scholarly journals G protein-coupled receptor kinase 5 (GRK5) contributes to impaired cardiac function and immune cell recruitment in post-ischemic heart failure

Author(s):  
Claudio de Lucia ◽  
Laurel A Grisanti ◽  
Giulia Borghetti ◽  
Michela Piedepalumbo ◽  
Jessica Ibetti ◽  
...  

Abstract Aims  Myocardial infarction (MI) is the most common cause of heart failure (HF) worldwide. G protein-coupled receptor kinase 5 (GRK5) is upregulated in failing human myocardium and promotes maladaptive cardiac hypertrophy in animal models. However, the role of GRK5 in ischemic heart disease is still unknown. In this study, we evaluated whether myocardial GRK5 plays a critical role post-MI in mice and included the examination of specific cardiac immune and inflammatory responses. Methods and results  Cardiomyocyte-specific GRK5 overexpressing transgenic mice (TgGRK5) and non-transgenic littermate control (NLC) mice as well as cardiomyocyte-specific GRK5 knockout mice (GRK5cKO) and wild type (WT) were subjected to MI and, functional as well as structural changes together with outcomes were studied. TgGRK5 post-MI mice showed decreased cardiac function, augmented left ventricular dimension and decreased survival rate compared to NLC post-MI mice. Cardiac hypertrophy and fibrosis as well as fetal gene expression were increased post-MI in TgGRK5 compared to NLC mice. In TgGRK5 mice, GRK5 elevation produced immuno-regulators that contributed to the elevated and long-lasting leukocyte recruitment into the injured heart and ultimately to chronic cardiac inflammation. We found an increased presence of pro-inflammatory neutrophils and macrophages as well as neutrophils, macrophages and T-lymphocytes at 4-days and 8-weeks respectively post-MI in TgGRK5 hearts. Conversely, GRK5cKO mice were protected from ischemic injury and showed reduced early immune cell recruitment (predominantly monocytes) to the heart, improved contractility and reduced mortality compared to WT post-MI mice. Interestingly, cardiomyocyte-specific GRK2 transgenic mice did not share the same phenotype of TgGRK5 mice and did not have increased cardiac leukocyte migration and cytokine or chemokine production post-MI. Conclusions  Our study shows that myocyte GRK5 has a crucial and GRK-selective role on the regulation of leucocyte infiltration into the heart, cardiac function and survival in a murine model of post-ischemic HF, supporting GRK5 inhibition as a therapeutic target for HF.

Author(s):  
Liangpeng Li ◽  
Wenbin Fu ◽  
Xue Gong ◽  
Zhi Chen ◽  
Luxun Tang ◽  
...  

Abstract Aims G protein-coupled receptor kinase 4 (GRK4) has been reported to play an important role in hypertension, but little is known about its role in cardiomyocytes and myocardial infarction (MI). The goal of present study is to explore the role of GRK4 in the pathogenesis and progression of MI. Methods and results We studied the expression and distribution pattern of GRK4 in mouse heart after MI. GRK4 A486V transgenic mice, inducible cardiomyocyte-specific GRK4 knockout mice, were generated and subjected to MI with their control mice. Cardiac infarction, cardiac function, cardiomyocyte apoptosis, autophagic activity, and HDAC4 phosphorylation were assessed. The mRNA and protein levels of GRK4 in the heart were increased after MI. Transgenic mice with the overexpression of human GRK4 wild type (WT) or human GRK4 A486V variant had increased cardiac infarction, exaggerated cardiac dysfunction and remodelling. In contrast, the MI-induced cardiac dysfunction and remodelling were ameliorated in cardiomyocyte-specific GRK4 knockout mice. GRK4 overexpression in cardiomyocytes aggravated apoptosis, repressed autophagy, and decreased beclin-1 expression, which were partially rescued by the autophagy agonist rapamycin. MI also induced the nuclear translocation of GRK4, which inhibited autophagy by increasing HDAC4 phosphorylation and decreasing its binding to the beclin-1 promoter. HDAC4 S632A mutation partially restored the GRK4-induced inhibition of autophagy. MI caused greater impairment of cardiac function in patients carrying the GRK4 A486V variant than in WT carriers. Conclusion GRK4 increases cardiomyocyte injury during MI by inhibiting autophagy and promoting cardiomyocyte apoptosis. These effects are mediated by the phosphorylation of HDAC4 and a decrease in beclin-1 expression.


2005 ◽  
Vol 66 (3) ◽  
pp. 512-519 ◽  
Author(s):  
K LEINEWEBER ◽  
P ROHE ◽  
A BEILFUS ◽  
C WOLF ◽  
H SPORKMANN ◽  
...  

2012 ◽  
Vol 9 (4) ◽  
pp. e155-e162 ◽  
Author(s):  
Sarah M. Schumacher-Bass ◽  
Christopher J. Traynham ◽  
Walter J. Koch

2016 ◽  
Vol 118 (7) ◽  
pp. 1116-1124 ◽  
Author(s):  
Giuseppe Rengo ◽  
Gennaro Pagano ◽  
Pasquale Perrone Filardi ◽  
Grazia Daniela Femminella ◽  
Valentina Parisi ◽  
...  

2013 ◽  
Vol 34 (suppl 1) ◽  
pp. P1486-P1486
Author(s):  
G. Rengo ◽  
G. Pagano ◽  
G. Galasso ◽  
G. D. Femminella ◽  
D. Liccardo ◽  
...  

2018 ◽  
Vol 50 (6) ◽  
pp. 407-415 ◽  
Author(s):  
Michael D. Steury ◽  
Ho Jun Kang ◽  
Taehyung Lee ◽  
Peter C. Lucas ◽  
Laura R. McCabe ◽  
...  

G protein-coupled receptor kinase 2 (GRK2) is a serine/threonine kinase and plays a key role in different disease processes. Previously, we showed that GRK2 knockdown enhances wound healing in colonic epithelial cells. Therefore, we hypothesized that ablation of GRK2 would protect mice from dextran sodium sulfate (DSS)-induced acute colitis. To test this, we administered DSS to wild-type (GRK2+/+) and GRK2 heterozygous (GRK+/−) mice in their drinking water for 7 days. As predicted, GRK2+/− mice were protected from colitis as demonstrated by decreased weight loss (20% loss in GRK2+/+ vs. 11% loss in GRK2+/−). lower disease activity index (GRK2+/+ 9.1 vs GRK2+/− 4.1), and increased colon lengths (GRK2+/+ 4.7 cm vs GRK2+/− 5.3 cm). To examine the mechanisms by which GRK2+/− mice are protected from colitis, we investigated expression of inflammatory genes in the colon as well as immune cell profiles in colonic lamina propria, mesenteric lymph node, and in bone marrow. Our results did not reveal differences in immune cell profiles between the two genotypes. However, expression of inflammatory genes was significantly decreased in DSS-treated GRK2+/− mice compared with GRK2+/+. To understand the mechanisms, we generated myeloid-specific GRK2 knockout mice and subjected them to DSS-induced colitis. Similar to whole body GRK2 heterozygous knockout mice, myeloid-specific knockout of GRK2 was sufficient for the protection from DSS-induced colitis. Together our results indicate that deficiency of GRK2 protects mice from DSS-induced colitis and further suggests that the mechanism of this effect is likely via GRK2 regulation of inflammatory genes in the myeloid cells.


Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Liam M Casey ◽  
Jason Greenman ◽  
Frederick Aguilar ◽  
Olga Dunaevsky ◽  
Burns C Blaxall

G protein-coupled receptors (GPCRs) play crucial roles in normal heart function and dysregulated GPCR signaling contributes to heart failure (HF). Protease-activated receptors (PARs) are one class of GPCR expressed in the heart. Emerging evidence demonstrates that excessive PAR-1 signaling induces cardiac dysfunction. An important component of PAR signaling is ERK1/2, which is phosphorylated in response to PAR stimulation and can influence myocyte hypertrophy and survival. Cytoplasmic pERK1/2 accumulation depends in part on scaffolding complexes that assemble with internalized GPCRs. Activation of an internalization-defective PAR mutant leads to enhanced nuclear pERK1/2 accumulation upon stimulation. The nuclear/cytoplasmic distribution of pERK1/2 may be a key factor in determining the cellular effects of PAR stimulation as elevated nuclear pERK in cardiomyocytes is suggested to promote survival and physiological hypertrophy. Phosphorylation of PARs by G-protein coupled receptor kinase 3 (GRK3) is thought to promote receptor internalization, potentially influencing overall pERK1/2 accumulation and subcellular distribution. We have used a dominant negative form of GRK3 lacking the kinase domain (GRK3ct) to test the hypothesis that GRK3 influences PAR1 internalization and ERK1/2 phosphorylation. By measuring cell surface receptor levels we demonstrate that GRK3ct interferes with PAR1 internalization. Immunofluorescent staining and cellular fractionation techniques further show that GRK3ct enhances nuclear accumulation of pERK1/2 in COS-7 cells and adult mouse cardiomyocytes. Furthermore we find that GRK3ct overexpression in neonatal rat cardiomyocytes increases PAR1-activation induced physiologic hypertrophy. In summary these results may explain recent unpublished reports that mice overexpressing GRK3ct in the heart are protected against ischemic injury, a heart failure model that involves pathologic PAR signaling. Thus we conclude that following ischemic injury, reducing PAR1 internalization via interfering with endogenous GRK3 activity or promoting nuclear pERK accumulation might improve cardiac recovery.


Sign in / Sign up

Export Citation Format

Share Document