Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 162(4): 1242-1255.e11, 2022 04.
Article in English | MEDLINE | ID: mdl-34922945

ABSTRACT

BACKGROUND & AIMS: Acinar to ductal metaplasia is the prerequisite for the initiation of Kras-driven pancreatic ductal adenocarcinoma (PDAC), and candidate genes regulating this process are emerging from genome-wide association studies. The adaptor protein p130Cas emerged as a potential PDAC susceptibility gene and a Kras-synthetic lethal interactor in pancreatic cell lines; however, its role in PDAC development has remained largely unknown. METHODS: Human PDAC samples and murine KrasG12D-dependent pancreatic cancer models of increasing aggressiveness were used. p130Cas was conditionally ablated in pancreatic cancer models to investigate its role during Kras-induced tumorigenesis. RESULTS: We found that high expression of p130Cas is frequently detected in PDAC and correlates with higher histologic grade and poor prognosis. In a model of Kras-driven PDAC, loss of p130Cas inhibits tumor development and potently extends median survival. Deletion of p130Cas suppresses acinar-derived tumorigenesis and progression by means of repressing PI3K-AKT signaling, even in the presence of a worsening condition like pancreatitis. CONCLUSIONS: Our observations finally demonstrated that p130Cas acts downstream of Kras to boost the PI3K activity required for acinar to ductal metaplasia and subsequent tumor initiation. This demonstrates an unexpected driving role of p130Cas downstream of Kras through PI3K/AKT, thus indicating a rational therapeutic strategy of targeting the PI3K pathway in tumors with high expression of p130Cas.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Crk-Associated Substrate Protein , Pancreatic Neoplasms , Acinar Cells/pathology , Adenocarcinoma/pathology , Animals , Carcinogenesis , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , Crk-Associated Substrate Protein/metabolism , Genome-Wide Association Study , Humans , Metaplasia/pathology , Mice , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms
2.
Front Cell Dev Biol ; 9: 729093, 2021.
Article in English | MEDLINE | ID: mdl-34708040

ABSTRACT

p130Cas/BCAR1 is an adaptor protein devoid of any enzymatic or transcriptional activity, whose modular structure with various binding motifs, allows the formation of multi-protein signaling complexes. This results in the induction and/or maintenance of signaling pathways with pleiotropic effects on cell motility, cell adhesion, cytoskeleton remodeling, invasion, survival, and proliferation. Deregulation of p130Cas/BCAR1 adaptor protein has been extensively demonstrated in a variety of human cancers in which overexpression of p130Cas/BCAR1 correlates with increased malignancy. p140Cap (p130Cas associated protein), encoded by the SRCIN1 gene, has been discovered by affinity chromatography and mass spectrometry analysis of putative interactors of p130Cas. It came out that p140Cap associates with p130Cas not directly but through its interaction with the Src Kinase. p140Cap is highly expressed in neurons and to a lesser extent in epithelial tissues such as the mammary gland. Strikingly, in vivo and in vitro analysis identified its tumor suppressive role in breast cancer and in neuroblastoma, showing an inverse correlation between p140Cap expression in tumors and tumor progression. In this review, a synopsis of 15 years of research on the role of p130Cas/BCAR1 and p140Cap/SRCIN1 in breast cancer will be presented.

3.
Cell Death Differ ; 27(2): 790-807, 2020 02.
Article in English | MEDLINE | ID: mdl-31285546

ABSTRACT

Neuroblastoma is the most common extra-cranial pediatric solid tumor, responsible for 13-15% of pediatric cancer death. Its intrinsic heterogeneity makes it difficult to target for successful therapy. The adaptor protein p140Cap/SRCIN1 negatively regulates tumor cell features and limits breast cancer progression. This study wish to assess if p140Cap is a key biological determinant of neuroblastoma outcome. RNAseq profiles of a large cohort of neuroblastoma patients show that SRCIN1 mRNA levels are an independent risk factor inversely correlated to disease aggressiveness. In high-risk patients, CGH+SNP microarray analysis of primary neuroblastoma identifies SRCIN1 as frequently altered by hemizygous deletion, copy-neutral loss of heterozygosity, or disruption. Functional experiments show that p140Cap negatively regulates Src and STAT3 signaling, affects anchorage-independent growth and migration, in vivo tumor growth and spontaneous lung metastasis formation. p140Cap also increases sensitivity of neuroblastoma cells to doxorubicin and etoposide treatment, as well as to a combined treatment with chemotherapy drugs and Src inhibitors. Our functional findings point to a causal role of p140Cap in curbing the aggressiveness of neuroblastoma, due to its ability to impinge on specific molecular pathways, and to sensitize cells to therapeutic treatment. This study provides the first evidence that the SRCIN1/p140Cap adaptor protein is a key player in neuroblastoma as a new independent prognostic marker for patient outcome and treatment. Altogether, these data highlight the potential clinical impact of SRCIN1/p140Cap expression in neuroblastoma tumors, in terms of reducing cytotoxic effects of chemotherapy, one of the main issues for pediatric tumor treatment.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Biomarkers, Tumor/metabolism , Lung Neoplasms/secondary , Neuroblastoma/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Biomarkers, Tumor/genetics , Cell Proliferation , Cell Survival , Humans , Infant , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neuroblastoma/diagnosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
5.
Sci Rep ; 9(1): 17729, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31758081

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Sci Rep ; 9(1): 3089, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816273

ABSTRACT

The ErbB2 receptor tyrosine kinase is overexpressed in approximately 15-20% of breast tumors and associated with aggressive disease and poor clinical outcome. p130Cas represents a nodal scaffold protein regulating cell survival, migration and proliferation in normal and pathological contexts. p130Cas overexpression in ErbB2 human breast cancer correlates with poor prognosis and metastasis formation. Recent data indicate that p130Cas association to ErbB2 protects ErbB2 from degradation, thus enhancing tumorigenesis. Therefore, inhibiting p130Cas/ErbB2 interaction might represent a new therapeutic strategy to target breast cancer. Here we demonstrate by performing Molecular Modeling, Molecular Dynamics, dot blot, ELISA and fluorescence quenching experiments, that p130Cas binds directly to ErbB2. Then, by structure-based virtual screening, we identified two potential inhibitors of p130Cas/ErbB2 interaction. Their experimental validation was performed in vitro and in ErbB2-positive breast cancer cellular models. The results highlight that both compounds interfere with p130Cas/ErbB2 binding and significantly affect cell proliferation and sensitivity to Trastuzumab. Overall, this study identifies p130Cas/ErbB2 complex as a potential breast cancer target revealing new therapeutic perspectives for protein-protein interaction (PPI).


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Crk-Associated Substrate Protein/metabolism , Drug Discovery , Protein Binding/drug effects , Receptor, ErbB-2/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cloning, Molecular , Drug Resistance, Neoplasm/drug effects , Escherichia coli/genetics , Female , HEK293 Cells , Humans , Trastuzumab/therapeutic use
7.
Cell Commun Signal ; 16(1): 73, 2018 11 03.
Article in English | MEDLINE | ID: mdl-30390666

ABSTRACT

BACKGROUND: p130 Crk-associated substrate (p130CAS; also known as BCAR1) is a scaffold protein that modulates many essential cellular processes such as cell adhesion, proliferation, survival, cell migration, and intracellular signaling. p130Cas has been shown to be highly expressed in a variety of human cancers of epithelial origin. However, few data are available regarding the role of p130Cas during normal epithelial development and homeostasis. METHODS: To this end, we have generated a genetically modified mouse in which p130Cas protein was specifically ablated in the epidermal tissue. RESULTS: By using this murine model, we show that p130Cas loss results in increased cell proliferation and reduction of cell adhesion to extracellular matrix. In addition, epidermal deletion of p130Cas protein leads to premature expression of "late" epidermal differentiation markers, altered membrane E-cadherin/catenin proteins localization and aberrant tyrosine phosphorylation of E-cadherin/catenin complexes. Interestingly, these alterations in adhesive properties in absence of p130Cas correlate with abnormalities in progenitor cells balance resulting in the amplification of a more committed cell population. CONCLUSION: Altogether, these results provide evidence that p130Cas is an important regulator of epidermal cell fate and homeostasis.


Subject(s)
Cell Adhesion , Cell Differentiation , Crk-Associated Substrate Protein/deficiency , Crk-Associated Substrate Protein/genetics , Epidermis/metabolism , Gene Deletion , Homeostasis/genetics , Animals , Cell Proliferation , Extracellular Matrix/metabolism , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Phenotype
8.
Cell Commun Signal ; 16(1): 90, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30477510

ABSTRACT

Following publication of the original article [1], the authors reported an error in the name of the 11th author. The author's name was incorrectly published as "Vincenzo Calautti", instead of "Enzo Calautti".

10.
Sci Rep ; 7(1): 1145, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28442738

ABSTRACT

ErbB2 overexpression is detected in approximately 20% of breast cancers and is correlated with poor survival. It was previously shown that the adaptor protein p130Cas/BCAR1 is a crucial mediator of ErbB2 transformation and that its overexpression confers invasive properties to ErbB2-positive human mammary epithelial cells. We herein prove, for the first time, that the transcriptional repressor Blimp1 is a novel mediator of p130Cas/ErbB2-mediated invasiveness. Indeed, high Blimp1 expression levels are detected in invasive p130Cas/ErbB2 cells and correlate with metastatic status in human breast cancer patients. The present study, by using 2D and 3D breast cancer models, shows that the increased Blimp1 expression depends on both MAPK activation and miR-23b downmodulation. Moreover, we demonstrate that Blimp1 triggers cell invasion and metastasis formation via its effects on focal adhesion and survival signaling. These findings unravel the previously unidentified role that transcriptional repressor Blimp1 plays in the control of breast cancer invasiveness.


Subject(s)
Breast Neoplasms/pathology , Crk-Associated Substrate Protein/metabolism , Gene Expression Regulation , Neoplasm Invasiveness , Positive Regulatory Domain I-Binding Factor 1/metabolism , Receptor, ErbB-2/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice
11.
Nat Commun ; 8: 14797, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28300085

ABSTRACT

The docking protein p140Cap negatively regulates tumour cell features. Its relevance on breast cancer patient survival, as well as its ability to counteract relevant cancer signalling pathways, are not fully understood. Here we report that in patients with ERBB2-amplified breast cancer, a p140Cap-positive status associates with a significantly lower probability of developing a distant event, and a clear difference in survival. p140Cap dampens ERBB2-positive tumour cell progression, impairing tumour onset and growth in the NeuT mouse model, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. One major mechanism is the ability of p140Cap to interfere with ERBB2-dependent activation of Rac GTPase-controlled circuitries. Our findings point to a specific role of p140Cap in curbing the aggressiveness of ERBB2-amplified breast cancers and suggest that, due to its ability to impinge on specific molecular pathways, p140Cap may represent a predictive biomarker of response to targeted anti-ERBB2 therapies.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptor, ErbB-2/genetics , rac GTP-Binding Proteins/genetics
12.
Gastric Cancer ; 20(4): 629-639, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27995483

ABSTRACT

BACKGROUND: Most metastatic gastrointestinal stromal tumors (GISTs) develop resistance to the first-line imatinib treatment. Recently, increased vessel density and angiogenic markers were reported in GISTs with a poor prognosis, suggesting that angiogenesis is implicated in GIST tumor progression and resistance. The purpose of this study was to investigate the relationship between tumor vasculature and imatinib resistance in different GIST mouse models using a noninvasive magnetic resonance imaging (MRI) functional approach. METHODS: Immunodeficient mice (n = 8 for each cell line) were grafted with imatinib-sensitive (GIST882 and GIST-T1) and imatinib-resistant (GIST430) human cell lines. Dynamic contrast-enhanced MRI (DCE-MRI) was performed on GIST xenografts to quantify tumor vessel permeability (K trans) and vascular volume fraction (v p). Microvessel density (MVD), permeability (mean dextran density, MDD), and angiogenic markers were evaluated by immunofluorescence and western blot assays. RESULTS: Dynamic contrast-enhanced magnetic resonance imaging showed significantly increased vessel density (P < 0.0001) and permeability (P = 0.0002) in imatinib-resistant tumors compared to imatinib-sensitive ones. Strong positive correlations were observed between MRI estimates, K trans and v p, and their related ex vivo values, MVD (r = 0.78 for K trans and r = 0.82 for v p) and MDD (r = 0.77 for K trans and r = 0.94 for v p). In addition, higher expression of vascular endothelial growth factor receptors (VEGFR2 and VEFGR3) was seen in GIST430. CONCLUSIONS: Dynamic contrast-enhanced magnetic resonance imaging highlighted marked differences in tumor vasculature and microenvironment properties between imatinib-resistant and imatinib-sensitive GISTs, as also confirmed by ex vivo assays. These results provide new insights into the role that DCE-MRI could play in GIST characterization and response to GIST treatment. Validation studies are needed to confirm these findings.


Subject(s)
Drug Resistance, Neoplasm , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/pathology , Neovascularization, Pathologic/diagnostic imaging , Animals , Antineoplastic Agents , Cell Line, Tumor , Contrast Media , Heterografts , Humans , Imatinib Mesylate , Magnetic Resonance Imaging/methods , Male , Mice
13.
Oncotarget ; 7(4): 4442-53, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26716506

ABSTRACT

Overexpression of the ErbB2/HER2 receptor tyrosine kinase occurs in up to 20% of human breast cancers and correlates with aggressive disease. Several efficacious targeted therapies, including antibodies and kinase inhibitors, have been developed but the occurring of resistance to these agents is often observed. New therapeutic agents targeting the endocytic recycling and intracellular trafficking of membrane in tumor cells overexpressing ErbB2 are actually in clinical development. Nevertheless the mechanisms underlying ErbB2 downregulation are still obscure. We have previously demonstrated that the overexpression of the p130Cas adaptor protein in ErbB2 positive breast cancer, promotes tumor aggressiveness and progression. Here we demonstrate that lowering p130Cas expression in breast cancer cells is sufficient to induce ErbB2 degradation by autophagy. Conversely, p130Cas overexpression protects ErbB2 from degradation by autophagy. Furthermore, this autophagy-dependent preferential degradation of ErbB2 in absence of p130Cas is due to an increased ErbB2 ubiquitination. Indeed, the overexpression of p130Cas impairs ErbB2 ubiquitination by inhibiting the binding of Cbl and CHIP E3 ligases to ErbB2. Finally, our results indicate that p130Cas-dependent ErbB2 protection from degradation by autophagy may alter the sensitivity to the humanized monoclonal antibody trastuzumab. Consistently, in human ErbB2 positive breast cancers that develop resistance to trastuzumab, p130Cas expression is significantly increased suggesting that elevated levels of p130Cas can be involved in trastuzumab resistance.


Subject(s)
Autophagy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Crk-Associated Substrate Protein/metabolism , Drug Resistance, Neoplasm , Receptor, ErbB-2/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cell Proliferation , Crk-Associated Substrate Protein/genetics , Female , Humans , Immunoenzyme Techniques , Immunoprecipitation , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Protein Stability , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trastuzumab/pharmacology , Tumor Cells, Cultured
14.
Oncotarget ; 6(31): 30859-75, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26356564

ABSTRACT

High-grade epithelial ovarian cancer (HGEOC) is a clinically diverse and molecularly heterogeneous disease comprising subtypes with distinct biological features and outcomes. The receptor tyrosine kinases, expressed by EOC cells, and their ligands, present in the microenvironment, activate signaling pathways, which promote EOC cells dissemination. Herein, we established a molecular link between the presence of Gas6 ligand in the ascites of HGEOCs, the expression and activation of its receptor Axl in ovarian cancer cell lines and biopsies, and the progression of these tumors. We demonstrated that Gas6/Axl signalling converges on the integrin ß3 pathway in the presence of the adaptor protein p130Cas, thus inducing tumor cell adhesion to the extracellular matrix and invasion. Accordingly, Axl and p130Cas were significantly co-expressed in HGEOC samples. Clinically, we identified an Axl-associated signature of 62 genes able to portray the HGEOCs with the shortest overall survival. These data biologically characterize a group of HGEOCs and could help guide a more effective therapeutic approach to be taken for these patients.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Ascites/metabolism , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Ovarian Epithelial , Cell Adhesion , Cell Line, Tumor , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Integrin beta3/genetics , Integrin beta3/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Grading , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/mortality , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Predictive Value of Tests , Protein Interaction Maps , Proto-Oncogene Proteins/metabolism , RNA Interference , Receptor Protein-Tyrosine Kinases/metabolism , Survival Analysis , Time Factors , Transfection , Treatment Outcome , Axl Receptor Tyrosine Kinase
15.
Gene ; 562(1): 1-7, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25727852

ABSTRACT

BCAR1 (also known as p130Cas/BCAR1) is an adaptor protein that belongs to the CAS family of scaffold proteins. In the past years, increasing evidence has demonstrated the ability of p130Cas/BCAR1 to activate signaling originating from mechanical stimuli, cell-extracellular matrix (ECM) adhesion and growth factor stimulation cascades during normal development and disease in various biological models. In this review we will specifically discuss the more recent data on the contribution of p130Cas/BCAR1 in the regulation of tissue homeostasis and its potential implications in pathological conditions.


Subject(s)
Crk-Associated Substrate Protein/genetics , Gene Expression Regulation , Morphogenesis/genetics , Neoplasms/genetics , Animals , Cell Adhesion , Cell Movement , Crk-Associated Substrate Protein/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Extracellular Matrix , Homeostasis , Humans , Mechanotransduction, Cellular , Mice , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity , Phosphorylation , Protein Structure, Tertiary
16.
Cell Cycle ; 14(9): 1426-37, 2015.
Article in English | MEDLINE | ID: mdl-25723869

ABSTRACT

The proteins of the Dbl family are guanine nucleotide exchange factors (GEFs) of Rho GTPases and are known to be involved in cell growth regulation. Alterations of the normal function of these proteins lead to pathological processes such as developmental disorders, neoplastic transformation, and tumor metastasis. We have previously demonstrated that expression of Dbl oncogene in lens epithelial cells modulates genes encoding proteins involved in epithelial-mesenchymal-transition (EMT) and induces angiogenesis in the lens. Our present study was undertaken to investigate the role of Dbl oncogene in epithelial cells transformation, providing new insights into carcinoma progression.To assess how Dbl oncogene can modulate EMT, cell migration, morphogenesis, and expression of pro-apoptotic and angiogenic factors we utilized bi- and 3-dimensional cultures of MCF-10 A cells. We show that upon Dbl expression MCF-10 A cells undergo EMT. In addition, we found that Dbl overexpression sustains Cdc42 and Rac activation inducing morphological alterations, characterized by the presence of lamellipodia and conferring a high migratory capacity to the cells. Moreover, Dbl expressing MCF-10 A cells form altered 3D structures and can induce angiogenesis by producing proangiogenic factors such as CCL2. These results support a role for Dbl oncogene in epithelial cell differentiation and transformation and suggest the relevance of GEF deregulation in tumor onset and progression.


Subject(s)
Acinar Cells/enzymology , Angiogenic Proteins/metabolism , Breast Neoplasms/enzymology , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/enzymology , Epithelial-Mesenchymal Transition , Guanine Nucleotide Exchange Factors/metabolism , Mammary Glands, Human/enzymology , Proto-Oncogene Proteins/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Apoptosis , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation , Cell Line , Cell Movement , Cell Shape , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chemokine CCL2/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Guanine Nucleotide Exchange Factors/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Neovascularization, Physiologic , Proto-Oncogene Proteins/genetics , Signal Transduction , Transfection , Up-Regulation , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism
17.
J Pathol ; 234(2): 152-63, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24615293

ABSTRACT

Morgana/CHP-1 is a ubiquitously expressed protein able to inhibit ROCK II kinase activity. We have previously demonstrated that morgana haploinsufficiency leads to multiple centrosomes, genomic instability, and higher susceptibility to tumour development. While a large fraction of human cancers has shown morgana down-regulation, a small subset of tumours was shown to express high morgana levels. Here we demonstrate that high morgana expression in different breast cancer subtypes correlates with high tumour grade, mitosis number, and lymph node positivity. Moreover, morgana overexpression induces transformation in NIH-3T3 cells and strongly protects them from various apoptotic stimuli. From a mechanistic point of view, we demonstrate that morgana causes PTEN destabilization, by inhibiting ROCK activity, hence triggering the PI3K/AKT survival pathway. In turn, morgana down-regulation in breast cancer cells that express high morgana levels increases PTEN expression and leads to sensitization of cells to chemotherapy.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction/physiology , rho-Associated Kinases/metabolism , Animals , Breast Neoplasms/pathology , Centrosome/pathology , Down-Regulation/physiology , Female , Humans , Mice , Molecular Chaperones , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism
18.
Stem Cells ; 32(3): 754-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24123662

ABSTRACT

Signals downstream of Akt can either favor or oppose stem cell (SC) maintenance, but how this dual role can be achieved is still undefined. Using human limbal keratinocyte stem cells (LKSCs), a SC type used in transplantation therapies for corneal regeneration, we show that Akt signaling is prominent in SC populations both in vivo and in vitro, and that Akt1 promotes while Akt2 opposes SC self-renewal. Noteworthy, loss of Akt2 signaling enhances LKSC maintenance ex vivo, whereas Akt1 depletion anticipates SC exhaustion. Mechanistically, the antagonistic functions of Akt1 and Akt2 in SC control are mainly dictated by their differential subcellular distribution, being nuclear Akt2 selectively implicated in FOXO inhibition. Akt2 downregulation favors LKSC maintenance as a result of a gain of FOXO functions, which attenuates the mechanistic target of rapamycin complex one signaling via tuberous sclerosis one gene induction, and promotes growth factor signaling through Akt1. Consistently, Akt2 deficiency also enhances limbal SCs in vivo. Thus, our findings reveal distinct roles for nuclear versus cytosolic Akt signaling in normal epithelial SC control and suggest that the selective Akt2 inhibition may provide novel pharmacological strategies for human LKSC expansion in therapeutic settings and mechanistic research.


Subject(s)
Cell Nucleus/enzymology , Forkhead Transcription Factors/metabolism , Keratinocytes/cytology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology , TOR Serine-Threonine Kinases/metabolism , 3T3 Cells , Adult , Animals , Cell Proliferation , Clone Cells , Enzyme Activation , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Humans , Isoenzymes/metabolism , Limbus Corneae/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/deficiency , Repressor Proteins/metabolism , Signal Transduction , Stem Cells/enzymology , Transcription, Genetic
19.
Breast Cancer Res ; 16(5): 443, 2014.
Article in English | MEDLINE | ID: mdl-25606587

ABSTRACT

The members of the Cas protein family (p130Cas/BCAR1, Nedd9/HEF1, EFS and CASS4) are scaffold proteins required for the assembly of signal transduction complexes in response to several stimuli, such as growth factors, hormones and extracellular matrix components. Given their ability to integrate and coordinate multiple signalling events, Cas proteins have emerged as crucial players in the control of mammary cell proliferation, survival and differentiation. More importantly, it has been found that alterations of their expression levels result in aberrant signalling cascades, which promote initiation and progression of breast cancer. Based on the increasing data from in vitro, mouse model and clinical studies, in this review we will focus on two Cas proteins, p130Cas/BCAR1 and Nedd9, and their coupled signalling pathways, to examine their role in mammary cell transformation and in the acquirement of invasiveness and drug resistance of breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Crk-Associated Substrate Protein/physiology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Drug Resistance, Neoplasm , Female , Humans , Mammary Glands, Human/pathology , Neoplasm Invasiveness , Signal Transduction
20.
Am J Cancer Res ; 3(3): 290-301, 2013.
Article in English | MEDLINE | ID: mdl-23841028

ABSTRACT

p140Cap is an adaptor protein that negatively controls tumor cell properties, by inhibiting in vivo tumor growth and metastasis formation. Our previous data demonstrated that p140Cap interferes with tumor growth and impairs invasive properties of cancer cells inactivating signaling pathways, such as the tyrosine kinase Src or E-cadherin/EGFR cross-talk. In breast cancer p140Cap expression inversely correlates with tumor malignancy. p140Cap is composed of several conserved domains that mediate association with specific partners. Here we focus our attention on two domains of p140Cap, the TER (Tyrosine Enriched Region) which includes several tyrosine residues, and the CT (Carboxy Terminal) which contains a proline rich sequence, involved in binding to SH2 and SH3 domains, respectively. By generating stable cell lines expressing these two proteins, we demonstrate that both TER and CT domains maintain the ability to associate the C-terminal Src kinase (Csk) and Src, to inhibit Src activation and Focal adhesion kinase (Fak) phosphorylation, and to impair in vitro and in vivo tumor cell features. In particular expression of TER and CT proteins in cancer cells inhibits in vitro and in vivo growth and directional migration at a similar extent of the full length p140Cap protein. Moreover, by selective point mutations and deletion we show that the ability of the modules to act as negative regulators of cell migration and proliferation mainly resides on the two tyrosines (Y) inserted in the EPLYA and EGLYA sequences in the TER module and in the second proline-rich stretch contained in the CT protein. Gene signature of cells expressing p140Cap, TER or CT lead to the identification of a common pattern of 105 down-regulated and 128 up-regulated genes, suggesting that the three proteins can act through shared pathways. Overall, this work highlights that the TER and CT regions of p140Cap can efficiently suppress tumor cell properties, opening the perspective that short, defined p140Cap regions can have therapeutic effects.

SELECTION OF CITATIONS
SEARCH DETAIL
...