Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Oncogene ; 35(27): 3485-94, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-26500063

RESUMEN

Cellular senescence is an antiproliferative response with essential functions in tumor suppression and tissue homeostasis. Here we show that SIX1, a member of the SIX family of homeobox transcriptional factors, is a novel repressor of senescence. Our data show that SIX1 is specifically downregulated in fibroblasts upon oncogenic stress and other pro-senescence stimuli, as well as in senescent skin premalignant lesions. Silencing of SIX1 in human fibroblasts suffices to trigger senescence, which is mediated by p16INK4A and lacks a canonical senescence-associated secretory phenotype. Interestingly, SIX1-associated senescence is further characterized by the expression of a set of development and differentiation-related genes that significantly overlap with genes associated with SIX1 in organogenesis or human tumors, and show coincident regulation in oncogene-induced senescence. Mechanistically, we show that gene regulation by SIX1 during senescence is mediated, at least in part, by cooperation with Polycomb repressive complexes. In summary, our results identify SIX1, a key development regulator altered in human tumors, as a critical repressor of cellular senescence, providing a novel connection between senescence, differentiation and tumorigenesis.


Asunto(s)
Diferenciación Celular/genética , Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Animales , Western Blotting , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fibroblastos/citología , Perfilación de la Expresión Génica/métodos , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Ratones de la Cepa 129 , Papiloma/genética , Papiloma/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
2.
Oncogene ; 34(8): 951-64, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24632622

RESUMEN

Epithelial-mesenchymal transition (EMT) has been associated with increased aggressiveness and acquisition of migratory properties providing tumor cells with the ability to invade into adjacent tissues. Downregulation of E-cadherin, a hallmark of EMT, is mediated by several transcription factors (EMT-TFs) that act also as EMT inducers, among them, Snail1 and the bHLH transcription factor E47. We previously described lysyl oxidase-like 2 (LOXL2), a member of the lysyl oxidase family, as a Snail1 regulator and EMT inducer. Here we show that LOXL2 is also an E47-interacting partner and functionally collaborates in the repression of E-cadherin promoter. Loss and gain of function analyses combined with in vivo studies in syngeneic breast cancer models demonstrate the participation of LOXL2 and E47 in tumor growth and their requirement for lung metastasis. Furthermore, LOXL2 and E47 contribute to early steps of metastatic colonization by cell and noncell autonomous functions regulating the recruitment of bone marrow progenitor cells to the lungs and by direct transcriptional regulation of fibronectin and cytokines TNFα, ANG-1 and GM-CSF. Moreover, fibronectin and GM-CSF proved to be necessary for LOXL2/E47-mediated modulation of tumor growth and lung metastasis.


Asunto(s)
Aminoácido Oxidorreductasas/fisiología , Cadherinas/genética , Metástasis de la Neoplasia/genética , Factor de Transcripción 3/fisiología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Movimiento Celular/genética , Células Cultivadas , Perros , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos
3.
Oncogene ; 34(20): 2609-20, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25023702

RESUMEN

The presence of hypoxic regions in solid tumors is an adverse prognostic factor for patient outcome. Here, we show that hypoxia induces the expression of Ephrin-A3 through a novel hypoxia-inducible factor (HIF)-mediated mechanism. In response to hypoxia, the coding EFNA3 mRNA levels remained relatively stable, but HIFs drove the expression of previously unknown long noncoding (lnc) RNAs from EFNA3 locus and these lncRNA caused Ephrin-A3 protein accumulation. Ephrins are cell surface proteins that regulate diverse biological processes by modulating cellular adhesion and repulsion. Mounting evidence implicates deregulated ephrin function in multiple aspects of tumor biology. We demonstrate that sustained expression of both Ephrin-A3 and novel EFNA3 lncRNAs increased the metastatic potential of human breast cancer cells, possibly by increasing the ability of tumor cells to extravasate from the blood vessels into surrounding tissue. In agreement, we found a strong correlation between high EFNA3 expression and shorter metastasis-free survival in breast cancer patients. Taken together, our results suggest that hypoxia could contribute to metastatic spread of breast cancer via HIF-mediated induction of EFNA3 lncRNAs and subsequent Ephrin-A3 protein accumulation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Sitios Genéticos , Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/biosíntesis , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Efrina-A3/genética , Efrina-A3/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Pez Cebra
4.
Oncogene ; 33(15): 1945-53, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-23604125

RESUMEN

ING proteins have an essential role in the control of a variety of cellular functions whose deregulation is associated with tumor formation and dissemination, such as proliferation, apoptosis, senescence or invasion. Accordingly, loss of function of ING proteins is a frequent event in many types of human tumors. In this report, we have studied the function of ING4, a member of the ING family of tumor suppressors, in the context of normal, non-transformed primary fibroblasts. We show that ING4 negatively regulates cell proliferation in this cell type. The antiproliferative action of ING4 requires its ability to recognize chromatin marks, it is p53-dependent at least in part, and it is lost in an ING4 cancer-associated mutant. Gene expression analysis shows that ING4 regulates the expression and release of soluble factors of the chemokine family. The secretory phenotype regulated by ING4 in primary fibroblasts displays a selective paracrine effect on proliferation, fostering the division of tumor cells, while inhibiting division in primary fibroblasts. Consistently, ING4-expressing fibroblasts promoted tumor growth in vivo in co-injection tumorigenesis assays. Collectively, our results show that ING4 not only can regulate the proliferation of primary non-transformed human fibroblasts, but also orchestrates a secretory phenotype in these cells that promotes tumor cell proliferation in vitro and in vivo. These findings support a critical role for ING4 expression in normal cells in the non-cell-autonomous regulation of tumor growth.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Western Blotting , Proteínas de Ciclo Celular/genética , Línea Celular , Inmunoprecipitación de Cromatina , Citocinas/metabolismo , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Xenoinjertos , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética
5.
Oncogene ; 32(4): 403-13, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22430211

RESUMEN

In the western world, endometrial carcinoma (EC) is the most common cancer of the female genital tract. The annual incidence has been estimated at 10-20 per 100,000 women. Two clinicopathological variants are recognized: the estrogen related (type I, endometrioid) and the non-estrogen related (type II, non-endometrioid).The clinicopathological differences are paralleled by specific genetic alterations, with type I showing microsatellite instability and mutations in phosphatase and tensin homologue deleted on chromosome 10, PIK3CA, K-RAS and CTNNB1 (ß-catenin), and type II exhibiting TP53 mutations and chromosomal instability. Some non-endometrioid carcinomas probably arise from pre-existing endometrioid carcinomas as a result of tumor progression and, not surprisingly, some tumors exhibit combined or mixed features at the clinical, pathological and molecular levels. In EC, apoptosis resistance may have a role in tumor progression. Understanding pathogenesis at the molecular level is essential in identifying biomarkers for successful targeted therapies. In this review, the genetic changes of endometrial carcinogenesis are discussed in the light of the morphological features of the tumors and their precursors.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Animales , Progresión de la Enfermedad , Femenino , Humanos
6.
Clin. transl. oncol. (Print) ; 14(10): 715-720, oct. 2012. ilus
Artículo en Inglés | IBECS | ID: ibc-127006

RESUMEN

Endometrial cancer (EC) is the most common gynecologic malignancy of the female genital tract and the fourth most common neoplasia in women. In EC, myometrial invasion is considered one of the most important prognostic factors. For this process to occur, epithelial tumor cells need to undergo an epithelial to mesenchymal transition (EMT), either transiently or stably, and to differing degrees. This process has been extensively described in other types of cancer but has been poorly studied in EC. In this review, several features of EMT and the main molecular pathways responsible for triggering this process are investigated in relation to EC. The most common hallmarks of EMT have been found in EC, either at the level of E-cadherin loss or at the induction of its repressors, as well as other molecular alterations consistent with the mesenchymal phenotype-like L1CAM and BMI-1 up-regulation. Pathways including progesterone receptor, TGFβ, ETV5 and microRNAs are deeply related to the EMT process in EC (AU)


Asunto(s)
Humanos , Femenino , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/genética
7.
Oncogene ; 31(16): 2049-61, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21927030

RESUMEN

Somatostatin receptors (sst1-5) are present in different types of tumors, where they inhibit key cellular processes such as proliferation and invasion. Although ssts are densely expressed in breast cancer, especially sst2, their role and therapeutic potential remain uncertain. Recently, we identified a new truncated sst5 variant, sst5TMD4, which is related to the abnormal response of certain pituitary tumors to treatment with somatostatin analogs. Here, we investigated the possible role of sst5TMD4 in breast cancer. This study revealed that sst5TMD4 is absent in normal mammary gland, but is abundant in a subset of poorly differentiated human breast tumors, where its expression correlated to that of sst2. Moreover, in the MCF-7 breast cancer model cell, sst5TMD4 expression increased malignancy features such as invasion and proliferation abilities (both in cell cultures and nude mice). This was likely mediated by sst5TMD4-induced increase in phosphorylated extracellular signal-regulated kinases 1 and 2 and p-Akt levels, and cyclin D3 and Arp2/3 complex expression, which also led to mesenchymal-like phenotype. Interestingly, sst5TMD4 interacts physically with sst2 and thereby alters its signaling, enabling disruption of sst2 inhibitory feedback and providing a plausible basis for our findings. These results suggest that sst5TMD4 could be involved in the pathophysiology of certain types of breast tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Variación Genética , Receptores de Somatostatina/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , MAP Quinasa Quinasa 1/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Pronóstico , Somatostatina/fisiología
8.
Clin. transl. oncol. (Print) ; 10(12): 777-785, dic. 2008.
Artículo en Inglés | IBECS | ID: ibc-123557

RESUMEN

Advances in the analysis of expression profiles, using genomic techniques, have revealed the high heterogeneity present in breast cancers. These approaches have served to identify different breast cancer subgroups with specific molecular characteristics that could sub-classify these tumours as carcinomas expressing hormone receptors, denominated Luminal subtype, and tumours with negative expression of hormone receptors, the Basal and HER2+ phenotypes. Therefore, during recent years, identification of markers characteristic of each subtype has been the focus of many research groups. All of these breast tumour subtypes probably have specific clinical and morphological features; however, this hypothesis needs to be confirmed by analysing more homogenous series. Although this "new" classification has limitations, it could be useful in the clinical practice, allowing not only a more accurate prognosis in breast cancer patients but also a selective treatment for each predefined subtype (AU)


No disponible


Asunto(s)
Humanos , Femenino , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/diagnóstico , Células Madre Neoplásicas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Clasificación/métodos , Glándulas Mamarias Humanas/anatomía & histología , Glándulas Mamarias Humanas/patología , Modelos Biológicos , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Diagnóstico Molecular
9.
Oncogene ; 27(55): 6958-69, 2008 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19029937

RESUMEN

The epithelial-to-mesenchymal transition (EMT) is a crucial process in tumour progression providing tumour cells with the ability to escape from the primary tumour, to migrate to distant regions and to invade tissues. EMT requires a loss of cell-cell adhesion and apical-basal polarity, as well as the acquisition of a fibroblastoid motile phenotype. Several transcription factors have emerged in recent years that induce EMT, with important implications for tumour progression. However, their effects on cell polarity remain unclear. Here, we have re-examined the data available related to the effect of EMT related transcription factors on epithelial cell plasticity, focusing on their impact on cell polarity. Transcriptional and post-transcriptional regulatory mechanisms mediated by several inducers of EMT, in particular the ZEB and Snail factors, downregulate the expression and/or functional organization of core polarity complexes. We also summarize data on the expression of cell polarity genes in human tumours and analyse genetic interactions that highlight the existence of complex regulatory networks converging on the regulation of cell polarity by EMT inducers in human breast carcinomas. These recent observations provide new insights into the relationship between alterations in cell polarity components and EMT in cancer, opening new avenues for their potential use as therapeutic targets to prevent tumour progression.


Asunto(s)
Polaridad Celular/genética , Epitelio/fisiología , Regulación de la Expresión Génica/fisiología , Mesodermo/fisiología , Neoplasias/genética , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Desdiferenciación Celular/genética , Epitelio/metabolismo , Redes Reguladoras de Genes/fisiología , Humanos , Mesodermo/metabolismo , Modelos Biológicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/fisiología , Neoplasias/patología
10.
Oncogene ; 27(37): 5033-44, 2008 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-18454173

RESUMEN

It has been recently shown that cannabinoids, the active components of marijuana and their derivatives, inhibit cell cycle progression of human breast cancer cells. Here we studied the mechanism of Delta(9)-tetrahydrocannabinol (THC) antiproliferative action in these cells, and show that it involves the modulation of JunD, a member of the AP-1 transcription factor family. THC activates JunD both by upregulating gene expression and by translocating the protein to the nuclear compartment, and these events are accompanied by a decrease in cell proliferation. Of interest, neither JunD activation nor proliferation inhibition was observed in human non-tumour mammary epithelial cells exposed to THC. We confirmed the importance of JunD in THC action by RNA interference and genetic ablation. Thus, in both JunD-silenced human breast cancer cells and JunD knockout mice-derived immortalized fibroblasts, the antiproliferative effect exerted by THC was significantly diminished. Gene array and siRNA experiments support that the cyclin-dependent kinase inhibitor p27 and the tumour suppressor gene testin are candidate JunD targets in cannabinoid action. In addition, our data suggest that the stress-regulated protein p8 participates in THC antiproliferative action in a JunD-independent manner. In summary, this is the first report showing not only that cannabinoids regulate JunD but, more generally, that JunD activation reduces the proliferation of cancer cells, which points to a new target to inhibit breast cancer progression.


Asunto(s)
Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Dronabinol/farmacología , Proteínas Proto-Oncogénicas c-jun/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
11.
Oncogene ; 27(34): 4690-701, 2008 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-18408755

RESUMEN

Snai1 (Snail) and Snai2 (Slug), the two main members of Snail family factors, are important mediators of epithelial-mesenchymal transitions and involved in tumor progression. We recently reported that Snai1 plays a major role in tumor growth, invasion and metastasis, but the contribution of Snai2 to tumorigenesis is not yet well understood. To approach this question we have silenced Snai2 and/or Snai1 by stable RNA interference in two independent mouse skin carcinoma (HaCa4 and CarB) cell lines. We demonstrate that Snai2 knockdown has a milder effect, but collaborates with Snai1 silencing in reduction of tumor growth potential of either carcinoma cell line when injected into nude mice. Importantly, Snai1 or Snai2 silencing dramatically influences the metastatic ability of squamous carcinoma HaCa4 cells, inducing a strong reduction in liver and lung distant metastasis. However, only Snai1 knockdown has an effective action on invasiveness and fully abolishes tumor cell dissemination into the spleen. These results demonstrate that Snai1 and Snai2 collaborate on primary tumor growth and specifically contribute to site-specific metastasis of HaCa4 cells. These data also indicate that Snai1 is the major regulator of local invasion, supporting a hierarchical participation of both factors in the metastatic process.


Asunto(s)
Carcinoma/patología , Proliferación Celular , Metástasis de la Neoplasia/genética , Neoplasias Cutáneas/patología , Factores de Transcripción/fisiología , Animales , Biomarcadores de Tumor/metabolismo , Carcinoma/genética , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Interferente Pequeño/farmacología , Neoplasias Cutáneas/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/antagonistas & inhibidores , Trasplante Heterólogo , Carga Tumoral/genética
12.
Oncogene ; 26(20): 2902-13, 2007 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-17099726

RESUMEN

Among microtubule-targeting agents, docetaxel has received recent interest owing to its good therapeutic index. Clinical trials have underlined its potential for the treatment of advanced breast cancer, although little is known about its molecular mode of action in this context. We characterized the molecular changes induced by docetaxel in two well-known human breast carcinoma cell lines. Two mechanisms of action according to drug concentration were suggested by a biphasic sensitivity curve, and were further validated by cell morphology, cell cycle and cell death changes. Two to four nanomolar docetaxel induced aberrant mitosis followed by late necrosis, and 100 nM docetaxel induced mitotic arrest followed by apoptosis. Passing through mitosis phase was a requirement for hypodiploidy to occur, as shown by functional studies in synchronized cells and by combining docetaxel with the proteasome inhibitor MG132. Transcriptional profiling showed differences according to cell line and docetaxel concentration, with cell cycle, cell death and structural genes commonly regulated in both cell lines. Although p53 targets were mainly induced with low concentration of drug in MCF7 cells, its relevance in the dual mechanism of docetaxel cytotoxicity was ruled out by using an isogenic shp53 cell line. Many of the genes shown in this study may contribute to the dual mechanism by which docetaxel inhibits the growth of breast cancer cells at different concentrations. These findings provide a basis for rationally enhancing docetaxel therapy, considering lower concentrations, and better drug combinations.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Carcinoma/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Mitosis/efectos de los fármacos , Taxoides/farmacología , Antineoplásicos/farmacología , Apoptosis/genética , Neoplasias de la Mama/patología , Carcinoma/patología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Diploidia , Docetaxel , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Mitosis/genética , Células Tumorales Cultivadas
13.
J Pathol ; 208(5): 708-13, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16429393

RESUMEN

Some low-grade endometrioid carcinomas arise from a background of endometrioid tumours of borderline malignancy. To determine the molecular mechanisms involved in the initiation of endometrioid carcinoma, the present study investigated whether the genetic alterations reported in these tumours (mutations in PTEN, KRAS, and beta-catenin genes, and microsatellite instability) are already present in endometrioid tumours of borderline malignancy. Eight endometrioid tumours of borderline malignancy were studied. By immunohistochemistry, beta-catenin was expressed in the nuclei of all tumours, suggesting the presence of stabilizing beta-catenin mutations. By mutational analysis, five different beta-catenin mutations were found in seven of eight cases (90%), affecting codons 32, 33, and 37. In contrast, only one tumour harboured a PTEN mutation, which affected codon 130. Neither KRAS mutations nor microsatellite instability was detected. A review of the literature indicated that beta-catenin mutations are characteristic of well-differentiated endometrioid carcinomas, since they were present in nearly 60% of grade I but in less of 3% of grade III tumours. In conclusion, the present study identifies beta-catenin mutation as a nearly constant molecular alteration in borderline endometrioid tumours, whereas PTEN and KRAS mutations and microsatellite instability are very infrequent. The findings in the present study, and previously reported data, strongly suggest that beta-catenin mutation is an early event in endometrioid ovarian carcinogenesis, and that it is involved in the development of low-grade endometrioid tumours.


Asunto(s)
Carcinoma Endometrioide/genética , Mutación , Neoplasias Ováricas/genética , beta Catenina/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Análisis Mutacional de ADN/métodos , ADN de Neoplasias/genética , Femenino , Humanos , Repeticiones de Microsatélite , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , beta Catenina/metabolismo , Proteínas ras
15.
Cancer ; 92(11): 2829-36, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11753956

RESUMEN

BACKGROUND: Microsatellite instability (MI) is frequent in endometrial carcinomas (ECs), but its occurrence in ovarian tumors is uncertain. Microsatellite instability positive ECs frequently are associated with frameshift mutations in coding mononucleotide tracts in IGFIIR, BAX, hMSH6, and hMSH3. METHODS: DNA from 52 consecutive patients with ovarian tumors (10 benign, 7 borderline, and 35 malignant) was obtained from neoplastic and normal tissue. After preliminary results, the series was expanded by including 41 additional, previously selected, endometrioid and clear cell carcinomas. Microsatellite instability analysis was assessed by evaluating three (CA)n dinucleotide repeats (D2S123, D5S346, D17S250) and two mononucleotide tracts (BAT 25 and BAT 26). Frameshift mutations at coding mononucleotide repeats (IGFIIR, TGF beta II, BAX, hMSH6, and hMSH3) were investigated by single-strand conformation polymorphism analysis and DNA sequencing. MLH-1 methylation was assessed by methylation specific PCR. RESULTS: Microsatellite instability was identified in only 2 of the 52 (3.8%) tumors of the initial series (1 endometrioid and 1 clear cell carcinoma). After expanding the initial series of 15 endometrioid and clear cell carcinomas with 41 additional endometrioid and clear cell carcinomas, MI was found in 7 of the total series of 56 endometrioid and clear cell carcinomas (12.5%). Frameshift mutations in coding mononucleotide tracts were detected in BAX (6 of 7), IGFIIR (1 of 7), and MSH3 (2 of 7). MLH-1 promoter hypermethylation was identified in three of six MI positive tumors. CONCLUSIONS: Microsatellite instability was infrequent in this series of ovarian tumors, and it was limited to endometrioid and clear cell carcinomas. Like EC, many ovarian carcinomas with MI follow the same process of MLH-1 promoter methylation and accumulation of mutations in coding mononucleotide tracts.


Asunto(s)
Repeticiones de Microsatélite/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras , Metilación de ADN , Proteínas de Unión al ADN/genética , Femenino , Mutación del Sistema de Lectura , Humanos , Homólogo 1 de la Proteína MutL , Proteína 3 Homóloga de MutS , Estadificación de Neoplasias , Proteínas Nucleares , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteína X Asociada a bcl-2
16.
Br J Dermatol ; 145(4): 576-81, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11703283

RESUMEN

BACKGROUND: beta-catenin functions in signal transduction in the Wnt signalling pathway, which has recently been implicated in hair follicle (HF) morphogenesis. beta-catenin gene mutations affecting exon 3 have been reported in a high percentage of human pilomatrixomas. However, the expression pattern of beta-catenin in human HFs and pilomatrixomas has not been reported. OBJECTIVES: To analyse immunohistochemically the expression pattern of beta-catenin in normal anagen HFs and in 40 human pilomatrixomas. METHODS: In 11 of these tumours we also studied exon 3 beta-catenin gene mutations by polymerase chain reaction and direct sequencing. As these mutations have been related to a replication error (RER) phenotype in other tumour types, we explored whether or not this association also occurs in pilomatrixomas. RESULTS: beta-catenin was expressed in the cell membranes of the outer and inner root sheaths and in matrix cells located at the base and periphery of the HF bulb. However, central matrix cells that differentiate into cortical cells, cortical and cuticular cells expressed beta-catenin in the nucleus, suggesting a role in signal transduction. In addition, some fibroblasts of the dermal papilla also showed nuclear expression of beta-catenin. All 40 analysed pilomatrixomas showed intense nuclear and cytoplasmic beta-catenin expression in proliferating matrix (basaloid) cells. In areas of maturation, transitional cells mainly showed cytoplasmic and membranous expression of beta-catenin, while only a few cells retained nuclear expression. Shadow or ghost cells did not show beta-catenin expression. Three of 11 tumours (26%) had beta-catenin mutations. All three had the same heterozygote mis-sense mutation: a G to T change affecting the first nucleotide at codon 32 (D32Y). None of the 11 tumours studied had a positive RER phenotype. CONCLUSIONS: Present and previous studies suggest that the Wnt/beta-catenin/Tcf-Lef pathway is activated in normal matrix cells of the HF to induce differentiation to the hair shaft. Additionally, the beta-catenin mutation in matrix cells of the HF stabilizes beta-catenin protein, which translocates into the nucleus, where it activates of gene transcription together with lymphoid enhancer factor-1 producing pilomatrixoma. These mutations occur without an underlying defect in DNA mismatch repair.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Enfermedades del Cabello/metabolismo , Mutación Missense , Proteínas de Neoplasias/metabolismo , Pilomatrixoma/metabolismo , Neoplasias Cutáneas/metabolismo , Transactivadores , Adolescente , Adulto , Niño , Proteínas del Citoesqueleto/genética , Análisis Mutacional de ADN , Replicación del ADN , ADN de Neoplasias/genética , Femenino , Enfermedades del Cabello/genética , Folículo Piloso/metabolismo , Humanos , Masculino , Repeticiones de Microsatélite , Proteínas de Neoplasias/genética , Pilomatrixoma/genética , Reacción en Cadena de la Polimerasa , Neoplasias Cutáneas/genética , beta Catenina
17.
Mod Pathol ; 14(7): 650-4, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11454996

RESUMEN

We have immunohistochemically investigated P-cadherin (P-CD) expression in a series of 210 infiltrating ductal carcinomas (IDC) in an attempt to assess the biological and prognostic relevance of P-CD in patients harboring IDCs. Although only 74/210 (35%) of IDCs expressed P-CD in >5% of tumor cells (P-CD-positive carcinomas), categorical analyses revealed that P-CD-positive IDCs were larger (26 +/- 21 cm versus 22 +/- 11 cm, P =.0568), of higher histological grade (P =.0001), and had more lymph node metastases (P =.0327) than P-CD-negative breast carcinomas. In addition, P-CD-positive tumors were negative for estrogen (P =.0001) and progesterone receptors (P =.0001) and showed reduced E-cadherin expression (P =.0276) more frequently than P-CD-negative tumors. Univariate analysis carried out in 171 patients demonstrated that P-CD expression was also an indicator of poor prognosis (chi(2) = 8.292, P =.004), extent of lymph node metastasis (chi(2) = 20.854, P =.0000), histological grade (chi(2) = 12.908, P =.0016), and negative progesterone receptors (chi(2) = 4.116, P =.042). However, only histological grade and nodal metastases emerged as independent prognostic markers in the multivariate analysis. These results suggest that although P-CD expression may be involved in the progression of IDCs, its value as an independent prognostic factor remains to be established.


Asunto(s)
Neoplasias de la Mama/patología , Cadherinas/análisis , Carcinoma Ductal de Mama/patología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Estudios de Cohortes , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia
18.
Am J Clin Pathol ; 116(1): 143-8, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11447745

RESUMEN

We studied 48 human papillomavirus (HPV)-16-positive squamous intraepithelial lesions (SILs) from HIV-negative patients (16 low-grade SILs [LSILs]; 32 high-grade SILs [HSILs]) and 13 HPV-16-positive SILs from HIV-positive patients with AIDS (1 LSIL; 12 HSILs). After HPV typing, the entire HPV-16 E6 coding region was amplified and sequenced in all samples. We detected 12 HPV-16 E6 prototypes and 4 variants among the LSILs in HIV-negative patients, and 15 HPV-16 E6 prototypes and 17 HPV-16 variants in the HSIL group. The most prevalent variant of SIL types was European 350G, present in 3 and 13 cases, respectively. In 3 HSILs and no LSILs we found mixed infection by an HPV-16 E6 prototype and a variant. Two variants (1 each in LSIL and HSIL) were of non-European lineage. The only LSIL in HIV-positive patients had an HPV-16 E6 prototype; in the HSILs, we found 8 HPV-16 E6-prototypes, 4 with mixed infection with HPV-31 and 4 variants, all European 350G. The higher proportion of HPV-16 E6 variants in HSIL than in LSIL in HIV-negative patients suggests a greater risk of progression. However, further studies are needed.


Asunto(s)
Variación Genética , Seronegatividad para VIH , Seropositividad para VIH/virología , Papillomaviridae/clasificación , Papillomaviridae/genética , Enfermedades del Cuello del Útero/virología , Adolescente , Adulto , Secuencia de Bases/genética , Femenino , Humanos , Persona de Mediana Edad
19.
Virchows Arch ; 438(5): 464-9, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11407474

RESUMEN

The activation of the adenomatous polyposis coli (APC)/beta-catenin/T-cell factor (Tcf) pathway due to beta-catenin gene mutation has been recently implicated in the development of some endometrial carcinomas. beta- and gamma-catenin are structurally and functionally related molecules that participate in cell adhesion and signal transduction. Nuclear accumulation of beta- and gamma-catenin have been related to the activation of the APC/beta-catenin/Tcf pathway. In this study, we investigate the immunohistochemical expression pattern (nuclear vs membranous) of beta- and gamma-catenin in 40 endometrial carcinomas and their correlation with clinicopathological features and microsatellite instability (MI) status. MI was detected at three or more loci in 12 tumors: 11 were endometrioid and one was non-endometrioid. Nuclear catenin expression was found in 13 carcinomas: ten carcinomas had nuclear beta-catenin expression and three carcinomas had nuclear gamma-catenin expression. The nuclear catenin expression pattern significantly correlated with the histological type, International Federation of Gynecology and Obstetrics (FIGO) grade, and the presence of a second neoplasm. Nuclear catenin expression was always observed in low-grade endometrioid carcinomas; it was also more frequently associated with a second carcinoma. No correlation was observed between the catenin expression pattern and the level of myometrial infiltration, stage, associated endometrial hyperplasia, the existence of a source of estrogenic stimulation, and MI. However, four of 13 endometrioid carcinomas in this series had both catenin nuclear expression and MI. These data suggest that at least two different neoplastic pathways can lead to endometrial carcinomas with an endometrioid phenotype. In one, MI would be a key event, while in the other, the APC/beta-catenin/Tcf signaling pathways could be activated. Probably, in some cases, both pathways could simultaneously occur.


Asunto(s)
Carcinoma Endometrioide/genética , Proteínas del Citoesqueleto/genética , Neoplasias Endometriales/genética , Regulación Neoplásica de la Expresión Génica , Repeticiones de Microsatélite/genética , Transactivadores , Carcinoma Endometrioide/química , Carcinoma Endometrioide/patología , Núcleo Celular/química , Núcleo Celular/patología , Proteínas del Citoesqueleto/biosíntesis , ADN de Neoplasias/análisis , Desmoplaquinas , Neoplasias Endometriales/química , Femenino , Humanos , Técnicas para Inmunoenzimas , Membranas Intracelulares/química , Membranas Intracelulares/patología , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , beta Catenina , gamma Catenina
20.
Diagn Mol Pathol ; 10(2): 116-22, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11385321

RESUMEN

beta-Catenin gene mutations and microsatellite instability (MI) have been reported in endometrioid ovarian carcinomas. In colon but not endometrial cancer, beta-catenin gene mutations are associated with a replication error phenotype and MI. In this study the authors investigate whether beta-catenin mutations and MI are two independent oncogenic pathways in endometrioid ovarian carcinomas. They also evaluate the usefulness of these molecular markers in determining the primary origin of simultaneous tumors in the ovary and endometrium. This study was performed on 26 patients diagnosed with primary endometrioid ovarian carcinoma, five of whom also had pathologically diagnosed primary synchronous endometrioid endometrial carcinoma. Immunohistochemical and molecular analyses indicated that there were 25 primary ovarian tumors with four primary synchronous endometrial cancers and one ovarian metastasis of a primary endometrial carcinoma. All studies were performed on formalin-fixed, paraffin-embedded tissue samples. The beta-catenin expression pattern (nuclear vs. membranous) was analyzed immunohistochemically. Mutations in exon 3 of the beta-catenin gene were studied by polymerase chain reaction, single-strand conformational polymorphism, and direct sequencing. MI status was established by studying BAT-26 and BAT-25 mononucleotide repeats. In the group with 21 single ovarian tumors, 18 (85%) had beta-catenin nuclear expression, eight (38%) had beta-catenin gene mutations (always associated with beta-catenin nuclear expression), and four (19%) had MI. Only one case (5%) had both beta-catenin gene mutations and MI. The mutations affected one of the serine/threonine residues targeted for phosphorylation by glycogen synthase kinase-3beta or adjacent residues. At codon 32, a GAC-to-TAC (D32Y) change was found; at codon 33, two TCT-to-TGT (S33C) changes were found; at codon 37, three TCT-to-TTT (S37F) changes and one TCT-to-TGT (S37C) change were found; and, lastly, one ACC-to-GCC change at codon 41 (T41A) was detected. Four of the 25 endometrioid ovarian carcinomas (16%) had an associated synchronous endometrial carcinoma. There was a higher percentage of beta-catenin mutations (n = 3, 75%) in synchronous ovarian carcinomas than in single ones, although with a similar percentage of MI (n = 1, 25%). beta-catenin mutations were S37C in two cases and D32G in one. One of the four endometrial carcinomas showed an S33C beta-catenin mutation, and two carcinomas had MI. None of the four tumors had both beta-catenin gene mutation and MI. beta-catenin gene mutations were always associated with a nuclear beta-catenin expression pattern, whereas MI was associated with a membranous pattern. In one patient both the ovarian and the endometrial carcinomas had beta-catenin gene mutations, in another patient both tumors showed MI, whereas in the remaining two patients the ovarian carcinomas showed beta-catenin gene mutations and the endometrial carcinomas showed MI. To summarize, the results of this study suggest that beta-catenin mutations and MI could represent two independent pathways in endometrioid ovarian carcinomas because they occur simultaneously very infrequently (in 5% of these cases). beta-catenin mutations are always associated with a nuclear beta-catenin expression pattern, whereas cases with a replication error -plus phenotype showed no abnormal beta-catenin subcellular localization. The study of the beta-catenin expression pattern, beta-catenin mutations, and MI, together with conventional clinicopathologic findings, could aid in distinguishing between the metastatic or independent origin of simultaneous endometrioid ovarian and endometrial carcinomas. Tumors with identical immunohistochemical and molecular features should therefore be considered to have a common origin.


Asunto(s)
Carcinoma Endometrioide/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Neoplasias Endometriales/genética , Neoplasias Primarias Múltiples/genética , Neoplasias Ováricas/genética , Transactivadores , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Endometrioide/química , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Proteínas del Citoesqueleto/análisis , ADN de Neoplasias/análisis , Neoplasias Endometriales/química , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Humanos , Inmunohistoquímica , Repeticiones de Microsatélite , Persona de Mediana Edad , Mutación , Neoplasias Primarias Múltiples/química , Neoplasias Primarias Múltiples/metabolismo , Neoplasias Primarias Múltiples/patología , Neoplasias Ováricas/química , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...