Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Med Chem ; 264: 115971, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38071795

RESUMEN

Pharmacological inhibition of dihydrofolate reductase (DHFR) is an established approach for treating a variety of human diseases, including foreign infections and cancer. However, treatment with classic DHFR inhibitors, such as methotrexate (MTX), are associated with negative side-effects and resistance mechanisms that have prompted the search for alternatives. The DHFR inhibitor pyrimethamine (Pyr) has compelling anti-cancer activity in in vivo models, but lacks potency compared to MTX, thereby requiring higher concentrations to induce therapeutic responses. The purpose of this work was to investigate structural analogues of Pyr to improve its in vitro and cellular activity. A series of 36 Pyr analogues were synthesized and tested in a sequence of in vitro and cell-based assays to monitor their DHFR inhibitory activity, cellular target engagement, and impact on breast cancer cell viability. Ten top compounds were identified, two of which stood out as potential lead candidates, 32 and 34. These functionalized Pyr analogues potently engaged DHFR in cells, at concentrations as low as 1 nM and represent promising DHFR inhibitors that could be further explored as potential anti-cancer agents.


Asunto(s)
Antineoplásicos , Antagonistas del Ácido Fólico , Neoplasias , Humanos , Pirimetamina/farmacología , Antagonistas del Ácido Fólico/farmacología , Antagonistas del Ácido Fólico/química , Metotrexato/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Biología , Tetrahidrofolato Deshidrogenasa/química
2.
Sci Rep ; 12(1): 22154, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36550267

RESUMEN

In a previous study, we showed that serine/threonine-protein kinase 4 (STK4) is involved in the control on proliferation and migration of endometrial cancer (EC) cells in vitro. In the present paper, we studied STK4 expression in EC tissues from a large cohort of patients to determine whether STK4 can serve as a marker for the aggressiveness and prognosis of EC. Tissue samples from patients with EC were examined for tumor type, grade, and stage. The STK4 protein expression in EC cells was assessed by immunohistochemistry and related to clinicopathological data of patients, such as progression and patient survival rate. The STK4 mRNA levels and its relation to the survival rate were analyzed also in publicly available databases. The STK4 gene expression was low at both, the mRNA and protein levels in EC, especially in serous tumors. Comparison of STK4 expression with the patient survival rate shows that the higher expression is associated with worse prognosis in serous EC, while no such dependence was found in endometrioid EC. Hence, the determination of the SKT4 expression pattern could be used as a putative prognostic marker for serous EC.


Asunto(s)
Carcinoma Endometrioide , Cistadenocarcinoma Seroso , Neoplasias Endometriales , Femenino , Humanos , Neoplasias Endometriales/patología , Carcinoma Endometrioide/patología , Endometrio/metabolismo , Pronóstico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Proteínas Serina-Treonina Quinasas/genética , Péptidos y Proteínas de Señalización Intracelular
3.
Cancer Control ; 29: 10732748221094797, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35533253

RESUMEN

BACKGROUND: Several markers of survival among endometrial cancer (EC) patients have been proposed, namely, the oncoprotein stathmin, RAF kinase inhibitor (RKIP), Cyclin A, GATA-binding protein 3 (GATA3), and growth and differentiation factor-15 (GDF-15). Their elevated expression correlated significantly with a high stage, serous papillary/clear cell subtypes, and aneuploidy. In a previous study, we reported the elevated expression of the serine/threonine protein kinase N1 (PKN1) in cancerous cells. In the present paper, we studied PKN1 expression in EC tissues from a large cohort of patients, to determine whether PKN1 can serve as a marker for the aggressiveness and prognosis of EC, and/or as a marker of survival among EC patients. METHODS: Tissue samples from EC patients were examined retrospectively for tumor type, tumor size, FIGO stage and grade, depth of invasion in the myometrium, and presence of lymph node metastasis. The PKN1 protein expression in EC cells was assessed by immunohistochemistry. PKN1 mRNA levels were analyzed in publicly available databases, using bioinformatic tools. RESULTS: We found that expression of PKN1 at the mRNA and proteins levels tended to increase in high-grade EC samples (P = .0001 and P = .06, respectively). In addition, patients with metastatic disease had higher PKN1 mRNA levels (P = .02). Moreover, patients with high PKN1 expression could be characterized by poorer survival. CONCLUSIONS: We have shown a trend of the higher PKN1 expression levels in EC patients with poor prognosis. Therefore, PKN1 might be considered as a candidate prognostic marker for EC.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Endometriales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Humanos , Pronóstico , Proteína Quinasa C , ARN Mensajero , Estudios Retrospectivos , Regulación hacia Arriba
4.
J Biol Chem ; 298(2): 101531, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34953855

RESUMEN

Cancer is often characterized by aberrant gene expression patterns caused by the inappropriate activation of transcription factors. Signal transducer and activator of transcription 3 (STAT3) is a key transcriptional regulator of many protumorigenic processes and is persistently activated in many types of human cancer. However, like many transcription factors, STAT3 has proven difficult to target clinically. To address this unmet clinical need, we previously developed a cell-based assay of STAT3 transcriptional activity and performed an unbiased and high-throughput screen of small molecules known to be biologically active in humans. We identified the antimicrobial drug pyrimethamine as a novel and specific inhibitor of STAT3 transcriptional activity. Here, we show that pyrimethamine does not significantly affect STAT3 phosphorylation, nuclear translocation, or DNA binding at concentrations sufficient to inhibit STAT3 transcriptional activity, suggesting a potentially novel mechanism of inhibition. To identify the direct molecular target of pyrimethamine and further elucidate the mechanism of action, we used a new quantitative proteome profiling approach called proteome integral solubility alteration coupled with a metabolomic analysis. We identified human dihydrofolate reductase as a target of pyrimethamine and demonstrated that the STAT3-inhibitory effects of pyrimethamine are the result of a deficiency in reduced folate downstream of dihydrofolate reductase inhibition, implicating folate metabolism in the regulation of STAT3 transcriptional activity. This study reveals a previously unknown regulatory node of the STAT3 pathway that may be important for the development of novel strategies to treat STAT3-driven cancers.


Asunto(s)
Antiinfecciosos , Pirimetamina , Factor de Transcripción STAT3 , Tetrahidrofolato Deshidrogenasa , Antiinfecciosos/química , Antiinfecciosos/farmacología , Línea Celular Tumoral , Ácido Fólico/metabolismo , Humanos , Proteoma/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo
5.
ACS Chem Biol ; 15(7): 1842-1851, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32412740

RESUMEN

Signal transducer and activator of transcription (STAT) proteins have important biological functions; however, deregulation of STAT signaling is a driving force behind the onset and progression of inflammatory diseases and cancer. While their biological roles suggest that STAT proteins would be valuable targets for developing therapeutic agents, STAT proteins are notoriously difficult to inhibit using small drug-like molecules, as they do not have a distinct inhibitor binding site. Despite this, a multitude of small-molecule STAT inhibitors have been proposed, primarily focusing on inhibiting STAT3 protein to generate novel cancer therapies. Demonstrating that inhibitors bind to their targets in cells has historically been a very challenging task. With the advent of modern target engagement techniques, such as the cellular thermal shift assay (CETSA), interactions between experimental compounds and their biological targets can be detected with relative ease. To investigate interactions between STAT proteins and inhibitors, we herein developed STAT CETSAs and evaluated known STAT3 inhibitors for their ability to engage STAT proteins in biological settings. While potent binding was detected between STAT proteins and peptidic STAT inhibitors, small-molecule inhibitors elicited variable responses, most of which failed to stabilize STAT3 proteins in cells and cell lysates. The described STAT thermal stability assays represent valuable tools for evaluating proposed STAT inhibitors.


Asunto(s)
Ácidos Aminosalicílicos/metabolismo , Óxidos S-Cíclicos/metabolismo , Péptidos/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Sulfonamidas/metabolismo , Línea Celular Tumoral , Calefacción , Humanos , Unión Proteica , Estabilidad Proteica
6.
J Pharm Biomed Anal ; 160: 80-88, 2018 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-30086509

RESUMEN

STAT3 protein is an established target for the development of new cancer therapeutic agents. Despite lacking a traditional binding site for small molecule inhibitors, many STAT3 inhibitors have been identified and explored for their anti-cancer activity. Because STAT3 signaling is mediated by protein-protein interactions, indirect methods are often employed to determine if proposed STAT3 inhibitors bind to STAT3 protein. While established STAT3 inhibition assays (such as the fluorescence polarization assay, electrophoretic mobility shift assay and ELISAs) have been used to identify novel inhibitors of STAT3 signaling, methods that directly assess STAT3 protein-inhibitor interactions could facilitate the development of novel inhibitors. In this context, we herein report new STAT3 binding assays based on differential scanning fluorimetry (DSF) and differential scanning light scattering (DSLS) to characterize interactions between STAT3 protein and inhibitors. Several peptide and small molecule STAT3 inhibitors have been evaluated, and new insight into how these compounds may interact with STAT3 is provided.


Asunto(s)
Desarrollo de Medicamentos/métodos , Fluorometría/métodos , Péptidos/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Sitios de Unión , Óxidos S-Cíclicos/química , Óxidos S-Cíclicos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Luz , Péptidos/química , Unión Proteica , Dominios Proteicos , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/aislamiento & purificación , Dispersión de Radiación , Temperatura
7.
J Cell Mol Med ; 22(1): 163-172, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28782224

RESUMEN

Prokineticin 1 (PROK1), a hypoxia-regulated angiogenic factor, has emerged as a crucial regulator of embryo implantation and placentation. Dysregulation of PROK1 has been linked to recurrent pregnancy loss, pre-eclampsia, foetal growth restriction and preterm birth. These pregnancy complications are common in women with obesity and polycystic ovary syndrome, i.e. conditions associated with insulin resistance and compensatory hyperinsulinaemia. We investigated the effect of insulin on PROK1 expression during in vitro decidualization. Endometrial stromal cells were isolated from six healthy, regularly menstruating women and decidualized in vitro. Insulin induced a significant dose-dependent up-regulation of PROK1 on both mRNA and protein level in decidualizing endometrial stromal cells. This up-regulation was mediated by hypoxia-inducible factor 1-alpha (HIF1α) via the phosphatidylinositol 3-kinase (PI3K) pathway. Furthermore, we demonstrated that PROK1 did not affect the viability, but significantly inhibited the migration of endometrial stromal cells and the migratory and invasive capacity of trophoblast cell lines. This in vitro study provides new insights into the regulation of PROK1 by insulin in human decidualizing endometrial stromal cells, the action of PROK1 on migration of endometrial stromal cells, as well as migration and invasion of trophoblasts. We speculate that hyperinsulinaemia may be involved in the mechanisms by which PROK1 is linked to placenta-related pregnancy complications.


Asunto(s)
Decidua/citología , Decidua/metabolismo , Hormonas Gastrointestinales/genética , Insulina/farmacología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Adolescente , Adulto , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Coriocarcinoma/patología , Femenino , Hormonas Gastrointestinales/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Trofoblastos/citología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Adulto Joven
8.
Cell Signal ; 37: 81-92, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28600192

RESUMEN

Glioblastoma (GBM) heterogeneity is the main obstacle to efficient treatment due to the existence of subpopulation of cells with increased tumorigenicity and network of tumor associated parenchymal cells in the tumor microenvironment. We previously demonstrated that mast cells (MCs) infiltrate mouse and human gliomas in response to variety of signals in a glioma grade-dependent manner. However, the role of MCs in glioma development and the mechanisms behind MCs-glioma cells interaction remain unidentified. In the present study, we show that MCs upon activation by glioma cells produce soluble factors including IL-6, which are documented to be involved in cancer-related activities. We observe 'tumor educated' MCs decrease glioma cell proliferation and migration, reduce self-renewal capacity and expression of stemness markers but in turn promote glioma cell differentiation. 'Tumor educated' MC derived mediators exert these effects via inactivation of STAT3 signaling pathway through GSK3ß down-regulation. We identified 'tumor educated' MC derived IL-6 as one of the contributors among the complex mixture of MCs mediators, to be partially involved in the observed MC induced biological effect on glioma cells. Thus, MC mediated abolition of STAT3 signaling hampers glioma cell proliferation and migration by suppressing their stemness and inducing differentiation via down-regulation of GSK3ß expression. Targeting newly identified inflammatory MC-STAT3 axis could contribute to patient tailored therapy and unveil potential future therapeutic opportunities for patients.


Asunto(s)
Movimiento Celular , Proliferación Celular , Glioma/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Mastocitos/metabolismo , Factor de Transcripción STAT3/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Glioma/metabolismo , Humanos , Interleucina-6/metabolismo , Mastocitos/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
9.
Oncotarget ; 8(15): 24815-24827, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28445977

RESUMEN

Serglycin is an intracellular proteoglycan with a unique ability to adopt highly divergent structures by glycosylation with variable types of glycosaminoglycans (GAGs) when expressed by different cell types. Serglycin is overexpressed in aggressive cancers suggesting its protumorigenic role. In this study, we explored the expression of serglycin in human glioma and its correlation with survival and immune cell infiltration. We demonstrate that serglycin is expressed in glioma and that increased expression predicts poor survival of patients. Analysis of serglycin expression in a large cohort of low- and high-grade human glioma samples reveals that its expression is grade dependent and is positively correlated with mast cell (MC) infiltration. Moreover, serglycin expression in patient-derived glioma cells is significantly increased upon MC co-culture. This is also accompanied by increased expression of CXCL12, CXCL10, as well as markers of cancer progression, including CD44, ZEB1 and vimentin.In conclusion, these findings indicate the importance of infiltrating MCs in glioma by modulating signaling cascades involving serglycin, CD44 and ZEB1. The present investigation reveals serglycin as a potential prognostic marker for glioma and demonstrates an association with the extent of MC recruitment and glioma progression, uncovering potential future therapeutic opportunities for patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Mastocitos/metabolismo , Mastocitos/patología , Proteoglicanos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Progresión de la Enfermedad , Humanos
10.
J Invest Dermatol ; 135(2): 400-410, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25207817

RESUMEN

In this study, we used proteomics and biological network analysis to evaluate the potential biological processes and components present in the identified proteins of biopsies from cutaneous leishmaniasis (CL) patients infected by Leishmania braziliensis in comparison with normal skin. We identified 59 proteins differently expressed in samples from infected and normal skin. Biological network analysis employing identified proteins showed the presence of networks that may be involved in the cell death mediated by cytotoxic T lymphocytes. After immunohistochemical analyses, the expression of caspase-9, caspase-3, and granzyme B was validated in the tissue and positively correlated with the lesion size in CL patients. In conclusion, this work identified differentially expressed proteins in the inflammatory site of CL, revealed enhanced expression of caspase-9, and highlighted mechanisms associated with the progression of tissue damage observed in lesions.


Asunto(s)
Leishmaniasis Cutánea/metabolismo , Proteoma , Caspasa 3/análisis , Caspasa 9/análisis , Granzimas/análisis , Humanos , Inmunohistoquímica , Leishmaniasis Cutánea/patología , Piel/patología
11.
Int J Oncol ; 46(1): 107-12, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339198

RESUMEN

Cervical cancer is the second most prevalent malignancy among women worldwide, and additional objective diagnostic markers for this disease are needed. Given the link between cancer development and alternative splicing, we aimed to analyze the splicing patterns of the PRDX2, RAB1A, RAB1B, RAB5A and RAB25 genes, which are associated with different cancers, in normal cervical tissue, preinvasive cervical lesions and invasive cervical tumors, to identify new objective diagnostic markers. Biopsies of normal cervical tissue, preinvasive cervical lesions and invasive cervical tumors, were subjected to rapid amplification of cDNA 3' ends (3' RACE) RT­PCR. Resulting PCR products were analyzed on agarose gels, gel­purified and sequenced. Normal cervical tissue, preinvasive cervical lesions and invasive cervical tumors contained one PCR product corresponding to full­length PRDX2, RAB5A and RAB25 transcripts. All tissues contained two RAB1A­specific PCR products corresponding to the full­length transcript and one new alternatively spliced RAB1A transcript. Invasive cervical tumors contained one PCR product corresponding to the full­length RAB1B transcript, while all normal cervical tissue and preinvasive cervical lesions contained both the full­length RAB1B transcript and three new alternatively spliced RAB1B transcripts. Alternative splicing of the RAB1A transcript occurs in all cervical tissues, while alternative splicing of the RAB1B transcript occurs in normal cervical tissue and in preinvasive cervical lesions; not in invasive cervical tumors.


Asunto(s)
Peroxirredoxinas/genética , Neoplasias del Cuello Uterino/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab1/genética , Proteínas de Unión al GTP rab5/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cuello del Útero/metabolismo , Cuello del Útero/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Invasividad Neoplásica , Peroxirredoxinas/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab1/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
12.
Biochem Biophys Res Commun ; 454(1): 202-9, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25450381

RESUMEN

Neuroblastoma develops through processes which include cellular dedifferentiation. Ability of tumors to form spheroids is one of the manifestations of dedifferentiation and carcinogenic transformation. To study mechanisms of dedifferentiation of neuroblastoma cells, we generated spheroids and performed a proteomics study to compare the spheroids with parental SK-N-BE2 cells. We observed that dedifferentiation induced extensive changes in the proteome profiles of the cells, which affected more than 30% of detected cellular proteins. Using mass spectrometry, we identified 239 proteins affected by dedifferentiation into spheroids as compared to the parental cells. These proteins represented such regulatory processes as transcription, cell cycle regulation, apoptosis, cell adhesion, metabolism, intracellular transport, stress response, and angiogenesis. A number of potent regulators of stemness, differentiation and cancer were detected as subnetworks formed by the identified proteins. Our validation tissue microarray study of 30 neuroblastoma cases confirmed that two of the identified proteins, DISC1 and DNA-PKcs, had their expression increased in advanced malignancies. Thus, our report unveiled extensive changes of the cellular proteome upon dedifferentiation of neuroblastoma cells, indicated top subnetworks and clusters of molecular mechanisms involved in dedifferentiation, and provided candidate biomarkers for clinical studies.


Asunto(s)
Desdiferenciación Celular/fisiología , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteoma/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proteína Quinasa Activada por ADN/metabolismo , Humanos , Espectrometría de Masas , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas Nucleares/metabolismo , Mapas de Interacción de Proteínas , Proteómica , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Análisis de Matrices Tisulares
13.
Onco Targets Ther ; 7: 1397-408, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25120372

RESUMEN

BACKGROUND: The response of cells to TGFß and EGF is mediated by a network of various intracellular regulators. The signaling crosstalk between different regulators is of key importance for tumorigenesis. The crosstalk may explain the modulation of cellular responses to the same regulator by another signaling molecule. As PKN1 - a serine/threonine kinase implicated in tumorigenesis - was identified as potential crosstalk node for TGFß and EGF signaling, the cellular functions that may be affected by PKN1 in a crosstalk of TGFß and EGF were explored. METHODS: To investigate the contribution of PKN1 to TGFß and EGF signaling, transiently PKN1-transfected HEC-1-A endometrial cancer cells were generated and subjected to treatment with TGFß1, EGF, and their combination. Proliferation, apoptosis, invasion, wound healing, and migration assays were performed. The impact of PKN1 on the expression and phosphorylation of intracellular proteins was monitored by immunoblotting. RESULTS: It was demonstrated that PKN1 modulated the responses of HEC-A-1 endometrial cancer cells to TGFß1 and EGF. PKN1 had an inhibitory effect on the stimulation of cell migration, and PKN1 kinase activity was required for the inhibitory effect of TGFß and EGF on cell proliferation and invasiveness. It was observed that phosphorylation of Smad2, FAK, and Erk1/2 correlated with responses of the cells to TGFß1 and EGF. CONCLUSION: PKN1 modulates TGFß- and EGF-dependent regulation of cell proliferation, migration, and invasiveness, and therefore is a component of the network signaling downstream of TGFß and EGF.

14.
Int J Oncol ; 45(2): 853-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24841766

RESUMEN

Transforming growth factor-ß (TGFß) and epidermal growth factor (EGF) are two potent regulators of tumorigenesis. Signaling cross-talk of TGFß and EGF employs a number of regulators which define the impact on cell physiology. MST1 has recently been reported as a regulator of tumorigenesis and differentiation. To investigate the role of mammalian sterile-like 1 (MST1) in TGFß and EGF signaling, we established transiently MST1­transfected HEC-1-A endometrial cancer cells, and subjected the cells to treatment with TGFß1, EGF and their combination. We report MST1 as a negative regulator of combined TGFß and EGF signaling. We observed that enhanced expression of MST1 inhibited the combined action of TGFß1 and EGF on cell invasiveness, migration and proliferation. Monitoring of the intracellular regulatory proteins showed that MST1 contribution to the TGFß-EGF cross-talk may involve focal adhesion kinase and E-cadherin, but not activation of Smad2. Our data unveiled the role of MST1 as a negative feedback for TGFß1­ and EGF­regulated cell invasiveness, migration and proliferation.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neoplasias Endometriales/patología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor Cross-Talk/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias Endometriales/metabolismo , Endometrio/metabolismo , Femenino , Humanos , Immunoblotting , Invasividad Neoplásica/patología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...