Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Biochim Biophys Acta ; 1859(8): 1034-42, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27321990

RESUMEN

Sequence-specific degradation of homologous mRNA is the main mechanism by which short-interfering RNAs (siRNAs) suppress gene expression. Generally, it is assumed that the mRNA fragments resulting from Ago2 cleavage are rapidly degraded, thus making the transcript translation-incompetent. However, the molecular mechanisms involved in the post-cleavage mRNA decay are not completely understood and the fate of cleavage intermediates has been poorly studied. Using specific siRNAs and short-hairpin RNAs (shRNAs) we show that the 5' and 3' mRNA cleavage fragments of human papilloma virus type 16 (HPV-16) E6/7 mRNA, over-expressed in cervical malignancies, are unevenly degraded. Intriguingly, the 5' mRNA fragment was more abundant and displayed a greater stability than the corresponding 3' mRNA fragment in RNAi-treated cells. Further analysis revealed that the 5' mRNA fragment was polysome-associated, indicating its active translation, and this was further confirmed by using tagged E7 protein to show that C-terminally truncated proteins were produced in treated cells. Overall, our findings provide new insight into the degradation of siRNA-targeted transcripts and show that RNAi can alter protein expression in cells as a result of preferential stabilization and translation of the 5' cleavage fragment. These results challenge the current model of siRNA-mediated RNAi and provide a significant step forward towards understanding non-canonical pathways of siRNA gene silencing.


Asunto(s)
Células Epiteliales/metabolismo , Silenciador del Gen , Proteínas E7 de Papillomavirus/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Animales , Línea Celular Tumoral , Cuello del Útero/metabolismo , Cuello del Útero/patología , Cuello del Útero/virología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Células HeLa , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Humanos , Ratones , Proteínas E7 de Papillomavirus/genética , Polirribosomas/genética , Polirribosomas/metabolismo , Estabilidad del ARN , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
2.
Invest New Drugs ; 28(5): 575-86, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19636513

RESUMEN

We previously identified the induction of senescence in melanoma cell lines sensitive to diterpene esters, indicating a therapeutic potential. Here we compared the cytostatic effects of two diterpene esters: the prototypic PKC-activating drug TPA (12-O-tetradecanoylphorbol-13-acetate), and the novel compound PEP008 (20-O-acetyl-ingenol-3-angelate) in cell lines derived from melanoma, breast cancer and colon cancer. The diterpene esters induced permanent growth arrest with characteristics of senescence in a subset of cell lines in all three solid tumor models at 100-1000 ng/ml. Use of the PKC inhibitor bisindolylmaleimide-l demonstrated that activation of PKC was required for growth arrest. Full genome expression profiling identified pivotal genes involved in DNA synthesis and cell cycle control down-regulated by treatment in all three sensitive tumor models. At the protein level, prolonged down-regulation of E2F-1 and proliferating cell nuclear antigen (PCNA), sustained expression of p21(WAF1/CIP1) and dephosphorylation of retinoblastoma (Rb) occurred in the sensitive cells. Additionally, the type II tumor suppressor HRASLS3, which has a role in mitogen-activated protein kinase (MAPK) pathway suppression, was constitutively elevated in cell lines resistant to the senescence effects compared to their sensitive counterparts. Together, these results demonstrate that both TPA and the novel PKC-activating drug PEP008 induce growth arrest with characteristics of senescence in solid tumor cell lines derived from a variety of tissue types, and by a similar mechanism. PKC-activating diterpene esters may therefore have therapeutic potential in a subset of breast cancer, colon cancer and melanoma tumors.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Diterpenos/farmacología , Activadores de Enzimas/farmacología , Ésteres/farmacología , Neoplasias/enzimología , Neoplasias/patología , Proteína Quinasa C/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/metabolismo , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Fosfolipasas A2 Calcio-Independiente , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , beta-Galactosidasa/metabolismo
3.
Mol Oncol ; 14(1): 22-41, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31733171

RESUMEN

Ultraviolet radiation-induced DNA mutations are a primary environmental driver of melanoma. The reason for this very high level of unrepaired DNA lesions leading to these mutations is still poorly understood. The primary DNA repair mechanism for UV-induced lesions, that is, the nucleotide excision repair pathway, appears intact in most melanomas. We have previously reported a postreplication repair mechanism that is commonly defective in melanoma cell lines. Here we have used a genome-wide approach to identify the components of this postreplication repair mechanism. We have used differential transcript polysome loading to identify transcripts that are associated with UV response, and then functionally assessed these to identify novel components of this repair and cell cycle checkpoint network. We have identified multiple interaction nodes, including global genomic nucleotide excision repair and homologous recombination repair, and previously unexpected MASTL pathway, as components of the response. Finally, we have used bioinformatics to assess the contribution of dysregulated expression of these pathways to the UV signature mutation load of a large melanoma cohort. We show that dysregulation of the pathway, especially the DNA damage repair components, are significant contributors to UV mutation load, and that dysregulation of the MASTL pathway appears to be a significant contributor to high UV signature mutation load.


Asunto(s)
Reparación del ADN/efectos de la radiación , Replicación del ADN/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Melanoma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Polirribosomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Línea Celular Tumoral , Replicación del ADN/efectos de la radiación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Estudio de Asociación del Genoma Completo , Humanos , Melanoma/genética , Melanoma/patología , Proteínas Asociadas a Microtúbulos/genética , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Polirribosomas/genética , Polirribosomas/efectos de la radiación , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño , RNA-Seq , Reparación del ADN por Recombinación , Rayos Ultravioleta , Regulación hacia Arriba
4.
Respir Res ; 10: 81, 2009 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-19723343

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a major public health problem. The aim of this study was to identify genes involved in emphysema severity in COPD patients.Gene expression profiling was performed on total RNA extracted from non-tumor lung tissue from 30 smokers with emphysema. Class comparison analysis based on gas transfer measurement was performed to identify differentially expressed genes. Genes were then selected for technical validation by quantitative reverse transcriptase-PCR (qRT-PCR) if also represented on microarray platforms used in previously published emphysema studies. Genes technically validated advanced to tests of biological replication by qRT-PCR using an independent test set of 62 lung samples.Class comparison identified 98 differentially expressed genes (p < 0.01). Fifty-one of those genes had been previously evaluated in differentiation between normal and severe emphysema lung. qRT-PCR confirmed the direction of change in expression in 29 of the 51 genes and 11 of those validated, remaining significant at p < 0.05. Biological replication in an independent cohort confirmed the altered expression of eight genes, with seven genes differentially expressed by greater than 1.3 fold, identifying these as candidate determinants of emphysema severity.Gene expression profiling of lung from emphysema patients identified seven candidate genes associated with emphysema severity including COL6A3, SERPINF1, ZNHIT6, NEDD4, CDKN2A, NRN1 and GSTM3.


Asunto(s)
Perfilación de la Expresión Génica , Pulmón/metabolismo , Proteínas/análisis , Enfisema Pulmonar/diagnóstico , Enfisema Pulmonar/metabolismo , Índice de Severidad de la Enfermedad , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad
5.
Mol Cancer ; 7: 75, 2008 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-18831746

RESUMEN

BACKGROUND: Barrett's esophagus (BE) is the metaplastic replacement of squamous with columnar epithelium in the esophagus, as a result of reflux. It is the major risk factor for the development of esophageal adenocarcinoma (EAC). Methylation of CpG dinucleotides of normally unmethylated genes is associated with silencing of their expression, and is common in EAC. This study was designed to determine at what stage, in the progression from BE to EAC, methylation of key genes occurs. RESULTS: We examined nine genes (APC, CDKN2A, ID4, MGMT, RBP1, RUNX3, SFRP1, TIMP3, and TMEFF2), frequently methylated in multiple cancer types, in a panel of squamous (19 biopsies from patients without BE or EAC, 16 from patients with BE, 21 from patients with EAC), BE (40 metaplastic, seven high grade dysplastic) and 37 EAC tissues. The methylation frequency, the percentage of samples that had any extent of methylation, for each of the nine genes in the EAC (95%, 59%, 76%, 57%, 70%, 73%, 95%, 74% and 83% respectively) was significantly higher than in any of the squamous groups. The methylation frequency for each of the nine genes in the metaplastic BE (95%, 28%, 78%, 48%, 58%, 48%, 93%, 88% and 75% respectively) was significantly higher than in the squamous samples except for CDKN2A and RBP1. The methylation frequency did not differ between BE and EAC samples, except for CDKN2A and RUNX3 which were significantly higher in EAC. The methylation extent was an estimate of both the number of methylated alleles and the density of methylation on these alleles. This was significantly greater in EAC than in metaplastic BE for all genes except APC, MGMT and TIMP3. There was no significant difference in methylation extent for any gene between high grade dysplastic BE and EAC. CONCLUSION: We found significant methylation in metaplastic BE, which for seven of the nine genes studied did not differ in frequency from that found in EAC. This is also the first report of gene silencing by methylation of ID4 in BE or EAC. This study suggests that metaplastic BE is a highly abnormal tissue, more similar to cancer tissue than to normal epithelium.


Asunto(s)
Adenocarcinoma/genética , Esófago de Barrett/genética , Metilación de ADN , Neoplasias Esofágicas/genética , Adenocarcinoma/patología , Esófago de Barrett/patología , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Perfilación de la Expresión Génica , Humanos
6.
Int J Cancer ; 123(1): 227-31, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18386818

RESUMEN

We previously showed that mice carrying an activated Cdk4 mutation together with melanocyte-specific mutant Hras (Cdk4(R24C/R24C)/TPras) develop melanoma spontaneously, but penetrance is increased and age of onset reduced after neonatal ultraviolet radiation (UVR) exposure. UVR-treated mice were more likely to develop multiple primary lesions, and these melanomas more often expressed Trp53, and less often expressed c-Myc, than melanomas from nonirradiated mice (Hacker et al., Cancer Res 2006;66:2946-52). These data suggest differences in mechanisms of tumorigenesis between melanomas developing spontaneously, or as a result of UVR exposure. To further delineate these differences, we compared global gene expression between spontaneous and UVR-induced melanomas from these mice using microarrays. We found 264 genes differentially expressed between these groups (ANOVA, p < 0.05). Selected candidate genes were validated using qRT-PCR, which confirmed upregulation of Gpr155 and Bmp7, and downregulation of Plagl1, Akap12 and Il18 in UVR-induced mouse melanomas. In humans, epidemiological studies suggest that there may be 2 predominant pathways to melanoma development. One characterized by chronic UVR exposure and which leads mainly to melanomas on sun-exposed sites; the other associated with low UVR exposure and leading predominantly to melanomas on less-exposed body sites. We found by immunohistochemical analysis that, comparing a series of human melanomas from the head (a chronically sun-exposed site; N = 82) with a set from the trunk (an intermittently exposed site; N = 65), the prevalence of IL-18 expression was significantly lower in melanomas on the head (16%) than on truncal melanomas (34%, p = 0.011). We conclude that loss of IL-18 is a marker of UVR-induced melanoma, both in animal models and humans.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Interleucina-18/metabolismo , Melanoma/metabolismo , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Melanoma/etiología , Melanoma/genética , Ratones , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-kit/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Clin Cancer Res ; 13(10): 2946-54, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17504995

RESUMEN

PURPOSE: Improving outcomes for early-stage lung cancer is a major research focus at present because a significant proportion of stage I patients develop recurrent disease within 5 years of curative-intent lung resection. Within tumor stage groups, conventional prognostic indicators currently fail to predict relapse accurately. EXPERIMENTAL DESIGN: To identify a gene signature predictive of recurrence in primary lung adenocarcinoma, we analyzed gene expression profiles in a training set of 48 node-negative tumors (stage I-II), comparing tumors from cases who remained disease-free for a minimum of 36 months with those from cases whose disease recurred within 18 months of complete resection. RESULTS: Cox proportional hazards modeling with leave-one-out cross-validation identified a 54-gene signature capable of predicting risk of recurrence in two independent validation cohorts of 55 adenocarcinomas [log-rank P=0.039; hazard ratio (HR), 2.2; 95% confidence interval (95% CI), 1.1-4.7] and 40 adenocarcinomas (log-rank P=0.044; HR, 3.3; 95% CI, 1.4-7.9). Kaplan-Meier log-rank analysis found that predicted poor-outcome groups had significantly shorter survival, and furthermore, the signature predicted outcome independently of conventional indicators of tumor stage and node stage. In a subset of earliest stage adenocarcinomas, generally expected to have good outcome, the signature predicted samples with significantly poorer survival. CONCLUSIONS: We describe a 54-gene signature that predicts the risk of recurrent disease independently of tumor stage and which therefore has potential to refine clinical prognosis for patients undergoing resection for primary adenocarcinoma of the lung.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/diagnóstico , Anciano , Femenino , Genes Relacionados con las Neoplasias , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Riesgo
8.
Cancer Res ; 66(6): 2946-52, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540642

RESUMEN

Human melanoma susceptibility is often characterized by germ-line inactivating CDKN2A (INK4A/ARF) mutations, or mutations that activate CDK4 by preventing its binding to and inhibition by INK4A. We have previously shown that a single neonatal UV radiation (UVR) dose delivered to mice that carry melanocyte-specific activation of Hras (TPras) increases melanoma penetrance from 0% to 57%. Here, we report that activated Cdk4 cooperates with activated Hras to enhance susceptibility to melanoma in mice. Whereas UVR treatment failed to induce melanomas in Cdk4(R24C/R24C) mice, it greatly increased the penetrance and decreased the age of onset of melanoma development in Cdk4(R24C/R24C)/TPras animals compared with TPras alone. This increased penetrance was dependent on the threshold of Cdk4 activation as Cdk4(R24C/+)/TPras animals did not show an increase in UVR-induced melanoma penetrance compared with TPras alone. In addition, Cdk4(R24C/R24C)/TPras mice invariably developed multiple lesions, which occurred rarely in TPras mice. These results indicate that germ-line defects abrogating the pRb pathway may enhance UVR-induced melanoma. TPras and Cdk4(R24C/R24C)/TPras tumors were comparable histopathologically but the latter were larger and more aggressive and cultured cells derived from such melanomas were also larger and had higher levels of nuclear atypia. Moreover, the melanomas in Cdk4(R24C/R24C)/TPras mice, but not in TPras mice, readily metastasized to regional lymph nodes. Thus, it seems that in the mouse, Hras activation initiates UVR-induced melanoma development whereas the cell cycle defect introduced by mutant Cdk4 contributes to tumor progression, producing more aggressive, metastatic tumors.


Asunto(s)
Cocarcinogénesis , Quinasa 4 Dependiente de la Ciclina/genética , Genes ras/genética , Melanoma Experimental/etiología , Melanoma Experimental/genética , Rayos Ultravioleta , Animales , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Melanoma Experimental/secundario , Ratones , Ratones Transgénicos , Mutación
9.
Melanoma Res ; 17(6): 380-6, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17992121

RESUMEN

Upregulation of the Wnt5a pathway has been reported in some cutaneous melanomas but its role in uveal melanoma has not been assessed. We thus sought to determine whether activation of the Wnt-signalling pathway occurred in uveal melanoma through upregulation of some of the key downstream effectors, and whether expression of these components was associated with tumour characteristics and clinical outcome. Expression of Wnt5a, MMP7, and beta-catenin was determined in 40 primary uveal melanomas by immunohistochemistry and correlated with patient prognosis. The proportion of cells immunoreactive for Wnt5a, MMP7, and beta-catenin was higher in tumours from patients with shorter survival and this difference was statistically significant for Wnt5a (P<0.01) and beta-catenin (P=0.02). These data show for the first time activation of the Wnt/beta-catenin-signalling pathway in uveal melanoma and suggest that components of this pathway might be useful prognostic markers as well as attractive therapeutic targets to treat this disease.


Asunto(s)
Metaloproteinasa 7 de la Matriz/metabolismo , Melanoma/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Neoplasias de la Úvea/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Humanos , Estimación de Kaplan-Meier , Melanoma/mortalidad , Neoplasias de la Úvea/mortalidad , Proteína Wnt-5a
10.
Melanoma Res ; 16(4): 285-96, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16845324

RESUMEN

Prognosis in patients with uveal melanoma is poor as approximately half of all tumors metastasize and currently there are no effective treatments for disseminated disease. Differences in invasiveness between uveal melanomas could therefore be of major significance regarding clinical outcome. To identify genes associated with invasive potential, we have used microarray expression profiling combined with phenotypic characterization of uveal melanoma and melanocyte cell lines to define a gene signature associated with cellular invasion. A panel of 14 uveal cell cultures was assessed using three assays of invasiveness: matrigel invasion chamber system, scratch wound closure and cell motility. We identified a set of 853 differentially expressed transcripts (Wilcoxon-Mann-Whitney test, P<0.01) that discriminated between samples with high or low invasive capacity based on a composite phenotype that takes into account behavior across all three assays. Of particular interest, expression of two members of the p21-activated kinase (PAK) family, PAK1 and PAK7, was elevated in the more invasive cultures. PAK1 has previously been shown to play a central role in regulating cell motility and invasiveness in other cell types, and increased expression has been observed in breast and colorectal carcinomas. Using small interfering RNA-mediated PAK1 knockdown, we showed up to a five-fold decrease in invasion through matrigel, indicating that elevated levels of PAK1 are associated with invasive potential in uveal melanoma. These data implicate PAK1 as a potential new target for therapy of these tumors.


Asunto(s)
Melanoma/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias de la Úvea/patología , Biomarcadores de Tumor/genética , Movimiento Celular , Colágeno/metabolismo , ADN de Neoplasias/análisis , Combinación de Medicamentos , Perfilación de la Expresión Génica , Humanos , Laminina/metabolismo , Melanoma/enzimología , Melanoma/genética , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Proteoglicanos/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Úvea/enzimología , Neoplasias de la Úvea/genética , Quinasas p21 Activadas
11.
Cancer Res ; 62(3): 875-80, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11830546

RESUMEN

The contribution of the UV component of sunlight to the development of skin cancer is widely acknowledged, although the molecular mechanisms that are disrupted by UV radiation (UVR) resulting in the loss of normal growth controls of the epidermal stem cell keratinocytes and melanocytes is still poorly understood. Alpha-melanocyte stimulating hormone (alpha-MSH), acting via its receptor MC1, has a key role in skin pigmentation and the melanizing response after exposure to UVR. The cell cycle inhibitor p16/CDKN2A also appears to have an important function in a cell cycle checkpoint response in skin after exposure to UVR. Both of these genes have been identified as risk factors in skin cancer, MC1R variants are associated with increased risk to both melanoma and nonmelanoma skin cancers, and p16/CDKN2A with increased risk of melanoma. Here we demonstrate that the increased expression of p16 after exposure to suberythemal doses of UVR is potentiated by alpha-MSH, a ligand for MC1R, and this effect is mimicked by cAMP, the intracellular mediator of alpha-MSH signaling via the MC1 receptor. This link between p16 and MC1R may provide a molecular basis for the increased skin cancer risk associated with MC1R polymorphisms.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Piel/metabolismo , Piel/efectos de la radiación , Rayos Ultravioleta , alfa-MSH/análogos & derivados , alfa-MSH/farmacología , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Técnicas de Cultivo de Órganos , Receptores de Corticotropina/fisiología , Receptores de Melanocortina , Transducción de Señal , Piel/efectos de los fármacos
12.
Pigment Cell Melanoma Res ; 29(3): 329-39, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26854966

RESUMEN

Melanomas have high levels of genomic instability that can contribute to poor disease prognosis. Here, we report a novel defect of the ATM-dependent cell cycle checkpoint in melanoma cell lines that promotes genomic instability. In defective cells, ATM signalling to CHK2 is intact, but the cells are unable to maintain the cell cycle arrest due to elevated PLK1 driving recovery from the arrest. Reducing PLK1 activity recovered the ATM-dependent checkpoint arrest, and over-expressing PLK1 was sufficient to overcome the checkpoint arrest and increase genomic instability. Loss of the ATM-dependent checkpoint did not affect sensitivity to ionizing radiation demonstrating that this defect is distinct from ATM loss of function mutations. The checkpoint defective melanoma cell lines over-express PLK1, and a significant proportion of melanomas have high levels of PLK1 over-expression suggesting this defect is a common feature of melanomas. The inability of ATM to impose a cell cycle arrest in response to DNA damage increases genomic instability. This work also suggests that the ATM-dependent checkpoint arrest is likely to be defective in a higher proportion of cancers than previously expected.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Puntos de Control del Ciclo Celular/efectos de la radiación , Inestabilidad Genómica/genética , Melanoma/genética , Melanoma/patología , Mutación/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa de Punto de Control 2/metabolismo , Inestabilidad Genómica/efectos de la radiación , Humanos , Melanoma/enzimología , Mitosis/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Radiación Ionizante , Regulación hacia Arriba/efectos de la radiación , Quinasa Tipo Polo 1
13.
Pigment Cell Melanoma Res ; 29(4): 444-52, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27166757

RESUMEN

Here we have carried out a multiparameter analysis using a panel of 28 immunohistochemical markers to identify markers of transformation from benign and dysplastic naevus to primary melanoma in three separate cohorts totalling 279 lesions. We have identified a set of eight markers that distinguish naevi from melanoma. None of markers or parameters assessed differentiated benign from dysplastic naevi. Indeed, the naevi clustered tightly in terms of their immunostaining patterns whereas primary melanomas showed more diverse staining patterns. A small subset of histopathologically benign lesions had elevated levels of multiple markers associated with melanoma, suggesting that these represent naevi with an increased potential for transformation to melanoma.


Asunto(s)
Biomarcadores/metabolismo , Transformación Celular Neoplásica/patología , Melanoma/patología , Nevo Pigmentado/patología , Neoplasias Cutáneas/patología , Transformación Celular Neoplásica/metabolismo , Humanos , Melanoma/metabolismo , Nevo Pigmentado/metabolismo , Pronóstico , Neoplasias Cutáneas/metabolismo , Análisis de Matrices Tisulares
14.
Oncogene ; 23(23): 4060-7, 2004 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-15048078

RESUMEN

We have used microarray gene expression profiling and machine learning to predict the presence of BRAF mutations in a panel of 61 melanoma cell lines. The BRAF gene was found to be mutated in 42 samples (69%) and intragenic mutations of the NRAS gene were detected in seven samples (11%). No cell line carried mutations of both genes. Using support vector machines, we have built a classifier that differentiates between melanoma cell lines based on BRAF mutation status. As few as 83 genes are able to discriminate between BRAF mutant and BRAF wild-type samples with clear separation observed using hierarchical clustering. Multidimensional scaling was used to visualize the relationship between a BRAF mutation signature and that of a generalized mitogen-activated protein kinase (MAPK) activation (either BRAF or NRAS mutation) in the context of the discriminating gene list. We observed that samples carrying NRAS mutations lie somewhere between those with or without BRAF mutations. These observations suggest that there are gene-specific mutation signals in addition to a common MAPK activation that result from the pleiotropic effects of either BRAF or NRAS on other signaling pathways, leading to measurably different transcriptional changes.


Asunto(s)
Melanoma/metabolismo , Mutación , Proteínas Proto-Oncogénicas c-raf/genética , Sustitución de Aminoácidos , Humanos , Melanoma/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas B-raf , Proteínas ras/genética
15.
Methods Mol Biol ; 280: 175-83, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15187253

RESUMEN

A short-term whole-skin organ culture model has been established to enable the investigation of cell cycle perturbations in epidermal layer cells following exposure to ultraviolet radiation (UVR). This model affords the opportunity to manipulate the growth and nutrient conditions, and to perform detailed biochemical and immunohistochemical analysis of skin cells in their normal epidermal layer microenvironment. The use of this model is described in this chapter.


Asunto(s)
Inmunohistoquímica/métodos , Piel/metabolismo , Bromodesoxiuridina/farmacología , Ciclo Celular , Colorantes/farmacología , Medios de Cultivo/farmacología , Epidermis/metabolismo , Formaldehído/metabolismo , Genes cdc , Humanos , Masculino , Técnicas de Cultivo de Órganos , Factores de Tiempo , Rayos Ultravioleta
16.
J Invest Dermatol ; 134(1): 150-158, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23842115

RESUMEN

A hallmark of cancer is genomic instability that is considered to provide the adaptive capacity of cancers to thrive under conditions in which the normal precursors would not survive. Recent genomic analysis has revealed a very high degree of genomic instability in melanomas, although the mechanism by which this instability arises is not known. Here we report that a high proportion (68%) of melanoma cell lines are either partially (40%) or severely (28%) compromised for the G2 phase decatenation checkpoint that normally functions to ensure that the sister chromatids are able to separate correctly during mitosis. The consequence of this loss of checkpoint function is a severely reduced ability to partition the replicated genome in mitosis and thereby increase genomic instability. We also demonstrate that decatenation is dependent on both TopoIIα and ß isoforms. The high incidence of decatenation checkpoint defect is likely to be a major contributor to the high level of genomic instability found in melanomas.


Asunto(s)
Genes cdc/genética , Inestabilidad Genómica/genética , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/genética , Fase G2/genética , Humanos , Mitosis/genética , ARN Interferente Pequeño/genética , Intercambio de Cromátides Hermanas/genética
17.
Cell Cycle ; 13(20): 3302-11, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25485510

RESUMEN

Whereas many components regulating the progression from S phase through G2 phase into mitosis have been identified, the mechanism by which these components control this critical cell cycle progression is still not fully elucidated. Cyclin A/Cdk2 has been shown to regulate the timing of Cyclin B/Cdk1 activation and progression into mitosis although the mechanism by which this occurs is only poorly understood. Here we show that depletion of Cyclin A or inhibition of Cdk2 during late S/early G2 phase maintains the G2 phase arrest by reducing Cdh1 transcript and protein levels, thereby stabilizing Claspin and maintaining elevated levels of activated Chk1 which contributes to the G2 phase observed. Interestingly, the Cyclin A/Cdk2 regulated APC/C(Cdh1) activity is selective for only a subset of Cdh1 targets including Claspin. Thus, a normal role for Cyclin A/Cdk2 during early G2 phase is to increase the level of Cdh1 which destabilises Claspin which in turn down regulates Chk1 activation to allow progression into mitosis. This mechanism links S phase exit with G2 phase transit into mitosis, provides a novel insight into the roles of Cyclin A/Cdk2 in G2 phase progression, and identifies a novel role for APC/C(Cdh1) in late S/G2 phase cell cycle progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cadherinas/metabolismo , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Fase G2/fisiología , Fase S/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos CD , Cadherinas/genética , Ciclina A/genética , Quinasa 2 Dependiente de la Ciclina/genética , Fase G2/genética , Humanos , Fase S/genética
18.
Pigment Cell Melanoma Res ; 27(5): 813-21, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24890688

RESUMEN

Melanoma cell lines are commonly defective for the G2-phase cell cycle checkpoint that responds to incomplete catenation of the replicated chromosomes. Here, we demonstrate that melanomas defective for this checkpoint response are less sensitive to genotoxic stress, suggesting that the defective cell lines compensated for the checkpoint loss by increasing their ability to cope with DNA damage. We performed an siRNA kinome screen to identify kinases responsible and identified PI3K pathway components. Checkpoint-defective cell lines were three-fold more sensitive to small molecule inhibitors of PI3K. The PI3K inhibitor PF-05212384 promoted apoptosis in the checkpoint-defective lines, and the increased sensitivity to PI3K inhibition correlated with increased levels of activated Akt. This work demonstrates that increased PI3K pathway activation is a necessary adaption for the continued viability of melanomas with a defective decatenation checkpoint.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Cutáneas/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Separación Celular , Daño del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Citometría de Flujo , Humanos , Melanoma/genética , Morfolinas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/metabolismo , Neoplasias Cutáneas/genética , Células Madre , Triazinas/química
19.
Pigment Cell Melanoma Res ; 26(6): 805-16, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23837768

RESUMEN

The ultraviolet radiation (UVR) component of sunlight is the major environmental risk factor for melanoma, producing DNA lesions that can be mutagenic if not repaired. The high level of mutations in melanomas that have the signature of UVR-induced damage indicates that the normal mechanisms that detect and repair this damage must be defective in this system. With the exception of melanoma-prone heritable syndromes which have mutations of repair genes, there is little evidence for somatic mutation of known repair genes. Cell cycle checkpoint controls are tightly associated with repair mechanisms, arresting cells to allow for repair before continuing through the cell cycle. Checkpoint signaling components also regulate the repair mechanisms. Defects in checkpoint mechanisms have been identified in melanomas and are likely to be responsible for increased mutation load in melanoma. Loss of the checkpoint responses may also provide an opportunity to target melanomas using a synthetic lethal approach to identify and inhibit mechanisms that compensate for the defective checkpoints.


Asunto(s)
Puntos de Control del Ciclo Celular , Reparación del ADN , Melanoma/patología , Melanoma/terapia , Terapia Molecular Dirigida , Puntos de Control del Ciclo Celular/efectos de la radiación , Reparación del ADN/efectos de la radiación , Humanos , Rayos Ultravioleta
20.
Front Pharmacol ; 3: 9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22347187

RESUMEN

Conventional chemotherapeutics target the proliferating fraction of cells in the patient's body, which will include the tumor cells, but are also toxic to actively proliferating normal tissues. Cellular stresses, such as those imposed by chemotherapeutic drugs, induce cell cycle checkpoint arrest, and currently approaches targeting these checkpoints are being explored to increase the efficacy and selectivity of conventional chemotherapeutic treatments. Loss of a checkpoint may also make cancer cells more reliant on other mechanisms to compensate for the loss of this function, and these compensatory mechanisms may be targeted using synthetic lethal approaches. Here we will discuss the utility of targeting checkpoint defects as novel anti-cancer therapies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA