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.
PLoS One ; 10(11): e0142787, 2015.
Article in English | MEDLINE | ID: mdl-26571308

ABSTRACT

α1a Adrenergic receptors (α1aARs) are the predominant AR subtype in human vascular smooth muscle cells (SMCs). α1aARs in resistance vessels are crucial in the control of blood pressure, yet the impact of naturally occurring human α1aAR genetic variants in cardiovascular disorders remains poorly understood. To this end, we present novel findings demonstrating that 3D cultures of vascular SMCs expressing human α1aAR-247R (247R) genetic variant demonstrate significantly increased SMC contractility compared with cells expressing the α1aAR-WT (WT) receptor. Stable expression of 247R genetic variant also triggers MMP/EGFR-transactivation dependent serum- and agonist-independent (constitutive) hyperproliferation and agonist-dependent hypertrophy of SMCs. Agonist stimulation reduces contractility Using pathway-specific inhibitors we determined that the observed hyperproliferation of 247R-expressing cells is triggered via ß-arrestin1/Src/MMP-2/EGFR/ERK-dependent mechanism. MMP-2-specific siRNA inhibited 247R-triggered hyperproliferation indicating MMP-2 involvement in 247R-triggered hyperproliferation in SMCs. ß-arrestin1-specific shRNA also inhibited 247R-triggered hyperproliferation but did not affect hypertrophy in 247R-expressing SMCs, indicating that agonist-dependent hypertrophy is independent of ß-arrestin1. Our data reveal that in different cardiovascular cells the same human receptor genetic variant can activate alternative modulators of the same signaling pathway. Thus, our findings in SMCs demonstrate that depending on the type of cells expressing the same receptor (or receptor variant), different target-specific inhibitors could be used to modulate aberrant hyperproliferative or hypertrophic pathways in order to restore normal phenotype.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/pharmacology , ErbB Receptors/genetics , Genetic Variation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/pathology , Receptors, Adrenergic, alpha-1/genetics , Transcriptional Activation/drug effects , Animals , Arrestins/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Hypertrophy , Matrix Metalloproteinases/metabolism , Models, Biological , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects , beta-Arrestins , src-Family Kinases/metabolism
2.
Cell Signal ; 26(9): 1985-97, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24835978

ABSTRACT

The role of naturally occurring human α1a-Adrenergic Receptor (α1aAR) genetic variants associated with cardiovascular disorders is poorly understood. Here, we present the novel findings that expression of human α1aAR-247R (247R) genetic variant in cardiomyoblasts leads to transition of cardiomyoblasts into a fibroblast-like phenotype, evidenced by morphology and distinct de novo expression of characteristic genes. These fibroblast-like cells exhibit constitutive, high proliferative capacity and agonist-induced hypertrophy compared with cells prior to transition. We demonstrate that constitutive, synergistic activation of EGFR, Src and ERK kinases is the potential molecular mechanism of this transition. We also demonstrate that 247R triggers two distinct EGFR transactivation-dependent signaling pathways: 1) constitutive Gq-independent ß-arrestin-1/Src/MMP/EGFR/ERK-dependent hyperproliferation and 2) agonist-induced Gq- and EGFR/STAT-dependent hypertrophy. Interestingly, in cardiomyoblasts agonist-independent hyperproliferation is MMP-dependent, but in fibroblast-like cells it is MMP-independent, suggesting that expression of α1aAR genetic variant in cardiomyocytes may trigger extracellular matrix remodeling. Thus, these novel findings demonstrate that EGFR transactivation by α1aAR-247R leads to hyperproliferation, hypertrophy and alterations in cardiomyoblasts, suggesting that these unique genetically-mediated alterations in signaling pathways and cellular function may lead to myocardial fibrosis. Such extracellular matrix remodeling may contribute to the genesis of arrhythmias in certain types of heart failure.


Subject(s)
Fibroblasts/cytology , Myoblasts, Cardiac/cytology , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Animals , Arrestins/antagonists & inhibitors , Arrestins/genetics , Arrestins/metabolism , Cell Line , Cell Proliferation/drug effects , Dipeptides/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/metabolism , Mutation , Phenylephrine/pharmacology , Phosphorylation , Quinazolines/pharmacology , Rats , Receptors, Adrenergic, alpha-1/chemistry , Receptors, Adrenergic, alpha-1/genetics , Transcriptional Activation , Tyrphostins/pharmacology , beta-Arrestin 1 , beta-Arrestins , src-Family Kinases/metabolism
3.
Rev Course Lect ; 2012: 30-34, 2012 May.
Article in English | MEDLINE | ID: mdl-26740973

ABSTRACT

Genomic variation is an important factor in why supposedly "similar" patients react differently to drugs, have different disease course(s), and varying clinical outcomes. This review provides an update on concepts in modern genomic medicine with an emphasis on clinically relevant study approaches, disease/drug pathway analysis, and recent pharmacogenomic findings. The application of genomic medicine and its importance for rapid diagnosis of disease-causing agents, as well as its clinical application in human disease diagnosis/treatment and in cardiovascular disease are discussed. In addition to direct clinical applications, modern genomic approaches also play an important role in elucidating new mechanisms of disease. Finally, the role of the National Institutes of Health national pharmacogenomics research network in codifying "bench to bedside" translation of genetic results that impact drug therapy will also be discussed.

4.
Proc Natl Acad Sci U S A ; 108(49): 19796-801, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22089237

ABSTRACT

We previously identified a naturally occurring human SNP, G247R, in the third intracellular loop of the α(1a)-adrenergic receptor (α(1a)-247R) and demonstrated that constitutive expression of α(1a)-247R results in twofold increased cell proliferation compared with WT. In the present study we elucidate molecular mechanisms and signal transduction pathways responsible for increased cell proliferation unique to α(1a)-247R, but not α(1a)-WT, α(1b), or α(1d)AR subtypes. We show that elevated levels of matrix metalloproteinase-7 (MMP7) and a disintegrin and metalloproteinase-12 (ADAM12) in α(1a)-247R-expressing cells are responsible for EGF receptor (EGFR) transactivation, downstream ERK activation, and increased cell proliferation; this pathway is confirmed using MMP, EGFR, and ERK inhibitors. We demonstrate that EGFR transactivation and downstream ERK activation depends on increased shedding of heparin-binding EGF. Finally, we demonstrate that knockdown of MMP7 or ß-arrestin1 by shRNAs results in attenuation of proliferation of cells expressing α(1a)-247R. Importantly, accelerated cell proliferation triggered by the α(1a)-247R is serum- and agonist-independent, providing unique evidence for constitutive active coupling to the ß-arrestin1/MMP/EGFR transactivation pathway by any G protein-coupled receptor. These findings raise the possibility of a previously unexplored mechanism for sympathetically mediated human hypertension triggered by a naturally occurring human genetic variant.


Subject(s)
ErbB Receptors/genetics , Mutation , Receptors, Adrenergic, alpha-1/genetics , Transcriptional Activation , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM12 Protein , Amino Acid Substitution , Animals , Arrestins/genetics , Arrestins/metabolism , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Humans , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase Inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Confocal , Phosphorylation/drug effects , Quinazolines/pharmacology , RNA Interference , Receptors, Adrenergic, alpha-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tyrphostins/pharmacology , beta-Arrestins
5.
J Biol Chem ; 285(14): 10487-96, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20097759

ABSTRACT

Phogrin is a transmembrane protein expressed in cells with stimulus-coupled peptide hormone secretion, including pancreatic beta cells, in which it is localized to the membrane of insulin-containing dense-core vesicles. By sequence, phogrin is a member of the family of receptor-like protein-tyrosine phosphatases, but it contains substitutions in conserved catalytic sequences, and no significant enzymatic activity for phogrin has ever been reported. We report here that phogrin is able to dephosphorylate specific inositol phospholipids, including phosphatidylinositol (PI) 3-phosphate and PI 4,5-diphosphate but not PI 3,4,5-trisphosphate. The phosphatidylinositol phosphatase (PIPase) activity of phogrin was measurable but low when evaluated by the ability of a catalytic domain fusion protein to hydrolyze soluble short-chain phosphatidylinositol phospholipids. Unlike most PIPases, which are cytoplasmic proteins that associate with membranes, mature phogrin is a transmembrane protein. When the transmembrane form of phogrin was overexpressed in mammalian cells, it reduced plasma membrane phosphatidylinositol 4,5-disphosphate levels in a dose-dependent manner. When purified and assayed in vitro, the transmembrane form had a specific activity of 142 mol/min/mol, 75-fold more active than the catalytic domain fusion protein and comparable with the specific activities of the other PIPases. The PIPase activity of phogrin depended on the catalytic site cysteine and correlated with effects on glucose-stimulated insulin secretion. We propose that phogrin functions as a phosphatidylinositol phosphatase that contributes to maintaining subcellular differences in levels of PIP that are important for regulating stimulus-coupled exocytosis of insulin.


Subject(s)
Hypoglycemic Agents/metabolism , Insulin/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Secretory Vesicles/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Exocytosis/physiology , Fluorescent Antibody Technique , Glucose/metabolism , Insulin Secretion , Insulinoma/genetics , Insulinoma/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Phosphotyrosine/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 8/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Mol Biol Cell ; 19(2): 563-71, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032585

ABSTRACT

TSPs 1 and 2 function as endogenous inhibitors of angiogenesis. Although thrombospondins (TSPs) have been shown to induce apoptosis in HMVECs, we reasoned that a homeostatic mechanism would also be needed to inhibit EC growth without causing cell death, e.g., in the maintenance of a normal vascular endothelium. HMVECs, cultured in low serum, responded to VEGF with an increase in [(3)H]thymidine incorporation that was inhibited by TSPs and was accompanied by decreases in the phosphorylation of Akt and MAPK, without an increase in apoptosis. RAP, an inhibitor of the low-density lipoprotein (LDL) family of endocytic receptors, and blocking antibodies to VLDLR were as effective as TSPs in the inhibition of thymidine uptake in response to VEGF, and the effects of these agents were not additive. Supportive evidence for the role of the VLDLR in mediating this inhibition was provided by the demonstration of a high-affinity interaction between TSPs and the VLDLR. We propose that TSP1 and TSP2, together with the VLDLR, initiate a nonapoptotic pathway for maintenance of the normal adult vascular endothelium in a quiescent state, similar to that invoked for the regulation of mitogenesis by PDGF, but involving signaling via the VLDLR rather than LRP1.


Subject(s)
Apoptosis , Cell Division , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Receptors, LDL/metabolism , Thrombospondin 1/metabolism , Thrombospondins/metabolism , Animals , Apoptosis/drug effects , CHO Cells , Cell Division/drug effects , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Humans , Immunoprecipitation , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , Thrombospondins/chemistry , Thrombospondins/isolation & purification , Thrombospondins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
7.
J Biol Chem ; 281(50): 38507-18, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17018525

ABSTRACT

The function of the NH(2)-terminal propeptide of type I procollagen (N-propeptide) is poorly understood. We now show that a recombinant trimeric N-propeptide interacts with transforming growth factor-beta1 and BMP2 and exhibits functional effects in stably transfected cells. The synthesis of N-propeptide by COS-7 cells results in an increase in phosphorylation of Akt and Smad3 and is associated with a marked reduction in type I procollagen synthesis and impairment in adhesion. In C2C12 cells, N-propeptide inhibits the osteoblastic differentiation induced by BMP2. Our data suggest that these effects are mediated by the interaction of N-propeptide with an intracellular receptor in the secretory pathway, because they are not observed when recombinant N-propeptide is added to the culture medium of either COS-7 or C2C12 cells. Both the binding of N-propeptide to cytokines and its functional properties are entirely dependent on the exon 2-encoded globular domain, and a mutation that substitutes a serine for a highly conserved cysteine in exon 2 abolishes its function. Our findings suggest that N-propeptide performs an important feedback regulatory function and provides a rationale for the prominence of a homotrimeric form of type I procollagen (alpha1 trimer) during vertebrate development.


Subject(s)
Collagen Type I/physiology , Animals , Cell Adhesion , Cell Line , Collagen Type I/chemistry , Collagen Type I/genetics , Mice , Mutation , Osteogenesis Imperfecta/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Smad2 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...