Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(20)2023 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-37895093

RESUMEN

Epithelial ovarian cancer (EOC) is the leading cause of death from gynecological cancers in Western countries. High-Grade Serous Ovarian Carcinoma (HGSOC) accounts for 60-70% of EOC and is the most aggressive subtype. Reduced PTPN13 expression levels have been previously correlated with worse prognosis in HGSOC. However, PTPN13's exact role and mechanism of action in these tumors remained to be investigated. To elucidate PTPN13's role in HGSOC aggressiveness, we used isogenic PTPN13-overexpressing clones of the OVCAR-8 cell line, which poorly expresses PTPN13, and also PTPN13 CRISPR/Cas9-mediated knockout/knockdown clones of the KURAMOCHI cell line, which strongly expresses PTPN13. We investigated their migratory and invasive capacity using a wound healing assay, their mesenchymal-epithelial transition (EMT) status using microscopy and RT-qPCR, and their sensitivity to chemotherapeutic drugs used for HGSOC. We found that (i) PTPN13 knockout/knockdown increased migration and invasion in KURAMOCHI cells that also displayed a more mesenchymal phenotype and increased expression of the SLUG, SNAIL, ZEB-1, and ZEB-2 EMT master genes; and (ii) PTPN13 expression increased the platinum sensitivity of HGSOC cells. These results suggest that PTPN13 might be a predictive marker of response to platinum salts in HGSOC.


Asunto(s)
Cistadenocarcinoma Seroso , Neoplasias Ováricas , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Transición Epitelial-Mesenquimal/genética , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Carcinoma Epitelial de Ovario/genética , Fenotipo , Línea Celular Tumoral , Proteína Tirosina Fosfatasa no Receptora Tipo 13/genética
2.
Biomolecules ; 11(2)2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33670716

RESUMEN

Spleen tyrosine kinase (SYK) can behave as an oncogene or a tumor suppressor, depending on the cell and tissue type. As pharmacological SYK inhibitors are currently evaluated in clinical trials, it is important to gain more information on the molecular mechanisms underpinning these opposite roles. To this aim, we reconstructed and compared its signaling networks using phosphoproteomic data from breast cancer and Burkitt lymphoma cell lines where SYK behaves as a tumor suppressor and promoter. Bioinformatic analyses allowed for unveiling the main differences in signaling pathways, network topology and signal propagation from SYK to its potential effectors. In breast cancer cells, the SYK target-enriched signaling pathways included intercellular adhesion and Hippo signaling components that are often linked to tumor suppression. In Burkitt lymphoma cells, the SYK target-enriched signaling pathways included molecules that could play a role in SYK pro-oncogenic function in B-cell lymphomas. Several protein interactions were profoundly rewired in the breast cancer network compared with the Burkitt lymphoma network. These data demonstrate that proteomic profiling combined with mathematical network modeling allows untangling complex pathway interplays and revealing difficult to discern interactions among the SYK pathways that positively and negatively affect tumor formation and progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Quinasa Syk/metabolismo , Neoplasias de la Mama/genética , Linfoma de Burkitt/genética , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Modelos Teóricos , Fosfoproteínas/metabolismo , Proteómica , Transducción de Señal/genética , Transducción de Señal/fisiología , Quinasa Syk/genética
3.
Biomolecules ; 10(12)2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33322542

RESUMEN

In this review article, we present the current knowledge on PTPN13, a class I non-receptor protein tyrosine phosphatase identified in 1994. We focus particularly on its role in cancer, where PTPN13 acts as an oncogenic protein and also a tumor suppressor. To try to understand these apparent contradictory functions, we discuss PTPN13 implication in the FAS and oncogenic tyrosine kinase signaling pathways and in the associated biological activities, as well as its post-transcriptional and epigenetic regulation. Then, we describe PTPN13 clinical significance as a prognostic marker in different cancer types and its impact on anti-cancer treatment sensitivity. Finally, we present future research axes following recent findings on its role in cell junction regulation that implicate PTPN13 in cell death and cell migration, two major hallmarks of tumor formation and progression.


Asunto(s)
Neoplasias/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Animales , Metilación de ADN/genética , Epigénesis Genética , Humanos , Modelos Biológicos , Neoplasias/genética , Transducción de Señal
4.
Theranostics ; 10(3): 1016-1032, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31938048

RESUMEN

Clinical data suggest that the protein tyrosine phosphatase PTPN13 exerts an anti-oncogenic effect. Its exact role in tumorigenesis remains, however, unclear due to its negative impact on FAS receptor-induced apoptosis. Methods: We crossed transgenic mice deleted for PTPN13 phosphatase activity with mice that overexpress human HER2 to assess the exact role of PTPN13 in tumor development and aggressiveness. To determine the molecular mechanism underlying the PTPN13 tumor suppressor activity we developed isogenic clones of the aggressive human breast cancer cell line MDA-MB-231 overexpressing either wild type or a catalytically-inactive mutant PTPN13 and subjected these to phosphoproteomic and gene ontology analyses. We investigated the PTPN13 consequences on cell aggressiveness using wound healing and Boyden chamber assays, on intercellular adhesion using videomicroscopy, cell aggregation assay and immunofluorescence. Results: The development, growth and invasiveness of breast tumors were strongly increased by deletion of the PTPN13 phosphatase activity in transgenic mice. We observed that PTPN13 phosphatase activity is required to inhibit cell motility and invasion in the MDA-MB-231 cell line overexpressing PTPN13. In vivo, the negative PTPN13 effect on tumor invasiveness was associated with a mesenchymal-to-epithelial transition phenotype in athymic mice xenografted with PTPN13-overexpressing MDA-MB-231 cells, as well as in HER2-overexpressing mice with wild type PTPN13, compared to HER2-overexpressing mice that lack PTPN13 phosphatase activity. Phosphoproteomic and gene ontology analyses indicated a role of PTPN13 in the regulation of intercellular junction-related proteins. Finally, protein localization studies in MDA-MB-231 cells and HER2-overexpressing mice tumors confirmed that PTPN13 stabilizes intercellular adhesion and promotes desmosome formation. Conclusions: These data provide the first evidence for the negative role of PTPN13 in breast tumor invasiveness and highlight its involvement in cell junction stabilization.


Asunto(s)
Neoplasias Mamarias Experimentales , Proteína Tirosina Fosfatasa no Receptora Tipo 13/fisiología , Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal , Femenino , Humanos , Uniones Intercelulares , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Invasividad Neoplásica , Trasplante de Neoplasias , Receptor ErbB-2/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
5.
Cancers (Basel) ; 11(12)2019 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-31817924

RESUMEN

While first discovered in immunoreceptor signaling, the Syk protein kinase behaves as a tumor and metastasis suppressor in epithelial cells. Its reduced expression in breast and other carcinomas is correlated with decreased survival and increased metastasis risk, but its action mechanism remains largely unknown. Using phosphoproteomics we found that Syk phosphorylated E-cadherin and α-, ß-, and p120-catenins on multiple tyrosine residues that concentrate at intercellular junctions. Increased Syk expression and activation enhanced E-cadherin/catenin phosphorylation, promoting their association and complex stability. In human breast cancer cells, Syk stimulated intercellular aggregation, E-cadherin recruitment and retention at adherens junctions, and promoted epithelial integrity, whereas it inhibited cell migration and invasion. Opposite effects were obtained with Syk knockdown or non-phosphorylatable mutant E-cadherin expression. Mechanistically, Syk stimulated the interaction of the E-cadherin/catenin complex with zonula occludens proteins and the actin cytoskeleton. Conditional Syk knockout in the lactating mouse mammary gland perturbed alveologenesis and disrupted E-cadherin localization at adherens junctions, corroborating the observations in cells. Hence, Syk is involved in the maintenance of the epithelial integrity of the mammary gland via the phosphorylation and stabilization of the E-cadherin/catenin adherens junction complex, thereby inhibiting cell migration and malignant tumor invasion.

6.
Proteomics ; 19(21-22): e1800450, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31472481

RESUMEN

Protein phosphorylation acts as an efficient switch controlling deregulated key signaling pathway in cancer. Computational biology aims to address the complexity of reconstructed networks but overrepresents well-known proteins and lacks information on less-studied proteins. A bioinformatic tool to reconstruct and select relatively small networks that connect signaling proteins to their targets in specific contexts is developed. It enables to propose and validate new signaling axes of the Syk kinase. To validate the potency of the tool, it is applied to two phosphoproteomic studies on oncogenic mutants of the well-known phosphatidyl-inositol 3-kinase (PIK3CA) and the unfamiliar Src-related tyrosine kinase lacking C-terminal regulatory tyrosine and N-terminal myristoylation sites (SRMS) kinase. By combining network reconstruction and signal propagation, comprehensive signaling networks from large-scale experimental data are built and multiple molecular paths from these kinases to their targets are extracted. Specific paths from two distinct PIK3CA mutants are retrieved, and their differential impact on the HER3 receptor kinase is explained. In addition, to address the missing connectivities of the SRMS kinase to its targets in interaction pathway databases, phospho-tyrosine and phospho-serine/threonine proteomic data are integrated. The resulting SRMS-signaling network comprises casein kinase 2, thereby validating its currently suggested role downstream of SRMS. The computational pipeline is publicly available, and contains a user-friendly graphical interface (http://doi.org/10.5281/zenodo.3333687).


Asunto(s)
Neoplasias/metabolismo , Proteómica , Transducción de Señal , Línea Celular Tumoral , Humanos , Mutación/genética , Proteínas de Neoplasias/metabolismo , Fosforilación , Interfaz Usuario-Computador
7.
Oncotarget ; 8(56): 95662-95673, 2017 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-29221157

RESUMEN

BACKGROUND: Chromosome 4q loss of heterozygosity (LOH) is frequently observed in high-grade serous ovarian carcinoma (HGSOC). However, this LOH has not been clearly associated with the inactivation of any tumor suppressor gene(s). As the tumor suppressor gene PTPN13 is located on chromosome 4q21, we investigated its expression in HGSOC. METHODS: PTPN13 protein expression was investigated by immunohistochemistry (IHC) in normal ovary epithelium and in 30 HGSOC samples, whereas PTPN13 mRNA expression was quantified by RT-PCR in another independent cohort of 28 HGSOC samples. Patients in both cohorts were followed for more than 8.5 years. RESULTS: PTPN13 protein expression was lower in one third of HGSOC samples compared with normal ovary epithelium. In both cohorts, high PTPN13 expression level (mRNA or protein) in the tumor was associated with favorable outcome and significantly longer survival (HR=0.27; p=0.0087 and HR=0.42; p=0.03, respectively). CONCLUSION: This study demonstrates, for the first time, that high PTPN13 expression level is a prognostic indicator of favorable outcome in patients with HGSOC. This finding, in conjunction with our previous mechanistic studies, suggests that PTPN13 loss, possibly by 4q LOH, enhances HGSOC aggressiveness and highlight the interest of studying PTPN13 signaling in HGSOC to identify new potential therapeutic targets.

8.
PLoS Comput Biol ; 13(3): e1005432, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28306714

RESUMEN

The ability to build in-depth cell signaling networks from vast experimental data is a key objective of computational biology. The spleen tyrosine kinase (Syk) protein, a well-characterized key player in immune cell signaling, was surprisingly first shown by our group to exhibit an onco-suppressive function in mammary epithelial cells and corroborated by many other studies, but the molecular mechanisms of this function remain largely unsolved. Based on existing proteomic data, we report here the generation of an interaction-based network of signaling pathways controlled by Syk in breast cancer cells. Pathway enrichment of the Syk targets previously identified by quantitative phospho-proteomics indicated that Syk is engaged in cell adhesion, motility, growth and death. Using the components and interactions of these pathways, we bootstrapped the reconstruction of a comprehensive network covering Syk signaling in breast cancer cells. To generate in silico hypotheses on Syk signaling propagation, we developed a method allowing to rank paths between Syk and its targets. We first annotated the network according to experimental datasets. We then combined shortest path computation with random walk processes to estimate the importance of individual interactions and selected biologically relevant pathways in the network. Molecular and cell biology experiments allowed to distinguish candidate mechanisms that underlie the impact of Syk on the regulation of cortactin and ezrin, both involved in actin-mediated cell adhesion and motility. The Syk network was further completed with the results of our biological validation experiments. The resulting Syk signaling sub-networks can be explored via an online visualization platform.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Quinasa Syk/metabolismo , Línea Celular Tumoral , Simulación por Computador , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Células MCF-7
9.
Anticancer Agents Med Chem ; 11(1): 78-88, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21235435

RESUMEN

Protein tyrosine phosphorylation plays a major role in many cellular functions implicated in cancer development and progression, but only a few of the known protein tyrosine phosphatases have yet been clearly classified as oncogenes or tumor suppressors. PTPL1 interacts with tumor-associated proteins, suggesting a link between PTPL1, the PTPN13 gene product, and tumorigenesis or cancer progression. However, the impact of PTPL1 on cancer is divided between its capacity to counteract the activity of oncogenic tyrosine kinases and its inhibitory interaction with the death receptor, Fas. In this manuscript, we review the PTPL1-interacting proteins implicated in cancer. In addition, we examine the phenotypic arguments concerning both the PTPL1/Fas interaction and the ability of PTPL1 to inhibit signaling from growth factor receptors or oncogenes with tyrosine kinase activity. Finally, we compare the alterations in expression and the genetic and epigenetic arguments supporting an oncogenic or an anti-oncogenic impact of PTPL1.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Animales , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 13/genética
10.
Cancer Res ; 70(12): 5116-26, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20501847

RESUMEN

The protein tyrosine phosphatase PTPL1/PTPN13, the activity of which is decreased through allelic loss, promoter methylation, or somatic mutations in some tumors, has been proposed as a tumor suppressor gene. Moreover, our recent clinical study identified PTPL1 expression level as an independent prognostic indicator of a favorable outcome for patients with breast cancer. However, how PTPL1 can affect tumor aggressiveness has not been characterized. Here, we first show that PTPL1 expression, assessed by immunohistochemistry, is decreased in breast cancer and metastasis specimens compared with nonmalignant tissues. Second, to evaluate whether PTPL1 plays a critical role in breast cancer progression, RNA interference experiments were performed in poorly tumorigenic MCF-7 breast cancer cells. PTPL1 inhibition drastically increased tumor growth in athymic mice and also enhanced several parameters associated with tumor progression, including cell proliferation on extracellular matrix components and cell invasion. Furthermore, the inhibition of Src kinase expression drastically blocked the effects of PTPL1 silencing on cell growth. In PTPL1 knockdown cells, the phosphorylation of Src on tyrosine 419 is increased, leading to the activation of its downstream substrates Fak and p130cas. Finally, substrate-trapping experiments revealed that Src tyrosine 419 is a direct target of the phosphatase. Thus, by identification of PTPL1 as the first phosphatase able to inhibit Src through direct dephosphorylation in intact cells, we presently describe a new mechanism by which PTPL1 inhibits breast tumor aggressiveness.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Familia-src Quinasas/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/genética , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Ratones , Ratones Desnudos , Invasividad Neoplásica , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 13/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 13/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal , Análisis de Matrices Tisulares , Familia-src Quinasas/antagonistas & inhibidores
11.
Int J Biochem Cell Biol ; 41(11): 2173-80, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19782949

RESUMEN

The insulin/insulin-like growth factor 1 (IGF-1) signaling pathway is a major regulator of adipose tissue growth and differentiation. We recently demonstrated that human protein tyrosine phosphatase (PTP) L1, a large cytoplasmic phosphatase also known as PTP-BAS/PTPN13/PTP-1E, is a negative regulator of IGF-1R/IRS-1/Akt pathway in breast cancer cells. This triggered us to investigate the potential role of PTPL1 in adipogenesis. To evaluate the implication of PTP-BL, the mouse orthologue of PTPL1, in adipose tissue biology, we analyzed PTP-BL mRNA expression in adipose tissue in vivo and during proliferation and differentiation of 3T3-L1 pre-adipocytes. To elucidate the role of PTP-BL and of its catalytic activity during adipogenesis we use siRNA techniques in 3T3-L1 pre-adipocytes, and mouse embryonic fibroblasts that lack wildtype PTP-BL and instead express a variant without the PTP domain (Delta P/Delta P MEFs). Here we show that PTP-BL is strongly expressed in white adipose tissue and that PTP-BL transcript and protein levels increase during proliferation and differentiation of 3T3-L1 pre-adipocytes. Strikingly, knockdown of PTP-BL expression in 3T3-L1 adipocytes caused a dramatic decrease in adipogenic gene expression levels (PPAR gamma, aP2) and lipid accumulation but did not interfere with the insulin/Akt pathway. Delta P/Delta P MEFs differentiate into the adipogenic lineage as efficiently as wildtype MEFs. However, when expression of either PTP-BL or PTP-BL Delta P was inhibited a dramatic reduction in the number of MEF-derived adipocytes was observed. These findings demonstrate a key role for PTP-BL in 3T3-L1 and MEF-derived adipocyte differentiation that is independent of its enzymatic activity.


Asunto(s)
Adipogénesis/genética , Regulación hacia Abajo/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 13/genética , Células 3T3-L1 , Adipocitos/citología , Adipocitos/enzimología , Animales , Diferenciación Celular , Proliferación Celular , Células Clonales , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 13/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo
12.
Int J Cancer ; 124(3): 638-43, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19004008

RESUMEN

Although it is well established that some protein tyrosine kinases have a prognostic value in breast cancer, the involvement of protein tyrosine phosphatases (PTPs) is poorly substantiated for breast tumors. Three of these enzymes (PTP-gamma, LAR, and PTPL1) are already known to be regulated by estrogens or their antagonists in human breast cancer cells. We used a real-time reverse transcriptase polymerase chain reaction method to test the expression levels of PTP-gamma, LAR and its neuronal isoform, and PTPL1 in a training set of RNA from 59 breast tumors. We sought correlations between levels of these molecular markers, current tumor markers, and survival. We then quantified the expression level of the selected phosphatase in 232 additional samples, resulting in a testing set of 291 breast tumor RNAs from patients with a median follow-up of 6.4 years. The Spearman nonparametric test revealed correlations between PTPL1 expression and differentiation markers. Cox univariate analysis of the overall survival studies demonstrated that PTPL1 is a prognostic factor [risk ratio (RR)=0.45], together with the progesterone receptor (PR) (RR=0.52) and node involvement (RR=1.58). In multivariate analyses, PTPL1 and PR retained their prognostic value (RRs of 0.48 and 0.55, respectively). This study demonstrates for the first time that PTPL1 expression level is an independent prognostic indicator of favorable outcome for patients with breast cancer. In conjunction with our mechanistic studies, this finding identifies PTPL1 as an important regulatory element of human breast tumor aggressiveness and sensitivity to treatments such as antiestrogens and antiaromatase.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Genes Supresores de Tumor , Proteína Tirosina Fosfatasa no Receptora Tipo 13/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Femenino , Expresión Génica , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Proyectos Piloto , Pronóstico , ARN Mensajero/análisis , Receptores de Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Cancer Res ; 67(14): 6806-13, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638892

RESUMEN

The protein tyrosine phosphatase (PTP) PTPL1/PTPN13 is a candidate tumor suppressor gene. Indeed, PTPL1 activity has been reported recently to be decreased through somatic mutations, allelic loss, or promoter methylation in some tumors. We showed previously that its expression was necessary for inhibition of Akt activation and induction of apoptosis by antiestrogens in breast cancer cells. Implications of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in cancer progression are now well established, and our study was therefore designed to define whether PTPL1 is sufficient to inhibit this pathway and, if so, to identify a direct substrate of this PTP, which may trigger a proapoptotic effect. We first show by complementary approaches that PTPL1 specifically dephosphorylates insulin receptor substrate-1 (IRS-1) in vitro and in cellulo. Next, our experiments using a dominant-negative mutant and RNA interference confirm the crucial role of PTPL1 in IRS-1 dephosphorylation. Finally, we report that PTPL1 expression is sufficient to block the IRS-1/PI3K/Akt signaling pathway, to inhibit the insulin-like growth factor-I effect on cell survival, and to induce apoptosis. Altogether, these data provide the first evidence for a direct positive role of the putative tumor suppressor gene PTPL1/PTPN13 on apoptosis and identify its target in the IRS-1/PI3K/Akt signaling pathway.


Asunto(s)
Apoptosis , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Dominio Catalítico , Línea Celular Tumoral , Fragmentación del ADN , Progresión de la Enfermedad , Células HeLa , Humanos , Proteínas Sustrato del Receptor de Insulina , Microscopía Fluorescente , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Plásmidos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 13 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
14.
J Cell Sci ; 118(Pt 22): 5393-403, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16280553

RESUMEN

Numerous scaffold proteins coordinate signals from the environment with actin-based protrusions during shape change and migration. Many scaffolds integrate signals from Rho-family GTPases to effect the assembly of specific actin structures. Here we investigate the mechanism of action MIM-B (missing in metastasis-B) on the actin cytoskeleton. MIM-B binds actin monomer through a WASP homology 2 motif, bundles actin filaments via an IRSp53/MIM domain, and is a long isoform of MIM, a proposed metastasis suppressor. We analysed the activity of MIM-B toward the actin cytoskeleton as well as its potential link to cancer metastasis. Endogenous MIM-B protein is widely expressed and its expression is maintained in various metastatic cell lines. MIM-B induces lamellipodia-like actin-rich protrusions. The IRSp53/MIM domain of MIM-B, as well as Rac activity are required to induce protrusions, but not the WASP homology 2 motif. MIM-B binds and activates Rac via its IRSp53/MIM domain, but this is not sufficient to induce lamellipodia. Finally, our data revealed that actin bundling and Rac-binding properties of MIM-B are not separable. Thus, MIM-B is unlikely to be a metastasis suppressor but acts as a scaffold protein that interacts with Rac, actin and actin-associated proteins to modulate lamellipodia formation.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Medio de Cultivo Libre de Suero/farmacología , Citoesqueleto/química , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Mutación , Proteínas de Neoplasias , Unión Proteica , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Alineación de Secuencia , Células 3T3 Swiss , Transfección
15.
J Cell Biol ; 168(3): 489-99, 2005 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-15668295

RESUMEN

The aspartyl-protease cathepsin D (cath-D) is overexpressed and hypersecreted by epithelial breast cancer cells and stimulates their proliferation. As tumor epithelial-fibroblast cell interactions are important events in cancer progression, we investigated whether cath-D overexpression affects also fibroblast behavior. We demonstrate a requirement of cath-D for fibroblast invasive growth using a three-dimensional (3D) coculture assay with cancer cells secreting or not pro-cath-D. Ectopic expression of cath-D in cath-D-deficient fibroblasts stimulates 3D outgrowth that is associated with a significant increase in fibroblast proliferation, survival, motility, and invasive capacity, accompanied by activation of the ras-MAPK pathway. Interestingly, all these stimulatory effects on fibroblasts are independent of cath-D proteolytic activity. Finally, we show that pro-cath-D secreted by cancer cells is captured by fibroblasts and partially mimics effects of transfected cath-D. We conclude that cath-D is crucial for fibroblast invasive outgrowth and could act as a key paracrine communicator between cancer and stromal cells, independently of its catalytic activity.


Asunto(s)
Catepsina D/fisiología , Movimiento Celular/fisiología , Fibroblastos/citología , Animales , Apoptosis/fisiología , Butadienos/farmacología , Catepsina D/genética , Catepsina D/metabolismo , Aumento de la Célula/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Endocitosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Humanos , Manosafosfatos/farmacología , Ratones , Microscopía Electrónica , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Nitrilos/farmacología , Comunicación Paracrina/fisiología , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , Transfección , Cicatrización de Heridas
16.
Crit Rev Oncol Hematol ; 52(1): 9-17, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15363463

RESUMEN

Protein tyrosine phosphatases (PTPs) consist of a large family of related enzymes, including the group of classical PTPs with its two main subgroups, the transmembrane receptor-type (RPTPs) and the intracellular or non-transmembrane PTPs. Published data on the expression and function of a panel of these enzymes in normal and cancerous breast tissues are discussed in this review. While most studies, albeit on different enzymes, have tended to agree on the evidence for an increased PTP expression in breast cancer, any connection between PTP expression and the enzymes' role in cancer development and progression remains largely open to interpretation. Concomitant increases of protein tyrosine kinase (PTK) and PTP activities in many cancers further indicate that a complex dysregulation in the balance of tyrosine phosphorylation could be responsible for major alterations in various cellular processes controlling tissue homeostasis. In particular, any relationship between the expression of PTPs and their specific diverse roles in the regulation of cell growth and apoptosis in breast cancer needs to be addressed in major fundamental, preclinical and clinical studies.


Asunto(s)
Neoplasias de la Mama/etiología , Proteínas Tirosina Fosfatasas/genética , Animales , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal
17.
Bull Cancer ; 91(4): 325-32, 2004 Apr.
Artículo en Francés | MEDLINE | ID: mdl-15242314

RESUMEN

The protein tyrosine phosphatase L1 (PTPL1), also known as FAP1, has two major types of remarkable structural domains, in addition to its catalytic unit: a FERM domain which is responsible for its localization at the apical pole of the cell plasma membrane and 5 PDZ domains suggestive of numerous possibilities of protein partners and consequently of a role as a cargo protein or an integrator between different signalling pathways. In fact, though it was initially suggested, in 1995, that this enzyme acts as an inhibitor of Fas death receptor several recent studies indicate that PTPL1 plays many other roles. It dephosphorylates Ephrin B (ligand of Eph, a receptor triggering angiogenesis and axonal guidance), it interacts with numerous proteins associated to cytoskeleton plasticity and it is implicated in cytokinesis. We have demonstrated that its expression is regulated by antiestrogens in mammary cancer and shown, with stable antisense transfectants, that PTPL1 plays a key role in the mediation of the inhibitory effects of these antagonists on growth factor signalling by impeding the IRS-I/PI3-K/Akt survival pathway. Altogether PTPL1 has to be regarded as a unique marker of mammary tumor response to antiestrogens and a potential therapeutic target to activate apoptotic stimuli in tumor cells.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Regulación Neoplásica de la Expresión Génica , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/farmacología , Supervivencia Celular , Citoesqueleto/metabolismo , Femenino , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 13 , Transducción de Señal
18.
J Cell Sci ; 116(Pt 12): 2519-30, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12766187

RESUMEN

PTPL1 is the largest known cytoplasmic protein tyrosine phosphatase (PTP) containing a FERM (four point-1, ezrin, radixin and moesin) domain. Enzyme localization and PTP-substrate specificity are thought to play crucial roles in the regulation of PTP activity, which determines their functions. Here we report that PTPL1 is predominantly localized at the apical face of plasma membrane enriched in dorsal microvilli when expressed in HeLa cells. By comparing localization of the full-length enzyme with its FERM domain or FERM-deleted PTPL1 construct, we first concluded that PTPL1-FERM domain is necessary and sufficient to address the wild-type enzyme at the membrane. Two potential phosphatidylinositol 4,5-biphosphate [PtdIns(4,5)P2]-binding motifs were identified within the PTPL1-FERM sequence. We further showed that mutation of both sites altered PTPL1 localization similarly to FERM domain deletion, and impaired its subcellular distribution as confirmed biochemically by cell-fractionation experiments. Using protein-lipid overlays, we demonstrated an interaction of the FERM domain of PTPL1 with PtdIns(4,5)P2, which was lost after mutation of potential PtdIns(4,5)P2-binding motifs. Moreover, neomycin, which masks PtdIns(4,5)P2 polar heads, was shown to decrease by 50% the association of PTPL1 with the cytoskeletal fraction. These results identify the crucial role of the FERM domain in PTPL1 intracellular targeting and demonstrate that localization of PTPL1 is regulated by phosphoinositide metabolism.


Asunto(s)
Membrana Celular/enzimología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Transporte de Proteínas/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Sitios de Unión/genética , Células COS , Compartimento Celular/genética , Citoesqueleto/genética , Células HeLa , Humanos , Microvellosidades/metabolismo , Datos de Secuencia Molecular , Mutación/genética , Neomicina/farmacología , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 13 , Homología de Secuencia de Aminoácido
19.
J Biol Chem ; 277(49): 47861-9, 2002 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-12354757

RESUMEN

Studies in Jurkat leukemia cells have suggested that protein-tyrosine phosphatase PTPL1/FAP-1 rescues Fas-induced cell death. However, we have previously shown that this enzyme triggers 4-hydroxytamoxifen-induced growth inhibition in human breast cancer cells. The present study addresses the role of PTPL1/FAP-1 in antiestrogen-regulated apoptotic effect and insulin-like growth factor-I survival action in MCF7 cells and further identifies the impacted signaling pathway. By terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling and cytoplasmic nucleosome enzyme-linked immunosorbent assay, we demonstrated that 4-hydroxytamoxifen-induced apoptosis was totally lost in PTPL1/FAP-1 antisense transfectants in which enzyme expression was abrogated, revealing the crucial role of this phosphatase in the apoptotic process in human breast cancer cells. Time-dependent expression of PTPL1/FAP-1 in MCF7 cells completely abolished the survival action of insulin-like growth factor-I. This effect occurred through a highly significant reduction in phosphatidylinositol 3-kinase/Akt pathway activation (80% reduction in phosphatidylinositol 3-kinase activity, 55% inhibition of Akt activation) accompanied by a 65% decrease in insulin receptor substrate-1 growth factor-induced tyrosine phosphorylation. These results provide the first evidence that PTPL1/FAP-1 has a key role in the apoptotic process in human breast cancer cells independent of Fas but associated with an early inhibition of the insulin receptor substrate-1/phosphatidylinositol 3-kinase pathway. Our data therefore suggest new therapeutic routes and strengthen the importance of identifying endogenous regulators and substrates of this phosphatase in breast tumors.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Western Blotting , Supervivencia Celular , Fragmentación del ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Etiquetado Corte-Fin in Situ , Proteínas Sustrato del Receptor de Insulina , Oligonucleótidos Antisentido/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 13 , Transducción de Señal , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Tirosina/metabolismo
20.
Int J Cancer ; 98(5): 698-706, 2002 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-11920638

RESUMEN

Resistance to the antiestrogen tamoxifen is the main stumbling block for the success of breast cancer therapy. We focused our study on cellular alterations induced by a prolonged treatment with the active tamoxifen metabolite hydroxytamoxifen (OHT). We show that a prolonged OHT treatment (for up to 7 days) led to a progressive increase in the level of phosphorylated p44/42 mitogen activated kinase (MAP kinase) induced by 10(-7) M TPA stimulation, without any significant change in the protein level. This effect was also observed in MCF-7 cells grown first in medium containing dextran-coated charcoal-treated FCS (DCC medium) for 20 days prior to OHT treatment, indicating a specific effect of the antiestrogen and not an effect of estrogen deprivation. It was prevented by cotreatment with estradiol and not observed in the estrogen receptor negative HeLa cell line, suggesting that it was mediated by the estrogen receptor. TPA induced phosphorylation of MEK1/2 was also raised by OHT treatment, without any change in their protein level or Raf-1 and H-Ras levels. When the MCF-7R OHT resistant cell line was grown in antiestrogen containing medium, the level of phosphorylated p44/42 MAP kinase was also high but reversed when the antiestrogen was removed. The 2 other MAP kinase, JNK and P38 pathways were not affected in the same way by OHT treatment. In conclusion, our data reveal that a prolonged OHT treatment, by increasing p44/42 MAPK activity, affects a key step in the growth control of MCF-7 cells, although not sufficiently to overcome the growth inhibitory effect of the drug.


Asunto(s)
Neoplasias de la Mama/enzimología , Estradiol/análogos & derivados , Proteínas Quinasas JNK Activadas por Mitógenos , Sistema de Señalización de MAP Quinasas/fisiología , Tamoxifeno/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Anisomicina/farmacología , Antibacterianos/farmacología , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Femenino , Fulvestrant , Genes ras/fisiología , Humanos , MAP Quinasa Quinasa 4 , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes/metabolismo , Tamoxifeno/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA