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.
PLoS One ; 12(2): e0171265, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28212443

RESUMEN

Pathogenic gene fusions have been identified in several histologic types of salivary gland neoplasia, but not previously in acinic cell carcinoma (AcCC). To discover novel gene fusions, we performed whole-transcriptome sequencing surveys of three AcCC archival cases. In one specimen we identified a novel HTN3-MSANTD3 gene fusion, and in another a novel PRB3-ZNF217 gene fusion. The structure of both fusions was consistent with the promoter of the 5' partner (HTN3 or PRB3), both highly expressed salivary gland genes, driving overexpression of full-length MSANTD3 or ZNF217. By fluorescence in situ hybridization of an expanded AcCC case series, we observed MSANTD3 rearrangements altogether in 3 of 20 evaluable cases (15%), but found no additional ZNF217 rearrangements. MSANTD3 encodes a previously uncharacterized Myb/SANT domain-containing protein. Immunohistochemical staining demonstrated diffuse nuclear MSANTD3 expression in 8 of 27 AcCC cases (30%), including the three cases with MSANTD3 rearrangement. MSANTD3 displayed heterogeneous expression in normal salivary ductal epithelium, as well as among other histologic types of salivary gland cancer though without evidence of translocation. In a broader survey, MSANTD3 showed variable expression across a wide range of normal and neoplastic human tissue specimens. In preliminary functional studies, engineered MSANTD3 overexpression in rodent salivary gland epithelial cells did not enhance cell proliferation, but led to significant upregulation of gene sets involved in protein synthesis. Our findings newly identify MSANTD3 rearrangement as a recurrent event in salivary gland AcCC, providing new insight into disease pathogenesis, and identifying a putative novel human oncogene.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma de Células Acinares/genética , Reordenamiento Génico , Neoplasias de las Glándulas Salivales/genética , Adulto , Animales , Carcinoma de Células Acinares/patología , Línea Celular Tumoral , Secuencia Conservada , Perfilación de la Expresión Génica , Fusión Génica , Humanos , Ratas , Neoplasias de las Glándulas Salivales/patología , Regulación hacia Arriba
2.
PLoS One ; 10(11): e0142061, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26556242

RESUMEN

NKX2-1, encoding a homeobox transcription factor, is amplified in approximately 15% of non-small cell lung cancers (NSCLC), where it is thought to drive cancer cell proliferation and survival. However, its mechanism of action remains largely unknown. To identify relevant downstream transcriptional targets, here we carried out a combined NKX2-1 transcriptome (NKX2-1 knockdown followed by RNAseq) and cistrome (NKX2-1 binding sites by ChIPseq) analysis in four NKX2-1-amplified human NSCLC cell lines. While NKX2-1 regulated genes differed among the four cell lines assayed, cell proliferation emerged as a common theme. Moreover, in 3 of the 4 cell lines, epidermal growth factor receptor (EGFR) was among the top NKX2-1 upregulated targets, which we confirmed at the protein level by western blot. Interestingly, EGFR knockdown led to upregulation of NKX2-1, suggesting a negative feedback loop. Consistent with this finding, combined knockdown of NKX2-1 and EGFR in NCI-H1819 lung cancer cells reduced cell proliferation (as well as MAP-kinase and PI3-kinase signaling) more than knockdown of either alone. Likewise, NKX2-1 knockdown enhanced the growth-inhibitory effect of the EGFR-inhibitor erlotinib. Taken together, our findings implicate EGFR as a downstream effector of NKX2-1 in NKX2-1 amplified NSCLC, with possible clinical implications, and provide a rich dataset for investigating additional mediators of NKX2-1 driven oncogenesis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Clorhidrato de Erlotinib/farmacología , Humanos , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor Nuclear Tiroideo 1 , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
3.
Nat Genet ; 46(7): 722-5, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24859340

RESUMEN

Here we report the discovery of oncogenic mutations in the Hedgehog and mitogen-activated protein kinase (MAPK) pathways in over 80% of ameloblastomas, locally destructive odontogenic tumors of the jaw, by genomic analysis of archival material. Mutations in SMO (encoding Smoothened, SMO) are common in ameloblastomas of the maxilla, whereas BRAF mutations are predominant in tumors of the mandible. We show that a frequently occurring SMO alteration encoding p.Leu412Phe is an activating mutation and that its effect on Hedgehog-pathway activity can be inhibited by arsenic trioxide (ATO), an anti-leukemia drug approved by the US Food and Drug Administration (FDA) that is currently in clinical trials for its Hedgehog-inhibitory activity. In a similar manner, ameloblastoma cells harboring an activating BRAF mutation encoding p.Val600Glu are sensitive to the BRAF inhibitor vemurafenib. Our findings establish a new paradigm for the diagnostic classification and treatment of ameloblastomas.


Asunto(s)
Ameloblastoma/genética , Neoplasias Maxilomandibulares/genética , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Receptores Acoplados a Proteínas G/genética , Ameloblastoma/tratamiento farmacológico , Ameloblastoma/patología , Antineoplásicos/farmacología , Trióxido de Arsénico , Arsenicales/farmacología , Proliferación Celular/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Indoles/farmacología , Neoplasias Maxilomandibulares/tratamiento farmacológico , Neoplasias Maxilomandibulares/patología , Óxidos/farmacología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Receptor Smoothened , Sulfonamidas/farmacología , Células Tumorales Cultivadas , Vemurafenib
4.
PLoS One ; 7(9): e46518, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029544

RESUMEN

The phosphoinositide 3-kinase (PI3K) signaling pathway is significantly altered in a wide variety of human cancers, driving cancer cell growth and survival. Consequently, a large number of PI3K inhibitors are now in clinical development. To begin to improve the selection of patients for treatment with PI3K inhibitors and to identify de novo determinants of patient response, we sought to identify and characterize candidate genomic and phosphoproteomic biomarkers predictive of response to the selective PI3K inhibitor, GDC-0941, using the NCI-60 human tumor cell line collection. In this study, sixty diverse tumor cell lines were exposed to GDC-0941 and classified by GI(50) value as sensitive or resistant. The most sensitive and resistant cell lines were analyzed for their baseline levels of gene expression and phosphorylation of key signaling nodes. Phosphorylation or activation status of both the PI3K-Akt signaling axis and PARP were correlated with in vitro response to GDC-0941. A gene expression signature associated with in vitro sensitivity to GDC-0941 was also identified. Furthermore, in vitro siRNA-mediated silencing of two genes in this signature, OGT and DDN, validated their role in modulating sensitivity to GDC-0941 in numerous cell lines and begins to provide biological insights into their role as chemosensitizers. These candidate biomarkers will offer useful tools to begin a more thorough understanding of determinants of patient response to PI3K inhibitors and merit exploration in human cancer patients treated with PI3K inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Indazoles/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Sulfonamidas/farmacología , Transcriptoma/efectos de los fármacos , Acetilglucosamina/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa de Punto de Control 2 , Análisis por Conglomerados , Técnicas de Silenciamiento del Gen , Humanos , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor ErbB-3/metabolismo , Transducción de Señal , Estadísticas no Paramétricas
5.
PLoS One ; 6(8): e23924, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21887346

RESUMEN

Pancreatic cancer is a deadly disease, and new therapeutic targets are urgently needed. We previously identified DNA amplification at 7q21-q22 in pancreatic cancer cell lines. Now, by high-resolution genomic profiling of human pancreatic cancer cell lines and human tumors (engrafted in immunodeficient mice to enrich the cancer epithelial fraction), we define a 325 Kb minimal amplicon spanning SMURF1, an E3 ubiquitin ligase and known negative regulator of transforming growth factor ß (TGFß) growth inhibitory signaling. SMURF1 amplification was confirmed in primary human pancreatic cancers by fluorescence in situ hybridization (FISH), where 4 of 95 cases (4.2%) exhibited amplification. By RNA interference (RNAi), knockdown of SMURF1 in a human pancreatic cancer line with focal amplification (AsPC-1) did not alter cell growth, but led to reduced cell invasion and anchorage-independent growth. Interestingly, this effect was not mediated through altered TGFß signaling, assayed by transcriptional reporter. Finally, overexpression of SMURF1 (but not a catalytic mutant) led to loss of contact inhibition in NIH-3T3 mouse embryo fibroblast cells. Together, these findings identify SMURF1 as an amplified oncogene driving multiple tumorigenic phenotypes in pancreatic cancer, and provide a new druggable target for molecularly directed therapy.


Asunto(s)
Amplificación de Genes/fisiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Ubiquitina-Proteína Ligasas/genética , Animales , Comunicación Celular , Línea Celular , Línea Celular Tumoral , Humanos , Hibridación Fluorescente in Situ , Ratones , Invasividad Neoplásica , Oncogenes
6.
Mol Oncol ; 4(3): 255-66, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20434415

RESUMEN

Breast cancer is a heterogeneous disease, appreciable by molecular markers, gene-expression profiles, and most recently, patterns of genomic alteration. In particular, genomic profiling has revealed three distinct patterns of DNA copy-number alteration: a "simple" type with few gains or losses of whole chromosome arms, an "amplifier" type with focal high-level DNA amplifications, and a "complex" type marked by numerous low-amplitude changes and copy-number transitions. The three patterns are associated with distinct gene-expression subtypes, and preferentially target different loci in the genome (implicating distinct cancer genes). Moreover, the different patterns of alteration imply distinct underlying mechanisms of genomic instability. The amplifier pattern may arise from transient telomere dysfunction, although new data suggest ongoing "amplifier" instability. The complex pattern shows similarity to breast cancers with germline BRCA1 mutation, which also exhibit "basal-like" expression profiles and complex-pattern genomes, implicating a possible defect in BRCA1-associated repair of DNA double-strand breaks. As such, targeting presumptive DNA repair defects represents a promising area of clinical investigation. Future studies should clarify the pathogenesis of breast cancers with amplifier and complex-pattern genomes, and will likely identify new therapeutic opportunities.


Asunto(s)
Neoplasias de la Mama/genética , Inestabilidad Genómica , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Aberraciones Cromosómicas , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo
7.
PLoS One ; 4(7): e6146, 2009 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-19582160

RESUMEN

BACKGROUND: Breast cancer cell lines have been used widely to investigate breast cancer pathobiology and new therapies. Breast cancer is a molecularly heterogeneous disease, and it is important to understand how well and which cell lines best model that diversity. In particular, microarray studies have identified molecular subtypes-luminal A, luminal B, ERBB2-associated, basal-like and normal-like-with characteristic gene-expression patterns and underlying DNA copy number alterations (CNAs). Here, we studied a collection of breast cancer cell lines to catalog molecular profiles and to assess their relation to breast cancer subtypes. METHODS: Whole-genome DNA microarrays were used to profile gene expression and CNAs in a collection of 52 widely-used breast cancer cell lines, and comparisons were made to existing profiles of primary breast tumors. Hierarchical clustering was used to identify gene-expression subtypes, and Gene Set Enrichment Analysis (GSEA) to discover biological features of those subtypes. Genomic and transcriptional profiles were integrated to discover within high-amplitude CNAs candidate cancer genes with coordinately altered gene copy number and expression. FINDINGS: Transcriptional profiling of breast cancer cell lines identified one luminal and two basal-like (A and B) subtypes. Luminal lines displayed an estrogen receptor (ER) signature and resembled luminal-A/B tumors, basal-A lines were associated with ETS-pathway and BRCA1 signatures and resembled basal-like tumors, and basal-B lines displayed mesenchymal and stem/progenitor-cell characteristics. Compared to tumors, cell lines exhibited similar patterns of CNA, but an overall higher complexity of CNA (genetically simple luminal-A tumors were not represented), and only partial conservation of subtype-specific CNAs. We identified 80 high-level DNA amplifications and 13 multi-copy deletions, and the resident genes with concomitantly altered gene-expression, highlighting known and novel candidate breast cancer genes. CONCLUSIONS: Overall, breast cancer cell lines were genetically more complex than tumors, but retained expression patterns with relevance to the luminal-basal subtype distinction. The compendium of molecular profiles defines cell lines suitable for investigations of subtype-specific pathobiology, cancer stem cell biology, biomarkers and therapies, and provides a resource for discovery of new breast cancer genes.


Asunto(s)
Neoplasias de la Mama/genética , Perfilación de la Expresión Génica , Secuencia de Bases , Western Blotting , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cartilla de ADN , Femenino , Genes BRCA1 , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Transcripción Genética
8.
PLoS Genet ; 4(5): e1000081, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18535672

RESUMEN

Pancreatobiliary cancers have among the highest mortality rates of any cancer type. Discovering the full spectrum of molecular genetic alterations may suggest new avenues for therapy. To catalogue genomic alterations, we carried out array-based genomic profiling of 31 exocrine pancreatic cancers and 6 distal bile duct cancers, expanded as xenografts to enrich the tumor cell fraction. We identified numerous focal DNA amplifications and deletions, including in 19% of pancreatobiliary cases gain at cytoband 18q11.2, a locus uncommonly amplified in other tumor types. The smallest shared amplification at 18q11.2 included GATA6, a transcriptional regulator previously linked to normal pancreas development. When amplified, GATA6 was overexpressed at both the mRNA and protein levels, and strong immunostaining was observed in 25 of 54 (46%) primary pancreatic cancers compared to 0 of 33 normal pancreas specimens surveyed. GATA6 expression in xenografts was associated with specific microarray gene-expression patterns, enriched for GATA binding sites and mitochondrial oxidative phosphorylation activity. siRNA mediated knockdown of GATA6 in pancreatic cancer cell lines with amplification led to reduced cell proliferation, cell cycle progression, and colony formation. Our findings indicate that GATA6 amplification and overexpression contribute to the oncogenic phenotypes of pancreatic cancer cells, and identify GATA6 as a candidate lineage-specific oncogene in pancreatobiliary cancer, with implications for novel treatment strategies.


Asunto(s)
Neoplasias del Sistema Biliar/genética , Factor de Transcripción GATA6/genética , Amplificación de Genes , Neoplasias Pancreáticas/genética , Animales , Neoplasias del Sistema Biliar/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Cromosomas Humanos Par 18/genética , Factor de Transcripción GATA6/metabolismo , Perfilación de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Ratones , Neoplasias Pancreáticas/metabolismo , Trasplante Heterólogo/patología , Trasplante Heterólogo/veterinaria
9.
Mol Oncol ; 2(4): 327-39, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19383354

RESUMEN

Breast cancer exhibits clinical and molecular heterogeneity, where expression profiling studies have identified five major molecular subtypes. The basal-like subtype, expressing basal epithelial markers and negative for estrogen receptor (ER), progesterone receptor (PR) and HER2, is associated with higher overall levels of DNA copy number alteration (CNA), specific CNAs (like gain on chromosome 10p), and poor prognosis. Discovering the molecular genetic basis of tumor subtypes may provide new opportunities for therapy. To identify the driver oncogene on 10p associated with basal-like tumors, we analyzed genomic profiles of 172 breast carcinomas. The smallest shared region of gain spanned just seven genes at 10p13, including calcium/calmodulin-dependent protein kinase ID (CAMK1D), functioning in intracellular signaling but not previously linked to cancer. By microarray, CAMK1D was overexpressed when amplified, and by immunohistochemistry exhibited elevated expression in invasive carcinomas compared to carcinoma in situ. Engineered overexpression of CAMK1D in non-tumorigenic breast epithelial cells led to increased cell proliferation, and molecular and phenotypic alterations indicative of epithelial-mesenchymal transition (EMT), including loss of cell-cell adhesions and increased cell migration and invasion. Our findings identify CAMK1D as a novel amplified oncogene linked to EMT in breast cancer, and as a potential therapeutic target with particular relevance to clinically unfavorable basal-like tumors.


Asunto(s)
Neoplasias de la Mama/patología , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Células Epiteliales/patología , Amplificación de Genes , Células Madre Mesenquimatosas/patología , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/análisis , Adhesión Celular/genética , Proliferación Celular , Cromosomas Humanos Par 10/genética , Femenino , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/clasificación , Invasividad Neoplásica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos
10.
Open Cancer J ; 2: 1-4, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20664806

RESUMEN

We have previously identified a structural variant of the α6 integrin (Laminin receptor) called α6p. The α6p variant is a 70 kDa form of the full-length α6 integrin (140 kDa) that remains paired with either the ß1 or ß4 subunit on the cell surface. α6p is produced by urokinase-type plasminogen activator (uPA), which removes the extracellular ß-barrel domain while the receptor is on the cell surface. The α6p integrin was present in human prostate cancer tissue but not in normal tissue and the cleavage of the α6 integrin extracellular domain promotes tumor cell invasion and migration on laminin. The objective of the present study was to determine whether the α6p integrin is observed in other models of carcinogenesis. Our results indicate detectable low levels of α6p in normal mouse skin, and comparatively elevated levels in mouse papillomas and squamous cell carcinomas induced by DMBA, TPA and MNNG treatments. Furthermore, we have found that α6p was present at high levels in skin melanomas of transgenic mice that over express activated Ha-ras under the control of the tyrosinase promoter. Finally, subcutaneous injection into athymic nude mice of a malignant mouse keratinocyte derived cell line (6M90) that is α6p negative, results in the development of tumors that contain α6p integrin. The latter results indicate that α6p is induced in vivo suggesting that the tumor microenvironment plays a major role in the production of α6p. Taken together, these data suggest that the cell surface cleavage of the α6 integrin may be a novel mechanism of integrin regulation and might be an important step during skin tissue remodeling and during carcinogenesis.

11.
Free Radic Biol Med ; 40(5): 863-75, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16520238

RESUMEN

We have used a keratinocyte in vivo/in vitro cell model to test the hypothesis that hydrogen peroxide acts as a signaling molecule, contributing to proliferation and tumorigenesis. A cell line, 6M90, that produces squamous cell carcinoma (SCC), has high levels of ROS and low levels of catalase. A new cell line, MTOC2, generated from parental 6M90 cells by introduction of a Tet-responsive catalase transgene, effectively expressed higher peroxisomal catalase. Increased catalase expression diminished constitutive ROS and enhanced viability after treatment with hydrogen peroxide. Protein tyrosine phosphatase activity was higher in the MTOC2 cells with high catalase, consistent with detection of a lower level of phosphorylation at tyrosine 1068 of the epidermal growth factor receptor (EGF-R). Transcription of downstream c-fos, AP-1 transactivation and cell proliferation were higher in the low catalase cells. An EGF-R inhibitor, AG1478, blocks the higher AP-1 transactivation and cell proliferation of the low catalase 6M90 cells. Tumorigenesis in SCID mice was greatly diminished in the high catalase cells. Our data suggest that hydrogen peroxide functions as a signaling molecule that can modulate activity of a protein tyrosine phosphatase/(s) resulting in phosphorylation of tryrosine/(s) on the EGF-R. Therefore, catalase acts as a tumor-suppressor gene in part by decreasing EGF-R signaling.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Catalasa/genética , Receptores ErbB/metabolismo , Genes Supresores de Tumor , Especies Reactivas de Oxígeno/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Catalasa/análisis , Catalasa/metabolismo , Línea Celular Tumoral , Proliferación Celular , Genes fos/genética , Humanos , Peróxido de Hidrógeno/toxicidad , Queratinocitos/efectos de los fármacos , Queratinocitos/enzimología , Queratinocitos/patología , Ratones , Ratones SCID , Fosforilación , Proteínas Tirosina Fosfatasas/metabolismo , Ratas , Factor de Transcripción AP-1/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Lett ; 231(2): 326-38, 2006 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-15893875

RESUMEN

We have previously developed an in vitro tumor progression model with mouse skin keratinocytes to study the molecular targets that mediate the tumor cell's progression from a benign to a malignant phenotype. The malignantly transformed cells were found to have elevated MAP kinase signaling and increases in AP-1, NFkappaB and cAMP response element (CRE) transcription factors activities compared to their benign counter-part. In this study, we showed that Rac1, a member of the Rho superfamily of small GTPases, functions as a key signaling molecule that mediates these malignant phenotypes. We used a doxycycline inducible system to express dominant negative Rac1 (N17 Rac1) in the squamous cell carcinomas producing 6M90 cell line. Conditional expression of the N17 Rac1 was able to decrease multiple markers of malignancy including: growth rate, colony formation, migration, invasion and most importantly, in vivo tumor growth. In addition, these phenotypic changes were accompanied by decreases in mitogenic signals, which include ERK1/2, JNK, and PI-3 kinase/Akt activation. Transactivation mediated by AP-1, NFkappaB, and CRE were also attenuated by expression of dominant negative Rac1. These observations led us to conclude that Rac1 signaling is required for the malignant phenotypes of the squamous cell carcinoma cells.


Asunto(s)
Transformación Celular Neoplásica , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Proteína de Unión al GTP rac1/metabolismo , Animales , Antibacterianos/farmacología , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/prevención & control , Movimiento Celular , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Doxiciclina/farmacología , Genes Dominantes , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/prevención & control , Tasa de Supervivencia , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
13.
Neoplasia ; 6(5): 440-8, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15548352

RESUMEN

Previous studies in our laboratory have shown that the elevation of reactive oxygen species levels and the repression of the antioxidant enzyme, catalase, played a critical role in the in vitro progression of benign papilloma cells to malignant carcinoma cells. Catalase message, protein levels, and activity levels were found to be downregulated in the malignantly progressed cells. The goal of this study is to further characterize the repression of catalase in malignant progression of mouse skin tumors. To validate the in vitro observations, we examined catalase expression in tumor samples generated by the multistep chemical carcinogenesis protocol. Higher levels of catalase mRNA and protein were observed in benign papillomas versus malignant carcinomas. Nuclear run-on analysis showed that catalase repression in the cultured malignant cells was transcription-dependent. Results from luciferase reporter assays indicated that malignant cells have lower catalase promoter activities than benign papilloma cells, in part through the Wilm's tumor suppressor 1 (WT1) binding site within the proximal promoter region. The WT1 protein levels were found to be inversely correlated with the observed catalase promoter activities, with higher levels observed in the malignant cells versus the benign cells. These results led us to conclude that WT1 is acting as a transcription repressor in catalase gene regulation during tumor progression.


Asunto(s)
Carcinoma de Células Escamosas/genética , Catalasa/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Papiloma/genética , Neoplasias Cutáneas/genética , Animales , Carcinoma de Células Escamosas/metabolismo , Catalasa/metabolismo , Línea Celular Tumoral , Dactinomicina/farmacología , Genes Reporteros/genética , Queratinas/análisis , Luciferasas/análisis , Luciferasas/genética , Ratones , Mutación/genética , Papiloma/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional , Proteínas Supresoras de Tumor/metabolismo , Proteínas WT1/metabolismo
14.
Mol Carcinog ; 37(3): 149-57, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12884366

RESUMEN

Resistance to apoptosis may be a critical phenotype that cells must acquire during skin carcinogenesis. The Akt kinase is a known upstream regulator of apoptosis in many cell types and has been shown to be activated by increased reactive oxygen species (ROS). We have previously demonstrated that two malignant variants (6M90 and 6R90) of the mouse keratinocyte 308 cell line have elevated ROS because of loss of catalase activity, and that this elevated ROS confers a growth advantage. We report here that in addition to a growth advantage, chronically increased ROS in the variants resulted in an increase in resistance to ultraviolet (UV) B-induced apoptosis. This resistance was due to basal increases of Akt phosphorylation in the malignant variants compared to the 308 cells. Modulation of ROS in 6M90 and 6R90 cells by catalase overexpression or antioxidant treatment resulted in decreased levels of Akt phosphorylation and subsequent loss of resistance to UVB-induced apoptosis. Conversely, treatment of 308 cells with hydrogen peroxide caused increases in Akt phosphorylation and increased apoptosis resistance. These results indicate that the chronically elevated ROS often observed in tumors may contribute to a malignant phenotype by keeping Akt in a phosphorylated state, resulting in increased apoptosis resistance.


Asunto(s)
Apoptosis/efectos de la radiación , Queratinocitos/efectos de la radiación , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Tolerancia a Radiación , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/farmacología , Catalasa/metabolismo , Células Cultivadas/efectos de los fármacos , Células Cultivadas/patología , Células Cultivadas/efectos de la radiación , Peróxido de Hidrógeno/farmacología , Queratinocitos/patología , Ratones , Oxidantes/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...