Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
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.
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
5.
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
6.
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
7.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...