Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 31(10): 2193-202, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21757658

ABSTRACT

OBJECTIVE: The investment of newly formed endothelial cell tubes with differentiated smooth muscle cells (SMC) is critical for appropriate vessel formation, but the underlying mechanisms remain unknown. We previously showed that depletion of focal adhesion kinase (FAK) in the nkx2.5 expression domain led to aberrant outflow tract (OFT) morphogenesis and strove herein to determine the cell types and mechanisms involved. METHODS AND RESULTS: We crossed fak(loxp) targeted mice with available Cre drivers to deplete FAK in OFT SMC (FAK(wnt) and FAK(nk)) or coronary SMC (FAK(cSMC)). In each case, depletion of FAK led to defective vasculogenesis that was incompatible with postnatal life. Immunohistochemical analysis of the mutant vascular structures revealed that FAK was not required for progenitor cell proliferation, survival, or differentiation into SMC but was necessary for subsequent SMC recruitment to developing vasculature. Using a novel FAK-null SMC culture model, we found that depletion of FAK did not influence SMC growth or survival, but blocked directional SMC motility and invasion toward the potent endothelial-derived chemokine, platelet-derived growth factor PDGFBB. FAK depletion resulted in unstable lamellipodial protrusions due to defective spatial-temporal activation of the small GTPase, Rac-1, and lack of Rac1-dependent recruitment of cortactin (an actin stabilizing protein) to the leading edge. Moreover, FAK null SMC exhibited a significant reduction in stimulated extracellular matrix degradation. CONCLUSIONS: FAK drives PDGFBB-stimulated SMC chemotaxis/invasion and is essential for SMC to appropriately populate the aorticopulmonary septum and the coronary vascular plexus.


Subject(s)
Chemotaxis , Focal Adhesion Kinase 1/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neovascularization, Physiologic , Animals , Aorta/embryology , Aorta/enzymology , Apoptosis , Becaplermin , Cell Proliferation , Cell Survival , Cells, Cultured , Chemotaxis/genetics , Coronary Vessels/embryology , Coronary Vessels/enzymology , Cortactin/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/embryology , Neovascularization, Physiologic/genetics , Neuropeptides/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Pseudopodia/enzymology , Pulmonary Artery/embryology , Pulmonary Artery/enzymology , Quail/embryology , RNA Interference , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
2.
Arterioscler Thromb Vasc Biol ; 28(12): 2115-22, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18787183

ABSTRACT

OBJECTIVE: Smooth muscle cell (SMC) differentiation is a dynamic process that must be tightly regulated for proper vascular development and to control the onset of vascular disease. Our laboratory previously reported that a specific focal adhesion kinase (FAK) inhibitor termed FRNK (FAK Related Non-Kinase) is selectively expressed in large arterioles when SMCs are transitioning from a synthetic to contractile phenotype and that FRNK inhibits FAK-dependent SMC proliferation and migration. Herein, we sought to determine whether FRNK expression modulates SMC phenotypes in vivo. METHODS AND RESULTS: We present evidence that FRNK(-/-) mice exhibit attenuated SM marker gene expression during postnatal vessel growth and after vascular injury. We also show that FRNK expression is regulated by transforming growth factor (TGF)-beta and that forced expression of FRNK in cultured cells induces serum- and TGF-beta-stimulated SM marker gene expression, whereas FRNK deletion or expression of a constitutively activated FAK variant attenuated SM gene transcription. CONCLUSIONS: These data highlight the possibility that extrinsic signals regulate the SMC gene profile, at least in part, by modulating the expression of FRNK and that tight regulation of FAK activity by FRNK is important for proper SMC differentiation during development and after vascular injury.


Subject(s)
Blood Vessels/cytology , Blood Vessels/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Animals , Blood Vessels/growth & development , Blood Vessels/injuries , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression/drug effects , Humans , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Protein-Tyrosine Kinases/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
3.
Circ Res ; 102(12): 1502-11, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18497331

ABSTRACT

Leupaxin is a LIM domain-containing adapter protein belonging to the paxillin family that has been previously reported to be preferentially expressed in hematopoietic cells. Herein, we identified leupaxin in a screen for focal adhesion kinase binding partners in aortic smooth muscle, and we show that leupaxin is enriched in human and mouse vascular smooth muscle and that leupaxin expression is dynamically regulated during development. In addition, our studies reveal that leupaxin can undergo cytoplasmic/nuclear shuttling and functions as an serum response factor cofactor in the nucleus. We found that leupaxin forms a complex with serum response factor and associates with CArG-containing regions of smooth muscle promoters and that ectopic expression of leupaxin induces smooth muscle marker gene expression in both 10T1/2 cells and rat aortic smooth muscle cells. Subsequent studies indicated that enhanced focal adhesion kinase activity (induced by fibronectin or expression of constitutively active focal adhesion kinase) attenuates the nuclear accumulation of leupaxin and limits the ability of leupaxin to enhance serum response factor-dependent gene transcription. Thus, these studies indicate that modulation of the subcellular localization of serum response factor cofactors is 1 mechanism by which extracellular matrix-dependent signals may regulate phenotypic switching of smooth muscle cells.


Subject(s)
Cell Adhesion Molecules/physiology , Myocytes, Smooth Muscle/metabolism , Phosphoproteins/physiology , Serum Response Element/physiology , Animals , Aorta/cytology , Aorta/embryology , Aorta/growth & development , Biological Transport , Cell Adhesion Molecules/pharmacology , Cell Differentiation , Cells, Cultured/drug effects , Coronary Vessels/cytology , Female , Focal Adhesion Kinase 1/physiology , Focal Adhesions/physiology , Gene Expression Regulation, Developmental , Humans , Mice , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myometrium/cytology , Organ Specificity , Phosphoproteins/pharmacology , Protein Interaction Mapping , Rats , Recombinant Fusion Proteins/physiology , Serum Response Element/drug effects , Serum Response Factor/physiology , Signal Transduction/physiology , Transcription Factors , Transcription, Genetic
4.
J Biol Chem ; 280(3): 2055-64, 2005 Jan 21.
Article in English | MEDLINE | ID: mdl-15542607

ABSTRACT

The Rac1/Cdc42 effector p21-activated kinase (PAK) is activated by various signaling cascades including receptor-tyrosine kinases and integrins and regulates a number of processes such as cell proliferation and motility. PAK activity has been shown to be required for maximal activation of the canonical Ras/Raf/MEK/ERK Map kinase signaling cascade, likely because of PAK co-activation of Raf and MEK. Herein, we found that adhesion signaling also stimulates an association between PAK1 and ERK1/2. PAK1 and ERK1/2 co-immunoprecipitated from rat aortic smooth muscle cells (SMC) plated on fibronectin, and the two proteins co-localized in membrane ruffles and adhesion complexes following PDGF-BB or sphingosine 1-phosphate treatment, respectively. Far Western analysis demonstrated a direct association between the two proteins, and peptide mapping identified an ERK2 binding site within the autoinhibitory domain of PAK1. Interestingly, deletion of a major ERK binding site in PAK attenuates activation of an ERK-dependent serum-responsive element (SRE)-luciferase reporter gene, indicating that association between PAK and ERK is required to facilitate ERK signaling. We also show that ERK2 phosphorylates PAK1 on Thr(212) in vitro and that Thr(212) is phosphorylated in smooth muscle cells following PDGF-BB treatment in an adhesion- and MEK/ERK-dependent fashion. Expression of a phosphomimic variant, PAK-T212E, does not alter ERK association, but markedly attenuates downstream ERK signaling. Taken together, these data suggest that PAK1 may facilitate ERK signaling by serving as a scaffold to recruit Raf, MEK, and ERK to adhesion complexes, and that subsequent growth factor-stimulated phosphorylation of PAK-Thr(212) by ERK may serve to provide a negative feedback signal to control coordinate activation of ERK by growth factor- and matrix-induced signals.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Threonine/metabolism , Animals , Base Sequence , Cells, Cultured , DNA Primers , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Rats , p21-Activated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...