Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Am J Physiol Heart Circ Physiol ; 318(5): H1162-H1175, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32216616

ABSTRACT

Nitric oxide (NO) and S-nitrosothiol (SNO) are considered cardio- and vasoprotective substances. We now understand that one mechanism in which NO/SNOs provide cardiovascular protection is through their direct inhibition of cardiac G protein-coupled receptor (GPCR) kinase 2 (GRK2) activity via S-nitrosylation of GRK2 at cysteine 340 (C340). This maintains GPCR homeostasis, including ß-adrenergic receptors, through curbing receptor GRK2-mediated desensitization. Previously, we have developed a knockin mouse (GRK2-C340S) where endogenous GRK2 is resistant to dynamic S-nitrosylation, which led to increased GRK2 desensitizing activity. This unchecked regulation of cardiac GRK2 activity resulted in significantly more myocardial damage after ischemic injury that was resistant to NO-mediated cardioprotection. Although young adult GRK2-C340S mice show no overt phenotype, we now report that as these mice age, they develop significant cardiovascular dysfunction due to the loss of SNO-mediated GRK2 regulation. This pathological phenotype is apparent as early as 12 mo of age and includes reduced cardiac function, increased cardiac perivascular fibrosis, and maladaptive cardiac hypertrophy, which are common maladies found in patients with cardiovascular disease (CVD). There are also vascular reactivity and aortic abnormalities present in these mice. Therefore, our data demonstrate that a chronic and global increase in GRK2 activity is sufficient to cause cardiovascular remodeling and dysfunction, likely due to GRK2's desensitizing effects in several tissues. Because GRK2 levels have been reported to be elevated in elderly CVD patients, GRK2-C340 mice can give insight into the aged-molecular landscape leading to CVD.NEW & NOTEWORTHY Research on G protein-coupled receptor kinase 2 (GRK2) in the setting of cardiovascular aging is largely unknown despite its strong established functions in cardiovascular physiology and pathophysiology. This study uses a mouse model of chronic GRK2 overactivity to further investigate the consequences of long-term GRK2 on cardiac function and structure. We report for the first time that chronic GRK2 overactivity was able to cause cardiac dysfunction and remodeling independent of surgical intervention, highlighting the importance of GRK activity in aged-related heart disease.


Subject(s)
Aging/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Heart Diseases/etiology , Heart/physiology , Myocardium/metabolism , Nitric Oxide/metabolism , Aging/metabolism , Animals , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Heart/growth & development , Heart/physiopathology , Heart Diseases/metabolism , Homeostasis , Male , Mice , Mutation
3.
JCI Insight ; 52019 08 08.
Article in English | MEDLINE | ID: mdl-31393858

ABSTRACT

Dual peroxisome proliferator-activated receptor (PPAR)α/γ agonists that were developed to target hyperlipidemia and hyperglycemia in type 2 diabetes patients, caused cardiac dysfunction or other adverse effects. We studied the mechanisms that underlie the cardiotoxic effects of a dual PPARα/γ agonist, tesaglitazar, in wild type and diabetic (leptin receptor deficient - db/db) mice. Mice treated with tesaglitazar-containing chow or high fat diet developed cardiac dysfunction despite lower plasma triglycerides and glucose levels. Expression of cardiac peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), which promotes mitochondrial biogenesis, had the most profound reduction among various fatty acid metabolism genes. Furthermore, we observed increased acetylation of PGC1α, which suggests PGC1α inhibition and lowered sirtuin 1 (SIRT1) expression. This change was associated with lower mitochondrial abundance. Combined pharmacological activation of PPARα and PPARγ in C57BL/6 mice reproduced the reduction of PGC1α expression and mitochondrial abundance. Resveratrol-mediated SIRT1 activation attenuated tesaglitazar-induced cardiac dysfunction and corrected myocardial mitochondrial respiration in C57BL/6 and diabetic mice but not in cardiomyocyte-specific Sirt1-/- mice. Our data shows that drugs, which activate both PPARα and PPARγ lead to cardiac dysfunction associated with PGC1α suppression and lower mitochondrial abundance likely due to competition between these two transcription factors.


Subject(s)
Heart Failure/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisomes/metabolism , Sirtuin 1/metabolism , Alkanesulfonates/adverse effects , Animals , Blood Glucose , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , PPAR alpha/agonists , PPAR gamma/agonists , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phenylpropionates/adverse effects , Receptors, Leptin/metabolism , Sirtuin 1/genetics , Transcription Factors , Transcriptome
4.
Expert Opin Ther Targets ; 23(3): 201-214, 2019 03.
Article in English | MEDLINE | ID: mdl-30701991

ABSTRACT

INTRODUCTION: One in every four deaths in the United States is attributed to cardiovascular disease, hence the development and employment of novel and effective therapeutics are necessary to improve the quality of life and survival of affected patient. Pathological hypertrophy is a maladaptive response by the heart to relieve wall stress that could result from cardiovascular disease. Maladaptive hypertrophy can lead to further disease progression and complications such as heart failure; hence, efforts to target hypertrophy to prevent and treat further morbidity and mortality are necessary. Areas covered: This review summarizes the compelling literature that describes the mechanistic role of GRK2 and GRK5 in maladaptive cardiac hypertrophy; it examines the approaches to inhibit these kinases in hypertrophic animal models and furthermore, it assesses the potential of GRK2 and GRK5 as therapeutic targets for hypertrophy. Expert opinion: GRK2 and GRK5 are novel therapeutic targets for pathological hypertrophy and may have added benefits of ameliorating morbidity and mortality. Despite the lesser researched role of GRK2 in cardiac hypertrophy, it may be the advantageous strategy for treating cardiac hypertrophy because of its role in other maladaptive pathways. Anti-GRK2 therapy optimization and the discovery and development of specific GRK2 and GRK5 small-molecule inhibitors is necessary for the eventual application of successful, effective therapeutics.


Subject(s)
Cardiomegaly/drug therapy , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 5/antagonists & inhibitors , Animals , Cardiomegaly/complications , Cardiomegaly/physiopathology , Disease Models, Animal , Disease Progression , Drug Development , Drug Discovery , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Humans
5.
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
6.
PPAR Res ; 2015: 835985, 2015.
Article in English | MEDLINE | ID: mdl-26713088

ABSTRACT

Over 5 million people in the United States suffer from the complications of heart failure (HF), which is a rapidly expanding health complication. Disorders that contribute to HF include ischemic cardiac disease, cardiomyopathies, and hypertension. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family. There are three PPAR isoforms: PPARα, PPARγ, and PPARδ. They can be activated by endogenous ligands, such as fatty acids, as well as by pharmacologic agents. Activators of PPARs are used for treating several metabolic complications, such as diabetes and hyperlipidemia that are directly or indirectly associated with HF. However, some of these drugs have adverse effects that compromise cardiac function. This review article aims to summarize the current basic and clinical research findings of the beneficial or detrimental effects of PPAR biology on myocardial function.

7.
Clin Cancer Res ; 21(7): 1652-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25617424

ABSTRACT

PURPOSE: To investigate the roles of melanoma-associated macrophages in melanoma resistance to BRAF inhibitors (BRAFi). EXPERIMENTAL DESIGN: An in vitro macrophage and melanoma cell coculture system was used to investigate whether macrophages play a role in melanoma resistance to BRAFi. The effects of macrophages in tumor resistance were examined by proliferation assay, cell death assay, and Western blot analyses. Furthermore, two mouse preclinical models were used to validate whether targeting macrophages can increase the antitumor activity of BRAFi. Finally, the number of macrophages in melanoma tissues was examined by immunohistochemistry. RESULTS: We demonstrate that in BRAF-mutant melanomas, BRAFi paradoxically activate the mitogen-activated protein kinase (MAPK) pathway in macrophages to produce VEGF, which reactivates the MAPK pathway and stimulates cell growth in melanoma cells. Blocking the MAPK pathway or VEGF signaling then reverses macrophage-mediated resistance. Targeting macrophages increases the antitumor activity of BRAFi in mouse and human tumor models. The presence of macrophages in melanomas predicts early relapse after therapy. CONCLUSIONS: Our findings demonstrate that macrophages play a critical role in melanoma resistance to BRAFi, suggesting that targeting macrophages will benefit patients with BRAF-mutant melanoma.


Subject(s)
Drug Resistance, Neoplasm/immunology , Macrophages/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Blotting, Western , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Macrophages/drug effects , Macrophages/immunology , Melanoma/immunology , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy/adverse effects , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering , Transfection , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
8.
J Invest Dermatol ; 134(4): 1108-1118, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24157461

ABSTRACT

Tumor-associated macrophages (TAMs) have essential roles in tumor progression and metastasis. Tumor cells recruit myeloid progenitors and monocytes to the tumor site, where they differentiate into TAMs; however, this process is not well studied in humans. Here we show that human CD7, a T-cell and NK cell receptor, is highly expressed by monocytes and macrophages. Expression of CD7 decreases in M-CSF-differentiated macrophages and in melanoma-conditioned medium-induced macrophages (MCMI/Mφ) in comparison to monocytes. A ligand for CD7, SECTM1 (secreted and transmembrane protein 1), is highly expressed in many tumors, including melanoma cells. We show that SECTM1 binds to CD7 and significantly increases monocyte migration by activation of the PI3K (phosphatidylinositol 3'-kinase) pathway. In human melanoma tissues, tumor-infiltrating macrophages expressing CD7 are present. These melanomas, with CD7-positive inflammatory cell infiltrations, frequently highly express SECTM1, including an N-terminal, soluble form, which can be detected in the sera of metastatic melanoma patients but not in normal sera. Taken together, our data demonstrate that CD7 is present on monocytes and tumor macrophages and that its ligand, SECTM1, is frequently expressed in corresponding melanoma tissues, possibly acting as a chemoattractant for monocytes to modulate the melanoma microenvironment.


Subject(s)
Antigens, CD7/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Monocytes/cytology , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Cell Differentiation , Cell Movement , Cell Proliferation , Chemotactic Factors/chemistry , Culture Media, Conditioned/chemistry , Disease Progression , Humans , Interferon-alpha/metabolism , Ligands , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Macrophages/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/metabolism , Neoplasm Metastasis , Proteasome Inhibitors/chemistry , Protein Structure, Tertiary , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...