Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Genomics Proteomics ; 19(4): 490-502, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35732325

RESUMEN

BACKGROUND/AIM: Angiopoietin-like protein 4 (ANGPTL4) is a multifunctional signaling protein implicated in carbohydrate metabolism, inflammation, cancer growth and progression, anoikis resistance, angiogenesis, and metastasis. However, signaling pathways of ANGPTL4 in cholangiocarcinoma (CCA) remain unknown. The aim of this study was to explore ANGPTL4-related signaling proteins and pathways associated with CCA biology. MATERIALS AND METHODS: ANGPTL4 of CCA cells was silenced by small interfering RNA (siRNA) with scramble control and ANGPTL4-related signaling proteins were investigated using mass spectrometry, bioinformatics tools and molecular docking. RESULTS: Among the 321 differentially expressed proteins, 151 were down-regulated. Among them, bioinformatic analyses revealed that ANGPTL4 interacts with DNA-dependent protein kinase catalytic subunit (PRKDC) and 60S ribosomal protein L21 (RPL21) via AKT serine/threonine kinase 1 (AKT1), mechanistic target of rapamycin kinase (MTOR) and ribosomal protein L5 (RPL5). Online database analysis showed that mRNA and protein expression levels of ANGPTL4-related signaling proteins were significantly higher in CCA than in normal tissues. Moreover, a high mRNA expression level was associated with high tumor grade (p<0.0001) and lymph node metastasis (p<0.0001). CONCLUSION: The signaling pathway of ANGPTL4 in CCA progression might be regulated by PRKDC and RPL21. Furthermore, high expression of ANGPTL4-related signaling proteins has potential to be used in clinical prognosis.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina , Neoplasias de los Conductos Biliares , Colangiocarcinoma , Proteína 4 Similar a la Angiopoyetina/genética , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Humanos , Simulación del Acoplamiento Molecular , ARN Mensajero/genética , ARN Interferente Pequeño , Transducción de Señal
2.
PeerJ ; 10: e12750, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35070505

RESUMEN

BACKGROUND: Genetic alterations in ARID1A were detected at a high frequency in cholangiocarcinoma (CCA). Growing evidence indicates that the loss of ARID1A expression leads to activation of the PI3K/AKT pathway and increasing sensitivity of ARID1A-deficient cells for treatment with the PI3K/AKT inhibitor. Therefore, we investigated the association between genetic alterations of ARID1A and the PI3K/AKT pathway and evaluated the effect of AKT inhibition on ARID1A-deficient CCA cells. METHODS: Alterations of ARID1A, PI3K/AKT pathway-related genes, clinicopathological data and overall survival of 795 CCA patients were retrieved from cBio Cancer Genomics Portal (cBioPortal) databases. The association between genetic alterations and clinical data were analyzed. The effect of the AKT inhibitor (MK-2206) on ARID1A-deficient CCA cell lines and stable ARID1A-knockdown cell lines was investigated. Cell viability, apoptosis, and expression of AKT signaling were analyzed using an MTT assay, flow cytometry, and Western blots, respectively. RESULTS: The analysis of a total of 795 CCA samples revealed that ARID1A alterations significantly co-occurred with mutations of EPHA2 (p < 0.001), PIK3CA (p = 0.047), and LAMA1 (p = 0.024). Among the EPHA2 mutant CCA tumors, 82% of EPHA2 mutant tumors co-occurred with ARID1A truncating mutations. CCA tumors with ARID1A and EPHA2 mutations correlated with better survival compared to tumors with ARID1A mutations alone. We detected that 30% of patients with PIK3CA driver missense mutations harbored ARID1A-truncated mutations and 60% of LAMA1-mutated CCA co-occurred with truncating mutations of ARID1A. Interestingly, ARID1A-deficient CCA cell lines and ARID1A-knockdown CCA cells led to increased sensitivity to treatment with MK-2206 compared to the control. Treatment with MK-2206 induced apoptosis in ARID1A-knockdown KKU-213A and HUCCT1 cell lines and decreased the expression of pAKTS473 and mTOR. CONCLUSION: These findings suggest a dependency of ARID1A-deficient CCA tumors with the activation of the PI3K/AKT-pathway, and that they may be more vulnerable to selective AKT pathway inhibitors which can be used therapeutically.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Proteínas Proto-Oncogénicas c-akt , Fosfatidilinositol 3-Quinasas/genética , Colangiocarcinoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Conductos Biliares Intrahepáticos/metabolismo , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética
3.
EXCLI J ; 19: 154-166, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194362

RESUMEN

The incidence of cholangiocarcinoma (CCA) has risen in many countries, but there is still no appropriate screening and treatment available. The growing number of microarray data published todays can be a powerful resource for the discovery of biomarkers to tackle challenges in the management of CCA. This study analyzed multiple microarray datasets to identify the common transcriptional networks in CCA and select a possible biomarker for functional study in CCA cell lines. A systematic searching identified 4 microarray datasets from Gene Expression Omnibus (GEO) repository and PubMed articles. Differential expression analysis between tumor and normal tissues was performed in each dataset. In order to characterize the common expression pattern, differentially expressed genes (DEGs) from all datasets were combined and visualized by hierarchical clustering and heatmap. Gene enrichment analysis performed in each cluster revealed that over-expressed DEGs were enriched in cell cycle, cell migration and response to cytokines while under-expressed DEGs were enriched in metabolic processes such as oxidation-reduction, lipid, and drug. To explain tumor characteristics, genes enriched in cell migration and response to cytokines were further investigated. Among these genes, CCL20 was selected for functional study because its role has never been studied in CCA. Moreover, its signaling may be regulated by disrupting its only receptor, CCR6. Treatment with recombinant CCL20 induced higher cell migration and increased expression of N-cad. In contrast, knockdown of CCR6 by siRNA reduced cell migration ability and decreased N-cadherin level. Altogether, these results suggested the contribution of CCL20/CCR6 signaling in cell migration through epithelial-mesenchymal transition process. Thus, CCL20/CCR6 signaling might be a target for the management of CCA.

4.
Heliyon ; 6(1): e03255, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32051864

RESUMEN

Anoikis resistance is a critical feature involved in tumor progression and chemoresistance. Finding approaches to improve the effect of chemotherapy on anoikis-resistant cancer cells is therefore critically important. In this study, we examined the effects of curcumin in anoikis-resistant cholangiocarcinoma (CCA) cells, including HuCCT1 and TFK-1 that were anchorage-independently cultured (AI-cells) using poly (2-hydroxyethyl methacrylate). The AI-CCA cells were treated with curcumin alone or in combination with anti-cancer agents and their responses to each treatment were determined by cell viability assay. Gene expression in AI-cells was determined by quantitative real-time PCR. The potential involvement of angiopoietin-like 4 (ANGPTL4) in anoikis resistance was examined by gene knockdown. It was found that AI-cells tended to resist anti-cancer agents tested, especially AI-HuCCT1, which significantly resisted gemcitabine and suberoylanilide hydroxamic acid (SAHA). Curcumin alone significantly inhibited viability and colony formation of AI-cells. Moreover, curcumin combination significantly enhanced the treatment effect of SAHA on AI-HuCCT1 and AI-TFK-1 cells. Gene expression analysis revealed that ANGPTL4 was markedly upregulated in AI-CCA cells and its knockdown tended to sensitize AI-cells to cell death and treatments. In addition, curcumin treatment decreased phosphorylated STAT3 and expression levels of Mcl-1, HDACs and ANGPTL4. Altogether, these findings reveal the beneficial property of curcumin to potentiate chemotherapeutic effects on anoikis-resistant CCA cells, which might suggest the potential use of curcumin for cancer treatment.

5.
Immunol Invest ; 48(1): 11-26, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30321079

RESUMEN

Human γδ T lymphocytes play a role in the immune system defense against cancer. Their broad anti-cancer activity against different types of cancers makes them outstanding candidates for cancer immunotherapy. An issue of recent interest is whether their antigen presentation features are similar to mature dendritic cells. The antigen-presenting cell (APC)-like phenotype and function of γδ T lymphocytes have been confirmed in many clinical trials. In this study, to support the strong role played by Vγ9Vδ2 T cells against cancer, we provide evidence that Vγ9Vδ2 T cells activated with chronic myeloid leukemia (CML) cell lysate antigens can efficiently express an APC phenotype and function. Vγ9Vδ2 T cells derived from normal peripheral blood mononuclear cells were activated with tumor cell lysate, and the tumor-activated Vγ9Vδ2 T cells could recognize and kill CML through their cytotoxic activity. In conclusion, the Vγ9Vδ2 T cells activated by cancer cell lysate showed APC characteristics, and this may greatly increase interest in investigating their therapeutic potential in hematologic malignancies. Abbreviations: CML: chronic myeloid leukemia; APC: antigen-presenting cell; TCR: T cell receptor; MHC: major histocompatibility complex; N-BPs: nitrogen-containing bisphosphonates; IPP: isopentenyl pyrophosphate; PBMC: peripheral blood mononuclear cells; NKG2D: natural killer receptor group 2, member D; TRAIL: tumor necrosis factor-related apoptosis-inducing ligand.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva/métodos , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Linfocitos T/inmunología , Presentación de Antígeno , Células Presentadoras de Antígenos/trasplante , Antígenos de Neoplasias/inmunología , Diferenciación Celular , Línea Celular , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Leucocitos Mononucleares , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/trasplante
6.
Asian Pac J Cancer Prev ; 18(2): 473-477, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28345832

RESUMEN

Background: Metformin is an oral anti-diabetic agent that has been widely prescribed for treatment of type II diabetes. Anti-cancer properties of metformin have been revealed for numerous human malignancies including cholangiocarcinoma (CCA) with anti-proliferative effects in vitro. However, effects on CCA cell migration and invasion have not been fully investigated. The present study aimed to explore the inhibitory effects of metformin on motility, migration and invasion of the CCA cell line HuCCT1, and examine molecular mechanisms underlying metformin effects. Methods: HuCCT1 cells were exposed to increasing doses of metformin. Viability and growth of HuCCT1 cells were assessed by MTS and colony formation assays, respectively. Motility, migration and invasion of metformin-treated HuCCT1 cells were determined in vitro using wound healing, transwell migration and matrigel invasion assays. Expression of signaling molecules and epithelial-mesenchymal transition (EMT) markers was assessed by Western blotting. Results: It was observed that metformin significantly decreased HuCCT1 cell viability and colony formation. The agent also markedly reduced wound closure, migration and invasion of HuCCT1 cells. Furthermore, metformin exposure resulted in decreased STAT3 activation and down-regulation of anti-apoptotic protein Bcl-2 and Mcl-1 expression. In addition, it upregulated the expression of E-cadherin, while downregulating that of N-cadherin, Snail, and MMP-2. Conclusion: These results demonstrated inhibitory effects of metformin on CCA cell migration and invasion, possibly involving the STAT3 pathway and reversal of EMT markers expression. They further suggest that metformin may be useful for CCA management.

7.
Parasitol Int ; 66(4): 426-431, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27989833

RESUMEN

Protein 14-3-3s are abundant phospho-serine/threonine binding proteins, which are highly conserved among eukaryotes. Members of this protein family mediate metabolism and signal transduction networks through binding to hundreds of other protein partners. Protein 14-3-3s have been studied in other species of parasitic helminthes, but little is known about this protein in the carcinogenic liver fluke Opisthorchis viverrini. In this study, we identified and characterized protein 14-3-3s of O. viverrini. Seven protein 14-3-3 encoded sequences were retrieved from the O. viverrini genome database. Multiple alignment and phylogenetic analysis were performed. Two isoforms (protein 14-3-3 zeta and protein 14-3-3 epsilon) that have been previously found in the excretory-secretory (ES) products of O. viverrini were produced as recombinant protein in E. coli and the proteins were then used to immunize mice to obtain specific antibodies. Western blot analysis showed that both proteins were detected in all obtainable developmental stages of O. viverrini and the ES products. Immunolocalization revealed that both isoforms were expressed throughout tissues and organs except the gut epithelium. The highest expression was observed in testes especially in developing spermatocytes, suggesting their role in spermatogenesis. Prominent expression was also detected on tegumental surface of the parasite and on epical surface of bile duct epithelium indicates their additional role in host-parasite interaction. These findings indicate that protein 14-3-3s play important role in the life cycle of the parasite and might be involved in the pathogenesis of O. viverrini infection.


Asunto(s)
Proteínas 14-3-3/genética , Expresión Génica , Proteínas del Helminto/genética , Opisthorchis/fisiología , Proteínas 14-3-3/química , Proteínas 14-3-3/metabolismo , Secuencia de Aminoácidos , Animales , Escherichia coli/genética , Proteínas del Helminto/química , Proteínas del Helminto/metabolismo , Ratones , Ratones Endogámicos BALB C , Opisthorchis/genética , Organismos Modificados Genéticamente/genética , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Transcriptoma
8.
J Hepatobiliary Pancreat Sci ; 23(7): 397-405, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27107220

RESUMEN

BACKGROUND: Anoikis is a specialized cell apoptosis triggered by loss of cell adhesion. Acquisition of anoikis resistance is a key cancer hallmark that has been associated with metastasis of human malignancies. Cholangiocarcinoma (CCA) is a highly metastatic cancer of the bile duct, in which mechanisms underlying its resistance to anoikis has not been well elucidated. METHODS: HuCCT1 and Huh28 were utilized as in vitro models to examine biological characteristics of anoikis resistant-CCA cells. Expression and activation of signaling molecules were determined by western blotting. RESULTS: We demonstrated that a significant proportion of CCA cells including HuCCT1 and Huh28 resisted anoikis induced by anchorage-independent condition. Notably, anoikis-resistant CCA cells, especially HuCCT1, substantially increased invasive potential and became resistant to gemcitabine, which is the standard chemotherapeutic agent for CCA. Mechanistically, the anoikis-resistant HuCCT1 cells markedly increased activation of signal transducer and activator of transcription 3 (STAT3) and concomitantly upregulated anti-apoptotic proteins, Mcl-1 and Bcl-2. Inhibition of STAT3 by AG 490 significantly induced HuCCT1 cells to undergo anoikis. CONCLUSIONS: Taken together, our findings uncover the aggressive properties of CCA cells that overcome anoikis, and reveal the involvement of STAT3 signaling pathway in anoikis resistance in CCA. Targeting STAT3 may restore anoikis and prevent metastasis of the disease.


Asunto(s)
Anoicis/efectos de los fármacos , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Factor de Transcripción STAT3/genética , Tirfostinos/farmacología , Anoicis/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/patología , Western Blotting , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Desoxicitidina/farmacología , Humanos , Fosforilación , Valores de Referencia , Transducción de Señal , Gemcitabina
9.
Drug Des Devel Ther ; 9: 2033-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25897210

RESUMEN

PURPOSE: Fork head box M1 (FoxM1) is an oncogenic transcription factor frequently elevated in numerous cancers, including cholangiocarcinoma (CCA). A growing body of evidence documents its diverse functions contributing to tumorigenesis and cancer progression. As such, discovery of agents that can target FoxM1 would be valuable for the treatment of CCA. The quinoline-based compounds, namely clioquinol (CQ) and nitroxoline (NQ), represent a new class of anticancer drug. However, their efficacy and underlying mechanisms have not been elucidated in CCA. In this study, anticancer activities and inhibitory effects of CQ and NQ on FoxM1 signaling were explored using CCA cells. METHODS: The effects of CQ and NQ on cell viability and proliferation were evaluated using the colorimetric 3-(4,5-dimethylthiazol-2yl)-5-(3-carboxymethoxyphenyl)-(4-sulfophenyl)-2H-tetrazolium (MTS assay). Colony formation and cell migration affected by CQ and NQ were investigated using a clonogenic and a wound healing assay, respectively. To demonstrate the agents' effects on FoxM1 signaling, expression levels of the target genes were quantitatively determined using real-time polymerase chain reaction. RESULTS: CQ and NQ significantly inhibited cell survival of HuCCT1 and Huh28 in a dose- and a time-dependent fashion. Further investigations using the rapidly proliferating HuCCT1 cells revealed significant suppression of cell proliferation and colony formation induced by low doses of the compounds. Treatment of CQ and NQ repressed expression of cyclin D1 but enhanced expression of p21. Most importantly, upon CQ and NQ treatment, expression of oncogenic FoxM1 was markedly decreased concomitant with downregulation of various FoxM1's downstream targets including cdc25b, CENP-B, and survivin. In addition, the compounds distinctly impaired HuCCT1 migration as well as inhibited expression of matrix metalloproteinase (MMP)-2 and MMP-9. CONCLUSION: Collectively, this study reports for the first time the anticancer effects of CQ and NQ against CCA cells, and highlights new insights into the mechanism of actions of the quinoline-based compounds to disrupt FoxM1 signaling.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Clioquinol/farmacología , Factores de Transcripción Forkhead/antagonistas & inhibidores , Nitroquinolinas/farmacología , Quinolinas/química , Antineoplásicos/química , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colangiocarcinoma/patología , Clioquinol/química , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Nitroquinolinas/química , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Factores de Tiempo
10.
Nat Genet ; 45(12): 1474-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24185513

RESUMEN

The impact of different carcinogenic exposures on the specific patterns of somatic mutation in human tumors remains unclear. To address this issue, we profiled 209 cholangiocarcinomas (CCAs) from Asia and Europe, including 108 cases caused by infection with the liver fluke Opisthorchis viverrini and 101 cases caused by non-O. viverrini-related etiologies. Whole-exome sequencing (n = 15) and prevalence screening (n = 194) identified recurrent somatic mutations in BAP1 and ARID1A, neither of which, to our knowledge, has previously been reported to be mutated in CCA. Comparisons between intrahepatic O. viverrini-related and non-O. viverrini-related CCAs demonstrated statistically significant differences in mutation patterns: BAP1, IDH1 and IDH2 were more frequently mutated in non-O. viverrini CCAs, whereas TP53 mutations showed the reciprocal pattern. Functional studies demonstrated tumor suppressive functions for BAP1 and ARID1A, establishing the role of chromatin modulators in CCA pathogenesis. These findings indicate that different causative etiologies may induce distinct somatic alterations, even within the same tumor type.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/parasitología , Conductos Biliares Intrahepáticos , Colangiocarcinoma/genética , Colangiocarcinoma/parasitología , Exoma/genética , Fasciola hepatica , Fascioliasis/complicaciones , Animales , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Mutación , Análisis de Secuencia de ADN , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética
11.
Sci Transl Med ; 5(197): 197ra101, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23926199

RESUMEN

Aristolochic acid (AA), a natural product of Aristolochia plants found in herbal remedies and health supplements, is a group 1 carcinogen that can cause nephrotoxicity and upper urinary tract urothelial cell carcinoma (UTUC). Whole-genome and exome analysis of nine AA-associated UTUCs revealed a strikingly high somatic mutation rate (150 mutations/Mb), exceeding smoking-associated lung cancer (8 mutations/Mb) and ultraviolet radiation-associated melanoma (111 mutations/Mb). The AA-UTUC mutational signature was characterized by A:T to T:A transversions at the sequence motif A[C|T]AGG, located primarily on nontranscribed strands. AA-induced mutations were also significantly enriched at splice sites, suggesting a role for splice-site mutations in UTUC pathogenesis. RNA sequencing of AA-UTUC confirmed a general up-regulation of nonsense-mediated decay machinery components and aberrant splicing events associated with splice-site mutations. We observed a high frequency of somatic mutations in chromatin modifiers, particularly KDM6A, in AA-UTUC, demonstrated the sufficiency of AA to induce renal dysplasia in mice, and reproduced the AA mutational signature in experimentally treated human renal tubular cells. Finally, exploring other malignancies that were not known to be associated with AA, we screened 93 hepatocellular carcinoma genomes/exomes and identified AA-like mutational signatures in 11. Our study highlights an unusual genome-wide AA mutational signature and the potential use of mutation signatures as "molecular fingerprints" for interrogating high-throughput cancer genome data to infer previous carcinogen exposures.


Asunto(s)
Ácidos Aristolóquicos/efectos adversos , Carcinógenos/análisis , Genoma Humano/genética , Mutación/efectos de los fármacos , Mutación/genética , Animales , Ácidos Aristolóquicos/análisis , Carcinógenos/toxicidad , Línea Celular Tumoral , Humanos , Enfermedades Renales/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Mutágenos/análisis , Mutágenos/toxicidad , Neoplasias/genética , Empalme del ARN/genética , Neoplasias Urológicas/genética , Urotelio/patología
12.
Nat Genet ; 44(6): 690-3, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22561520

RESUMEN

Opisthorchis viverrini-related cholangiocarcinoma (CCA), a fatal bile duct cancer, is a major public health concern in areas endemic for this parasite. We report here whole-exome sequencing of eight O. viverrini-related tumors and matched normal tissue. We identified and validated 206 somatic mutations in 187 genes using Sanger sequencing and selected 15 genes for mutation prevalence screening in an additional 46 individuals with CCA (cases). In addition to the known cancer-related genes TP53 (mutated in 44.4% of cases), KRAS (16.7%) and SMAD4 (16.7%), we identified somatic mutations in 10 newly implicated genes in 14.8-3.7% of cases. These included inactivating mutations in MLL3 (in 14.8% of cases), ROBO2 (9.3%), RNF43 (9.3%) and PEG3 (5.6%), and activating mutations in the GNAS oncogene (9.3%). These genes have functions that can be broadly grouped into three biological classes: (i) deactivation of histone modifiers, (ii) activation of G protein signaling and (iii) loss of genome stability. This study provides insight into the mutational landscape contributing to O. viverrini-related CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/parasitología , Colangiocarcinoma/genética , Colangiocarcinoma/parasitología , Fascioliasis/complicaciones , Adulto , Anciano , Proteínas de Unión al ADN/genética , Exoma , Femenino , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Mutación , Receptores Inmunológicos/genética , Análisis de Secuencia de ADN
13.
Genome Biol ; 12(9): R96, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-21955916

RESUMEN

BACKGROUND: Well differentiated papillary mesothelioma of the peritoneum (WDPMP) is a rare variant of epithelial mesothelioma of low malignancy potential, usually found in women with no history of asbestos exposure. In this study, we perform the first exome sequencing of WDPMP. RESULTS: WDPMP exome sequencing reveals the first somatic mutation of E2F1, R166H, to be identified in human cancer. The location is in the evolutionarily conserved DNA binding domain and computationally predicted to be mutated in the critical contact point between E2F1 and its DNA target. We show that the R166H mutation abrogates E2F1's DNA binding ability and is associated with reduced activation of E2F1 downstream target genes. Mutant E2F1 proteins are also observed in higher quantities when compared with wild-type E2F1 protein levels and the mutant protein's resistance to degradation was found to be the cause of its accumulation within mutant over-expressing cells. Cells over-expressing wild-type E2F1 show decreased proliferation compared to mutant over-expressing cells, but cell proliferation rates of mutant over-expressing cells were comparable to cells over-expressing the empty vector. CONCLUSIONS: The R166H mutation in E2F1 is shown to have a deleterious effect on its DNA binding ability as well as increasing its stability and subsequent accumulation in R166H mutant cells. Based on the results, two compatible theories can be formed: R166H mutation appears to allow for protein over-expression while minimizing the apoptotic consequence and the R166H mutation may behave similarly to SV40 large T antigen, inhibiting tumor suppressive functions of retinoblastoma protein 1.


Asunto(s)
Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Mesotelioma/genética , Mutación , Peritoneo/patología , Adulto , Proliferación Celular , Inmunoprecipitación de Cromatina , Análisis Mutacional de ADN/métodos , Factor de Transcripción E2F1/metabolismo , Exoma , Femenino , Humanos , Mesotelioma/patología , Plásmidos/genética , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Estabilidad Proteica , Transfección , Células Tumorales Cultivadas
14.
Nature ; 469(7331): 539-42, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21248752

RESUMEN

The genetics of renal cancer is dominated by inactivation of the VHL tumour suppressor gene in clear cell carcinoma (ccRCC), the commonest histological subtype. A recent large-scale screen of ∼3,500 genes by PCR-based exon re-sequencing identified several new cancer genes in ccRCC including UTX (also known as KDM6A), JARID1C (also known as KDM5C) and SETD2 (ref. 2). These genes encode enzymes that demethylate (UTX, JARID1C) or methylate (SETD2) key lysine residues of histone H3. Modification of the methylation state of these lysine residues of histone H3 regulates chromatin structure and is implicated in transcriptional control. However, together these mutations are present in fewer than 15% of ccRCC, suggesting the existence of additional, currently unidentified cancer genes. Here, we have sequenced the protein coding exome in a series of primary ccRCC and report the identification of the SWI/SNF chromatin remodelling complex gene PBRM1 (ref. 4) as a second major ccRCC cancer gene, with truncating mutations in 41% (92/227) of cases. These data further elucidate the somatic genetic architecture of ccRCC and emphasize the marked contribution of aberrant chromatin biology.


Asunto(s)
Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Neoplasias Pancreáticas/genética
15.
Int J Oncol ; 35(2): 287-95, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19578742

RESUMEN

Cholangiocarcinoma (CCA) represents a model of tumor development after long-term inflammation which causes DNA damage or impairs DNA repair mechanism. AID and GANP, both appearing in antigen-driven B cells, are involved in affinity maturation of the immunoglobulin V-region with increased somatic mutation. A normal cholangiocyte line showed the induction of AID transcripts after stimulation with TNF-alpha, whereas ganp transcripts appeared constitutively in this cell line. Next, we examined the expression of AID and GANP in clinical CCA specimens to obtain information whether their expression levels are associated with the malignant grade of CCA. AID expression was similarly detected in the clinical cases of both well-differentiated and poorly-differentiated CCAs. On the contrary, GANP expression was detected in CCA cells at a higher level in the nucleus of poorly-differentiated CCAs with shorter survivals than in that of well-differentiated CCAs. The high and low cases of nuclear GANP expression showed no change in the frequency of the TP53 mutations, however, further investigation by in vitro experiment demonstrated that the high GANP expression caused the increased number of gammaH2AX foci after DNA damage by ionizing-irradiation. These results suggest that GANP is involved in regulation of DNA repair mechanism and the abnormal over-expression of GANP together with AID might be associated with rigorous DNA damage, potentially causing the malignant development of CCAs during long-term inflammation.


Asunto(s)
Acetiltransferasas/fisiología , Neoplasias de los Conductos Biliares/etiología , Conductos Biliares Intrahepáticos , Colangiocarcinoma/etiología , Citidina Desaminasa/fisiología , Región Variable de Inmunoglobulina/genética , Inflamación/complicaciones , Acetiltransferasas/análisis , Acetiltransferasas/genética , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/inmunología , Línea Celular Tumoral , Colangiocarcinoma/genética , Colangiocarcinoma/inmunología , Citidina Desaminasa/análisis , Citidina Desaminasa/genética , Daño del ADN , Genes p53 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mutación , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...