Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
ACS Cent Sci ; 8(1): 57-66, 2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35106373

RESUMEN

Optical control has enabled functional modulation in cell culture with unparalleled spatiotemporal resolution. However, current tools for in vivo manipulation are scarce. Here, we design and implement a genuine on-off optochemical probe capable of achieving hematopoietic control in zebrafish. Our photopharmacological approach first developed conformationally strained visible light photoswitches (CS-VIPs) as inhibitors of the histone methyltransferase MLL1 (KMT2A). In blood homeostasis MLL1 plays a crucial yet controversial role. CS-VIP 8 optimally fulfils the requirements of a true bistable functional system in vivo under visible-light irradiation, and with unprecedented stability. These properties are exemplified via hematopoiesis photoinhibition with a single isomer in zebrafish. The present interdisciplinary study uncovers the mechanism of action of CS-VIPs. Upon WDR5 binding, CS-VIP 8 causes MLL1 release with concomitant allosteric rearrangements in the WDR5/RbBP5 interface. Since our tool provides on-demand reversible control without genetic intervention or continuous irradiation, it will foster hematopathology and epigenetic investigations. Furthermore, our workflow will enable exquisite photocontrol over other targets inhibited by macrocycles.

2.
PLoS One ; 7(9): e44951, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028699

RESUMEN

The human cysteine dioxygenase 1 (CDO1) gene is a non-heme structured, iron-containing metalloenzyme involved in the conversion of cysteine to cysteine sulfinate, and plays a key role in taurine biosynthesis. In our search for novel methylated gene promoters, we have analyzed differential RNA expression profiles of colorectal cancer (CRC) cell lines with or without treatment of 5-aza-2'-deoxycytidine. Among the genes identified, the CDO1 promoter was found to be differentially methylated in primary CRC tissues with high frequency compared to normal colon tissues. In addition, a statistically significant difference in the frequency of CDO1 promoter methylation was observed between primary normal and tumor tissues derived from breast, esophagus, lung, bladder and stomach. Downregulation of CDO1 mRNA and protein levels were observed in cancer cell lines and tumors derived from these tissue types. Expression of CDO1 was tightly controlled by promoter methylation, suggesting that promoter methylation and silencing of CDO1 may be a common event in human carcinogenesis. Moreover, forced expression of full-length CDO1 in human cancer cells markedly decreased the tumor cell growth in an in vitro cell culture and/or an in vivo mouse model, whereas knockdown of CDO1 increased cell growth in culture. Our data implicate CDO1 as a novel tumor suppressor gene and a potentially valuable molecular marker for human cancer.


Asunto(s)
Cisteína-Dioxigenasa/genética , Silenciador del Gen , Genes Supresores de Tumor , Neoplasias/genética , Regiones Promotoras Genéticas/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisteína-Dioxigenasa/deficiencia , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/efectos de los fármacos
3.
PLoS One ; 5(2): e9003, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20140245

RESUMEN

Aerobic glycolysis and mitochondrial dysfunction are common features of aggressive cancer growth. We observed promoter methylation and loss of expression in neurofilament heavy polypeptide (NEFH) in a significant proportion of primary esophageal squamous cell carcinoma (ESCC) samples that were of a high tumor grade and advanced stage. RNA interference-mediated knockdown of NEFH accelerated ESCC cell growth in culture and increased tumorigenicity in vivo, whereas forced expression of NEFH significantly inhibited cell growth and colony formation. Loss of NEFH caused up-regulation of pyruvate kinase-M2 type and down-regulation of pyruvate dehydrogenase, via activation of the Akt/beta-catenin pathway, resulting in enhanced aerobic glycolysis and mitochondrial dysfunction. The acceleration of glycolysis and mitochondrial dysfunction in NEFH-knockdown cells was suppressed in the absence of beta-catenin expression, and was decreased by the treatment of 2-Deoxyglucose, a glycolytic inhibitor, or API-2, an Akt inhibitor. Loss of NEFH activates the Akt/beta-catenin pathway and increases glycolysis and mitochondrial dysfunction. Cancer cells with methylated NEFH can be targeted for destruction with specific inhibitors of deregulated downstream pathways.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Proteínas de Neurofilamentos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , beta Catenina/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Clorpropamida/análogos & derivados , Clorpropamida/farmacología , Metilación de ADN , Desoxiglucosa/farmacología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Citometría de Flujo , Expresión Génica , Glucólisis/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Proteínas de Neurofilamentos/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Trasplante Heterólogo , Carga Tumoral , beta Catenina/genética
4.
Exp Mol Pathol ; 87(1): 36-41, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19341726

RESUMEN

Expression of STAT3/pSTAT3 in colorectal cancer (CRC) patients of Indian origin was studied to assess its significance in early detection and apoptosis regulation. Colorectal tissues with malignant lesions were STAT3/pSTAT3 positive in 66% of the cases and among these positive cases, well differentiated, moderately differentiated and poorly differentiated cancers were 86%, 60% and 0% respectively. All CRC specimens studied were immunoreactive with anti-carcinoembryonic antigen antibody. Cells purified from CRC tissues exhibited greater STAT3/pSTAT3 reactivity than peripheral blood mononuclear cells (PBMC) from healthy individuals, which served as control. apoptotic index (AI) was comparatively low in tissue specimens with STAT3/pSTAT3 expression. CRC cells with a comparatively less number of apoptotic cells, expressed a minimum number of Caspase-3 positive cells (4.73%), in comparison to healthy-PBMC (12.63%). CRC cells with high STAT3/pSTAT3 staining had cells with greater percentage of Bcl2 reactivity (23.05%), but less positivity with Caspase3 antibody (2.05%). Overall data suggests that CRC population was STAT3/pSTAT3 immunoreactive in a stage specific manner and STAT3 protects cancerous colorectal epithelial cells from apoptosis. Bcl-2, Cyclin D1 and Caspase-3 control the activity of apoptosis regulator, STAT3.


Asunto(s)
Apoptosis/fisiología , Neoplasias Colorrectales , Factor de Transcripción STAT3/metabolismo , Adulto , Antígeno Carcinoembrionario/metabolismo , Caspasa 3/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Transcripción STAT3/genética , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Adulto Joven
5.
Cell Cycle ; 7(18): 2846-55, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18769144

RESUMEN

We previously found that the pro-apoptotic DNA damaging agent, cisplatin, mediated the proteasome-dependent degradation of Delta Np63 alpha associated with its increased phosphorylated status. Since Delta Np63 alpha usually plays an opposite role to p53 and TAp63 in human cancers, we tested the notion that phosphorylation events induced by DNA damage would affect the protein degradation of Delta Np63 alpha in HNSCC cells upon cisplatin exposure. We found that Delta Np63 alpha is phosphorylated in the time-dependent fashion at the following positions: S385, T397 and S466, which were surrounded by recognition motifs for ATM, CDK2 and p70s6K kinases, respectively. We showed that chemical agents or siRNA inhibiting the activity of ATM, CDK2 and p70s6K kinases blocked degradation of Delta Np63 alpha in HNSCC cells after cisplatin exposure. Site-specific mutagenesis of Delta Np63 alpha residues targeted for phosphorylation by ATM, CDK2 or p70s6k led to dramatic modulation of Delta Np63 alpha degradation. Finally, we demonstrated that the Delta Np63 alpha protein is a target for direct in vitro phosphorylation by ATM, CDK2 or p70s6K. Our results implicate specific kinases, and target phosphorylation sites in the degradation of Delta Np63 alpha following DNA damage.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Neoplasias de Cabeza y Cuello/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Cisplatino/farmacología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Datos de Secuencia Molecular , Mutación/genética , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Fosfotreonina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Factores de Tiempo , Transactivadores/química , Factores de Transcripción , Proteínas Supresoras de Tumor/química
7.
Clin Cancer Res ; 14(12): 3754-60, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559593

RESUMEN

PURPOSE: Prostate cancer is a major cause of cancer death among men and the development of new biomarkers is important to augment current detection approaches. EXPERIMENTAL DESIGN: We identified hypermethylation of the ssDNA-binding protein 2 (SSBP2) promoter as a potential DNA marker for human prostate cancer based on previous bioinformatics results and pharmacologic unmasking microarray. We then did quantitative methylation-specific PCR in primary prostate cancer tissues to confirm hypermethylation of the SSBP2 promoter, and analyzed its correlation with clinicopathologic data. We further examined SSBP2 expression in primary prostate cancer and studied its role in cell growth. RESULTS: Quantitative methylation-specific PCR results showed that the SSBP2 promoter was hypermethylated in 54 of 88 (61.4%) primary prostate cancers versus 0 of 23 (0%) in benign prostatic hyperplasia using a cutoff value of 120. Furthermore, we found that expression of SSBP2 was down-regulated in primary prostate cancers and cancer cell lines. Hypermethylation of the SSBP2 promoter and its expression were closely associated with higher stages of prostate cancer. Reactivation of SSBP2 expression by the demethylating agent 5-aza-2'-deoxycytidine in prostate cancer cell lines confirmed epigenetic inactivation as one major mechanism of SSBP2 regulation. Moreover, forced expression of SSBP2 inhibited prostate cancer cell proliferation in the colony formation assay and caused cell cycle arrest. CONCLUSION: SSBP2 inhibits prostate cancer cell proliferation and seems to represent a novel prostate cancer-specific DNA marker, especially in high stages of human prostate cancer.


Asunto(s)
Adenocarcinoma/genética , Proliferación Celular , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Neoplasias de la Próstata/genética , Adenocarcinoma/patología , Anciano , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Análisis Mutacional de ADN , Decitabina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Estadificación de Neoplasias , Hiperplasia Prostática/genética , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas
8.
Mod Pathol ; 21(8): 979-91, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18487995

RESUMEN

The glycosylphosphatidylinositol transamidase complex (GPIT) consists of five subunits: PIG-U, PIG-T, GPAA1, PIG-S and GPI8, and is important in attaching GPI anchors to target proteins. On the basis of our previous reports incriminating PIG-U as an oncogene in bladder cancer and PIG-T and GPAA1 as oncogenes in breast cancer, we evaluated the expression pattern of the GPIT subunits in 19 different human cancers at both mRNA and protein levels. In general, our results demonstrate a more frequent expression of GPIT subunits in cancers than in normal. Among the 19 anatomic sites compared; breast, ovary and uterus showed consistent evidence of overexpression of specific GPIT subunits. There was also overexpression of PIG-U and GPI8 in lymphoma. In addition, non-small cell lung carcinoma showed significant overexpression of the GPIT subunits as compared to small cell lung carcinoma and normal lung tissue. Also, deregulation of specific GPIT subunits was seen in various other cancers. Forced overexpression of two GPIT subunits; PIG-S and GPI8 alone or in combination induced increased proliferation and invasion of breast cancer cells. Collectively, our study defines a trend involving the deregulated expression and the functional contribution of the GPIT subunits in various cancers with potential implications in diagnosis, prognosis and therapeutic intervention.


Asunto(s)
Aciltransferasas/genética , Aciltransferasas/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias/enzimología , Neoplasias/genética , Proliferación Celular , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Invasividad Neoplásica , Neoplasias/patología
9.
Mol Cancer Res ; 6(5): 851-61, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18505929

RESUMEN

PELP1 (proline-rich, glutamic acid-rich, and leucine-rich protein-1) is a potential proto-oncogene that functions as a coregulator of estrogen receptor (ER), and its expression is deregulated during breast cancer progression. Emerging evidence suggests growth factor signaling crosstalk with ER as one possible mechanism by which breast tumors acquire resistance to therapy. In this study, we examined mechanisms by which growth factors modulate PELP1 functions, leading to activation of ER. Using in vivo labeling assays, we have found that growth factors promote phosphorylation of PELP1. Utilizing a panel of substrate-specific phosphorylated antibodies, we discovered that growth factor stimulation promotes phosphorylation of PELP1 that is recognized by a protein kinase A (PKA) substrate-specific antibody. Accordingly, growth factor-mediated PELP1 phosphorylation was effectively blocked by PKA-specific inhibitor H89. Utilizing purified PKA enzyme and in vitro kinase assays, we obtained evidence of direct PELP1 phosphorylation by PKA. Using deletion and mutational analysis, we identified PELP1 domains that are phosphorylated by PKA. Interestingly, site-directed mutagenesis of the putative PKA site in PELP1 compromised growth factor-induced activation and subnuclear localization of PELP1 and also affected PELP1-mediated transactivation function. Utilizing MCF-7 cells expressing a PELP1 mutant that cannot be phosphorylated by PKA, we provide mechanistic insights by which growth factor signaling regulates ER transactivation in a PELP1-dependent manner. Collectively, these findings suggest that growth factor signals promote phosphorylation of ER coactivator PELP1 via PKA pathway, and such modification may have functional implications in breast tumors with deregulated growth factor signaling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptores de Estrógenos/metabolismo , Transactivadores/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Proteínas Co-Represoras , Eliminación de Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isoquinolinas/farmacología , Mutagénesis Sitio-Dirigida , Fosforilación , Proto-Oncogenes Mas , Transducción de Señal , Sulfonamidas/farmacología , Factores de Transcripción
10.
Cell Cycle ; 7(9): 1231-7, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18418053

RESUMEN

DeltaNp63alpha, the dominant negative isoform of the p63 family is an essential survival factor in head and neck squamous cell carcinoma. This isoform has been shown to be down regulated in response to several DNA damaging agents, including cisplatin. But little is understood about the post-translational protein stability of DeltaNp63alpha. In this present study we demonstrate for the first time that DeltaNp63alpha physically interacts with U-box-type E4 ubiquitin ligase UFD2a. UFD2a stabilizes DeltaNp63alpha, and ubiquitylation of DeltaNp63alpha is attenuated by UFD2a both in the presence and absence of cisplatin. Ectopic expression of UFD2a increased the half-life of DeltaNp63alpha in association with a significant enhancement of the repressive transcriptional activity of DeltaNp63alpha. Downregulation of endogenous UFD2a by RNAi resulted in degradation of DeltaNp63alpha. Taken together, our current study provides an insight onto the regulation of DeltaNp63alpha protein levels in response to cisplatin and also suggests that UFD2a might play an important role in the regulation of cisplatin mediated cell death mediated by p63.


Asunto(s)
Carcinoma/metabolismo , Cisplatino/farmacología , Neoplasias de Cabeza y Cuello/metabolismo , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Humanos , Mutación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Interferencia de ARN/fisiología , Elementos Reguladores de la Transcripción/efectos de los fármacos , Elementos Reguladores de la Transcripción/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/efectos de los fármacos , Transactivadores/genética , Factores de Transcripción , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitina-Proteína Ligasas , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
11.
Biochem Biophys Res Commun ; 370(1): 38-43, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18346456

RESUMEN

The pathogenesis of breast cancer involves multiple genetic and epigenetic events. In this study, we report an epigenetic alteration of DFNA5 in human breast cancer. DFNA5 gene was silenced in breast cancer cell lines that were methylated in the DFNA5 promoter, and restored by treatment with the demethylating agent, 5-aza-dC, and gene knock-down of DFNA5 increased cellular invasiveness in vitro. The mRNA expression of DFNA5 in breast cancer tissues was down-regulated as compared to normal tissues. Moreover, the DFNA5 promoter was found to be methylated in primary tumor tissues with high frequency (53%, 18/34). Quantitative methylation-specific PCR of DFNA5 clearly discriminated primary breast cancer tissues from normal breast tissues (15.3%, 2/13). Moreover, methylation status of DFNA5 was correlated with lymph node metastasis in breast cancer patients. Our data implicate DFNA5 promoter methylation as a novel molecular biomarker in human breast cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Metilación de ADN , Epistasis Genética , Regulación Neoplásica de la Expresión Génica , Receptores de Estrógenos/genética , Adulto , Anciano , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Humanos , Metástasis Linfática , Persona de Mediana Edad , Invasividad Neoplásica , ARN Mensajero , Receptores de Estrógenos/antagonistas & inhibidores
12.
Prostate ; 68(4): 418-26, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18196513

RESUMEN

BACKGROUND: DNA methylation has emerged as a promising biomarker for prostate cancer detection. In this report, we screened 36 candidate genes generated by a bioinformatic analysis of the human genome, and found that the melanoma cell adhesion molecule (MCAM) was an excellent candidate for cancer-specific methylation in prostate cancer. METHODS: Direct sequencing of bisulfite-treated genomic DNA, conventional methylation-specific PCR (MSP), real-time quantitative methylation-specific PCR, immunohistochemistry, colony formation assay, and statistical analysis. RESULTS: We found that the melanoma cell adhesion molecule (MCAM) gene promoter was specifically methylated in prostate cancer cell lines and primary prostate cancer (PCa) but not in non-neoplastic prostate (BPH) tissues by direct sequencing of bisulfite-treated genomic DNA and conventional methylation-specific PCR (MSP). Further analysis with quantitative MSP showed greater hypermethylation of the MCAM promoter (80%, 70/88) in primary prostate cancer compared to 12.5% (3/24) in BPH. Prostatic intraepithelial neoplasias (PIN), potential precursors of prostate carcinoma, showed an intermediate methylation rate of 23% (7/30). We further observed that MCAM promoter methylation was directly correlated with tumor stage (pT3+pT4) (P = 0.001) and Gleason score (P = 0.018) in primary prostate carcinoma. CONCLUSIONS: Our results suggest that MCAM promoter hypermethylation deserves further attention as a potential diagnostic prostatic DNA marker in human prostate cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Metilación de ADN , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Adulto , Anciano , Secuencia de Bases , Antígeno CD146/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Pruebas Genéticas , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias/métodos , Próstata/patología , Próstata/fisiología
13.
BJU Int ; 101(1): 113-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17941920

RESUMEN

OBJECTIVE: To investigate cell division cycle 91-like 1 (CDC91L1; also called phosphatidylinositol glycan class U, PIG-U) expression in bladder cancer at both the mRNA and protein levels, and to study its clinical and prognostic significance, as CDC91L1 was recently identified as a new oncogene in human bladder cancer and its role in the biological behaviour of bladder cancer is largely unknown. PATIENTS AND METHODS: In all, 73 bladder tumours and 14 samples of normal bladder urothelium were studied by reverse-transcription polymerase chain reaction (PCR), real-time quantitative PCR and immunohistochemistry. RESULTS: The normalized CDC91L1 mRNA copy number in tumours was significantly greater than in normal controls (P < 0.05). There was overexpression of CDC91L1 mRNA in 30.1% (22/73) of the bladder tumours compared with the normal urothelium. At the protein level, 75.3% (55/73) of the bladder tumours and two of 14 of the normal urothelium had high expression of CDC91L1 protein, which is statistically significant (P < 0.001). The correlation between CDC91L1 protein and tumour grade, and muscle invasion of tumour was significant (both P < 0.05). In addition to tumour extent and tumour grade, CDC91L1 protein was an independent predictor of recurrence for superficial bladder cancer and had a trend to predict tumour progression. CONCLUSIONS: CDC91L1 (PIG-U) plays a role in the development of bladder urothelial cell carcinoma. CDC91L1 protein might be a potential biomarker for prediction of recurrence and a therapeutic target in bladder cancer.


Asunto(s)
Aciltransferasas/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia/prevención & control , Estadificación de Neoplasias , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Urotelio/patología
14.
Int J Cancer ; 121(9): 1994-2000, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17620329

RESUMEN

NMDA receptor Type 2B (NMDAR2B) is a candidate TSG first identified in esophageal squamous cell carcinoma (ESCC). To evaluate NMDAR2B methylation in gastric cancer progression, we performed quantitative methylation-specific PCR (MSP), RT-PCR and immnunohistochemistry (IHC) in primary gastric tissues and colony formation assays in gastric cancer cell lines. We found that the expression of NMDAR2B was reactivated by the demethylating agent, 5-aza-2'-deoxycytidine, with or without trichostatin A in gastric cancer cell lines. Moreover, inactivation of NMDAR2B was found to be closely correlated with promoter methylation status in gastric cell lines and primary gastric tumors. IHC data also showed that NMDAR2B was specifically expressed in gastric epithelial cells and its expression was diminished or absent in gastric cancer epithelium. Quantitative analysis of NMDAR2B promoter methylation showed 61% (17/28) hypermethylation in primary gastric tumors versus 5% (1/20) in normal gastric tissues from nongastric cancer patients. Forced over-expression of NMDAR2B in gastric cancer cell lines significantly inhibited cell colony formation. Taken together, the above results suggest that NMDAR2B methylation is a common and important biologically relevant event in gastric cancer progression.


Asunto(s)
Metilación de ADN , Receptores de N-Metil-D-Aspartato/genética , Neoplasias Gástricas/genética , Biomarcadores de Tumor , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Epitelio/metabolismo , Epitelio/patología , Regulación Neoplásica de la Expresión Génica , Salud , Humanos , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
15.
Cancer Res ; 67(9): 4123-9, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17456585

RESUMEN

Promoter hypermethylation is one of the common mechanisms leading to gene silencing in various human cancers. Using a combination of pharmacologic unmasking and microarray techniques, we identified 59 candidate hypermethylated genes, including LOXL1, a lysyl oxidase-like gene, in human bladder cancer cells. We further showed that LOXL1 and LOXL4 are commonly silenced genes in human bladder cancer cells, and this silence is predominantly related to promoter methylation. We also found LOXL1 and LOXL4 gene methylation and loss of expression in primary bladder tumors. In addition, somatic mutations were identified in LOXL4, but not in LOXL1 in bladder cancer. Moreover, reintroduction of LOXL1 and LOXL4 genes into human bladder cancer cells leads to a decrease of colony formation ability. Further studies indicated that the overexpression of LOXL1 and LOXL4 could antagonize Ras in activating the extracellular signal-regulated kinase (ERK) signaling pathway. Thus, our current study suggests for the first time that lysyl oxidase-like genes can act as tumor suppressor genes and exert their functions through the inhibition of the Ras/ERK signaling pathway in human bladder cancer.


Asunto(s)
Aminoácido Oxidorreductasas/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/genética , Proteínas ras/antagonistas & inhibidores , Aminoácido Oxidorreductasas/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Citoplasma/enzimología , Metilación de ADN/efectos de los fármacos , Decitabina , Epigénesis Genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Humanos , Sistema de Señalización de MAP Quinasas , Mutación , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Proteína-Lisina 6-Oxidasa , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Proteínas ras/metabolismo
16.
Tumour Biol ; 26(3): 147-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15970649

RESUMEN

Insulin and insulin-like growth factors (IGFs) are major determinants of proliferation and apoptosis, thereby playing a significant role in carcinogenesis. Epidemiological evidence associates high levels of INS and IGFs with an increased risk of cancer. Polymorphism of the genes involved in insulin-signaling pathways has been associated with a variable risk for neoplasms in different ethnic and environmental backgrounds. In this study, using PCR-RFLP-based assays, we investigated the distribution of genetic polymorphism in INS and IGF-2 genes in tobacco chewing-mediated oral cancer patients (n = 60) and healthy controls (n = 45) of Indian ethnic origin. The genotyping was performed for +1127 INS-Pst1 in INS and +3580 IGF-2-Msp1 in IGF-2. The frequencies of the IGF-2 genotypes AG, GG and AA found in oral cancer patients were 0.68, 0.2 and 0.12, respectively, whereas in noncancer controls these frequencies were 0.27, 0.71 and 0.02. Frequencies of each allele, i.e. CT, TT and CC of INS gene, were found to be nearly equal in the tumor (0.22, 0.75 and 0.03) as well as the normal (0.27, 0.67 and 0.06) population. A significant difference was observed in genotypic frequencies of IGF-2 and INS in the Indian ethnic population as compared to the Caucasian, African and Hispanic populations. Polymorphism at +1127 INS-Pst1 locus of INS gene does not show an implication in oral cancer, whereas the genotype AG or AA at +3580 IGF-2-Msp1 locus of IGF-2 is associated with progression and increased risk of oral cancer. From our study we can conclude that single nucleotide polymorphisms in the IGF-2 gene can be used as a marker for prediction of the risk of oral carcinogenesis.


Asunto(s)
Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/genética , Predisposición Genética a la Enfermedad , Factor II del Crecimiento Similar a la Insulina/genética , Neoplasias de la Boca/etiología , Neoplasias de la Boca/genética , Polimorfismo Genético , Tabaco sin Humo/efectos adversos , Estudios de Casos y Controles , Etnicidad , Genotipo , Humanos , Hipoglucemiantes , India , Insulina/genética , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción
17.
Am J Hematol ; 79(1): 1-7, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15849769

RESUMEN

The frequency of p73 mutation is low in hematologic malignancies as well as solid tumors. In the present study, we scanned for mutations in the exons 4, 5, 6, and 7 of p73, as well as methylation of the CpG island in the untranslated region of exon 1, in 100 de novo AML patients. Four patients showed mutation in exon 5 and one in exon 6, and none of the patients showed mutation in exons 4 and 7. None of the patients showed p73 gene methylation. The expression level of p73 mRNA was also examined in 40 AML samples using reverse transcriptase-polymerase chain reaction. Only six AML patients showed p73 mRNA expression, as analyzed by RT-PCR analysis. However, p73 over-expression in 30% of patients was demonstrated by immunocytochemistry and Western blot analysis. Further, mutation of p73 has been correlated with p73 mRNA and p73 protein status. The results show the presence of over-expressed p73 mRNA and protein in the samples with mutated p73 gene. Thus, it is presumed that mutation of p73 might lead to production of defective p73 protein and p73 mRNA, and this might have a role in the process of leukemogenesis of AML. This report is the first demonstrating the presence of mutations in p73 gene in acute myelogenous leukemia.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Leucemia Mieloide Aguda/genética , Mutación , Proteínas Nucleares/genética , Adulto , Anciano , Secuencia de Bases , Metilación de ADN , Cartilla de ADN , Exones/genética , Neoplasias Hematológicas/genética , Humanos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple , Proteína Tumoral p73 , Proteínas Supresoras de Tumor
18.
Oral Oncol ; 39(3): 213-21, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12618193

RESUMEN

The present review aims to analyze the information available regarding the molecular mechanisms of Oral Carcinogenesis and explore the future directions where the field of Cancer Biology is venturing. Oncologists have excellently followed the proverb "Necessity is the mother of Invention". The desire to be more precise and comprehensive in their studies has led to the invention of some of the most innovative techniques like laser capture microdissection, comparative genomic hybridization, microarrays, and protein chips etc. Various Biotech companies and Cancer Institutes are on a hunt for anti-cancer drugs and molecular markers for cancers. These revolutionary approaches and the new breed of Oncologists have made the field very exciting and have generated the hope that finally the war against cancer would be won. In the end it is urged that the lead taken in other cancers like colon, breast, leukemia will be emulated in oral cancer. This is expected to provide a molecular blueprint for HNSCC, thus helping to identify suitable markers for the early detection of pre-neoplastic lesions, as well as novel targets for its pharmacological intervention.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias de la Boca/genética , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/terapia , Transformación Celular Neoplásica/genética , Predicción , Técnicas Genéticas , Humanos , Neoplasias de la Boca/irrigación sanguínea , Neoplasias de la Boca/terapia , Neovascularización Patológica/fisiopatología , Proteómica
19.
Med Sci Monit ; 8(11): RA258-67, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12444391

RESUMEN

The present review is an attempt to summarize the important advances made during the last decade in the molecular approach to oral cancer and its application for early, sensitive diagnosis, effective treatment, and improved prognosis. Cancer of the oral cavity is more prevalent in developing countries, where many people are addicted to tobacco chewing and maintain poor oral hygiene. Despite extensive research on the biological and molecular aspects of oral SCC, the problems of local-regional recurrence and distant metastasis still persist. Among the more pressing problems in clinical management is the lack of early detection, due to the absence of a potential diagnostic marker. Oncologists are now more aware of the challenges associated with the treatment of cancer of the oral cavity, and survival percentages are improving significantly. More trials are need in the area of improved surgical procedures, variations in dosages of radiotherapy, and the use of various combinations of chemotherapeutic agents with minimal side effects. Moreover, progress in the elucidation of the molecular genetic changes that lead to the development of these tumors should soon bring novel diagnostic and therapeutic procedures into clinical practice. The case of ONYX-015 is one example of success, which has shown the great potential in Phase-I and II clinical trials. Finally, the legislator should also impose some restrictions and bans on the easy availability of various forms of tobacco.


Asunto(s)
Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/etiología , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/terapia , Progresión de la Enfermedad , Humanos , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/terapia
20.
Cancer ; 94(9): 2393-400, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12015764

RESUMEN

BACKGROUND: The Jak/Stat signaling pathway transmits signals from many cytokines and growth factor receptors to target genes in the nucleus. Constitutive activation of Stat-3 recently has been observed in many tumor cells, and dysregulation of the Stat signaling pathway has been proposed to be implicated in malignant transformation. In the current study for the first time to the authors's knowledge, the expression of STAT-3 was analyzed in various stages and sites of squamous cell carcinoma of the head and neck (HNSCC). METHODS: Tissue samples from 90 patients of tobacco chewing-mediated HNSCC representing various stages, sites, and differentiation states were selected for studying STAT-3 protein and RNA expression. In vivo localization of STAT-3 was studied by immunohistochemistry of paraffin embedded sections. The presence of STAT-3 and its phophorylated and activated form pSTAT-3 was checked by Western blotting. mRNA expression was analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR). Apoptosis analysis was conducted by in situ ENA nick end labeling assay and hematoxylin and eosin staining. RESULTS: Overall, 58.9% of HNSCC tumors showed very high Stat-3 protein accumulation, and 23.3% showed intermediate accumulation whereas 17.8% of HNSCC tumors were negative for Stat-3. No Stat-3 was detected in normal samples, and only one of eight premalignant lesions showed intermediate Stat-3 accumulation. On immunoblotting, very high protein accumulation was detected in T1 and T2 classification, moderate in T3 and T4 (P = 0.033, chi-square test), whereas no Stat-3 was detected in normal samples. Similar trend also was found in Stat-3 mRNA expression by RT-PCR analysis which was high in T1 and T2 (early stages), moderate in T3 and T4 (late stages), and no expression in normal samples. The mean apoptotic indices were 1.75, 1.88, and 1.66 for normal, premalignant lesions, and HNSCC cases, respectively. CONCLUSIONS: Stat-3 activation is an early event in head and neck carcinogenesis though its role in blocking the apoptosis in vivo in solid tumors was not observed.


Asunto(s)
Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/genética , Proteínas de Unión al ADN , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/genética , Tabaco sin Humo/efectos adversos , Transactivadores , Apoptosis , Western Blotting , Proteínas de Unión al ADN/análisis , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/genética , ARN Mensajero/análisis , Factor de Transcripción STAT3 , Transactivadores/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA