Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 112(2): 357-64, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25321194

RESUMEN

BACKGROUND: SET and MYND domain-containing protein 2 (SMYD2) is a lysine methyltransferase for histone H3, p53 and Rb and inhibits their transactivation activities. In this study, we tested whether SMYD2 (1q42) acts as a cancer-promoting factor by being overexpressed in gastric cancer. METHODS: We analysed 7 gastric cancer cell lines and 147 primary tumor samples of gastric cancer, which were curatively resected in our hospital. RESULTS: SET and MYND domain-containing protein 2 was detected in these cell lines (five out of seven cell lines; 71.4%) and primary tumor samples (fifty-six out of one hundred and forty-seven cases; 38.1%). Knockdown of SMYD2 using specific small interfering RNA inhibited proliferation, migration and invasion of SMYD2-overexpressing cells in a TP53 mutation-independent manner. Overexpression of SMYD2 protein correlated with larger tumor size, more aggressive lymphatic invasion, deeper tumor invasion and higher rates of lymph node metastasis and recurrence. Patients with SMYD2-overexpressing tumours had a worse overall rate of survival than those with non-expressing tumours (P=0.0073, log-rank test) in an intensity and proportion score-dependent manner. Moreover, multivariate analysis demonstrated that SMYD2 was independently associated with worse outcome (P=0.0021, hazard ratio 4.25 (1.69-10.7)). CONCLUSIONS: These findings suggest that SMYD2 has a crucial role in tumor cell proliferation by its overexpression and highlight its usefulness as a prognostic factor and potential therapeutic target in gastric cancer.


Asunto(s)
Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Neoplasias Gástricas/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Modelos de Riesgos Proporcionales , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/cirugía , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética
2.
Nat Genet ; 5(2): 151-7, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8252040

RESUMEN

From chromosomal region 17q21.3, where a tumour suppressor gene(s) for breast and ovarian cancers is thought to be present, we have isolated a novel gene from a cosmid clone that revealed somatic rearrangements in two breast cancers. The gene (MDC) encodes a 524-amino acid metalloprotease-like, disintegrin-like and cysteine-rich protein with sequence similarity to members of the snake-venom metalloprotease/disintegrin family and guinea-pig sperm-surface protein PH-30. These proteins have been implicated in cell-cell or cell-extracellular matrix interactions. Rearrangements in both tumours involve multiple exons and disrupt the coding region of the new MDC.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 17 , Reordenamiento Génico , Metaloendopeptidasas/genética , Péptidos/genética , Ponzoñas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , ADN Complementario , Desintegrinas , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
3.
Science ; 270(5239): 1166-70, 1995 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-7502040

RESUMEN

A member of the inwardly rectifying potassium channel family was cloned here. The channel, called BIR (Kir6.2), was expressed in large amounts in rat pancreatic islets and glucose-responsive insulin-secreting cell lines. Coexpression with the sulfonylurea receptor SUR reconstituted an inwardly rectifying potassium conductance of 76 picosiemens that was sensitive to adenosine triphosphate (ATP) (IKATP) and was inhibited by sulfonylureas and activated by diazoxide. The data indicate that these pancreatic beta cell potassium channels are a complex composed of at least two subunits--BIR, a member of the inward rectifier potassium channel family, and SUR, a member of the ATP-binding cassette superfamily. Gene mapping data show that these two potassium channel subunit genes are clustered on human chromosome 11 at position 11p15.1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Canales de Potasio de Rectificación Interna , Canales de Potasio/química , Canales de Potasio/metabolismo , Potasio/metabolismo , Receptores de Droga/química , Adenosina Trifosfato/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Mapeo Cromosómico , Cromosomas Humanos Par 11 , Clonación Molecular , Cricetinae , Diazóxido/farmacología , Humanos , Islotes Pancreáticos/metabolismo , Canales KATP , Ratones , Datos de Secuencia Molecular , Canales de Potasio/genética , Ratas , Receptores de Droga/metabolismo , Rubidio/metabolismo , Compuestos de Sulfonilurea/farmacología , Receptores de Sulfonilureas
4.
Oncogene ; 26(8): 1178-87, 2007 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-16924234

RESUMEN

Anaplastic thyroid cancer (ATC) is one of the most lethal of all human tumors, but cytogenetic information concerning ATC is extremely limited. Using our in-house array-based comparative genomic hybridization and 14 ATC cell lines with further fluorescence in situ hybridization analysis, we demonstrated amplification of the DUSP26 gene, known by another report as MAP kinase phosphatase-8. DUSP26 was overexpressed in ATC cell lines and primary ATC tumor samples. When overexpressed, either exogenously or endogenously, DUSP26 promoted growth of the ATC cells. DUSP26 encodes a protein containing a dual-specificity phosphatase domain that can dephosphorylate itself. DUSP26 effectively dephosphorylates p38 and has a little effect on extracellular signal-regulated kinase in ATC cells. DUSP26 protein formed a physical complex with p38, and promoted survival of ATC cells by inhibiting p38-mediated apoptosis. Our findings suggest that DUSP26 may act as an oncogene in ATC, and might be a useful diagnostic marker and therapeutic target of this disease.


Asunto(s)
Carcinoma/patología , Amplificación de Genes , Proteínas Tirosina Fosfatasas/genética , Neoplasias de la Tiroides/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Apoptosis/genética , Carcinoma/enzimología , Carcinoma/genética , Caspasa 3/genética , Línea Celular Tumoral , Proliferación Celular , Clonación Molecular , Fosfatasas de Especificidad Dual , Humanos , Hibridación Fluorescente in Situ , Fosfatasas de la Proteína Quinasa Activada por Mitógenos , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , ARN Interferente Pequeño/farmacología , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/genética , Proteínas Quinasas p38 Activadas por Mitógenos/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Oncogene ; 26(8): 1110-21, 2007 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-17146433

RESUMEN

To identify target genes for the hemizygous deletions of chromosome 13 that are recurrently observed in malignant gliomas, we performed genome-wide DNA copy-number analysis using array-based comparative genomic hybridization and gene expression analysis using an oligonucleotide-array. The response gene to complement 32 (RGC32) at 13q14.11 was identified as a deletion target, and its expression was frequently silenced in glioma cell lines compared with normal brain. Levels of RGC32 mRNA tended to decrease toward higher grades of primary astrocytomas, especially in tumors with mutations of p53. Expression of RGC32 mRNA was dramatically increased by exogenous p53 in a p53-mutant glioma cell line, and also by endogenous p53 in response to DNA damage in p53+/+ colon-cancer cells, but not in isogenic p53-/- cells. Chromatin immunoprecipitation and reporter assays demonstrated binding of endogenous p53 protein to the promoter region of the RGC32 gene, implying p53-dependent transcriptional activity. Transiently and stably overexpressed RGC32 suppressed the growth of glioma cells, probably owing to induction of G2/M arrest. Immunocytochemical analysis revealed a concentration of RGC32 protein at the centrosome during mitosis. RGC32 formed a protein complex with polo-like kinase 1 and was phosphorylated in vitro. These observations implied a novel mechanism by which p53 might negatively regulate cell-cycle progression by way of this newly identified transcriptional target. Our results provide the first evidence that RGC32 might be a possible tumor-suppressor for glioma, that it is directly induced by p53, and that it mediates the arrest of mitotic progression.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cromosomas Humanos Par 13/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Proteínas Musculares/genética , Proteínas del Tejido Nervioso/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/metabolismo , División Celular , Inmunoprecipitación de Cromatina , Deleción Cromosómica , Daño del ADN , Fase G2 , Eliminación de Gen , Glioma/química , Humanos , Mitosis , Proteínas Musculares/análisis , Proteínas Musculares/metabolismo , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Elementos de Respuesta , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/metabolismo , Quinasa Tipo Polo 1
6.
Oncogene ; 26(44): 6456-68, 2007 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-17438526

RESUMEN

Epigenetic alterations and the resulting inactivation of tumor suppressor genes often contribute to the development of various cancers. To identify novel candidates that may be silenced by aberrant methylation in esophageal squamous-cell carcinoma (ESCC), we analysed ESCC cell lines by a recently developed method known as bacterial artificial chromosome array-based methylated CpG island amplification (BAMCA), and selected candidates through BAMCA-assisted strategy. In the course of this program, we identified frequent CpG methylation-dependent silencing of the gene encoding cellular retinoic acid binding protein 1 (CRABP1) in our panel of ESCC cell lines. Expression of CRABP1 mRNA was restored in gene-silenced ESCC cells after treatment with 5-aza 2'-deoxycytidine. The DNA methylation status of the CRABP1 CpG island with clear promoter activity correlated inversely with expression of this gene. CpG methylation of CRABP1 was frequently observed in primary ESCC tissues as well. Restoration of CRABP1 expression in ESCC cells lacking the protein reduced cell growth by inducing arrest at G(0)-G(1), whereas knockdown of the gene in cells expressing CRABP1 promoted cell growth. Among 113 primary ESCC tumors, the absence of immunoreactive CRABP1 was significantly associated with de-differentiation of cancer cells and with distant lymph-node metastases in the patients. These results indicate that CRABP1 appears to have a tumor-suppressor function in esophageal epithelium, and its epigenetic silencing may play a pivotal role during esophageal carcinogenesis. Its expression status in biopsies or resected tumors might serve as an index for identifying ESCC patients for whom combined therapeutic modalities would be recommended.


Asunto(s)
Carcinoma de Células Escamosas/genética , Metilación de ADN , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Receptores de Ácido Retinoico/genética , Azacitidina/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Proliferación Celular , Epigénesis Genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Humanos , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Receptores de Ácido Retinoico/metabolismo
7.
Br J Surg ; 95(5): 611-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18311747

RESUMEN

BACKGROUND: Patterns of cancer recurrence hold the key to prognosis after curative resection. This retrospective study aimed to identify a predictor and therapeutic candidate for aggressive recurrence of hepatocellular carcinoma (HCC). METHODS: Primary HCC tissues from 107 patients who had curative resection were analysed. Genome-wide gene expression profiles were investigated using a microarray technique, and clustering analysis was carried out based on the first diagnosis of recurrence according to the Milan criteria. Immunohistochemical expression and array-based comparative genomic hybridization (array-CGH) were also assessed. RESULTS: Microarray analysis revealed overexpression of Aurora kinase B, a chromosome passenger protein kinase, as the most significant predictor of the aggressive recurrence of HCC. Aurora kinase B protein expression was significantly associated with aggressive recurrence (P < 0.001) and prognosis (P < 0.001). Multivariable analysis identified Aurora kinase B as the only independent predictor of aggressive recurrence of HCC (P = 0.031). Array-CGH analysis showed that genomic instability was closely related to Aurora kinase B expression (P = 0.011). CONCLUSION: Aurora kinase B is an effective predictor of aggressive HCC recurrence, in relation to the genomic instability. It might be worth considering as a molecular target for the adjuvant therapy of HCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/cirugía , Hepatectomía/mortalidad , Neoplasias Hepáticas/cirugía , Recurrencia Local de Neoplasia/diagnóstico , Proteínas Serina-Treonina Quinasas/metabolismo , Aurora Quinasa B , Aurora Quinasas , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/mortalidad , Supervivencia sin Enfermedad , Expresión Génica , Inestabilidad Genómica , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/mortalidad , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , ARN Neoplásico/análisis , Estudios Retrospectivos
8.
Int J Oral Maxillofac Surg ; 37(11): 1047-53, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18621506

RESUMEN

Cellular inhibitor-of-apoptosis protein 1 (cIAP-1) is a member of the inhibitor-of- apoptosis protein family, which predominantly regulates apoptosis. It has been suggested that expression of cIAP-1 correlates with certain clinicopathological features. The possible significance of cIAP-1 expression in cervical lymph node metastasis of tongue squamous cell carcinoma (SCC) is investigated. Seventy-five tongue SCCs were analyzed by immunohistochemistry. cIAP-1 immunoreactivity patterns were nuclear in 38 (51%), cytoplasmic in 47 (63%), and concurrent in 37 (49%) cases. Nuclear, cytoplasmic and concurrent cIAP-1 immunoreactions were significantly correlated with lymph node metastasis in tongue SCCs (P=0.0011, 0.0012, and 0.0006, respectively). The cleaved caspase-3, which is a marker of tumor apoptosis, and Ki-67 index, which is a marker of tumor proliferation, were immunohistochemically examined in 21 tongue SCCs with concurrent nuclear and cytoplasmic cIAP-1 expression and with metastasis, and in 23 tongue SCCs without concurrent nuclear and cytoplasmic cIAP-1 expression and without metastasis. Concurrent cIAP-1 expression was inversely correlated with caspase-3 (P=0.0066), but was positively correlated with Ki-67 expression (P=0.0028). The mode of invasion was associated with lymph node metastasis (P=0.014) and differentiation (P=0.013), but was not correlated with cIAP-1 expression. There was no statistically significant correlation between nuclear or cytoplasmic cIAP-1 expression and the clinicopathological factors of gender, age, clinical stage or differentiation. These results suggest that both patterns of cIAP-1 are useful markers for predicting cervical lymph node metastasis in tongue SCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Metástasis Linfática/patología , Neoplasias de la Lengua/secundario , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/metabolismo , Caspasa 3/metabolismo , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Distribución Tisular , Neoplasias de la Lengua/metabolismo , Adulto Joven
9.
Oncogene ; 25(49): 6554-62, 2006 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-16715143

RESUMEN

Homozygous loss in the genomic sequence, a mechanism for inactivating tumor-suppressor genes (TSGs) in cancer, has been used as a tag for the identification of novel TSGs, and array-based comparative genomic hybridization (array-CGH) has a great potential for high-throughput identification of this change. We identified a homozygous loss of the very-low-density lipoprotein receptor (VLDLR) gene (9p24.2) from genome-wide screening for copy-number alterations in 32 gastric cancer (GC) cell lines using array-CGH. Although previous reports demonstrated mRNA or protein expression of VLDLR in various cancers including GC, the association between genomic losses or epigenetic silencing of this gene and carcinogenesis has never been reported before. Homozygous deletion of VLDLR was also seen in primary GCs, albeit infrequently, and about half of GC cell lines showed lost or reduced VLDLR expression. The VLDLR expression was restored in gene-silenced GC cells after treatment with 5-aza 2'-deoxycytidine. According to methylation analyses, hypermethylation of the VLDLR promoter region, which all of GC lines without its expression showed, occurred in some primary GCs. Restoration of VLDLR type I expression in GC cells reduced colony formation. These results suggest that not only the expression of VLDLR but also genetic or epigenetic silencing of this gene may contribute to tumor formation and be involved in gastric carcinogenesis.


Asunto(s)
Carcinoma/genética , Epigénesis Genética , Eliminación de Gen , Silenciador del Gen , Receptores de LDL/genética , Neoplasias Gástricas/genética , Biopsia , Carcinoma/metabolismo , Carcinoma/cirugía , Proliferación Celular , Transformación Celular Neoplásica , Cromosomas Humanos Par 9 , Islas de CpG , Metilación de ADN , Homocigoto , Humanos , Hibridación de Ácido Nucleico/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Regiones Promotoras Genéticas , Receptores de LDL/metabolismo , Neoplasias Gástricas/cirugía , Células Tumorales Cultivadas
10.
Mol Cell Biol ; 17(1): 469-81, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8972228

RESUMEN

Heat shock transcription factors (HSFs) mediate the inducible transcriptional response of genes that encode heat shock proteins and molecular chaperones. In vertebrates, three related HSF genes (HSF1 to -3) and the respective gene products (HSFs) have been characterized. We report the cloning and characterization of human HSF4 (hHSF4), a novel member of the hHSF family that shares properties with other members of the HSF family yet appears to be functionally distinct. hHSF4 lacks the carboxyl-terminal hydrophobic repeat which is shared among all vertebrate HSFs and has been suggested to be involved in the negative regulation of DNA binding activity. hHSF4 is preferentially expressed in the human heart, brain, skeletal muscle, and pancreas. Transient transfection of hHSF4 in HeLa cells, which do not express hHSF4, results in a constitutively active DNA binding trimer which, unlike other members of the HSF family, lacks the properties of a transcriptional activator. Constitutive overexpression of hHSF4 in HeLa cells results in reduced expression of the endogenous hsp70, hsp90, and hsp27 genes. hHSF4 represents a novel hHSF that exhibits tissue-specific expression and functions to repress the expression of genes encoding heat shock proteins and molecular chaperones.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Factores de Transcripción/genética , Secuencia de Aminoácidos , Secuencia de Bases , Núcleo Celular/química , Mapeo Cromosómico , Cromosomas Humanos Par 16/genética , Clonación Molecular , ADN Complementario/genética , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Genes/genética , Células HeLa , Factores de Transcripción del Choque Térmico , Humanos , Datos de Secuencia Molecular , Especificidad de Órganos , ARN Mensajero/análisis , Proteínas Recombinantes de Fusión , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Transactivadores , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo
11.
Mol Cell Biol ; 20(19): 7319-31, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10982849

RESUMEN

The expression of CLC-K1 and CLC-K2, two kidney-specific CLC chloride channels, is transcriptionally regulated on a tissue-specific basis. Previous studies have shown that a GA element near their transcriptional start sites is important for basal and cell-specific activities of the CLC-K1 and CLC-K2 gene promoters. To identify the GA-binding proteins, the human kidney cDNA library was screened by a yeast one-hybrid system. A novel member of the Cys2-His2 zinc finger gene designated KKLF (for "kidney-enriched Krüppel-like factor") and the previously isolated MAZ (for "myc-associated zinc finger protein") were cloned. KKLF was found to be abundantly expressed in the liver, kidneys, heart, and skeletal muscle, and immunohistochemistry revealed the nuclear localization of KKLF protein in interstitial cells in heart and skeletal muscle, stellate cells, and fibroblasts in the liver. In the kidneys, KKLF protein was localized in interstitial cells, mesangial cells, and nephron segments, where CLC-K1 and CLC-K2 were not expressed. A gel mobility shift assay revealed sequence-specific binding of recombinant KKLF and MAZ proteins to the CLC-K1 GA element, and the fine-mutation assay clarified that the consensus sequence for the KKLF binding site was GGGGNGGNG. In a transient-transfection experiment, MAZ had a strong activating effect on transcription of the CLC-K1-luciferase reporter gene. On the other hand, KKLF coexpression with MAZ appeared to block the activating effect of MAZ. These results suggest that a novel set of zinc finger proteins may help regulate the strict tissue- and nephron segment-specific expression of the CLC-K1 and CLC-K2 channel genes through their GA cis element.


Asunto(s)
Proteínas de Transporte de Anión , Proteínas Portadoras/fisiología , Canales de Cloruro/genética , Regulación de la Expresión Génica , Proteínas de la Membrana , Proteínas Nucleares , Regiones Promotoras Genéticas , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Dedos de Zinc/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Canales de Cloruro/metabolismo , Clonación Molecular , Colágeno/biosíntesis , Colágeno/genética , ADN Complementario/genética , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Fibroblastos/metabolismo , Genes , Genes Reporteros , Humanos , Factores de Transcripción de Tipo Kruppel , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Nefritis Intersticial/metabolismo , Nefronas/metabolismo , Especificidad de Órganos , Unión Proteica , Ratas , Proteínas Recombinantes de Fusión/fisiología , Secuencias Reguladoras de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Activación Transcripcional , Transfección
12.
Oncogene ; 36(30): 4267-4276, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28346428

RESUMEN

L-asparaginase has been used for more than three decades in acute lymphoblastic leukemia (ALL) patients and remains an essential drug in the treatment of ALL. Poor response to L-asparaginase is associated with increased risk of therapeutic failure in ALL. However, both the metabolic perturbation and molecular context of L-asparaginase-treated ALL cells has not been fully elucidated. Here we identify that treatment with L-asparaginase results in metabolic shutdown via the reduction of both glycolysis and oxidative phosphorylation, accompanied by mitochondrial damage and activation of autophagy. The autophagy is involved in reducing reactive oxygen species (ROS) level by eliminating injured mitochondria. Inhibition of autophagy enhances L-asparaginase-induced cytotoxicity and overcomes the acquired resistance to L-asparaginase in ALL cells. The ROS-p53-positive feedback loop is an essential mechanism of this synergistic cytotoxicity. Thus, our findings provide the rationale for the future development of combined treatment of L-asparaginase and anti-autophagy drug in ALL patients.


Asunto(s)
Antineoplásicos/farmacología , Asparaginasa/farmacología , Autofagia/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Estrés Fisiológico/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Cancer Res ; 59(3): 511-5, 1999 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9973190

RESUMEN

We used comparative genomic hybridization to study malignant fibrous histiocytomas (MFHs) from 19 patients to detect changes in the copy number of DNA sequences, along entire chromosomes. Together with losses and gains in various chromosomal regions, distinct high-level amplifications were found at six loci (4q12-21, 8p21-pter, 8q24.1-qter, 9q12-13, 12p11.2-pter, and 15q11.2-15), suggesting that those regions may contain unknown (proto) oncogenes. We focused on the 8p amplicon, where detailed characterization allowed us to determine that the minimal common amplified region lay between markers D8S1819 and D8S550 at 8p23.1. A novel gene designated MASL1 (MFH-amplified sequences with leucine-rich tandem repeats 1) was isolated from within this narrowly defined region. Expression of the MASL1 gene was enhanced significantly in MFH tumors bearing the 8p amplicon. The primary structure of its deduced product revealed an ATP/GTP-binding site, three leucine zipper domains, and a leucine-rich tandem repeat, all of which are important structural or functional elements for interactions among proteins related to the cell cycle. These features suggest that overexpression of MASL1 might well be oncogenic with respect to MFH.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cromosomas Humanos Par 8 , Proteínas de Unión al ADN/genética , Histiocitoma Fibroso Benigno/genética , Proteínas Oncogénicas , Oncogenes , Adulto , Anciano , Secuencia de Aminoácidos , Southern Blotting , Proteínas de Ciclo Celular/biosíntesis , Aberraciones Cromosómicas , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , ADN de Neoplasias/metabolismo , Proteínas de Unión al ADN/biosíntesis , Femenino , Amplificación de Genes , Histiocitoma Fibroso Benigno/metabolismo , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Cancer Res ; 56(7): 1629-34, 1996 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8603412

RESUMEN

We previously reported definition of a region containing a putative tumor suppressor gene for esophageal squamous cell carcinoma within an approximately 4-cM genomic segment at 9q31-q32. We have investigated this region further using six new microsatellite markers isolated from yeast artificial chromosome clones covering the deleted region and have narrowly defined the commonly deleted region to a segment between two loci, KM9.1 and D9S177. On the basis of the contig map of cosmid and yeast artificial chromosome clones, we estimate the physical size of the region of interest to be about 200 kb. Because the distal 9q region also has been implicated as the site of a tumor suppressor gene(s) related to squamous cell carcinomas of other tissues, our map provides useful information for attempts to identify a common gene for carcinomas of this cell type.


Asunto(s)
Carcinoma de Células Escamosas/genética , Deleción Cromosómica , Cromosomas Humanos Par 9 , Neoplasias Esofágicas/genética , Genes Supresores de Tumor , Secuencia de Bases , Mapeo Cromosómico , Humanos , Datos de Secuencia Molecular
15.
Cancer Res ; 61(18): 6629-34, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11559525

RESUMEN

Amplification of chromosomal DNA is thought to be one of the mechanisms that activate cancer-related genes in tumors. In a recent study, we identified high copy-number amplification at 11q21-q23 in cell lines derived from esophageal squamous cell carcinomas (ESCs) using comparative genomic hybridization. Because 11q21-q23 amplification has been reported in tumors of various other types as well, gene(s) associated with tumor progression may lie within this chromosomal region. To identify the most likely target(s) for amplification at 11q21-q23, we determined the extent of the amplicon by fluorescence in situ hybridization and then analyzed ESC cell lines for expression levels of 11 known genes and one uncharacterized transcript present within the 1.8-Mb commonly amplified region. Only cIAP1, a member of the IAP (antiapoptotic) gene family, was consistently overexpressed in cell lines that showed amplification. Additionally, the cIAP1 protein was overexpressed in the primary tumors from which those cell lines had been established. The ESC cell lines with cIAP1 amplification were resistant to apoptosis induced by chemotherapeutic reagents. An increase in cIAP1 copy number was also detected in 4 of 42 (9.5%) primary ESC tumors that were not related to the cell lines examined. Because inhibition of apoptosis seems to be an important feature of carcinogenesis, cIAP1 is likely to be a target for 11q21-23 amplification and may be involved in the progression of ESC, as well as other malignancies.


Asunto(s)
Carcinoma de Células Escamosas/genética , Cromosomas Humanos Par 11/genética , Neoplasias Esofágicas/genética , Proteínas/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Mapeo Cromosómico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Amplificación de Genes , Humanos , Hibridación Fluorescente in Situ , Proteínas Inhibidoras de la Apoptosis , Biosíntesis de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas
16.
Cancer Res ; 60(17): 4735-9, 2000 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10987278

RESUMEN

In a recent study, we identified frequent amplification of DNA copy number at chromosome 9p23-24 in cell lines derived from esophageal squamous cell carcinomas (ESCs), using comparative genomic hybridization. Because amplified regions often harbor oncogenes and/or other tumor-associated genes, and because 9p23-24 amplification had been reported in various other types of cancers, we used fluorescence in situ hybridization and Southern blot analysis to map the 9p23-24 amplicon. We then screened target genes/transcripts present within this amplicon by Northern blotting. With this strategy, we successfully cloned a novel gene, designated gene amplified in squamous cell carcinoma 1 (GASC1), that was amplified and overexpressed in several ESC cell lines. The deduced amino acid sequence of GASC1 contains two PHD-finger motifs and a PX domain. PHD-finger motifs are found in nuclear proteins that participate in chromatin-mediated transcriptional regulation and are present in a number of proto-oncogenes. Our findings suggest that overexpressed GASC1 may play an important role in the development and/or progression of various types of cancer including ESC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Cromosomas Humanos Par 9/genética , Neoplasias Esofágicas/genética , Proteínas de Neoplasias , Oncogenes/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Southern Blotting , Clonación Molecular , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , Amplificación de Genes , Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Histona Demetilasas con Dominio de Jumonji , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico , ARN Mensajero/genética , Células Tumorales Cultivadas
17.
Cancer Res ; 53(14): 3382-5, 1993 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8100738

RESUMEN

Using 11 restriction fragment length polymorphism markers, we examined loss of heterozygosity on the long arm of chromosome 17, where one or more genes responsible for hereditary breast and ovarian cancers may be present, in sporadic forms of 94 ovarian and 246 breast cancers. Loss of heterozygosity was observed in 33 of 84 (39.3%) ovarian and in 88 of 214 (41.1%) breast cancers that were informative with at least one marker. Detailed deletion mapping of chromosome 17q in these cancers identified two distinct, commonly deleted regions. One was located between 17q12 and 17q21.3 and the other between 17q25.1 and 17q25.3. In breast cancers, the proximal commonly deleted region was between two loci defined by markers CI17-701 and CI17-730 at 17q21.3, which are 2.4 cM apart. This segment overlaps the region that includes the putative gene for hereditary breast and ovarian carcinomas. The results suggest that at least two tumor suppressor genes associated with sporadic ovarian and breast cancers are present on chromosome 17q and that one of them may be the same gene that is responsible for the hereditary form.


Asunto(s)
Neoplasias de la Mama/genética , Deleción Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 17 , Neoplasias Ováricas/genética , Adenocarcinoma/genética , Adenocarcinoma Mucinoso/genética , Femenino , Marcadores Genéticos , Humanos , Menopausia , Polimorfismo de Longitud del Fragmento de Restricción
18.
Cancer Res ; 52(21): 6083-7, 1992 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-1394235

RESUMEN

We previously demonstrated that the 11q23 breakpoint region, designated the RCK locus, of the RC-K8 B-lymphoma cell line with t(11;14)(q23;q32) is centromeric to PBGD, while breakpoints of infantile leukemia cell lines with t(11;19)(q23;p13) are detectable by pulsed-field gel electrophoresis with the CD3D probe. In the present study, using a probe within 1.0 kilobase of the t(11;14) breakpoint, we isolated a partial complementary DNA clone for the putative RCK gene, which detects a 7.5-kilobase mRNA. Sequence analysis predicted a novel protein of 472 amino acids which demonstrated sequence homology to a translation initiation factor/helicase family. We also isolated a phage clone from the CD3D/G yeast artificial chromosome clone (yB22B2) which detects 11- and 12-kilobase mRNAs, most likely for the MLL/ALL-1 gene associated t(4;11)(q21;q23) and t(11;19)(q23;p13) translocations. By pulsed-field gel electrophoresis after NotI digestion, this recombinant clone is on a 96-kilobase fragment, while RCK and PBGD probes are on a more telomeric 690-kilobase NotI fragment. These results, altogether, suggested that two different genes, RCK and MLL/ALL-1, are associated with 11q23 translocation of hematopoietic tumors.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 14 , Cromosomas Humanos Par 19 , Cromosomas Humanos Par 4 , Leucemia Bifenotípica Aguda/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Translocación Genética/genética , Secuencia de Aminoácidos , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Células Tumorales Cultivadas
19.
Oncogene ; 35(40): 5304-5316, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27041563

RESUMEN

Metastasis is a critical factor contributing to poor prognosis in cancer, but the underlying mechanisms of metastasis are still poorly understood. We established a highly metastatic cell subline (HOC313-LM) derived from an oral squamous cell carcinoma cell line (HOC313) for uncovering the mechanisms of metastasis, and identified deoxyhypusine synthase (DHPS) as a metastasis-associated gene within the specific amplification at 19p13.2-p13.13 in HOC313-LM. DHPS-mediated hypusine-modification of eukaryotic translation factor 5A facilitated the translation of RhoA, resulting in the activation of the RhoA signaling pathway and leading to not only increased cell motility, invasion and metastasis of cancer cells in vitro, but also increased tumor growth in vivo. Moreover, the use of N1-Guanyl-1,7-diaminoheptane, a DHPS inhibitor, resulted in a significant decrease in tumor formation in vivo. In patients with esophageal squamous cell carcinoma (ESCC), overexpression of DHPS in ESCC tumors was significantly associated with worse recurrence-free survival, and correlated with distant metastasis. The elucidation of these molecular mechanisms within the hypusine cascade suggests opportunities for novel therapeutic targets in SCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias Esofágicas/tratamiento farmacológico , Lisina/análogos & derivados , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Proteína de Unión al GTP rhoA/genética , Adulto , Anciano , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Diaminas/administración & dosificación , Progresión de la Enfermedad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisina/biosíntesis , Lisina/genética , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
20.
Oncogene ; 10(9): 1731-8, 1995 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-7753550

RESUMEN

Previously, we cloned two gene fragments encoding novel protein tyrosine phosphatases, termed PTP-U1 and PTP-U2. Here, we report the full-length sequence, expression, and chromosomal localization of the PTP-U2 gene. The cDNA for PTP-U2, which was obtained from a human normal kidney library, predicts a protein of 1216 amino acids, -140 kDa, that contains a single transmembrane domain and a single intracellular catalytic domain. The extracellular domain of PTP-U2 contains 14 putative N-glycosylation sites and eight repeats of fibronectin type III-like motif. These data suggest that PTP-U2 is structurally similar to HPTP beta and DPTP10D, which have been reported previously. Northern blot analysis revealed that there were two different transcripts for PTP-U2. In kidney and brain, gene expression of PTP-U2 was detected as a 5.4 kb mRNA and in lung and placenta as 3.5 kb. The 3.5 kb transcript was also detected in human leukemia cell lines (eg., U937). Interestingly, its gene expression was enhanced by various differentiation-inducing agents, such as phorbol ester, dihydroxy vitamin D3, retinoic acid, and dimethyl sulfoxide. The bacterially expressed PTP-U2 fusion protein exhibited intrinsic tyrosine phosphatase activity. The PTP-U2 gene was assigned to chromosome 12p13.2-p13.3.


Asunto(s)
Proteínas Tirosina Fosfatasas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Cromosomas Humanos Par 12 , Clonación Molecular , Expresión Génica/efectos de los fármacos , Humanos , Hibridación Fluorescente in Situ , Riñón/enzimología , Datos de Secuencia Molecular , ARN Mensajero/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Acetato de Tetradecanoilforbol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA