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










Publication year range
1.
PLoS One ; 12(3): e0175061, 2017.
Article in English | MEDLINE | ID: mdl-28362832

ABSTRACT

In vitro and in vivo evidence has indicated that the tumor suppressor, p53, may play a significant role in the regulation of atherosclerotic plaque formation. In vivo studies using global knockout mice models, however, have generated inconclusive results that do not address the roles of p53 in various cell types involved in atherosclerosis. In this study, we have specifically ablated p53 in vascular smooth muscle cells (VSMC) in the ApoE-/- mouse model to investigate the roles of p53 in VSMC in atherosclerotic plaque formation and stability. We found that p53 deficiency in VSMC alone did not affect the overall size of atherosclerotic lesions. However, there was a significant increase in the number of p53-/- VSMC in the fibrous caps of atherosclerotic plaques in the early stages of plaque development. Loss of p53 results in migration of VSMC at a faster rate using wound healing assays and augments PDGF-induced formation of circular dorsal ruffles (CDR), known to be involved in cell migration and internalization of surface receptors. Furthermore, aortic VSMC from ApoE-/- /p53-/- mice produce significantly more podosomes and are more invasive. We conclude that p53-/- VSMC are enriched in the fibrous caps of lesions at early stages of plaque formation, which is caused in part by an increase in VSMC migration and invasion as shown by p53-/- VSMC in culture having significantly higher rates of migration and producing more CDRs and invasive podosomes.


Subject(s)
Atherosclerosis/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Plaque, Atherosclerotic/genetics , Tumor Suppressor Protein p53/genetics
2.
Cancers (Basel) ; 7(1): 96-111, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25575302

ABSTRACT

Mesenchymal cells employ actin-based membrane protrusions called podosomes and invadopodia for cross-tissue migration during normal human development such as embryogenesis and angiogenesis, and in diseases such as atherosclerosis plaque formation and cancer cell metastasis. The Akt isoforms, downstream effectors of phosphatidylinositol 3 kinase (PI3K), play crucial roles in cell migration and invasion, but their involvement in podosome formation and cell invasion is not known. In this study, we have used Akt1 and/or Akt2 knockout mouse embryonic fibroblasts and Akt3-targeted shRNA to determine the roles of the three Akt isoforms in Src and phorbol ester-induced podosome formation, and extracellular matrix (ECM) digestion. We found that deletion or knockdown of Akt1 significantly reduces Src-induced formation of podosomes and rosettes, and ECM digestion, while suppression of Akt2 has little effect. In contrast, Akt3 knockdown by shRNA increases Src-induced podosome/rosette formation and ECM invasion. These data suggest that Akt1 promotes, while Akt3 suppresses, podosome formation induced by Src, and Akt2 appears to play an insignificant role. Interestingly, both Akt1 and Akt3 suppress, while Akt2 enhances, phorbol ester-induced podosome formation. These data show that Akt1, Akt2 and Akt3 play different roles in podosome formation and ECM invasion induced by Src or phorbol ester, thus underscoring the importance of cell context in the roles of Akt isoforms in cell invasion.

3.
PLoS One ; 9(9): e108257, 2014.
Article in English | MEDLINE | ID: mdl-25247424

ABSTRACT

The tumor suppressor, p53, negatively regulates cell migration and invasion in addition to its role in apoptosis, cell cycle regulation and senescence. Here, we study the roles of p53 in PDGF-induced circular dorsal ruffle (CDR) formation in rat aortic smooth muscle (RASM) cells. In primary and immortalized RASM cells, up-regulation of p53 expression or increase in activity with doxorubicin inhibits CDR formation. In contrast, shRNA-knockdown of p53 or inhibition of its activity with pifithrin α promotes CDR formation. p53 acts by up-regulating PTEN expression, which antagonizes Rac and Cdc42 activation. Both lipid and protein phosphatase activities of PTEN are required for maximal suppression of CDR, but the lipid activity clearly plays the dominant role. N-WASP, the downstream effector of Cdc42, is the major positive contributor to CDR formation in RASM, and is an indirect target of p53. The Rac effector, WAVE2, appears to also play a minor role, while WAVE1 has no significant effect in CDR formation. In sum, we propose that p53 suppresses PDGF-induced CDR formation in RASM cells by upregulating PTEN leading mainly to the inhibition of the Cdc42-N-WASP pathway.


Subject(s)
Aorta/metabolism , Down-Regulation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Cell Line , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Rats , Tumor Suppressor Protein p53/genetics , Up-Regulation
4.
Cell Adh Migr ; 8(3): 205-14, 2014.
Article in English | MEDLINE | ID: mdl-24714032

ABSTRACT

Cell invasion of the extracellular matrix is prerequisite to cross tissue migration of tumor cells in cancer metastasis, and vascular smooth muscle cells in atherosclerosis. The tumor suppressor p53, better known for its roles in the regulation of cell cycle and apoptosis, has ignited much interest in its function as a suppressor of cell migration and invasion. How p53 and its gain-of-function mutants regulate cell invasion remains a puzzle and a challenge for future studies. In recent years, podosomes and invadopodia have also gained center stage status as veritable apparatus specialized in cell invasion. It is not clear, however, whether p53 regulates cell invasion through podosomes and invadopodia. In this review, evidence supporting a negative role of p53 in podosomes formation in vascular smooth muscle cells will be surveyed, and signaling nodes that may mediate this regulation in other cell types will be explored.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Extracellular Matrix/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology
5.
J Cell Physiol ; 228(2): 416-27, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22740332

ABSTRACT

Podosomes are adhesive structures on the ventral surface of cells that invade and degrade the extracellular matrix. Recently, we reported that phorbol 12,13-dibutyrate (PDBu), a protein kinase C (PKC) activator, induced podosome formation in normal human bronchial epithelial (NHBE) cells, and atypical PKCζ regulated MMP-9 recruitment to podosomes for its release and activation. The objective of this study was to explore signaling pathways that are involved in PKC activation-induced podosome formation and matrix degradation. Herein, we found that PDBu increased phosphorylation of PI3K p85, Akt, Src, ERK1/2, and JNK. Inhibitors for PI3K, Akt, and Src suppressed PDBu-induced podosome formation and matrix degradation. In contrast, blockers for MEK/ERK or JNK did not inhibit podosome formation but reduced proteolytic activity of podosomes. Inhibition of PKCζ activity with its pseudosubstrate peptide (PS)-inhibited PDBu-induced phosphorylation of MEK/ERK and JNK. On the other hand, inhibition of MEK/ERK or JNK pathway did not affect PKCζ phosphorylation, but reduced the recruitment of PKCζ and MMP-9 to podosomes. We conclude that PKCζ may regulate MEK/ERK and JNK phosphorylation and in turn activated MEK/ERK and JNK may regulate the proteolytic activity of PDBu-induced podosomes by influencing the recruitment of PKCζ and MMP-9 to podosomes.


Subject(s)
Bronchi/enzymology , MAP Kinase Signaling System , Matrix Metalloproteinase 9/metabolism , Protein Kinase C/metabolism , Respiratory Mucosa/enzymology , Bronchi/drug effects , Carcinogens/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Humans , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Respiratory Mucosa/drug effects
6.
J Cell Sci ; 124(Pt 10): 1739-51, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21525036

ABSTRACT

Invadopodia are actin-rich membrane protrusions that promote extracellular matrix degradation and invasiveness of tumor cells. Src protein-tyrosine kinase is a potent inducer of invadopodia and tumor metastases. Cdc42-interacting protein 4 (CIP4) adaptor protein interacts with actin regulatory proteins and regulates endocytosis. Here, we show that CIP4 is a Src substrate that localizes to invadopodia in MDA-MB-231 breast tumor cells expressing activated Src (MDA-SrcYF). To probe the function of CIP4 in invadopodia, we established stable CIP4 knockdown in MDA-SrcYF cell lines by RNA interference. Compared with control cells, CIP4 knockdown cells degrade more extracellular matrix (ECM), have increased numbers of mature invadopodia and are more invasive through matrigel. Similar results are observed with knockdown of CIP4 in EGF-treated MDA-MB-231 cells. This inhibitory role of CIP4 is explained by our finding that CIP4 limits surface expression of transmembrane type I matrix metalloprotease (MT1-MMP), by promoting MT1-MMP internalization. Ectopic expression of CIP4 reduces ECM digestion by MDA-SrcYF cells, and this activity is enhanced by mutation of the major Src phosphorylation site in CIP4 (Y471). Overall, our results identify CIP4 as a suppressor of Src-induced invadopodia and invasion in breast tumor cells by promoting endocytosis of MT1-MMP.


Subject(s)
Breast Neoplasms/metabolism , Endocytosis/physiology , Matrix Metalloproteinase 14/metabolism , Microtubule-Associated Proteins/genetics , cdc42 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Female , HEK293 Cells , Humans , Microtubule-Associated Proteins/metabolism , Minor Histocompatibility Antigens , Neoplasm Invasiveness , Transfection
7.
Cell Adh Migr ; 5(2): 144-9, 2011.
Article in English | MEDLINE | ID: mdl-21164280

ABSTRACT

The p53 transcription factor, discovered in 1979 ( 1;2) , is well known as a potent suppressor of tumor development by inhibiting cell cycle progression, and promoting senescence or apoptosis, when the genome is compromised or under oncogenic stress ( 3) . Accumulating evidence has pointed to an alternative role of p53 in the curtailment of tumor progression and colonization of secondary sites by negatively regulating tumor cell metastasis ( 4;5) . Recently, we have found that p53 suppresses Src-induced formation of podosomes and associated invasive phenotypes in fibroblasts and vascular smooth muscle cells (VSMC) ( 6;7) . In this review, I will focus on some recent studies that have identified p53 as a suppressor of cell migration and invasion in general, and VSMC podosome formation and ECM degradation in particular.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Actins/genetics , Actins/metabolism , Animals , Apoptosis , Calmodulin-Binding Proteins/genetics , Cell Communication , Cell Movement , Cortactin/genetics , Cortactin/metabolism , Cytoskeleton/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Genes, p53 , Genes, src , Humans , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Rats , Rosette Formation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics
8.
Cell Cycle ; 9(22): 4450-4, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21084866

ABSTRACT

We have recently identified mutually antagonizing signaling pathways that regulate podosome formation and invasive phenotypes in Src-transformed vascular smooth muscle cells and fibroblasts. Cross-talks between the anti-invasion p53-PTEN, and the pro-invasion Src-Stat3 and Src-PI3K-Akt pathways serve as a check and balance that dictates the outcome of either an invasive or non-invasive phenotype. Using a retrovirus vector encoding PTEN phosphatase mutants that retain either protein- or lipid-phosphatase activity on a Src(Y527F)background, we report here that both lipid- and protein-phosphatase activities of PTEN contribute to the suppression of Src-induced podosome formation and associated invasive phenotypes in rat aortic smooth muscle cells. This data suggests that p53 up-regulation of PTEN inhibits cell invasion via a two-prong mechanism: inactivating podosome agonists by its protein-phosphatase activity on the one hand, and antagonising the PI3K-Akt pathway by its lipid-phosphatase activity on the other.


Subject(s)
PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Movement , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Rats , STAT3 Transcription Factor/metabolism , Signal Transduction , Up-Regulation , src-Family Kinases/metabolism
9.
Mol Cell Biol ; 30(23): 5545-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20937775

ABSTRACT

Podosomes are transient cell surface structures essential for degradation of extracellular matrix during cell invasion. Protein kinase C (PKC) is involved in the regulation of podosome formation; however, the roles of individual PKC isoforms in podosome formation and proteolytic function are largely unknown. Recently, we reported that PDBu, a PKC activator, induced podosome formation in normal human bronchial epithelial cells. Here, we demonstrate that phorbol-12,13-dibutyrate (PDBu)-induced podosome formation is mainly mediated through redistribution of conventional PKCs, especially PKCα, from the cytosol to the podosomes. Interestingly, although blocking atypical PKCζ did not affect PDBu-induced podosome formation, it significantly reduced matrix degradation at podosomes. Inhibition of PKCζ reduced recruitment of matrix metalloprotease 9 (MMP-9) to podosomes and its release and activation. Downregulation of MMP-9 by small interfering RNA (siRNA) or neutralization antibody also significantly reduced matrix degradation. The regulatory effects of PKCζ on matrix degradation and recruitment of MMP-9 to podosomes were PKCζ kinase activity dependent. PDBu-induced recruitment of PKCζ and MMP-9 to podosomes was blocked by inhibition of novel PKC with rottlerin or PKCδ siRNA. Our data suggest that multiple PKC isozymes form a signaling cascade that controls podosome formation and dynamics and MMP-9 recruitment, release, and activation in a coordinated fashion.


Subject(s)
Cell Surface Extensions/enzymology , Matrix Metalloproteinase 9/metabolism , Protein Kinase C/metabolism , Base Sequence , Biological Transport, Active/drug effects , Cell Surface Extensions/drug effects , Cell Surface Extensions/ultrastructure , Cells, Cultured , DNA Primers/genetics , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/ultrastructure , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA, Small Interfering/genetics , Signal Transduction
10.
Mol Cell Biol ; 30(21): 4980-95, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20733006

ABSTRACT

We have recently shown that Src induces the formation of podosomes and cell invasion by suppressing endogenous p53, while enhanced p53 strongly represses the Src-induced invasive phenotype. However, the mechanism by which Src and p53 play antagonistic roles in cell invasion is unknown. Here we show that the Stat3 oncogene is a required downstream effector of Src in inducing podosome structures and related invasive phenotypes. Stat3 promotes Src phenotypes through the suppression of p53 and the p53-inducible protein caldesmon, a known podosome antagonist. In contrast, enhanced p53 attenuates Stat3 function and Src-induced podosome formation by upregulating the tumor suppressor PTEN. PTEN, through the inactivation of Src/Stat3 function, also stabilizes the podosome-antagonizing p53/caldesmon axis, thereby further enhancing the anti-invasive potential of the cell. Furthermore, the protein phosphatase activity of PTEN plays a major role in the negative regulation of the Src/Stat3 pathway and represses podosome formation. Our data suggest that cellular invasiveness is dependent on the balance between two opposing forces: the proinvasive oncogenes Src-Stat3 and the anti-invasive tumor suppressors p53-PTEN.


Subject(s)
Cell Movement/physiology , PTEN Phosphohydrolase/physiology , STAT3 Transcription Factor/physiology , Tumor Suppressor Protein p53/physiology , src-Family Kinases/physiology , 3T3 Cells , Animals , Base Sequence , Calmodulin-Binding Proteins/antagonists & inhibitors , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/physiology , Cell Line , Cell Movement/genetics , DNA Primers/genetics , Gene Knockdown Techniques , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/physiology , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 10/physiology , Matrix Metalloproteinase Inhibitors , Mice , Models, Biological , Mutant Proteins/genetics , Mutant Proteins/physiology , Myocytes, Smooth Muscle/physiology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , PTEN Phosphohydrolase/genetics , Phenotype , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , src-Family Kinases/genetics
12.
Mol Cell Biol ; 29(11): 3088-98, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19349302

ABSTRACT

The tumor-suppressive role of p53 at the level of tumor initiation is well documented. It has also been shown previously that p53 acts against tumor progression/metastasis. However, its role in modulating cell migration and invasion leading to metastasis is poorly understood. In this study, using vascular smooth muscle cells and NIH 3T3 fibroblast cells, we have shown that p53 potently suppresses Src-induced podosome/rosette formation, extracellular matrix digestion, cell migration, and invasion. The overexpression of exogenous wild-type p53 or the activation of the endogenous p53 function suppresses, while the short hairpin RNA-mediated knockdown of p53 expression or the pageing of its function exacerbates, Src-induced migratory and invasive phenotypes. We have also found that p53 expression and function are downregulated in cells stably transformed with constitutively active Src that exhibit aggressive invasive properties. Lastly, p53 upregulates the expression of caldesmon, an actin-binding protein that has been shown to be an inhibitor of podosome/invadopodium formation. The ability of p53 to suppress Src phenotypes in transformed cells was largely abolished by knocking down caldesmon. This study reports a novel molecular mechanism (caldesmon), as well as a structural basis (podosomes/rosettes), to show how p53 can act as an anti-motility/invasion/metastasis agent.


Subject(s)
Calmodulin-Binding Proteins/genetics , Cell Movement , Proto-Oncogene Proteins pp60(c-src)/metabolism , Pseudopodia/enzymology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics , Animals , Blood Vessels/cytology , Cell Movement/drug effects , Collagen/metabolism , Drug Combinations , Enzyme Activation/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Laminin/metabolism , Mice , Microfilament Proteins/metabolism , Models, Biological , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , NIH 3T3 Cells , Proteoglycans/metabolism , Pseudopodia/drug effects , Rats , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation/drug effects
13.
J Cell Physiol ; 218(2): 366-75, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18932175

ABSTRACT

Spreading and migration of the basal cells neighboring a wound is essential for airway epithelial repair. To gain insight into the molecular mechanisms that govern these cellular processes, we asked whether normal human airway epithelial cells can form podosomes, a cellular structure discovered from cancer and mesenchymal cells that controls migration and invasion. Herein, we report that phorbol-12, 13-dibutyrate (PDBu), a protein kinase C activator, induced reorganization of cytoskeletal structure in primary normal human bronchial epithelial cells, and in normal human airway epithelial BEAS2B cells. Z-stack scanning confocal microscopy showed that PDBu-induced podosome-like structures contain actin-rich columns that arise from the ventral surface of the cell, and also revealed the presence of circular ruffles/waves at the dorsal cell surface. The molecular components of these cytoskeletal structures were determined with immunofluorescent staining. Using in situ zymography, we demonstrated that PDBu-induced podosomes were capable of degrading fibronectin-gelatin-sucrose matrix. PDBu also increased epithelial cell invasion across Transwell chamber. Podosomes and circular dorsal ruffles may be important for epithelial cell migration and invasion, thus contributing to respiratory epithelial repair and regeneration.


Subject(s)
Bronchi/cytology , Cell Membrane Structures/drug effects , Cell Membrane Structures/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Cell Line , Cell Membrane Structures/enzymology , Cell Movement/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epithelial Cells/enzymology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinases/metabolism , Protein-Tyrosine Kinases/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Vinculin/metabolism
14.
Eur J Cell Biol ; 86(4): 189-206, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17343955

ABSTRACT

Cells degrade extracellular matrix (ECM) barriers at focal locations by the formation of membrane protrusions called invadopodia. Polymerization of the actin cytoskeleton is critical to the extension of these processes into the ECM. We used a short interference RNA/rescue strategy to investigate the role of cortactin in the formation of Src-induced invadopodia in 3T3 fibroblasts, and subsequent degradation of the ECM. Cortactin-depleted cells did not form invadopodia or degrade the ECM. Functional invadopodia were restored in cortactin-depleted cells by expression of full-length cortactin, and fragments that contained the intact actin-binding repeats. Mutation of the three Src-targeted Tyr sites to Phe caused a loss in its rescuing ability, while mutation of the Erk phosphorylation sites had little effect on invadopodia formation. Interestingly, knock-down of cortactin did not affect the formation of lamellipodia and only slightly attenuated random cell motility. Our data shows that formation of functional invadopodia requires interaction between cortactin and filamentous actin, while interaction with SH3- and NTA-binding partners plays a less significant role. Furthermore, phosphorylation of cortactin by Src, but not by Erk, is essential for functional invadopodia formation. These results also suggest that cortactin plays a different role in invadopodia-dependent ECM degradation and lamellipodia formation in cell movement.


Subject(s)
Cell Surface Extensions/metabolism , Cortactin/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , src Homology Domains , src-Family Kinases/metabolism , Actins/metabolism , Animals , Cell Line, Transformed , Cell Movement , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cortactin/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mutation , NIH 3T3 Cells , Neoplasm Invasiveness , Phenotype , Phosphorylation , Pseudopodia/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Time Factors , Transfection , src Homology Domains/genetics , src-Family Kinases/genetics
15.
Arch Biochem Biophys ; 456(2): 183-93, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16854367

ABSTRACT

Cortactin is an F-actin binding protein that is enriched in dynamic cytoskeletal organelles such as podosomes, membrane ruffles, and lamellipodia. We have shown previously that Src-phosphorylation of cortactin is not required for its translocation to phorbol-ester induced podosomes in A7r5 aortic smooth muscle cells, but may be important for stability and turnover of podosomes. However, little is known of the role of Ser/Thr kinases in the regulation of cortactin. Here, we report that p21-associated kinase (PAK), which plays a crucial role in the formation of podosome and membrane ruffles, is able to phosphorylate cortactin in vitro. The predominant phosphorylation site is located at Ser113 in the first actin-binding repeat. Phosphorylation by PAK is not required for the translocation of cortactin to podosomes, lamellipodia, or membrane ruffles in A7r5 smooth muscle cells. However, binding of cortactin to F-actin is significantly reduced by PAK-phosphorylation. Taken together, these results suggest a role for PAK-phosphorylation of cortactin in the regulation of the dynamics of branched actin filaments in dynamic cytoskeletal organelles.


Subject(s)
Cortactin/chemistry , Cortactin/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cells, Cultured , Enzyme Activation , Phosphorylation , Protein Binding , Rats , p21-Activated Kinases
16.
Cell Motil Cytoskeleton ; 63(9): 543-62, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16800003

ABSTRACT

Caldesmon is believed to be one of the key regulators for actin dynamics and thereby cell polarity, membrane extension, and cell motility. We have shown previously that stress fiber formation and cell movement are severely impaired in the cells expressing human fibroblast caldesmon fragment defective in Ca2+/CaM binding sites. Both Ser458 and Ser489, adjacent to the Ca2+/CaM-binding sites, are phosphorylated by p21-activated kinase (PAK) in vitro. Here we report that Ser458 is phosphorylated in response to cell movement. We substituted Ser458 and Ser489 on C-terminal caldesmon (CaD39) with alanine or glutamic acid to mimic under-phosphorylated (CaD39-PAKA) or constitutively phosphorylated (CaD39-PAKE) caldesmon. In vitro, CaD39-PAKE, but not CaD39-PAKA, fails to inhibit myosin ATPase activity and exhibits reduced binding to Ca2+/CaM. When stably expressed in Chinese Hamster Ovary cells, both CaD39-PAKA and CaD39-PAKE incorporate into stress fibers and localize to the leading edge of the migrating cell. Expression of CaD39-PAKE, but not CaD39-PAKA, fails to protect stress fibers from cytochalasin depolymerization. However, both mutations inhibit cell polarization and lead to defects in membrane extension and cell migration. We conclude that phosphorylation of caldesmon by PAK is a dynamic process required to regulate actin dynamics and membrane protrusions in wound-induced cell migration.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Cell Movement , Protein Serine-Threonine Kinases/metabolism , Pseudopodia/physiology , Alanine/chemistry , Animals , CHO Cells , Calmodulin-Binding Proteins/chemistry , Cells, Cultured , Cricetinae , Fibroblasts/cytology , Gene Expression , Glutamic Acid/chemistry , Humans , Mutation/genetics , Phosphorylation , Stress Fibers/metabolism , Wound Healing/physiology , p21-Activated Kinases
17.
J Cell Sci ; 119(Pt 9): 1691-702, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16595550

ABSTRACT

Podosomes are highly dynamic actin-based structures commonly found in motile and invasive cells such as macrophages, osteoclasts and vascular smooth muscle cells. Here, we have investigated the role of caldesmon, an actin-binding protein, in the formation of podosomes in aortic smooth muscle A7r5 cells induced by the phorbol ester PDBu. We found that endogenous low molecular weight caldesmon (l-caldesmon), which was normally localised to actin-stress fibres and membrane ruffles, was recruited to the actin cores of PDBu-induced podosomes. Overexpression of l-caldesmon in A7r5 cells caused dissociation of actin-stress fibres and disruption of focal adhesion complexes, and significantly reduced the ability of PDBu to induce podosome formation. By contrast, siRNA interference of caldesmon expression enhanced PDBu-induced formation of podosomes. The N-terminal fragment of l-caldesmon, CaD40, which contains the myosin-binding site, did not label stress fibres and was not translocated to PDBu-induced podosomes. Cad39, the C-terminal fragment housing the binding sites for actin, tropomyosin and calmodulin, was localised to stress fibres and was translocated to podosomes induced by PDBu. The caldesmon mutant, CadCamAB, which does not interact with Ca2+/calmodulin, was not recruited to PDBu-induced podosomes. These results show that (1) l-caldesmon is an integral part of the actin-rich core of the podosome; (2) overexpression of l-caldesmon suppresses podosome formation, whereas siRNA knock-down of l-caldesmon facilitates its formation; and (3) the actin-binding and calmodulin-binding sites on l-caldesmon are essential for the translocation of l-caldesmon to the podosomes. In summary, this data suggests that caldesmon may play a role in the regulation of the dynamics of podosome assembly and that Ca2+/calmodulin may be part of a regulatory mechanism in podosome formation.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Actins/metabolism , Animals , Aorta/cytology , Calmodulin/metabolism , Calmodulin-Binding Proteins/genetics , Cell Line , Cytoskeleton/metabolism , Fluorescent Dyes/metabolism , Myocytes, Smooth Muscle/drug effects , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stress Fibers/metabolism , Stress Fibers/ultrastructure
18.
Exp Cell Res ; 312(6): 760-9, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16434035

ABSTRACT

Cortactin, a multi-domain scaffolding protein involved in actin polymerization, is enriched in podosomes induced by phorbol ester in vascular smooth muscle cells. We generated several functional and truncation mutants of cortactin to probe the roles of various protein interaction domains in the regulation of the dynamics of podosome formation. At the onset of podosome genesis, cortactin clustered near the ends of stress fibers that appeared to act as nucleation platforms onto which the actin polymerization machinery assembled. Translocation of cortactin to these pre-podosome clusters required the intact N-WASp-binding SH3 domain. Overexpression of the C-terminal third of cortactin containing the intact SH3 domain inhibited podosome formation presumably by sequestering of N-WASp and prevented cortactin clustering. Subsequent assembly of the actin-rich core of podosomes required translocation of additional cortactin to the actin core, a process that required the actin-binding repeats, but not the Arp2/3-binding N-terminal acidic region nor the SH3 domain. These results suggest that the SH3 domain and the actin-binding repeat region are involved, respectively, in the early and late stages of podosome formation process.


Subject(s)
Actins/metabolism , Cell Membrane Structures/physiology , Cortactin/physiology , Actins/drug effects , Animals , Binding Sites , Cell Membrane Structures/metabolism , Cells, Cultured , Cortactin/genetics , Cortactin/metabolism , Liver/cytology , Liver/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Mutagenesis, Site-Directed , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rats
19.
Am J Physiol Cell Physiol ; 290(2): C463-71, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16162656

ABSTRACT

Cortactin, a predominant substrate of Src family kinases, plays an important role in Arp2/3-dependent actin polymerization in lamellipodia and membrane ruffles and was recently shown to be enriched in podosomes induced by either c-Src or phorbol ester. However, the mechanisms by which cortactin regulates podosome formation have not been determined. In this study, we showed that cortactin is required for podosome formation, using siRNA knockdown of cortactin expression in smooth muscle A7r5 cells. Treatment with phorbol ester or expression of constitutively active c-Src induced genesis of cortactin-containing podosomes as well as increase in phosphorylation of cortactin at Y421 and Y466, the Src phosphorylation sites on cortactin. The Src kinase inhibitor SU-6656 significantly inhibited formation of podosomes induced by phorbol ester and phosphorylation of cortactin, whereas PKCalpha inhibitor did not affect podosome formation in c-Src-transfected cells. Unexpectedly, expression of cortactin mutants containing Y421F, Y421D, Y466F, or Y466D mutated sites did not affect podosome formation or cortactin translocation to podosomes, although endogenous tyrosine-phosphorylated cortactin at Y421 and Y466 was present in podosomes. Our data indicate that 1) PKCalpha acts upstream of Src in phosphorylation of cortactin and podosome formation in smooth muscle cells; 2) expression of cortactin is essential for genesis of podosomes; 3) phosphorylation at Y421 and Y466 is not required for translocation of cortactin to podosomes, although phosphorylation at these sites appears to be enriched in podosomes; and 4) tyrosine phosphorylation of cortactin may be involved in regulation of stability and turnover of podosomes, rather than targeting this protein to the site of podosome formation.


Subject(s)
Cell Surface Extensions/metabolism , Cortactin/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Tyrosine/metabolism , Animals , Carcinogens/metabolism , Cell Line , Cell Shape , Cortactin/genetics , Indoles/metabolism , Maleimides/metabolism , Microscopy, Fluorescence , Phorbol 12,13-Dibutyrate/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , RNA, Small Interfering/metabolism , Rats , Sulfonamides/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
20.
Am J Physiol Cell Physiol ; 289(4): C898-907, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15944209

ABSTRACT

Remodeling of the vascular smooth muscle cytoskeleton is essential for cell motility involved in the development of diseases such as arteriosclerosis and restenosis. The p21-activated kinase (PAK), which is an effector of the Rho GTPases Rac and Cdc42, has been shown to be involved in cytoskeletal remodeling and cell motility. We show herein that expression of cytoskeletally active constructs of PAK1 is able to induce the formation of dynamic, podosome-like F-actin columns in the A7r5 vascular smooth muscle cell line. Most of these actin columns appear at the junctions between stress fibers and focal adhesions and contain several known podosomal protein markers, such as cortactin, Arp2/3, alpha-actinin, and vinculin. The kinase activity of PAK plays a role in the regulation of the turnover rates of these actin columns but is not essential for their formation. The ability of PAK to interact with the PAK-interacting exchange factor (PIX) but not with Rac or Cdc42, however, is required for the formation of the actin columns as well as for the translocation of PIX and G protein-coupled receptor kinase-interacting protein (GIT) to focal adhesions adjacent to the actin columns. These findings suggest that interaction between PAK and PIX, as well as the recruitment of PIX and GIT to focal adhesions, plays an important role in the formation of actin columns that resemble podosomes induced by phorbol ester in vascular smooth muscle cells.


Subject(s)
Actin Cytoskeleton/physiology , Cell Cycle Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Muscle, Smooth, Vascular/ultrastructure , Protein Serine-Threonine Kinases/physiology , Animals , Cell Line , Rats , Rho Guanine Nucleotide Exchange Factors , p21-Activated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...