Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Pathologica ; 116(1): 32-45, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38482673

ABSTRACT

Introduction: Endometrial carcinoma (EC) is the commonest gynecological cancer affecting women in Western populations. To predict patient risk, the 2020 edition of the World Health Organization (WHO) Classification of Tumors of the Female Genital Tract stressed the importance of integrated histo-molecular classification of the disease. This survey analysis poses attention on the most frequently used immunohistochemical and molecular markers adopted in daily categorization of ECs in European laboratories. Methods: We analyzed data collected through questionnaires administered to 40 Italian, 20 Spanish, 3 Swiss and 6 United Kingdom (UK) laboratories. We collected information regarding daily practice in EC evaluation, specifically concerning mismatch repair status (MMR) and microsatellite instability (MSI). Summary and descriptive statistical analyses were carried out to evaluate the current practice of each laboratory. Results: The results show that MMR status is mainly evaluated by using immunohistochemistry (IHC) on most EC samples. The most frequent approach for the analysis of MMR status is IHC of four proteins (PMS2, MSH6, MSH2, MLH1). MSI analysis by molecular methods is uncommon but useful as a supplemental tool in specific conditions. MLH1 promoter hypermethylation and BRAF V600 mutations analysis are performed in case of negative expression of MLH1/PMS2. Other markers (mainly p53 followed by POLE and PTEN) are investigated in particular in Spain and Switzerland in a consistent number of cases. Conclusion: Guidelines consultation and standardization of laboratory procedures are efficient means for EC prognostic risk stratification and improving the quality of care.


Subject(s)
Endometrial Neoplasms , Female , Humans , Mismatch Repair Endonuclease PMS2 , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Biomarkers , Europe
2.
Pathologica ; 114(3): 189-198, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35775705

ABSTRACT

Objective: Endometrial carcinoma (EC) is the most common gynecological malignant disease in high income countries. The 2020 edition of the World Health Organization (WHO) Classification of Tumors of the Female Genital Tract underlines the important clinical implications of the new integrated histo-molecular classification system, in order to correctly define the specific prognostic risk group. This survey analysis will focus on the most commonly adopted immunohistochemical and molecular biomarkers used in daily clinical characterization of a diagnosed endometrial carcinoma in Italian labs. Methods: An evaluation questionnaire was distributed to 41 Italian pathology laboratories. Normal habits in EC evaluation, especially regarding mismatch repair status (MMR) and microsatellite instability (MSI), were collected. A summary and a descriptive statistical analysis were used to show the current practice of each laboratory. Results: The analysis of MMR status by immunohistochemistry (IHC) is carried out on the majority of all EC samples. The most frequent strategy for the analysis of MMR status in EC is IHC of four proteins (PMS2, MSH6, MSH2, MLH1). MSI analysis by molecular method in endometrial cancer is rarer and more restricted to some circumstances. Hypermethylation of the MLH1 promoter by methylation-specific PCR and pyrosequencing was analyzed in case of negative expression of MLH1/PMS2. Also, the analysis of p53 in EC is performed in the majority of cases. POLE mutational profiling is adopted only in a limited number of laboratories. Fifty-five percent of Italian laboratories refer to national/international guidelines when analyzing biomarkers in EC (among those, 45% use the ESGO Guidelines, 18% ASCO-CAP, 18% AIOM, 14% WHO, 5% British Association of Gynaecological Pathologist, 5% ESMO, 5% NCCN). Conclusions: Adoption of guidelines and standardization of pre-analytical and analytical procedures are effective tools for adequate EC prognostic risk stratification and high quality standard of care.


Subject(s)
Data Analysis , Endometrial Neoplasms , Female , Humans , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Mismatch Repair Endonuclease PMS2/metabolism , Surveys and Questionnaires , Practice Guidelines as Topic
3.
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
4.
Mol Cell Ther ; 3: 8, 2015.
Article in English | MEDLINE | ID: mdl-26464805

ABSTRACT

BACKGROUND: Medulloblastoma (MB) is the most common malignant childhood brain tumor with the propensity to disseminate at an early stage, and is associated with high morbidity. New treatment strategies are needed to improve cure rates and to reduce life-long cognitive and functional deficits associated with current therapies. Extracellular Vesicles (EVs) are important players in cell-to-cell communication in health and diseases. A clearer understanding of cell-to-cell communication in tumors can be achieved by studying EV secretion in medullospheres. This can reveal subtle modifications induced by the passage from adherent to non-adherent growth, as spheres may account for the adaptation of tumor cells to the mutated environment. METHODS: Formation of medullospheres from MB cell lines stabilized in adherent conditions was obtained through culture conditioning based on low attachment flasks and specialized medium. EVs collected by ultracentrifugation, in adherent conditions and as spheres, were subjected to electron microscopy, NanoSight measurements and proteomics. RESULTS: Interestingly, iron carrier proteins were only found in EVs shed by CSC-enriched tumor cell population of spheres. We used iron chelators when culturing MB cell lines as spheres. Iron chelators induced a decrease in number/size of spheres and in stem cell populations able to initiate in vitro spheres formation. CONCLUSIONS: This work suggests a not yet identified role of iron metabolism in MB progression and invasion and opens the possibility to use chelators as adjuvants in anti-tumoral chemotherapy.

5.
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
6.
Cell Cycle ; 12(15): 2409-22, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23839042

ABSTRACT

Understanding transcriptional changes during cancer progression is of crucial importance to develop new and more efficacious diagnostic and therapeutic approaches. It is well known that ErbB2 is overexpressed in about 25% of human invasive breast cancers. We have previously demonstrated that p130Cas overexpression synergizes with ErbB2 in mammary cell transformation and promotes ErbB2-dependent invasion in three-dimensional (3D) cultures of human mammary epithelial cells. Here, by comparing coding and non-coding gene expression profiles, we define the invasive signatures associated with concomitant p130Cas overexpression and ErbB2 activation in 3D cultures of mammary epithelial cells. Specifically, we have found that genes involved in amino acids synthesis (CBS, PHGDH), cell motility, migration (ITPKA, PRDM1), and angiogenesis (HEY1) are upregulated, while genes involved in inflammatory response (SAA1, S100A7) are downregulated. In parallel, we have shown that the expression of specific miRNAs is altered. Among these, miR-200b, miR-222, miR-221, miR-R210, and miR-424 are upregulated, while miR-27a, miR-27b, and miR-23b are downregulated. Overall, this study presents, for the first time, the gene expression changes underlying the invasive behavior following p130Cas overexpression in an ErbB2 transformed mammary cell model.


Subject(s)
Crk-Associated Substrate Protein/metabolism , Epithelial Cells/metabolism , Mammary Glands, Human/pathology , Receptor, ErbB-2/metabolism , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Movement , Epithelial Cells/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , Methionine Sulfoxide Reductases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Microfilament Proteins , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Phosphoglycerate Dehydrogenase/metabolism , S100 Calcium Binding Protein A7 , S100 Proteins/metabolism , Transcription Factors/metabolism , Transcriptome , Up-Regulation
7.
Stem Cells ; 31(7): 1422-33, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592522

ABSTRACT

It has recently been proposed that defective differentiation of mammary luminal progenitors predisposes to basal-like breast cancer. However, the molecular and cellular mechanisms involved are still unclear. Here, we describe that the adaptor protein p130Cas is a crucial regulator of mouse mammary epithelial cell (MMEC) differentiation. Using a transgenic mouse model, we show that forced p130Cas overexpression in the luminal progenitor cell compartment results in the expansion of luminal cells, which aberrantly display basal cell features and reduced differentiation in response to lactogenic stimuli. Interestingly, MMECs overexpressing p130Cas exhibit hyperactivation of the tyrosine kinase receptor c-Kit. In addition, we demonstrate that the constitutive c-Kit activation alone mimics p130Cas overexpression, whereas c-Kit downregulation is sufficient to re-establish proper differentiation of p130Cas overexpressing cells. Overall, our data indicate that high levels of p130Cas, via abnormal c-Kit activation, promote mammary luminal cell plasticity, thus providing the conditions for the development of basal-like breast cancer. Consistently, p130Cas is overexpressed in human triple-negative breast cancer, further suggesting that p130Cas upregulation may be a priming event for the onset of basal-like breast cancer.


Subject(s)
Crk-Associated Substrate Protein/metabolism , Mammary Glands, Animal/cytology , Mammary Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-kit/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cells, Cultured , Crk-Associated Substrate Protein/biosynthesis , Crk-Associated Substrate Protein/genetics , Female , Humans , Immunohistochemistry , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Pregnancy , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
8.
PLoS One ; 7(12): e49817, 2012.
Article in English | MEDLINE | ID: mdl-23239970

ABSTRACT

p130Cas adaptor protein regulates basic processes such as cell cycle control, survival and migration. p130Cas over-expression has been related to mammary gland transformation, however the in vivo consequences of p130Cas over-expression during mammary gland morphogenesis are not known. In ex vivo mammary explants from MMTV-p130Cas transgenic mice, we show that p130Cas impairs the functional interplay between Epidermal Growth Factor Receptor (EGFR) and Estrogen Receptor (ER) during mammary gland development. Indeed, we demonstrate that p130Cas over-expression upon the concomitant stimulation with EGF and estrogen (E2) severely impairs mammary morphogenesis giving rise to enlarged multicellular spherical structures with altered architecture and absence of the central lumen. These filled acinar structures are characterized by increased cell survival and proliferation and by a strong activation of Erk1/2 MAPKs and Akt. Interestingly, antagonizing the ER activity is sufficient to re-establish branching morphogenesis and normal Erk1/2 MAPK activity. Overall, these results indicate that high levels of p130Cas expression profoundly affect mammary morphogenesis by altering epithelial architecture, survival and unbalancing Erk1/2 MAPKs activation in response to growth factors and hormones. These results suggest that alteration of morphogenetic pathways due to p130Cas over-expression might prime mammary epithelium to tumorigenesis.


Subject(s)
Crk-Associated Substrate Protein , Epidermal Growth Factor , Estrogens , Mammary Glands, Animal , Animals , Cell Transformation, Neoplastic , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism , Epidermal Growth Factor/metabolism , Epithelium/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Morphogenesis/genetics , Organ Culture Techniques , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
9.
Breast Cancer Res ; 14(5): R137, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23098208

ABSTRACT

INTRODUCTION: Intrinsic plasticity of breast carcinoma cells allows them to undergo a transient and reversible conversion into mesenchymal cells to disseminate into distant organs, where they can re-differentiate to an epithelial-like status to form a cohesive secondary mass. The p130Cas scaffold protein is overexpressed in human ER+ and HER2+ breast cancer where it contributes to cancer progression, invasion and resistance to therapy. However, its role in regulating mesenchymal aggressive breast cancer cells remains to be determined. The aim of this study was to investigate the molecular and functional involvement of this adaptor protein in breast cancer cell plasticity. METHODS: We used silencing strategies and rescue experiments to evaluate phenotypic and biochemical changes from mesenchymal to epithelial traits in breast tumor cell lines. In the mouse A17 cell model previously related to mesenchymal cancer stem cells and basal-like breast cancer, we biochemically dissected the signaling pathways involved and performed functional in vivo tumor growth ability assays. The significance of the signaling platform was assessed in a human setting through the use of specific inhibitors in aggressive MDA-MB-231 subpopulation LM2-4175 cells. To evaluate the clinical relevance of the results, we analyzed publicly available microarray data from the Netherlands Cancer Institute and from the Koo Foundation Sun Yat-Sen Cancer Center. RESULTS: We show that p130Cas silencing induces loss of mesenchymal features, by downregulating Vimentin, Snail, Slug and Twist transcriptional factors, resulting in the acquirement of epithelial-like traits. Mechanistically, p130Cas controls Cyclooxygenase-2 transcriptional expression, which in turn contributes to p130Cas-dependent maintenance of mesenchymal phenotype. This cascade of events also compromises in vivo tumor growth through inhibition of cell signaling controlling cell cycle progression. c-Src and JNK kinases are sequential players in p130Cas/ Cyclooxygenase-2 axis and their pharmacological inhibition is sufficient to downregulate Cyclooxygenase-2 leading to an epithelial phenotype. Finally, in silico microarray data analysis indicates that p130Cas and Cyclooxygenase-2 concomitant overexpression predicts poor survival and high probability of breast tumor recurrence. CONCLUSIONS: Overall, these data identify a new p130Cas/Cyclooxygenase-2 axis as a crucial element in the control of breast tumor plasticity, opening new therapeutic strategies leading to inhibition of these pathways in aggressive breast carcinoma.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crk-Associated Substrate Protein/metabolism , Cyclooxygenase 2/metabolism , Animals , Breast Neoplasms/genetics , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Crk-Associated Substrate Protein/genetics , Cyclooxygenase 2/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Models, Biological , Phenotype , Quantitative Trait, Heritable , src-Family Kinases/metabolism
10.
Am J Cancer Res ; 1(5): 663-73, 2011.
Article in English | MEDLINE | ID: mdl-21994904

ABSTRACT

The assembly of molecular hubs upon integrin and growth factor stimulation represents a preferential way to transduce signals throughout the cell. Among the intracellular kinases that are responsive to integrin and growth factor activation, Src Family Kinases (SFKs) are crucial regulators of cell migration and invasion. Increasing evidence highlight the importance of adaptor proteins in these processes, based on their ability to create signalling platforms that control downstream signals. Among these adaptors we will discuss the molecular features of p130Cas and p140Cap proteins in terms of regulation of cell migration and invasion in normal and transformed cells.

11.
Eur J Cell Biol ; 90(2-3): 237-48, 2011.
Article in English | MEDLINE | ID: mdl-20961652

ABSTRACT

ErbB2 over-expression is detected in approximately 25% of invasive breast cancers and is strongly associated with poor patient survival. We have previously demonstrated that p130Cas adaptor is a crucial mediator of ErbB2 transformation. Here, we analysed the molecular mechanisms through which p130Cas controls ErbB2-dependent invasion in three-dimensional cultures of mammary epithelial cells. Concomitant p130Cas over-expression and ErbB2 activation enhance PI3K/Akt and Erk1/2 MAPK signalling pathways and promote invasion of mammary acini. By using pharmacological inhibitors, we demonstrate that both signalling cascades are required for the invasive behaviour of p130Cas over-expressing and ErbB2 activated acini. Erk1/2 MAPK and PI3K/Akt signalling triggers invasion through distinct downstream effectors involving mTOR/p70S6K and Rac1 activation, respectively. Moreover, in silico analyses indicate that p130Cas expression in ErbB2 positive human breast cancers significantly correlates with higher risk to develop distant metastasis, thus underlying the value of the p130Cas/ErbB2 synergism in regulating breast cancer invasion. In conclusion, high levels of p130Cas favour progression of ErbB2-transformed cells towards an invasive phenotype.


Subject(s)
Breast Neoplasms/metabolism , Crk-Associated Substrate Protein/metabolism , Receptor, ErbB-2/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Crk-Associated Substrate Protein/biosynthesis , Enzyme Activation , Female , Humans , Imaging, Three-Dimensional , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Oncogene Protein v-akt/metabolism , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics
12.
PLoS One ; 5(11): e14131, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21152434

ABSTRACT

BACKGROUND: Mounting clinical and experimental evidence suggests that the shift of carcinomas towards a mesenchymal phenotype is a common paradigm for both resistance to therapy and tumor recurrence. However, the mesenchymalization of carcinomas has not yet entered clinical practice as a crucial diagnostic paradigm. METHODOLOGY/PRINCIPAL FINDINGS: By integrating in silico and in vitro studies with our epithelial and mesenchymal tumor models, we compare herein crucial molecular pathways of previously described carcinoma-derived mesenchymal tumor cells (A17) with that of both carcinomas and other mesenchymal phenotypes, such as mesenchymal stem cells (MSCs), breast stroma, and various types of sarcomas. We identified three mesenchymal/stromal-signatures which A17 cells shares with MSCs and breast stroma. By using a recently developed computational approach with publicly available microarray data, we show that these signatures: 1) significantly relates to basal-like breast cancer subtypes; 2) significantly relates to bone metastasis; 3) are up-regulated after hormonal treatment; 4) predict resistance to neoadjuvant therapies. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that mesenchymalization is an intrinsic property of the most aggressive tumors and it relates to therapy resistance as well as bone metastasis.


Subject(s)
Bone Neoplasms/genetics , Breast Neoplasms/genetics , Gene Expression Profiling , Mesoderm/metabolism , Stromal Cells/metabolism , Animals , Blotting, Western , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cluster Analysis , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Drug Resistance, Neoplasm/genetics , Epithelial Cells/metabolism , Female , Humans , Mesenchymal Stem Cells/metabolism , Mesoderm/pathology , Mice , Neoadjuvant Therapy/methods , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stromal Cells/pathology
13.
Adv Exp Med Biol ; 674: 43-54, 2010.
Article in English | MEDLINE | ID: mdl-20549939

ABSTRACT

Integrin signaling has a critical function in organizing cells in tissues during both embryonic development and tissue repair. Following their binding to the extracellular ligands, the intracellular signaling pathways triggered by integrins are directed to two major functions: organization of the actin cytoskeleton and regulation of cell behaviour including survival, differentiation and growth. Basic research conducted in the past twelve years has lead to remarkable breakthroughs in this field. Integrins are catalytically inactive and translate positional cues into biochemical signals by direct and/or functional association with intracellular adaptors, cytosolic tyrosine kinases or growth factor and cytokine receptors. The purpose of this chapter is to highlight recent experimental and conceptual advances in integrin signaling with particular emphasis on the ability of integrins to regulate Fak/Src family kinases (SFKs) activation and the cross-talk with soluble growth factors receptors and cytokines.


Subject(s)
Integrins/metabolism , Signal Transduction/physiology , Actins/metabolism , Animals , Cell Differentiation/physiology , Cell Survival/physiology , Cytoskeleton/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Cytokine/metabolism , src-Family Kinases/metabolism
14.
FASEB J ; 24(10): 3796-808, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20505116

ABSTRACT

The ErbB2 oncogene is often overexpressed in breast tumors and associated with poor clinical outcome. p130Cas represents a nodal scaffold protein regulating cell survival, migration, and proliferation in normal and pathological cells. The functional role of p130Cas in ErbB2-dependent breast tumorigenesis was assessed by its silencing in breast cancer cells derived from mouse mammary tumors overexpressing ErbB2 (N202-1A cells), and by its reexpression in ErbB2-transformed p130Cas-null mouse embryonic fibroblasts. We demonstrate that p130Cas is necessary for ErbB2-dependent foci formation, anchorage-independent growth, and in vivo growth of orthotopic N202-1A tumors. Moreover, intranipple injection of p130Cas-stabilized siRNAs in the mammary gland of Balbc-NeuT mice decreases the growth of spontaneous tumors. In ErbB2-transformed cells, p130Cas is a crucial component of a functional molecular complex consisting of ErbB2, c-Src, and Fak. In human mammary cells, MCF10A.B2, the concomitant activation of ErbB2, and p130Cas overexpression sustain and strengthen signaling, leading to Rac1 activation and MMP9 secretion, thus providing invasive properties. Consistently, p130Cas drives N202-1A cell in vivo lung metastases colonization. These results demonstrate that p130Cas is an essential transducer in ErbB2 transformation and highlight its potential use as a novel therapeutic target in ErbB2 positive human breast cancers.


Subject(s)
Cell Transformation, Neoplastic , Crk-Associated Substrate Protein/physiology , Genes, erbB-2 , Mammary Neoplasms, Experimental/pathology , Animals , Cell Line, Tumor , Crk-Associated Substrate Protein/genetics , Female , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , RNA, Small Interfering , Signal Transduction
15.
J Neurochem ; 103(4): 1344-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17680992

ABSTRACT

Neuroblastoma (NB) and Ewing's sarcoma (ES) represent the most common extracranial solid tumors of childhood. Heat shock proteins (HSP) are elevated in cancer cells and their over-expression was correlated to drug-resistance. In this work we identified the HSP by a sensitive proteomic analysis of NB and ES cell lines, then, we studied the HSP response to doxorubicin. Some identified HSP were constitutively more expressed in NB than in ES cells. Doxorubicin-stimulated HSP response only in NB cells. Quercetin was found to inhibit HSP expression depleting heat shock factor 1 (HSF1) cellular stores. Quercetin caused a higher anti-proliferative effect in NB (IC(50): 6.9 +/- 5.8 mumol/L) than in ES cells (IC(50): 85.5 +/- 53.1 mumol/L). Moreover, quercetin caused a very pronounced doxorubicin sensitizing effect in NB cells (241 fold IC(50) decrease) and a moderate effect in ES cells. HSP involvement in NB cells sensitization was confirmed by the silencing of HSF1. Quercetin treatment and HSF1 silencing increased the pro-apoptotic effect of doxorubicin. In conclusion, the higher HSP levels, observed in NB cells, did not confer increased resistance to doxorubicin; on the contrary, HSP inhibition by quercetin or gene silencing caused higher sensitization to doxorubicin. These results may have a potential application in the treatment of NB.


Subject(s)
Doxorubicin/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/biosynthesis , Neuroblastoma/metabolism , Quercetin/pharmacology , Sarcoma, Ewing/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Silencing , Heat-Shock Proteins/genetics , Humans , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Quercetin/therapeutic use , Sarcoma, Ewing/genetics
16.
Cancer Res ; 66(9): 4672-80, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651418

ABSTRACT

To investigate the mechanisms through which p130Cas adaptor protein is linked to tumorigenesis, we generated mouse mammary tumor virus (MMTV)-p130Cas mice overexpressing p130Cas in the mammary gland. MMTVp130Cas transgenic mice are characterized by extensive mammary epithelial hyperplasia during development and pregnancy and by delayed involution at the end of lactation. These phenotypes are associated with activation of Src kinase, extracellular signal-regulated kinase 1/2, mitogen-activated protein kinase, and Akt pathways, leading to an increased rate of proliferation and a decreased apoptosis. A double-transgenic line derived from crossing MMTV-p130Cas with MMTV-HER2-Neu mice expressing the activated form of the HER2-Neu oncogene develops multifocal mammary tumors with a significantly shorter latency than the HER2-Neu parental strain alone. Mammary epithelial cells isolated from tumors of double-transgenic mice display increased tyrosine phosphorylation, c-Src, and Akt activation compared with cells derived from HER2-Neu tumors. In addition, p130Cas down-regulation by RNA interference increases apoptosis in HER2-Neu-expressing cells, indicating that p130Cas regulates cell survival. Consistently with the double-transgenic mice model, p130Cas is overexpressed in a significant subset of human breast cancers and high levels of p130Cas in association with HER2 expression correlate with elevated proliferation. These findings provide evidences for a role of p130Cas as a positive regulator of both proliferation and survival in normal and transformed mammary epithelial cells. Its overexpression contributes to HER2-Neu-induced breast tumorigenesis, thus identifying this protein as a putative target for clinical therapy.


Subject(s)
Cell Transformation, Neoplastic/pathology , Crk-Associated Substrate Protein/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Receptor, ErbB-2/physiology , Animals , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle , Cell Transformation, Neoplastic/genetics , Crk-Associated Substrate Protein/biosynthesis , Crk-Associated Substrate Protein/genetics , Female , Genes, erbB-2 , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , RNA Interference , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Signal Transduction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...