Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 110(14): E1301-10, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23509284

ABSTRACT

Triple-negative breast cancer (TNBC) accounts for ∼20% of cases and contributes to basal and claudin-low molecular subclasses of the disease. TNBCs have poor prognosis, display frequent mutations in tumor suppressor gene p53 (TP53), and lack targeted therapies. The MET receptor tyrosine kinase is elevated in TNBC and transgenic Met models (Met(mt)) develop basal-like tumors. To investigate collaborating events in the genesis of TNBC, we generated Met(mt) mice with conditional loss of murine p53 (Trp53) in mammary epithelia. Somatic Trp53 loss, in combination with Met(mt), significantly increased tumor penetrance over Met(mt) or Trp53 loss alone. Unlike Met(mt) tumors, which are histologically diverse and enriched in a basal-like molecular signature, the majority of Met(mt) tumors with Trp53 loss displayed a spindloid pathology with a distinct molecular signature that resembles the human claudin-low subtype of TNBC, including diminished claudins, an epithelial-to-mesenchymal transition signature, and decreased expression of the microRNA-200 family. Moreover, although mammary specific loss of Trp53 promotes tumors with diverse pathologies, those with spindloid pathology and claudin-low signature display genomic Met amplification. In both models, MET activity is required for maintenance of the claudin-low morphological phenotype, in which MET inhibitors restore cell-cell junctions, rescue claudin 1 expression, and abrogate growth and dissemination of cells in vivo. Among human breast cancers, elevated levels of MET and stabilized TP53, indicative of mutation, correlate with highly proliferative TNBCs of poor outcome. This work shows synergy between MET and TP53 loss for claudin-low breast cancer, identifies a restricted claudin-low gene signature, and provides a rationale for anti-MET therapies in TNBC.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Claudins/metabolism , Disease Models, Animal , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/deficiency , Animals , Cells, Cultured , Female , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Transgenic , Microarray Analysis , Proto-Oncogene Proteins c-met/genetics
2.
Mol Cell Biol ; 29(11): 3018-32, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19289496

ABSTRACT

Hepatocyte growth factor (HGF), the ligand for the Met receptor tyrosine kinase, induces epithelial cell dispersal, invasion, and morphogenesis, events that require remodeling of the actin cytoskeleton. The scaffold protein Gab1 is essential for these biological responses downstream from Met. We have identified p21-activated kinase 4 (Pak4) as a novel Gab1-interacting protein. We show that in response to HGF, Gab1 and Pak4 associate and colocalize at the cell periphery within lamellipodia. The association between Pak4 and Gab1 is dependent on Gab1 phosphorylation but independent of Pak4 kinase activity. The interaction is mediated through a region in Gab1, which displays no homology to known Gab1 interaction motifs and through the guanine exchange factor-interacting domain of Pak4. In response to HGF, Gab1 and Pak4 synergize to enhance epithelial cell dispersal, migration, and invasion, whereas knockdown of Pak4 attenuates these responses. A Gab1 mutant unable to recruit Pak4 fails to promote epithelial cell dispersal and an invasive morphogenic program in response to HGF, demonstrating a physiological requirement for Gab1-Pak4 association. These data demonstrate a novel association between Gab1 and Pak4 and identify Pak4 as a key integrator of cell migration and invasive growth downstream from the Met receptor.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Proto-Oncogene Proteins c-met/metabolism , p21-Activated Kinases/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Cell Communication/drug effects , Cell Line , Cell Movement/drug effects , Dogs , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Hepatocyte Growth Factor/pharmacology , Humans , Morphogenesis/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Pseudopodia/drug effects , Pseudopodia/enzymology , p21-Activated Kinases/chemistry
3.
J Cell Physiol ; 214(3): 694-705, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17894413

ABSTRACT

Gab1 and Gab2 are conserved scaffolding proteins that amplify and integrate signals stimulated by many growth factor receptors including the Met receptor. Gab1 acts to diversify the signal downstream from Met through the recruitment of multiple signaling proteins, and is essential for epithelial morphogenesis. However, whereas Gab1 and Gab2 are both expressed in epithelial cells, Gab2 fails to support a morphogenic response. We demonstrate that Gab1 and Gab2 are divergent in their function whereby Gab1, but not Gab2, promotes lamellipodia formation, and is localized to the membrane of lamellipodia upon Met activation. We have identified activation of ERK1/2 as a requirement for lamellipodia formation. Moreover, activated ERK1/2 are localized to lamellipodia in Gab1 expressing cells but not in cells that overexpress Gab2. By structure-function studies, we identify that enhanced membrane localization conferred through the addition of a myristoylation signal, together with the addition of the direct Met binding motif (MBM) from Gab1, are required to promote lamellipodia and confer a morphogenic signaling response to Gab2. Moreover, the morphogenesis competent myristoylated Gab2MBM promotes localization of activated ERK1/2 to the leading edge of lamellipodia in a similar manner to Gab1. Hence, subcellular localization of the Gab scaffold, as well as the ability of Gab to interact directly with the Met receptor, are both essential components of the morphogenic signaling response which involves lamellipodia formation and the localization of ERK1/2 activation in membrane ruffles.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Cell Shape , Epithelial Cells/cytology , Proto-Oncogene Proteins c-met/metabolism , Pseudopodia/enzymology , Amino Acid Motifs , Animals , Cell Line , Chickens , Dogs , Enzyme Activation , Epithelial Cells/enzymology , ErbB Receptors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction
4.
Mol Cell Biol ; 25(21): 9632-45, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227611

ABSTRACT

The Met receptor tyrosine kinase (RTK) regulates epithelial remodeling, dispersal, and invasion and is deregulated in many human cancers. It is now accepted that impaired down-regulation, as well as sustained activation, of RTKs could contribute to their deregulation. Down-regulation of the Met receptor involves ligand-induced internalization, ubiquitination by Cbl ubiquitin ligases, and lysosomal degradation. Here we report that a ubiquitination-deficient Met receptor mutant (Y1003F) is tumorigenic in vivo. The Met Y1003F mutant is internalized, and undergoes endosomal trafficking with kinetics similar to the wild-type Met receptor, yet is inefficiently targeted for degradation. This results in sustained activation of Met Y1003F and downstream signals involving the Ras-mitogen-activated protein kinase pathway, cell transformation, and tumorigenesis. Although Met Y1003F undergoes endosomal trafficking and localizes with the cargo-sorting protein Hrs, it is unable to induce phosphorylation of Hrs. Fusion of monoubiquitin to Met Y1003F is sufficient to decrease Met receptor stability and prevent sustained MEK1/2 activation. In addition, this rescues Hrs tyrosine phosphorylation and decreases transformation in a focus-forming assay. These results demonstrate that Cbl-dependent ubiquitination is dispensable for Met internalization but is critical to target the Met receptor to components of the lysosomal sorting machinery and to suppress its inherent transforming activity.


Subject(s)
Cell Transformation, Neoplastic , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-met/physiology , Ubiquitin/metabolism , Animals , Cell Line , Dogs , Endosomal Sorting Complexes Required for Transport , Endosomes/physiology , Enzyme Activation , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/metabolism , MAP Kinase Signaling System/physiology , Mice , Mutation , Phosphorylation , Protein Transport/physiology , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , ras Proteins/physiology
5.
Mol Biol Cell ; 16(2): 550-61, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15548598

ABSTRACT

Activation of the hepatocyte growth factor receptor Met induces a morphogenic response and stimulates the formation of branching tubules by Madin-Darby canine kidney (MDCK) epithelial cells in three-dimensional cultures. A constitutively activated ErbB2/Neu receptor, NeuNT, promotes a similar invasive morphogenic program in MDCK cells. Because both receptors are expressed in breast epithelia, are associated with poor prognosis, and hepatocyte growth factor (HGF) is expressed in stroma, we examined the consequence of cooperation between these signals. We show that HGF disrupts NeuNT-induced epithelial morphogenesis, stimulating the breakdown of cell-cell junctions, dispersal, and invasion of single cells. This correlates with a decrease in junctional proteins claudin-1 and E-cadherin, in addition to the internalization of the tight junction protein ZO-1. HGF-induced invasion of NT-expressing cells is abrogated by pretreatment with a pharmacological inhibitor of the mitogen-activated protein kinase kinase (MEK) pathway, which restores E-cadherin and ZO-1 at cell-cell junctions, establishing the involvement of MEK-dependent pathways in this process. These results demonstrate that physiological signals downstream from the HGF/Met receptor synergize with ErbB2/Neu to enhance the malignant phenotype, promoting the breakdown of cell-cell junctions and enhanced cell invasion. This is particularly important for cancers where ErbB2/Neu is overexpressed and HGF is a physiological growth factor found in the stroma.


Subject(s)
Cell Movement , Epithelial Cells/cytology , Genes, erbB-2 , Hepatocyte Growth Factor/metabolism , Morphogenesis , Animals , Anoikis , Blotting, Western , Cadherins/metabolism , Cell Line , Claudin-1 , Collagen/metabolism , Dogs , Drug Combinations , Extracellular Matrix/metabolism , Laminin/metabolism , Membrane Proteins/metabolism , Microscopy, Confocal , Models, Biological , Phosphoproteins/metabolism , Precipitin Tests , Proteoglycans/metabolism , Receptor, ErbB-2/metabolism , Tight Junctions , Zonula Occludens-1 Protein
6.
Proc Natl Acad Sci U S A ; 101(8): 2345-50, 2004 Feb 24.
Article in English | MEDLINE | ID: mdl-14983012

ABSTRACT

The etiology and progression of a variety of human malignancies are linked to the deregulation of receptor tyrosine kinases (RTKs). To define the role of RTK-dependent signals in various oncogenic processes, we have previously engineered RTK oncoproteins that recruit either the Shc or Grb2 adaptor proteins. Although these RTK oncoproteins transform cells with similar efficiencies, fibroblasts expressing the Shc-binding RTK oncoproteins induced tumors with short latency (approximately 7 days), whereas cells expressing the Grb2-binding RTK oncoproteins induced tumors with delayed latency (approximately 24 days). The early onset of tumor formation correlated with the ability of cells expressing the Shc-binding RTK oncoproteins to produce vascular endothelial growth factor (VEGF) in culture and an angiogenic response in vivo. Consistent with this, treatment with a VEGF inhibitor, VEGF-Trap, blocked the in vivo angiogenic and tumorigenic properties of these cells. The importance of Shc recruitment to RTKs for the induction of VEGF was further demonstrated by using mutants of the Neu/ErbB2 RTK, where the Shc, but not Grb2, binding mutant induced VEGF. Moreover, the use of fibroblasts derived from ShcA-deficient mouse embryos, demonstrated that Shc was essential for the induction of VEGF by the Met/hepatocyte growth factor RTK oncoprotein and by serum-derived growth factors. Together, our findings identify Shc as a critical angiogenic switch for VEGF production downstream from the Met and ErbB2 RTKs.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/genetics , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Blotting, Northern , Cloning, Molecular , Female , Fibroblasts/physiology , Humans , Mice , Mice, Nude , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
7.
Mol Biol Cell ; 14(4): 1691-708, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12686619

ABSTRACT

The hepatocyte growth factor receptor tyrosine kinase Met promotes cell dissociation and the inherent morphogenic program of epithelial cells. In a search for substrates downstream from Met, we have previously identified the Grb2-associated binder-1 (Gab1) as critical for the morphogenic program. Gab1 is a scaffold protein that acts to diversify the signal downstream from the Met receptor through its ability to couple with multiple signal transduction pathways. Gab1 contains a pleckstrin homology (PH) domain with specificity for phosphatidylinositol 3,4,5-trisphosphate. The phospholipid binding capacity of the Gab1 PH domain is required for the localization of Gab1 at sites of cell-cell contact in colonies of epithelial cells and for epithelial morphogenesis, suggesting that PH domain-dependent subcellular localization of Gab1 is a prerequisite for function. We have investigated the requirement for membrane localization of Gab1 for biological activity. We show that substitution of the Gab1 PH domain with the myristoylation signal from the c-Src protein is sufficient to replace the Gab1 PH domain for epithelial morphogenesis. The membrane targeting of Gab1 enhances Rac activity in the absence of stimulation and switches a nonmorphogenic noninvasive response to epidermal growth factor to a morphogenic invasive program. These results suggest that the subcellular localization of Gab1 is a critical determinant for epithelial morphogenesis and invasiveness.


Subject(s)
Phosphoproteins/metabolism , Actins/metabolism , Animals , Blood Proteins/chemistry , CSK Tyrosine-Protein Kinase , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Dogs , Epidermal Growth Factor/pharmacology , Epithelium/drug effects , Epithelium/growth & development , Epithelium/metabolism , Kinetics , Morphogenesis , Mutation , Myristic Acid/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , src-Family Kinases
8.
Mol Biol Cell ; 13(6): 2132-46, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12058075

ABSTRACT

The Gab family of docking proteins (Gab1 and Gab2) are phosphorylated in response to various cytokines and growth factors. Gab1 acts to diversify the signal downstream from the Met receptor tyrosine kinase through the recruitment of multiple signaling proteins, and is essential for epithelial morphogenesis. To determine whether Gab1 and Gab2 are functionally redundant, we have examined the role of Gab2 in epithelial cells. Both Gab1 and Gab2 are expressed in epithelial cells and localize to cell-cell junctions. However, whereas overexpression of Gab1 promotes a morphogenic response, the overexpression of Gab2 fails to induce this response. We show that Gab2 recruitment to the Met receptor is dependent on the Grb2 adapter protein. In contrast, Gab1 recruitment to Met is both Grb2 dependent and Grb2 independent. The latter requires a novel amino acid sequence present in the Met-binding domain of Gab1 but not Gab2. Mutation of these residues in Gab1 impairs both association with the Met receptor and the ability of Gab1 to promote a morphogenic response, whereas their insertion into Gab2 increases Gab2 association with Met, but does not confer on Gab2 the ability to promote epithelial morphogenesis. We propose that the Grb2-independent recruitment of Gab proteins to Met is necessary but not sufficient to promote epithelial morphogenesis.


Subject(s)
Epithelial Cells/physiology , Morphogenesis/physiology , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-met/physiology , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Cell Line , DNA Primers , Dogs , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphorylation , Recombinant Fusion Proteins/metabolism , Transfection
9.
Oncogene ; 21(12): 1800-11, 2002 Mar 14.
Article in English | MEDLINE | ID: mdl-11896612

ABSTRACT

Many human cancers have been associated with the deregulation of receptor tyrosine kinases (RTK). However, the individual contribution of receptor-associated signaling proteins in cellular transformation and metastasis is poorly understood. To examine the role of RTK activated signal transduction pathways to processes involved in cell transformation, we have exploited the oncogenic derivative of the Met RTK (Tpr-Met). Unlike other RTKs, twin tyrosine residues in the carboxy-terminal tail of the Met oncoprotein and receptor are required for all biological and transforming activities, and a mutant lacking these tyrosines is catalytically active but non transforming. Using this mutant we have inserted oligonucleotide cassettes, each encoding a binding site for a specific signaling protein derived from other RTKs. We have generated variant forms of the Tpr-Met oncoprotein with the ability to bind individually to the p85 subunit of PI3'K, PLCgamma, or to the Grb2 or Shc adaptor proteins. Variants that recruit the Shc or Grb2 adaptor proteins generated foci of morphologically transformed fibroblast cells and induced anchorage-independent growth, scattering of epithelial cells and experimental metastasis. In contrast, variants that bind and activate PI3'K or PLCgamma failed to generate readily detectable foci. Although cell lines expressing the PI3'K variant grew in soft-agar, these cells were non metastatic. Using this unique RTK oncoprotein model, we have established that Grb2 or Shc dependent signaling pathways are sufficient for cell transformation and metastatic spread.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Transformation, Neoplastic/metabolism , Intramolecular Transferases/metabolism , Isoenzymes/metabolism , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteins/metabolism , Signal Transduction/physiology , Type C Phospholipases/metabolism , Animals , Blotting, Western , Cell Adhesion/physiology , Cell Line , Colony-Forming Units Assay , Cricetinae , DNA Primers , Enzyme Activation , Epithelial Cells/metabolism , GRB2 Adaptor Protein , Humans , Intramolecular Transferases/genetics , Isoenzymes/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis, Site-Directed , Neoplasm Metastasis , Oncogene Proteins, Fusion/genetics , Phosphatidylinositol 3-Kinases/genetics , Phospholipase C gamma , Phosphoproteins/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Proteins/genetics , Rats , Rats, Inbred F344 , Type C Phospholipases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...