirement of endogenous GRK5 for cardiac hypertrophy and HF development after chronic hypertrophic stimuli. Methods and Results: Mice with global deletion of GRK5 were subjected to transverse aortic constriction. At 12 weeks, these mice showed attenuated hypertrophy, remodeling, and hypertrophic gene transcription along with preserved cardiac function. Global GRK5 deletion also diminished hypertrophy and related gene expression due to chronic phenylephrine infusion. We then generated mice with conditional, cardiac-specific deletion of GRK5 that also demonstrated similar protection from pathological cardiac hypertrophy and HF after transverse aortic constriction. Conclusions: These results define myocyte GRK5 as a critical regulator of pathological cardiac growth after ventricular pressure overload, supporting its role as an endogenous (patho)-physiological HDAC kinase. Further, these results define GRK5 as a potential therapeutic target to limit HF development after hypertrophic stress. (Circ Res. 2012;111:1048–1053.) Key Words: G protein–coupled receptor kinases ◼ cardiac hypertrophy ◼ conditional transgenic mouse H eart failure (HF), a leading cause of death in the Western world, often occurs as an end phase of pathological myocardial hypertrophy.1 Hypertrophy is initially an adaptive response to stresses ranging from hypertension, valve disease, or cardiac injury.2,3 In an attempt to normalize wall stress, cardiomyocytes enlarge, sarcomeres reorganize, fibroblasts proliferate, and hypertrophic genes, including the so-called fetal gene program, are upregulated. If prolonged, these responses lead to chamber dilation, myocardial apoptosis, and HF.2,3 In pathological cardiac growth, the molecular pathways affecting transcription lie downstream of the nodal hypertrophic signal transducer, Gq.1 Among these complex pathways are the class II histone deacetylases (HDACs). Physiologically opposed to cardiac growth, the HDACs repress expression of key hypertrophic genes, primarily through inhibition of myocyte enhancer factor 2 (MEF2).3 Genetic deletion of the class II HDAC, HDAC5, sensitizes mice to cardiac stress, whereas murine deletion of MEF2 confers cardioprotection, decreasing pathological hypertrophy and attenuating upregulation of the fetal gene program.4 HDAC kinases control nuclear HDAC activity as phosphorylation of HDAC5 induces its nuclear export and MEF2 derepression.3–5 In This Issue, see p 947 Editorial, see p 957 We recently identified G protein-coupled receptor (GPCR) kinase-5 (GRK5) as a nuclear HDAC kinase, joining protein kinase D (PKD) and calmodulin-dependent kinase II (CaMKII) as HDAC-mediated facilitators of cardiac growth after hypertrophic stimuli.5–7 This represents a novel, non-GPCR cardiac role for this GRK. We showed that mice with cardiac overexpression of GRK5 demonstrate Gq-dependent nuclear translocation of GRK5, where it can phosphorylate HDAC5 and induce Original received May 8, 2012; revision received July 30, 2012; accepted August 1, 2012. In July 2012, the average time from submission to first decision for all original research papers submitted to Circulation Research was 11.2 days. From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.I.G., W.J.K.); Center for Translational Medicine (E.G., X.S., W.J.K.) and the Department of Pharmacology (W.J.K.), Temple University School of Medicine, Philadelphia, PA; and the Department of Medicine, Duke University Medical Center, Durham, NC (R.T.P.). This manuscript was sent to Elizabeth Murphy, Consulting Editor, for review by expert referees, editorial decision, and final disposition. The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA.112. 273367/-/DC1. Correspondence to Walter J. Koch, MD, Center for Translational Medicine, Temple University School of Medicine, 3500 N Broad St, MERB 941, Philadelphia, PA 19140. E-mail Walter.Koch@temple.edu © 2012 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.112.273367 Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 1048 Gold et al Non-standard Abbreviations and Acronyms AR CaMKII %EF GPCR GRK HDAC HF HW/BW KO LV LVIDs LVPWT MEF2 PBS PE PKD TAC WT adrenergic receptor calmodulin-dependent kinase II ejection fraction percent G protein–coupled receptor G protein–coupled receptor kinase histone deacetylase heart failure heart weight–to–body weight ratio knockout left ventricular systolic left ventricular internal diameter left ventricular posterior wall thickness myocyte enhancer factor 2 phospho-buffered saline phenylephrine protein kinase D transverse aortic constriction wild-type MEF2 activity.6 Moreover, cardiac GRK5-overexpressing mice displayed an intolerance to ventricular pressure overload with potentiated maladaptive hypertrophy and accelerated HF after transverse aortic constriction (TAC).6 Importantly, these transgenic studies did not address whether endogenous GRK5 plays a role in the hypertrophic response. In the present study, we used global and cardiomyocytespecific GRK5 knockout (KO) mice to directly address the importance of endogenous GRK5 in cardiac hypertrophy. We found that GRK5 in cardiomyocytes is absolutely required for hypertrophic responses after stress. Further, our data indicate that limiting GRK5 expression in the heart can protect against maladaptive cardiac growth and HF development. Methods Generation of GRK5cKO Mice Detailed mouse protocols are described in the Online Data Supplement Methods. Surgical Procedures and Echocardiography TAC methodology, minipump implantation, and determination of in vivo cardiac function and morphology are described in the Online Data Supplement and Methods. RNA Analysis Methods for RNA analysis are described in the Online Data Supplement. Immunoblot Analysis Detailed methods for heart subfractionation and immunoblotting are described in the Online Data Supplement. Results Global GRK5 Deletion Diminishes In Vivo Cardiac Hypertrophy We subjected male global GRK5 gene knockout mice (GRK5gKO)8 and littermate wild-type (WT) control mice to GRK5 in Pathological Cardiac Hypertrophy 1049 TAC. Constitutive GRK5 deletion attenuated pressure-overloadinduced cardiac growth seen in WT mice at 12 weeks after TAC (Figure 1A). Cardiac dimensions were measured serially by echocardiography to track development of hypertrophy and left ventricular (LV) dilatation over 12 weeks. WT mice showed a quick rise in LV posterior wall thickness (LVPWT), with a peak thickness of 1.99±0.05 mm at 4 weeks after TAC, which then decreased rapidly, indicative of adverse remodeling (Figure 1B). Interestingly, global GRK5 deletion significantly delays the initiation of cardiac hypertrophy after TAC, as GRK5gKO mice do reach a similar LVPWT (1.89±0.09 mm) as WT mice but not until 12 weeks after TAC (Figure 1B). At the end of 12 weeks, GRK5gKO mice had significantly less cardiac hypertrophy as determined by smaller heart weighten-to-enbody weight (HW/ BW) ratios (Figure 1C). Importantly, GRK5gKO mice show no signs of LV dilatation via echocardiographic measurements of systolic LV interior diameter (LVIDs), which were significantly increased in post-TAC WT mice (Figure 1D and Online Figure I). Further, post-TAC GRK5gKO mice showed preserved cardiac function as determined by ejection fraction percent (%EF) compared with post-TAC WT mice that had significant LV dysfunction (Figure 1E and Online Figure I). We have previously shown that elevated GRK5 plays a critical role in myocytes as an HDAC5 kinase, derepressing hypertrophy-related transcription.6 Upregulation of generalized hypertrophy markers, including those of the fetal gene program— atrial natriuretic factor (ANF), β-myosin heavy chain (βMHC), and procollagen, type Iα2 (Col1a2)—were significantly attenuated in the GRK5gKO mice compared with WT mice 12 weeks after TAC (Figure 1F). Loss of GRK5 expression globally also prevented the post-TAC upregulation of hypertrophic genes directly regulated by MEF29: brain natriuretic peptide (BNP), actin-α1 (Acta-1), and connective tissue growth factor (CTGF) (Figure 1G). Thus, deletion of GRK5 is protective against hypertrophy at the molecular level, consistent with the in vivo phenotype. Grk5gKO Mice Are Resistant to Phenylephrine-Dependent Hypertrophy Phenylephrine (PE), acting through α-adrenergic receptors (αARs), induces cardiomyocyte hypertrophy in vitro and in vivo. Previously, we have found that PE causes GRK5 nuclear translocation and increased MEF2 activity in myocytes.6 Therefore, we tested whether endogenous GRK5 was necessary for development of PE-induced cardiac hypertrophy. Male mice (WT and GRK5gKO) were treated with a subpressor dose of PE (35 mg/kg per day) or phospho-buffered saline (PBS) for 14 days via osmotic minipumps (Figure 2A). This period covers only an initial hypertrophy stage without decompensation, thereby testing the requirement of endogenous GRK5 in myocardial αAR-mediated hypertrophy. PE treatment caused cardiac growth in WT mice, with a significant 26.9±8% increase in HW/BW ratio at 2 weeks, whereas GRK5gKO mice subjected to PE had only an 11.2±7% increase in HW/BW, insignificant compared with PBS-treated GRK5gKO mice (Figure 2B). Two weeks of PE treatment does not change cardiac function, which is what we found for %EF (Figure 2C), although PE significantly altered morphology in WT mice (Figure 2D and 2E). Importantly, PE-treated GRK5gKO mice showed no changes in cardiac dimensions (Figure 2D, 2E). Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 1050 Circulation Research September 28, 2012 Figure 1. Attenuated hypertrophy seen in GRK5gKO mice after TAC. A, Hearts from WT and GRKgKO mice subjected to a sham operation or TAC (top). Histological sections stained are with Masson trichrome for ibrosis (middle; bottom). WT sham, n=6; WT TAC, n=9; gKO sham, n=6; gKO TAC, n=11. B, Systolic LVPWT measured serially by echocardiogram after sham or TAC operations. *P<0.05. C, HW/BW ratios were measured 12 weeks after TAC. *P<0.01. LVIDs (D) or %EF (E) as measured by echocardiogram at 12 weeks after TAC. *P<0.01. RT-PCR was used to measure mRNA expression of known markers of cardiac hypertrophy (F) and genes directly regulated by MEF2 (G); n=8, *P<0.05. As with post-TAC, GRK5 deletion significantly decreased PE-mediated upregulation of hypertrophy markers. Indeed, whereas PE led to robustly increased expression of our panel of hypertrophic genes in WT mice, no such upregulation was seen in GRK5gKO mice (Figure 2F and 2G). Overall, these data demonstrate that GRK5 plays a key role in cardiac hypertrophy downstream of PE, identical to post-TAC phenotype. 0.91±0.14; WT sham: 1.02±0.17) (Figure 3A and 3B). Similar results were seen in the non-nuclear fraction from WT mice that had received chronic infusion of PE. These mice showed significantly greater non-nuclear phosphorylated HDAC5 than PE-infused GRK5gKO mice and PBS-infused WT mice (Figure 3C), further reinforcing the role of endogenous GRK5 as an HDAC5 kinase. Global GRK5 Ablation Decreases Nuclear HDAC5 Export After Hypertrophic Stimulus The above results show that complete GRK5 ablation attenuates the cardiac hypertrophic response but does not address the specific role of myocyte GRK5 in maladaptation and post-TAC HF. Therefore, we developed conditional GRK5KO mice in which GRK5 deletion was cardiac-specific. We bred floxedGRK5 mice with transgenic mice expressing Cre-recombinase under control of the αMHC promoter.10 These conditional GRK5 KO (GRK5cKO) mice had >50% loss of cardiac GRK5 determined by either protein immunoblotting or RT-PCR (Online Figure II). As above, we stressed GRK5cKO mice and WT control mice (GRK5floxed) via TAC and studied these groups alongside sham-operated mice for 12 weeks (Figure 4A). Serial echocardiography showed significantly attenuated hypertrophy The diminished upregulation of genetic hypertrophy markers seen in GRK5gKO mice after TAC suggests that GRK5 can regulate cardiac gene transcription. This is probably due to the ability of GRK5 to phosphorylate HDAC5, inducing its export from the nucleus.6 Hence, we examined location and phosphorylation of HDAC5 after hypertrophic stress in GRK5gKO and WT mice. Hearts from the above experiments were subjected to subcellular fractionation. After TAC, WT mice showed a significantly increased amount of phosphorylated HDAC5 in the non-nuclear subcellular fraction compared with GRK5gKO TAC mice and WT sham mice (WT TAC: 1.94±0.13;GRK5gKO TAC: Cardiac-Specific Deletion of GRK5 Attenuates Hypertrophy After TAC Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 Gold et al GRK5 in Pathological Cardiac Hypertrophy 1051 Figure 2. PE induces less hypertrophy in GRK5gKO mice. A, Hearts from WT and GRK5 gKO mice subjected to 2 weeks of chronic infusion of PBS or subpressor PE (35 mg/kg per day) (top). Histological sections are stained with Masson trichrome for ibrosis (middle; bottom). WT PBS, n=8; WT PE, n=10; gKO PBS, n=9; gKO PE, n=11. B, HW/BW ratios for WT and GRK5gKO mice were measured after 2 weeks of chronic PBS or PE infusion. *P<0.05. %EF (C), LVIDs (D), and LVPWT (E) as measured by echocardiogram at 2 weeks. *P<0.05. RT-PCR was used to measure mRNA expression of known markers of cardiac hypertrophy (F) or MEF2-regulated genes (G); n=8, *P<0.05. in GRK5cKO mice with maximally increased cardiac mass at 12-weeks when WT mice are clearly decompensated after their peak hypertrophy 4 weeks after TAC (Figure 4B). This delayed compensatory hypertrophy led to a trend toward lower HW/BW ratio in GRK5cKO mice at 12 weeks after TAC compared with WT mice (Figure 4C). This effect is less robust than the GRK5gKO mice but no doubt due to incomplete GRK5 ablation. However, from in vivo functional studies, it is clear that the myocyte GRK5 loss protects these hearts from adverse LV remodeling and HF, as GRK5cKO mice displayed no increased LV dilatation at the study’s end, compared with significantly increased LVIDs in WT mice (Figure 4D and Online Figure III). This protection against HF was also evident in global in vivo cardiac function as 12-week post-TAC WT mice had a significant loss of LV %EF compared with sham WT mice, while there was absolutely no drop in %EF in 12-week post-TAC GRK5cKO mice (Figure 4E). Again, we examined our panel of hypertrophy-related genes. At 12 weeks after TAC, hearts from GRK5cKO mice showed significantly diminished upregulation of these common cardiac hypertrophy markers, including those of the fetal gene program (Figure 4F), and specific MEF2-regulated genes (Figure 4G). Overall, these data demonstrate that GRK5 expression in cardiomyocytes alone is required for WT molecular, functional, and morphological responses after TAC. Discussion Our results indicate an absolute requirement of cardiomyocyte GRK5 for normal hypertrophic responses and that this kinase plays a critical pathological role in ventricular decompensation and transition to HF after ventricular pressure overload. Importantly, merely decreasing cardiomyocyte GRK5 in mice blunts hypertrophic myocardial growth and prevents HF after TAC. Importantly, these data demonstrate that endogenous myocyte GRK5 plays a crucial role in adaptive and maladaptive hypertrophy and is required for WT response to stress. Thus, increased GRK5 in the failing human heart11 has pathological significance, since lowering GRK5 or inhibiting its activity appears to offer novel beneficial effects against maladaptive cardiac growth. These results in global and cardiac-specific GRK5KO mice, coupled with our previous results showing that GPCR-independent nuclear activity of GRK5 can facilitate hypertrophy,6 indicate that nuclear targeting of endogenous GRK5 as a class II HDAC kinase is physiologically significant in normal and abnormal cardiac growth after stress. Interestingly, loss of nuclear GRK5 activity can delay HF onset through slower cardiac growth, although its deletion does not completely ameliorate hypertrophy. This may be due to compensatory and discrete roles of the other known HDAC5 kinases, CAMKII, and PKD, Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 1052 Circulation Research September 28, 2012 Figure 3. Global GRK5 ablation decreases phosphorylated HDAC5 in the cytoplasm. A, Twelve weeks after TAC or a sham operation, hearts from WT and GRK5gKO mice were fractionated into non-nuclear and nuclear fractions. HDAC5 was immunoprecipitated from the non-nuclear fraction and immunoblotted for phosphorylated and total amounts (pHDAC5 and tHDAC5, respectively). B, Denistometric quantiication of pHDAC5 normalized to tHDAC. *P<0.01 versus all groups, n=6. C, Denistometric quantiication of pHDAC5 normalized to tHDAC5 after immunoprecipitation and immunoblotting for pHDAC5 in WT and GRK5gKO mouse hearts after 2 weeks of chronic PBS or PE infusion. *P<0.01 versus all groups, n=6. acting downstream of hypertrophic signaling.5,7 Each kinase has been shown to cause HDAC5 nuclear export after select receptor activation such as endothelin-1 receptor for CAMKII and αAR for PKD.5,12 We now can add GRK5 to the list of physiological HDAC kinases downstream of TAC and αAR stimulation. These distinct activators of HDAC kinases appear to underlie a complex network of parallel signaling converging on the same target, HDAC5. Overall, our data demonstrate that targeting only 1 Figure 4. Attenuated hypertrophy seen in GRK5cKO mice after TAC. A, Hearts from WT and GRK5cKO mice subjected to a sham operation or TAC (top). Histological sections are stained with Masson trichrome for ibrosis (middle; bottom). WT sham, n=9; WT TAC, n=14; cKO sham, n=8; cKO TAC, n=9. B, Systolic LVPWT measured serially by echocardiogram after sham or TAC operations. *P<0.05. C, HW/BW ratios for WT and GRK5cKO mice were measured 12 weeks after TAC. LVIDs (D) or %EF (E) as measured by echocardiogram at 12 weeks after TAC. *P<0.01. RT-PCR was used to measure mRNA expression of known markers of cardiac hypertrophy (F) and MEF2-regulated genes (G); n=8, *P<0.05. Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 Gold et al HDAC kinase can delay HF and may be advantageous in therapeutic intervention by allowing some signaling. Because GRK5 plays a dual role in the cardiomyocyte— membrane-associated GPCR desensitizing kinase and nuclear kinase facilitating transcription—the most critical activities for cardiac signaling and function are uncertain. Classic GRK5 activity toward β-adrenergic receptors (βARs) has been shown to induce transactivation of the cardioprotective epidermal growth factor receptor.13 A human polymorphism of GRK5 (Q41L) appears to increase βAR desensitization, protecting some HF patients chronically.14 Additionally, transgenic overexpression of Gαq caused a slight cardiac dilatation in GRK5gKO mice compared with WT controls.15 Therefore, there is some question as to whether the increased GRK5 is protective or injurious. In the present study, simply decreasing myocyte GRK5, either completely in GRK5gKO mice or significantly in GRK5cKO mice, attenuated cardiac hypertrophy and prevented pathogenesis of HF. We show that GRK5 ablation does not completely prevent hypertrophy but significantly delays it. Therefore, despite potential beneficial activities at the plasma membrane, removing cardiomyocyte GRK5 has a profound positive effect on outcomes after pressure overload and chronic α-adrenergic stress. Clearly, our data now indicate that GRK5, and, most likely, its nuclear HDAC kinase activity, represent a novel target to prevent maladaptive cardiac hypertrophy and protect against ventricular decompensation and HF. Acknowledgments We thank Zuping Qiu for excellent technical support. Sources of Funding This research was supported in part by National Institutes of Health grants P01 HL091799 (to W.J.K.) and P01 HL07544 Project 2 (to W.J.K.) and a Pre-Doctoral Fellowship Award from the American Heart Association Great Rivers Affiliate (to J.I.G.). Disclosures None. References 1. Dorn GW II, Force T. Protein kinase cascades in the regulation of cardiac hypertrophy. J Clin Invest. 2005;115:527–537. GRK5 in Pathological Cardiac Hypertrophy 1053 2. Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signaling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600. 3. McKinsey TA, Olson EN. Toward transcriptional therapies for the failing heart: chemical screens to modulate genes. J Clin Invest. 2005;115:538–546. 4. Chang S, McKinsey TA, Zhang CL, Richardson JA, Hill JA, Olson EN. Histone deacetylases 5 and 9 govern responsiveness of the heart to a subset of stress signals and play redundant roles in heart development. Mol Cell Biol. 2004;24:8467–8476. 5. Wu X, Zhang T, Bossuyt J, Li X, McKinsey TA, Dedman JR, Olson EN, Chen J, Brown JH, Bers DM. Local INSP3-dependent perinuclear Ca2+ signaling in cardiac myocyte excitation-transcription coupling. J Clin Invest. 2006;116:675–682. 6. Martini JS, Raake P, Vinge LE, DeGeorge BR Jr, Chuprun JK, Harris DM, Gao E, Eckhart AD, Pitcher JA, Koch WJ. Uncovering G proteinencoupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proc Natl Acad Sci U S A. 2008;105:12457–12462. 7. Vega RB, Harrison BC, Meadows E, Roberts CR, Papst PJ, Olson EN, McKinsey TA. Protein kinases C and D mediate agonist-dependent cardiac hypertrophy through nuclear export of histone deacetylase 5. Mol Cell Biol. 2004;24:8374–8385. 8. Gainetdinov RR, Bohn LM, Walker JK, Laporte SA, Macrae AD, Caron MG, Lefkowitz RJ, Premont RT. Muscarinic supersensitivity and impaired receptor desensitization in G protein–coupled receptor kinase 5-deficient mice. Neuron. 1999;24:1029–1036. 9. van Oort RJ, van Rooij E, Bourajjaj M, Schimmel J, Jansen MA, van der Nagel R, Doevendans PA, Schneider MD, van Echteld CJ, De Windt LJ. Mef2 activates a genetic program promoting chamber dilation and contractile dysfunction in calcineurin-induced heart failure. Circulation. 2006;114:298–308. 10. Raake PW, Vinge LE, Gao E, Boucher M, Rengo G, Chen X, DeGeorge BR Jr, Matkovich S, Houser SR, Most P, Eckhart AD, Dorn GW II, Koch WJ. G protein-ncoupled receptor kinase 2 ablation in cardiac myocytes before or after myocardial infarction prevents heart failure. Circ Res. 2008;103:413–422. 11. Dzimiri N, Muiya P, Andres E, Al-Halees Z. Differential functional expression of human myocardial g protein receptor kinases in left ventricular cardiac diseases. Eur J Pharmacol. 2004;489:167–177. 12. Bossuyt J, Chang CW, Helmstadter K, Kunkel MT, Newton AC, Campbell KS, Martin JL, Bossuyt S, Robia SL, Bers DM. Spatiotemporally distinct protein kinase d activation in adult cardiomyocytes in response to phenylephrine and endothelin. J Biol Chem. 2011;286:33390–33400. 13. 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. Betaarrestin-mediated beta1-adrenergic receptor transactivation of the EGFR confers cardioprotection. J Clin Invest. 2007;117:2445–2458. 14. Liggett SB, Cresci S, Kelly RJ, Syed FM, Matkovich SJ, Hahn HS, Diwan A, Martini JS, Sparks L, Parekh RR, Spertus JA, Koch WJ, Kardia SL, Dorn GW II. A grk5 polymorphism that inhibits beta-adrenergic receptor signaling is protective in heart failure. Nat Med. 2008;14:510–517. 15. Zhang Y, Matkovich SJ, Duan X, Gold JI, Koch WJ, Dorn GW II. Nuclear effects of G-protein receptor kinase 5 on histone deacetylase 5-regulated gene transcription in heart failure. Circ Heart Fail. 2001;4:659–668. Novelty and Significance What Is Known? • G protein-ncoupled receptor kinase 5 (GRK5) is upregulated in numerous models of heart failure (HF), as well as in the failing human heart. • GRK5 enters the nucleus and acts as a histone deacetylase 5 (HDAC5) kinase, increasing transcription of cardiac hypertrophy genes. • Increased nuclear GRK5 is pathological in the setting of chronic pressure overload. What New Information Does This Article Contribute? • Ablation of GRK5 significantly delays maladaptive cardiac remodeling and HF after chronic pressure overload or α-adrenergic receptor (αAR) stimulation. • Removing nuclear GRK5 by global ablation decreases HDAC5 export. • Deletion of GRK5 in cardiomyocytes alone significantly delays the onset of HF. Pathological cardiac hypertrophy, a process commonly ending in HF, occurs through activation of nodal signal transducer, Gq. Downstream of Gq, class II HDACs represses hypertrophic gene transcription. GRK5, a recently identified HDAC kinase, has been shown to be upregulated in human HF, although the is unknown. Here, we investigated the role of endogenous GRK5 in maladaptive cardiac remodeling. Our results show that global and cardiomyocyte-specific ablation of GRK5 significantly attenuates pathological cardiac hypertrophy, delaying HF onset. This cardioprotection may be attributed to decreased nuclear HDAC5 export after GRK5 deletion. Overall, this study suggests that endogenous nuclear GRK5 plays an injurious role in maladaptive hypertrophy and may represent a novel therapeutic target. Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 Determining the Absolute Requirement of G Protein−Coupled Receptor Kinase 5 for Pathological Cardiac Hypertrophy: Short Communication Jessica I. Gold, Erhe Gao, Xiying Shang, Richard T. Premont and Walter J. Koch Circ Res. 2012;111:1048-1053; originally published online August 2, 2012; doi: 10.1161/CIRCRESAHA.112.273367 Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Copyright © 2012 American Heart Association, Inc. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571 The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/content/111/8/1048 Data Supplement (unedited) at: http://circres.ahajournals.org/content/suppl/2012/08/02/CIRCRESAHA.112.273367.DC1.html Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: http://www.lww.com/reprints Subscriptions: Information about subscribing to Circulation Research is online at: http://circres.ahajournals.org//subscriptions/ Downloaded from http://circres.ahajournals.org/ by guest on June 13, 2016 Supporting Information Materials and Methods Generation of Conditional GRK5 knockout mice. . All animal procedures and experiments were performed in accordance with the guidelines of the Institutional Animal Care and Use Committee of Thomas Jefferson University. The GRK5/flox line was created as described in Gainetdinov et al, 19991, except that a Cre transfected, GRK5targeted ES cell line was selected for deletion of the TK-NEO marker cassette but maintenance of the exon 7/8 region of the GRK5 gene flanked by loxP sites. These GRK5/flox cells were microinjected into day 3.5 blastocysts (c57BL/6J) and implanted into pseudopregnant mice to generate founder chimeric pups. After germline transmission was verified, GRK5/flox mice were backcrossed with c57BL/6J mice for >12 generations. An initial cross between GRK5flox/flox and αMHC-Cre mice led to a cardiac-specific GRK5 deletion. This conditional deletion was maintained by continued breeding of the GRK5flox/flox with GRK5flox/flox/αMHC-Cre+/-. Only 8 week-old males from the above cross were utilized in our experiments. Genotyping was performed using specific PCR primers designed for the loxP site. TAC. Transverse aortic constriction was performed as described previously2. Briefly, 8 week-old mice were sedated in an isoflurane sedation box (induction 3% and maintenance 5%) and anesthetized to a surgical plane with an i.p. dose of ketamine (50mg/kg) and xylazine (2.5mg/kg). Anesthetized mice were intubated using a blunt 20gauge needed that was connected to a volume-cycle rodent ventilator on supplemental oxygen at a rate of 1 L/min and respiratory rate of 140 bpm/min. A midline cervical incision was made to expose the trachea, carotid arteries and rib cage. Aortic constriction was performed by tying a 7.0 nylon suture ligature against a 27-gauge needle that was promptly removed to yield a 0.4 mm constriction. Pressure gradients were determined by in vivo echocardiography of the transverse aorta and mice with gradients greater than 30 mmHg were used. Echocardiography. Echocardiography was performed as previously described3. To measure global cardiac function, echocardiography was performed at 8 weeks of age prior to TAC and at 1, 2, 4, 8, and 12 weeks post-TAC by use of the VisualSonics VeVo 770 imagingsystem with a 707 scan head in anesthetized animals (1.5% isoflurane, vol/vol). The internal diameter of the left ventricle was measure in the short-axis view from M-mode recordings in end diastole and end systole. At 1-week post-TAC, pressure gradients were determined by PW Doppler mode. Mini-osmotic pumps. Chronic infusion of phenylephrine (Sigma) was achieved using Alzet 2 week osmotic minipumps (model 2002, DURECT Corporation). Pumps were filled following the manufacturer’s specifications with sterile PBS, or PE (35µM/kg/day). Briefly, Mice were anesthetized with isofluorane (2.5% vol/vol) and pumps were implanted subcutaneously through a sub-scapular incision, which was then closed using 4.0 silk suture (Ethicon). The contents of the pumps were delivered at a rate of 0.5µl/hour for 2 weeks. Mice were followed by echocardiogram prior to pump implantation and then weekly until the end of the 2 week period when mice were euthanized. RNA Isolation and Semiquantitative PCR. RNA isolation and analysis was performed as previously described2. Total RNA isolation was performed using the Ultraspec RNA Isolation System (Biotecx), according to the manufacturer’s protocol and as previously 1 described2. After RNA isolation, cDNA was synthesized from 200ng of total RNA using the iScript cDNA Synthesis Kit (Bio-Rad Laboratories). Semiquantitative PCR was carried out on cDNA using iQ SYBR Green Supermix (Bio-Rad Laboratories) and 100 nM of the following gene-specific oligonucleotides—18S, ANF, βMHC, BNP, Col1a2, CTGF, and Acta-1. Quantitation was established by comparing 18s rRNA, which was similar between groups, for normalization. For each run, saturation of amplification cycles was controlled by the use of MyiQ software (version 1.0); subsequently, a melting curve was generated by heating the product to 95°C, cooling to and maintaining at 55°C for 20 seconds, and then slowly (0.5°C/s) heating to 95°C to determine the specificity of PCR products, which was then confirmed by gel electrophoresis. Immunoblot Analysis. Isolated cardiac tissue was first homogenized using a Dounce homogenizer in a buffer containing: 4mM Hepes, 320mM sucrose, 10mM KCL, 5mM EDTA, 2mg NaF, 8mg MgCl2, .1% Triton x-10, 1.094g DTT, and protease inhibitors. The homogenate was filtered through a 70µm cell filter. Total cell lysate was taken at this point. Then the lysate was subjected the same protocol as previously described for NRVM2. Following subcellular fractionation, 500 µg of non-nuclear protein was immunoprecipitated with protein A/G and anti-HDAC5 (sc-133106, Santa Cruz). Proteins were subjected to SDS-PAGE, and immunoblotted with antiphospho-HDAC5 (PA114187, Thermo Scientific), and anti-HDAC5 (sc-133106, Santa Cruz). Visualization of Western blot signals was performed using secondary antibodies coupled to Alexa Fluor 680 or 800 (Molecular Probes) on a LI-COR infrared imager (Odyssey). Pictures were processed by Odyssey version 1.2 infrared imaging software. All densitometry scans were carried out in the linear range of detection. Statistics. All the values in the text and figures are presented as mean ± SEM from given n sizes. Statistical significance of multiple treatments was determined by one-way ANOVA followed by the Bonferroni’s post hoc test when appropriate. Statistical significance between two groups was determined using the two-tailed Student’s t test. P values of <0.05 were considered significant. Supplemental References 1. Gainetdinov RR, Bohn LM, Walker JK, Laporte SA, Macrae AD, Caron MG, Lefkowitz RJ, Premont RT. 1999. Muscarinic supersensitivity and impaired receptor desensitization in G protein-coupled receptor kinase 5-deficient mice. Neuron. 24:1029-36 2. Martini JS, Raake P, Vinge LE, DeGeorge BR, Jr., Chuprun JK, Harris DM, Gao E, Eckhart AD, Pitcher JA, Koch WJ. 2008. Uncovering G protein-coupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proceedings of the National Academy of Sciences of the United States of America. 105:12457-62 3. Brinks H, Boucher M, Gao E, Chuprun JK, Pesant S, Raake PW, Huang ZM, Wang X, Qiu G, Gumpert A, Harris DM, Eckhart AD, Most P, Koch WJ. 2010. Level of Gprotein-coupled receptor kinase-2 determines myocardial ischemia/reperfusion injury via pro- and anti-apoptotic mechanisms. Circulation research. 107:1140-9 2 Online Supplemental Figure Legends Online Fig. I Representative Echocardiogram tracings from WT and GRK5gKO mice 12 weeks following sham or TAC operations Online Fig. II GRK5Flox/αMHC-Cre+ mice have a greater than 50% knockdown of GRK5. (A) To determine GRK5 expression in our αMHC-Cre system, hearts from GRK5Flox/αMHC-Cre- (WT) and GRK5Flox/αMHC-Cre+ (cKO) were homogenized and immunoblotted for GRK5 and GAPDH. (B) Quantitative analysis of immunoblots for GRK5 expression in the hearts of (A). n=4, *p<0.01. (C) Semi-quantitative PCR was used to assess GRK5 mRNA levels in the hearts of WT or cKO mice. n=6, *p<0.05 Online Fig. III Representative Echocardiogram tracings from WT and GRK5cKO mice 12 weeks following sham or TAC operations. 3 Online Supplemental Figure I 4 Online Supplemental Figure II 5 Online Supplemental Figure III 6 7