Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 109(12): 3116-24, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24169347

RESUMEN

BACKGROUND: Processing of microRNAs (miRNAs) is a highly controlled process. Deregulation of miRNA expression was observed in several types of cancer but changes in the miRNA-processing enzymes have not been analysed until today. In this study, we analysed Argonaute2 (AGO2, EIF2C2), as one main factor of the miRNA processing ensemble, in the context of cancer development, especially in melanoma. METHODS: We determined the AGO2 expression level in melanoma, as well as in other cancers, with biochemical approaches (qRT-PCR, western blot and immunofluorescence studies) and analysed the cell behaviour in migration assays. RESULTS: Specifically in melanoma, we revealed a strong reduction of AGO2 expression compared with primary melanocytes. The reduction of AGO2 expression was only found on protein level, whereas the mRNA level stayed unchanged hinting to post-transcriptional regulation. We could show that re-expression of AGO2 in melanoma leads to a strong improvement of regulatory effects due to increased functionality of small-interfering RNAs and short hairpin RNAs. CONCLUSION: We identified melanoma-specific downregulation of AGO2 and corresponding reduced RNAi efficiency. These findings will help to understand the molecular basis of malignant melanoma and can potentially lead to an improvement of therapeutic strategies.


Asunto(s)
Proteínas Argonautas/deficiencia , Proteínas Argonautas/genética , Melanoma/metabolismo , MicroARNs/genética , Proteínas Argonautas/biosíntesis , Proteínas Argonautas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Melanoma/genética , Melanoma/patología , MicroARNs/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transfección
2.
Br J Cancer ; 108(7): 1460-9, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23511560

RESUMEN

BACKGROUND: The role of melanoma inhibitory activity 2 (MIA2) was examined in human oral squamous cell carcinoma (OSCC). METHODS: MIA2 role was examined by immunohistochemistry of human OSCCs and knockdown studies using human 3 OSCC cell lines with MIA2 expression. RESULTS: MIA2 expression was observed in 62 (66.7%) of 93 OSCCs and was associated with tumour expansion and nodal metastasis. Melanoma inhibitory activity 2 expression was inversely correlated with intratumoral infiltration of lymphocytes. Invasion and anti-apoptotic survival were reduced by MIA2 knockdown in HSC3 cells. MOLT-3 lymphocytes infiltrating the HSC3 cell layer was enhanced by MIA2 knockdown or MIA2 depletion with the antibody. In HSC3 cells, MIA2 knockdown decreased the expressions of vascular endothelial growth factor (VEGF), VEGF-C, and VEGF-D. The downregulation of VEGF-C and -D was caused by inhibition of p38 and extracellular signal-regulated kinase (ERK)1/2, respectively. Melanoma inhibitory activity 2 was co-precipitated with integrin α4 andα5 in HSC3 cells. Integrin α4 knockdown decreased p38 phosphorylation and increased apoptosis, whereas integrin α5 knockdown decreased c-Jun N-terminal kinase (JNK) phosphorylation and apoptosis. Inhibition of JNK decreased apoptosis in the HSC3 cells. CONCLUSION: These findings suggest that the roles of MIA2 might be based on the variety of the integrins and the subtypes of mitogen-activated protein kinase.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Anciano , Antígenos de Neoplasias , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Integrinas/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias de la Boca/inmunología , Neoplasias de la Boca/patología , Proteínas de Neoplasias , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor C de Crecimiento Endotelial Vascular/biosíntesis , Factor D de Crecimiento Endotelial Vascular/biosíntesis
3.
Carcinogenesis ; 32(6): 935-43, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21459757

RESUMEN

Slits are a group of secreted glycoproteins that act as molecular guidance cues in cellular migration. Recently, several studies demonstrated that Slit-2 can operate as candidate tumour suppressor protein in various tissues. In this study, we show Slit-2 expression in basal cell layers of normal oral mucosa colocalized with P-cadherin expression. In contrast, there is a loss of Slit-2 and P-cadherin expression in mucosa of oral squamous cell carcinoma (OSCC). Our in vitro investigations reveal a correlation of P-cadherin and Slit-2 expression: OSCC cells with induced P-cadherin expression (PCI52_PC) display an increased Slit-2 expression. However, abrogating P-cadherin function with a function-blocking antibody decreases Slit-2 secretion confirming a direct link between P-cadherin and Slit-2. Moreover, experiments with OSCC cells show that Slit-2 interferes with a Wnt related signalling pathway, which in turn affects Slit-2 expression in a feedback loop. Functionally, transwell migration assays demonstrate a Slit-2 dose-dependent decrease of PCI52_PC cell migration. However, there is no influence on migration in mock control cells. Responsible for this migration block might be an interaction of P-cadherin with Roundabout (Robo)-3, a high affinity receptor of Slit-2. Indeed, proximity ligation assays exhibit P-cadherin/Robo-3 interactions on PCI52_PC cells. Additionally, we detect a modulation of this interaction by addition of recombinant Slit-2. Down-regulation of Robo-3 expression via small interfering RNA neutralizes Slit-2 induced migration block in PCI52_PC cells. In summary, our experiments show antitumorigenic effects of Slit-2 on P-cadherin expressing OSCC cells supposedly via modulation of Robo-3 interaction.


Asunto(s)
Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Movimiento Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Laríngeas/metabolismo , Neoplasias de la Boca/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Western Blotting , Cadherinas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Mucosa Bucal/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Proteínas del Tejido Nervioso/genética , Fosforilación , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Receptores de Superficie Celular , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
4.
Br J Cancer ; 105(2): 231-8, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21673687

RESUMEN

BACKGROUND: Malignant melanoma cells are known to have altered expression of growth factors compared with normal human melanocytes. These changes most likely favour tumour growth and progression, and influence tumour environment. The induction of transforming growth factor beta1, 2 and 3 as well as BMP4 and BMP7 expression in malignant melanoma has been reported before, whereas the expression of an important modulator of these molecules, connective tissue growth factor (CTGF), has not been investigated in melanomas until now. METHODS: Expression of CTGF was analysed in melanoma cell lines and tissue samples by qRT-PCR and immunohistochemistry. To determine the regulation of CTGF expression in malignant melanoma, specific siRNA was used. Additionally, migration, invasion and attachment assays were carried out. RESULTS: We were able to demonstrate that CTGF expression is upregulated in nine melanoma cell lines and in primary and metastatic melanoma in situ. The transcription factor HIF-1α was revealed as a positive regulator for CTGF expression. Melanoma cells, in which CTGF expression is diminished, show a strong reduction of migratory and invasive properties when compared with controls. Further, treatment of normal human epidermal melanocytes with recombinant CTGF leads to an increase of migratory and invasive behaviour of these cells. CONCLUSION: These results suggest that CTGF promotes melanoma cell invasion and migration and, therefore, has an important role in the progression of malignant melanoma.


Asunto(s)
Movimiento Celular/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/metabolismo , Línea Celular Tumoral , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Invasividad Neoplásica , ARN Interferente Pequeño/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
Osteoarthritis Cartilage ; 19(2): 206-12, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21134476

RESUMEN

OBJECTIVE: Recently, the transcription factor AP-2ɛ was shown to be a regulator of hypertrophy in cartilage and to be differentially expressed in osteoarthritis (OA). However, the only known target gene of AP-2ɛ up to date is integrin alpha10. To better characterize the function of AP-2ɛ in cartilage we screened for additional target genes. DESIGN: Promoter analysis, ChIP-assays and electrophoretic mobility shift assay were used to characterize the regulation of a new AP-2ɛ target gene in detail. RESULTS: In this study, we determined the chemokine CXCL1, already known to be important in osteoarthritis (OA), as a new target gene of AP-2ɛ. We could confirm that CXCL1 is expressed in chondrocytes and significantly over-expressed in OA-chondrocytes. Transient transfection of chondrocytes with an AP-2ɛ expression construct led to a significant increase of the CXCL1 mRNA level in these cells. We identified three potential AP-2 binding sites within the CXCL1 promoter and performed luciferase assays, indicating that an AP-2 binding motif (AP-2.2) ranging from position -135 to -144 bp relative to the translation start is responsive to AP-2ɛ. This result was further addressed by site-directed mutagenesis demonstrating that activation of the CXCL1 promoter by AP-2ɛ is exclusively dependent on AP-2.2. Chromatin immunoprecipitation and electromobility shift assays confirmed the direct binding of AP-2ɛ to the CXCL1 promoter in OA-chondrocytes at this site. CONCLUSION: These findings revealed CXCL1 as a novel target gene of AP-2ɛ in chondrocytes and support the important role of AP-2ɛ in cartilage.


Asunto(s)
Cartílago/metabolismo , Quimiocina CXCL1/metabolismo , Osteoartritis/metabolismo , Factor de Transcripción AP-2/metabolismo , Anciano , Células Cultivadas , Condrocitos/metabolismo , Electroforesis , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Factor de Transcripción AP-2/genética , Transfección
6.
Carcinogenesis ; 30(10): 1781-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19654099

RESUMEN

Cadherins belong to a family of Ca(2+)-dependent homophilic cell-cell adhesion proteins that are important for correct cellular localization and tissue integrity. They play a major role in the development and homeostasis of epithelial architecture. Recently, it has become more and more evident that P-cadherin contributes to the oncogenesis of many tumors. To analyze the role of P-cadherin in oral squamous cell carcinoma (OSCC), we used a cell line that was deficient of the classical cadherins, P-cadherin, E-cadherin and N-cadherin. This cell line was transfected with full-length P-cadherin (PCI52_PC). After overexpression of P-cadherin, PCI52_PC gained an epithelial-like brickstone morphology in contrast to the mock-transfected cells with a spindle-shaped mesenchymal morphology. Immunohistochemical analysis revealed a strong nuclear Snail staining in mock-transfected cells compared with a significantly reduced nuclear staining and translocation to the cytoplasm in P-cadherin-overexpressing cells. Interestingly, the effects triggered by P-cadherin overexpression could be reversed by transfecting the cells with an antisense P-cadherin plasmid construct. Additional investigations showed a reexpression of E-cadherin in all P-cadherin-transfected cell clones in contrast to the mock controls. Analyzing the signaling mechanism behind it, we found glycogen-synthase-kinase-3beta (GSK-3beta) bound to Snail in all cell clones. Furthermore, P-cadherin-overexpressing cell lines showed activated GSK-3beta that phosphorylated Snail leading to its cytoplasmic translocation. In summary, our results reveal P-cadherin as one major component in reconfiguring mesenchymal cells with epithelial features by triggering GSK-3beta-mediated inactivation and cytoplasmatic translocation of Snail in OSCC.


Asunto(s)
Cadherinas/toxicidad , Carcinoma de Células Escamosas/inducido químicamente , Glucógeno Sintasa Quinasa 3/metabolismo , Neoplasias de la Boca/inducido químicamente , Factores de Transcripción/metabolismo , Cadherinas/deficiencia , Cadherinas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , Amplificación de Genes , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Células HeLa , Humanos , Inmunohistoquímica , Cinética , Mesodermo/efectos de los fármacos , Mesodermo/fisiología , Neoplasias de la Boca/patología , Invasividad Neoplásica , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail , Transfección
8.
Br J Cancer ; 101(4): 551-6, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19638982

RESUMEN

Analysis of microRNA (miRNA) biogenesis and function is an area of research that started only recently but has subsequently accelerated tremendously. This is because of the impressive impact of miRNA-mediated gene regulation and the obvious potential of those tiny RNA molecules in future diagnostic and therapeutic applications. In this review, recent progress to reveal the role of miRNAs in the tumourigenesis of malignant melanoma, as well as future prospects of melanoma-related miRNA research, will be addressed.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Melanoma/genética , MicroARNs/genética , Neoplasias Cutáneas/genética , Animales , Perfilación de la Expresión Génica , Humanos
9.
Int J Immunopathol Pharmacol ; 22(3): 715-22, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19822088

RESUMEN

Changes in the expression of repellent factors, i.e., Netrins and their receptors, may be responsible for the invasive behavior of the synovial tissue cells in patients with rheumatoid arthritis (RA) and osteoarthritis (OA). This study was carried out to analyze the expression of Netrins and their receptors in synovial cells of patients with RA, OA, and control subjects without synovial inflammation. Quantitative RT-PCR was performed to measure the expression of Netrin-1, -3, -4, Neogenin, DCC, UNC5A-D. The influence of Netrin-1 on synovial fibroblasts (SF) was analyzed by determining proliferation, migration, and their ability to organize collagen. SF expressed all repellent factors of the Netrin family. When comparing SF of healthy donors to patients with RA and OA, a stronger expression of UNC5B (4 fold) and UNC5C (769 fold) in RA and OA was found, whereas expression of the other molecules revealed no significant differences. Treating the SF-cells with recombinant Netrin-1 resulted in inhibition of migration of RA- and OA-SFs whereas control cells were not affected. The stronger expression of UNC5B and UNC5C receptors might contribute to the disordered phenotype of RA- and OA-SFs. Addition of Netrin-1 reduces the migratory ability of SFs, potentially by repulsion, as seen in neuronal cells in embryonic development. Due to its function, Netrin-1 may constitute a novel target in the treatment of OA and RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Movimiento Celular , Fibroblastos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Osteoartritis/metabolismo , Receptores de Superficie Celular/metabolismo , Membrana Sinovial/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Cartílago/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Colágeno/metabolismo , Receptor DCC , Femenino , Fibroblastos/patología , Glicosaminoglicanos/metabolismo , Humanos , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Receptores de Netrina , Netrina-1 , Netrinas , Osteoartritis/genética , Osteoartritis/patología , Receptores de Superficie Celular/genética , Proteínas Recombinantes/metabolismo , Membrana Sinovial/patología , Proteínas Supresoras de Tumor/genética
10.
Gut ; 57(2): 243-51, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17881540

RESUMEN

BACKGROUND: Melanoma inhibitory activity 2 (MIA2) is a novel gene of the MIA gene family. The selective expression of MIA2 in hepatocytes is controlled by hepatocyte nuclear factor (HNF) 1 binding sites in the MIA2 promotor. In contrast, in most hepatocellular carcinomas (HCC) MIA2 expression is down-regulated or lost. AIM: In this study we examined the regulation and functional role of MIA2 in hepatocancerogenesis. METHODS AND RESULTS: In HCC cell lines and tissues HNF-1 expression was lower than in primary human hepatocytes (PHH) and corresponding non-tumorous tissue, respectively, and correlated significantly with the down-regulation of MIA2 expression. Re-expression of HNF-1 in HCC cells reinduced MIA2 in HCC cells to similar levels as found in PHH. Further, MIA2 was re-expressed in HCC cell lines by stable transfection, and the generated cell clones revealed a strongly reduced invasive potential and proliferation rate in vitro. In line with these findings treatment of HCC cells with recombinant MIA2 inhibited proliferation and invasion. In nude mice MIA2 re-expressing HCC cells grew significantly slower and revealed a less invasive growth pattern. Immunohistochemical analysis of a tissue microarray containing HCC and corresponding non-cancerous liver tissue of 85 patients confirmed reduced MIA2 expression in HCC. Furthermore, MIA2 negative HCC tissue showed a significantly higher Ki67 labelling index and loss of MIA2 expression correlated significantly with more advanced tumour stages. CONCLUSION: This study presents MIA2 as an inhibitor of HCC growth and invasion both in vitro and in vivo, and consequently, as a tumour suppressor of HCC. Further, our findings indicate a novel mechanism, how loss of HNF-1 expression in HCC affects tumorigenicity via down-regulation of MIA2.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas de la Matriz Extracelular/genética , Genes Supresores de Tumor , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias , Ciclo Celular , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Femenino , Factor Nuclear 1 del Hepatocito/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/antagonistas & inhibidores
11.
Oncogene ; 26(28): 4158-70, 2007 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-17173062

RESUMEN

Analyses of malignant melanomas revealed a strong expression of bone morphogenic proteins (BMPs) and their autocrine effect in promoting cell invasion and migration. Here, we report a paracrine effect of BMPs on the vascular network. Both BMP2 and BMP4 induced tube formation as well as the migratory efficiency of microvascular endothelial cells. Melanoma cells with reduced BMP activity attracted less endothelial cells in invasion assays than control cells. Furthermore, reduction of BMPs in melanoma cells had a strong effect on vasculogenic mimicry. Tube formation on matrigel was analysed for melanoma cells as well as in co-cultures of endothelial and melanoma cells. Melanoma cells with reduced BMP activity were not capable of forming cord-like structures by themselves and additionally inhibited tube formation of the endothelial cells. Genes involved in angiogenesis turned out to be strongly downregulated in these cell clones. Tumors derived from cells with impaired BMP activity showed reduced tumor growth or large necrotic areas owing to lack of angiogenesis in in vivo analyses.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Melanoma/irrigación sanguínea , Neovascularización Patológica , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Cartilla de ADN , Humanos , Inmunohistoquímica , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos
12.
Cell Death Differ ; 14(8): 1414-21, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17431427

RESUMEN

The p66shc protein governs oxidant stress and mammalian lifespan. Here, we identify melanoma inhibitory activity (MIA), a protein secreted by melanoma cells, as a novel binding partner and antagonist of p66shc. The N-terminal collagen homology-2 (CH2) domain of p66shc binds to the Src Homology-3 (SH3)-like domain of MIA in vitro. In cells, ectopically expressed MIA and p66shc colocalize and co-precipitate. MIA also co-precipitates with the CH2 domain of p66shc in vivo. MIA expression in vivo suppresses p66shc-stimulated increase in endogenous hydrogen peroxide (H(2)O(2)), and inhibits basal and H(2)O(2)-induced phosphorylation of p66shc on serine 36 and H(2)O(2)-induced death. In human melanoma cells expressing MIA, endogenous MIA and p66shc co-precipitate. Downregulation of MIA in melanoma cells increases basal and ultraviolet radiation (UVR)-induced phosphorylation of p66shc on serine 36, augments endogenous H(2)O(2) levels, and increases their susceptibility to UVR-induced death. These findings show that MIA binds to p66shc, and suggest that this interaction antagonizes phosphorylation and function of p66shc.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas de la Matriz Extracelular/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Sitios de Unión , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Proteínas de la Matriz Extracelular/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Melanoma/genética , Melanoma/patología , Ratones , Proteínas de Neoplasias/genética , Estrés Oxidativo , Fosforilación , Unión Proteica , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
13.
Curr Protein Pept Sci ; 9(3): 221-6, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18537677

RESUMEN

In the year 1994, the protein MIA (melanoma inhibitory activity) was found to be strongly expressed and secreted by malignant melanomas and subsequent studies revealed that MIA has an important function in melanoma development and invasion. Multidimensional NMR-spectroscopy and x-ray crystallography revealed that recombinant human MIA adopts a Src homology 3 (SH3) domain-like fold in solution, a structure with two perpendicular antiparallel three- and five-stranded beta-sheets. SH3 domains are protein modules that are found in many intracellular signalling proteins and mediate protein-protein interactions by binding to proline-rich peptide sequences. Unlike previously described protein structures with SH3 domain folds, MIA is a secreted single-domain protein of 12 kDa that contains an additional antiparallel beta-sheet and two disulfide bonds. Furthermore, the charge surrounding the canonical binding site differs from that of classical SH3 domains. The two disulfide bonds are crucial for correct folding and function as revealed by mutation analysis. Therefore, MIA appears to be the first extracellular protein adopting an SH3 domain-like fold. MIA was shown to interact with fibronectin, and MIA-interacting peptide ligands identified by phage display screening are similar to the consensus sequence of type III human fibronectin repeats, especially FN14. Interestingly, recent data revealed that MIA can also directly bind to integrin alpha 4 beta 1 and alpha 5 beta1 and that it modulates integrin activity negatively. These findings suggest an interesting role of the SH3-domain proteins in the extracellular compartment. Recently, MIA homologous proteins with a sequence identity of 44% and a sequence homology of approximately 80% were determined (TANGO, MIA-2, OTOR). This clearly suggests that this structural device is used more frequently, in processes ranging from developmental changes to the interference of cell attachment in the extracellular matrix. Detailed studies are necessary to determine the exact function of the MIA homologous proteins. It will be interesting to know whether additional protein families can be identified which are secreted and carry SH3 domain-like modules, in addition to elucidate what the specific physiological targets of this protein family are.


Asunto(s)
Proteínas de la Matriz Extracelular/química , Dominios Homologos src , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de la Matriz Extracelular/clasificación , Humanos , Melanoma/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína
14.
FEBS J ; 275(16): 4198-210, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18637117

RESUMEN

Cadherins belong to a family of homophilic cell-cell adhesion proteins that are responsible for the establishment of a precise cell architecture and tissue integrity. Moreover, experimental data suggest that loss of intercellular adhesion is inversely correlated with cellular differentiation. Furthermore, dedifferentiation is closely linked to tumor progression. Recently, we have shown that a secreted 50 kDa N-terminal fragment of P-cadherin plays a role in the progression of malignant melanoma. In this study, we have detected both the full-length and the truncated versions of P-cadherin in cell lysates of differentiated head and neck oral squamous cell carcinoma cell lines, whereas in cell lysates of dedifferentiated cell lines, we detected only the truncated 50 kDa version of P-cadherin. Treatment of the cell lines with a recombinantly expressed biotinylated, soluble 50 kDa form of the N-terminal part of P-cadherin revealed a major effect on cell aggregation and migration of oral squamous cell carcinoma cells. However, the 50 kDa N-terminal fragment of P-cadherin did not show any influence on cell proliferation in 2D and 3D cell culture. These results suggest that generation of truncated P-cadherin during the progression of oral squamous carcinoma attenuates tissue integrity, facilitates cellular separation, and leads to the acquisition of a more migratory phenotype.


Asunto(s)
Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/metabolismo , Cadherinas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Humanos , Masculino , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Eliminación de Secuencia
15.
Oncogene ; 37(7): 897-911, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29059159

RESUMEN

Malignant melanoma reveals rapidly increasing incidence and mortality rates worldwide. By now, BRAF inhibition is the standard therapy for advanced melanoma in patients carrying BRAF mutations. However, only approximately 50% of melanoma patients harbor therapeutically attackable BRAF mutations, and overall survival after treatment with BRAF inhibitors is modest. KRAS (Kirsten Rat sarcoma) proteins are acting upstream of BRAF and have a major role in human cancer. Recent approaches awaken the hope to use KRAS inhibition (KRASi) as a clinical tool. In this study, we identified wild-type KRAS as a novel therapeutic target in melanoma. KRASi functions synergistically with BRAF inhibition to reduce melanoma proliferation and to induce apoptosis independently of BRAF mutational status. Moreover, acquired resistance to BRAF inhibitors in melanoma is dependent on dynamic regulation of KRAS expression with subsequent AKT and extracellular-signal regulated kinase activation and can be overcome by KRASi. This suggests KRASi as novel approach in melanoma-alone or in combination with other therapeutic regimes.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Resistencia a Antineoplásicos/genética , Melanoma/patología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias Cutáneas/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Estudios de Seguimiento , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/genética , Ratones , Ratones Desnudos , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Melanoma Cutáneo Maligno
16.
Oncogene ; 25(2): 248-59, 2006 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-16132038

RESUMEN

E-cadherin is known to be an important molecule in epithelial-mesenchymal transition (EMT). Malignant transformation of melanocytes frequently attends with loss of E-cadherin expression and induction of expression of mesenchymal molecules like N-cadherin. The switch of the cadherin class is an interesting phenomenon of melanoma cells and in EMT in general. Therefore, we analysed the capacity of E-cadherin to regulate expression of N-cadherin in melanocytic cells. Our experiments revealed that melanoma cells downregulate endogenous N-cadherin expression after transient transfection of full-length E-cadherin, but also of the cytoplasmic domain of E-cadherin. Therefore, we concluded that the extracellular domain of E-cadherin and cell-cell contacts are not necessary for negative regulation of N-cadherin. Melanoma cells re-expressing full-length or cytoplasmatic E-cadherin have reduced NFkappaB activity in comparison to mock-transfected cells. Downregulation of NFkappaB activity, either directly or by re-expression of E-cadherin, led to a suppression of N-cadherin promoter activity and N-cadherin expression. Consequently, an NFkappaB-binding site in the N-cadherin promoter was characterized. In summary, our results suggest that N-cadherin is directly regulated by E-cadherin. Loss of E-cadherin induces NFkappaB activity and N-cadherin expression in tumorigenic EMT.


Asunto(s)
Cadherinas/genética , Cadherinas/metabolismo , Cadherinas/farmacología , Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Sitios de Unión , Western Blotting , Movimiento Celular , Regulación hacia Abajo , Humanos , Melanocitos/citología , Melanocitos/metabolismo , Melanoma/genética , Melanoma/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica/patología , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Transcripción Genética , Transfección
17.
Oncogene ; 25(17): 2546-57, 2006 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-16331256

RESUMEN

Gastric cancer (GC) is one of the most common malignancies worldwide. Genes expressed only in cancer tissue will be useful molecular markers for diagnosis and may also be good therapeutic targets. However, little is known about cancer-specific genes, at least in GC. In this study, we searched for GC-specific genes by serial analysis of gene expression (SAGE) data analysis and quantitative reverse transcription (RT)-PCR. Comparing GC SAGE libraries with those of various normal tissues in the SAGEmap database, we identified 54 candidate GC-specific genes. Quantitative RT-PCR analysis of these candidates revealed that APin protein (APIN), taxol resistance-associated gene 3 (TRAG3), cytochrome P450, family 2, subfamily W, polypeptide 1 (CYP2W1), melanoma inhibitory activity (MIA), matrix metalloproteinase-10 (MMP-10), dickkopf homolog 4 (DKK4), GW112, regenerating islet-derived family, member 4 (REGIV), and HORMA domain-containing 1 (HORMAD1) were expressed much more highly in GC than in 14 kinds of normal tissues. Immunohistochemical staining for MIA, MMP-10, and DKK4 was found in 47 (31.1%), 68 (45.0%), and two (1.3%) of 151 GCs, respectively, and staining for both MIA and MMP-10 was correlated with poor prognosis in advanced GC (P=0.0001 and 0.0141, respectively). Moreover, enzyme-linked immunosorbent assay showed high levels of MMP-10 (65/69, 94.2%) in serum samples from patients with GC. Levels of MIA were raised in a small proportion of serum samples from patients with GC (4/69, 5.8%). In Boyden chamber invasion assays, MIA-transfected GC cells were up to three times more invasive than cells transfected with empty vector. Taken together, these results suggest that MMP-10 is a good marker for the detection of GC and that MIA and MMP-10 are prognostic factors for GC. As expression of MIA and MMP-10 is narrowly restricted in cancer, these two molecules may be good therapeutic targets for GC.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Metaloendopeptidasas/genética , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Proteínas de la Matriz Extracelular , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 10 de la Matriz , Metaloendopeptidasas/sangre , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/sangre , Estadificación de Neoplasias , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/sangre , Neoplasias Gástricas/patología
18.
Oncogene ; 25(36): 5027-36, 2006 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-16568085

RESUMEN

The Dickkopf (DKK) genes were originally identified as factors inducing head formation in Xenopus. The genes code for inhibitors that are involved in Wnt signaling. We speculate that loss of DKK expression plays a role in development or progression of malignant melanoma. Thus, we evaluated melanoma cell lines and tissue samples of malignant melanoma for loss of DKK, especially DKK-3 transcription. We found that DKK-1, -2 and -3 were downregulated or lost in all cell lines and in most of the tumor samples analysed. Reduced DKK-3 expression occurred as early as in primary tumors detected by both immunohistochemical and reverse transcription-polymerase chain reaction RT-PCR analysis. Functional assays with stable DKK-3 transfected cell lines revealed that DKK-3 expression increased cell-cell adhesion and decreased cell migration. Further, downregulation of fibronectin, snail-1 and re-expression of E-cadherin was found in the DKK-3 expressing cell clones supporting a role of DKK-3 in tumor progression. Our studies thus indicate that loss of DKK-3 expression may contribute to melanoma progression.


Asunto(s)
Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Melanoma/genética , Proteínas Adaptadoras Transductoras de Señales , Adhesión Celular , Línea Celular Tumoral , Quimiocinas , Genes Supresores de Tumor , Humanos , Inmunohistoquímica , Melanoma/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Oncogene ; 25(1): 103-10, 2006 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-16170365

RESUMEN

The human gene Hugl-1 (Llgl/Lgl1) has significant homology to the Drosophila tumor suppressor gene lethal(2)giant larvae (lgl). The lgl gene codes for a cortical cytoskeleton protein, Lgl, that is involved in maintaining cell polarity and epithelial integrity. We speculate that Hugl-1 might play a role in epithelial-mesenchymal transition (EMT) and that loss of Hugl-1 expression plays a role in the development or progression of malignant melanoma. Thus, we evaluated melanoma cell lines and tissue samples of malignant melanoma for loss of Hugl-1 transcription. We found that Hugl-1 was downregulated or lost in all cell lines and in most of the tumor samples analysed, and that these losses were associated with advanced stage of the disease. Reduced Hugl-1 expression occurred as early as in primary tumors detected by both immunohistochemical and reverse transcription-polymerase chain reaction (RT-PCR) analysis. Functional assays with stable Hugl-1-transfected cell lines revealed that Hugl-1 expression increased cell adhesion and decreased cell migration. Further, downregulation of MMP2 and MMP14 (MT1-MMP) and re-expression of E-cadherin was found in the Hugl-1-expressing cell clones supporting a role of Hugl-1 in EMT. Our studies thus indicate that loss of Hugl-1 expression contributes to melanoma progression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteínas/metabolismo , Western Blotting , Cadherinas/biosíntesis , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proteínas del Citoesqueleto , Progresión de la Enfermedad , Regulación hacia Abajo , Epitelio/patología , Humanos , Inmunohistoquímica , Metaloproteinasa 14 de la Matriz , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasas de la Matriz/biosíntesis , Metaloproteinasas de la Matriz Asociadas a la Membrana , Melanoma/genética , Microscopía Fluorescente , Invasividad Neoplásica , Proteínas/genética , ARN/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular , Transcripción Genética , Transfección
20.
Int J Oncol ; 30(2): 499-507, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17203233

RESUMEN

The transforming growth factor-beta (TGF-beta) plays a pivotal role in the pathobiology of human gliomas: during carcinogenesis, it turns from a tumor suppressor to a tumor promoter. The traditional Smad pathway and the more recently discovered MAPK pathway are the most important pathways for TGF-beta related intracellular signal transduction mediating differential pathobiological effects. In this study, we investigated the effects of TGF-beta2 and the TGF-beta2 antisense phosphorothioate oligodeoxynucleotide (PTO) AS-11 on the functionality of both the Smad and MAPK pathways in high-grade gliomas. We aimed to correlate the imbalance between the pathways with differences in the behaviour of high-grade glioma cells. Gene and protein expression studies were used to detect levels of members of the Smad and MAPK pathways under regulation of TGF-beta2 and AS-11. Proliferation and migration assays were functional readouts for effects caused by these regulating tools. Gene arrays were used to detect yet unknown regulators of these functional effects. The Smad pathway was functional in the tested cell lines. Exogenous TGF-beta2 inhibited proliferation but enhanced migration. Smad 2 mRNA expression and activation were significantly reduced by incubation with AS-11. K-ras was reduced both in gene arrays and quPCR under treatment with AS-11, but there was no influence of K-ras down-regulation on the activity of ERK. Ubiquitination-related genes also were specifically down-regulated with AS-11. Our results indicate the involvement of K-ras in TGF-beta signaling in high-grade gliomas. ERK, which is a member of the MAPK pathway, was not influenced and seems to be activated through RAS independent cascades in glioma. These results suggest that combined antagonization of the TGF-beta and MAPK pathways might be a promising approach for glioma therapy. An imbalance between these two pathways might be responsible for TGF-beta switching to a tumor promoter protein in high-grade gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Modelos Biológicos , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Transducción de Señal , Factor de Crecimiento Transformador beta2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA