Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Biomicrofluidics ; 9(6): 064104, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26594265

RESUMEN

Circulating tumor cells (CTCs) are found in the blood of patients with cancer. Although these cells are rare, they can provide useful information for chemotherapy. However, isolation of these rare cells from blood is technically challenging because they are small in numbers. An integrated microfluidic chip, dubbed CTC chip, was designed and fabricated for conducting tumor cell isolation. As CTCs usually show multidrug resistance (MDR), the effect of MDR inhibitors on chemotherapeutic drug accumulation in the isolated single tumor cell is measured. As a model of CTC isolation, human prostate cancer cells were mixed with mouse blood cells and the label-free isolation of the tumor cells was conducted based on cell size difference. The major advantages of the CTC chip are the ability for fast cell isolation, followed by multiple rounds of single-cell measurements, suggesting a potential assay for detecting the drug responses based on the liquid biopsy of cancer patients.

3.
Oncogene ; 33(27): 3561-70, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23934189

RESUMEN

Caveolin-1 has a complex role in prostate cancer and has been suggested to be a potential biomarker and therapeutic target. As mature caveolin-1 resides in caveolae, invaginated lipid raft domains at the plasma membrane, caveolae have been suggested as a tumor-promoting signaling platform in prostate cancer. However, caveola formation requires both caveolin-1 and cavin-1 (also known as PTRF; polymerase I and transcript release factor). Here, we examined the expression of cavin-1 in prostate epithelia and stroma using tissue microarray including normal, non-malignant and malignant prostate tissues. We found that caveolin-1 was induced without the presence of cavin-1 in advanced prostate carcinoma, an expression pattern mirrored in the PC-3 cell line. In contrast, normal prostate epithelia expressed neither caveolin-1 nor cavin-1, while prostate stroma highly expressed both caveolin-1 and cavin-1. Utilizing PC-3 cells as a suitable model for caveolin-1-positive advanced prostate cancer, we found that cavin-1 expression in PC-3 cells inhibits anchorage-independent growth, and reduces in vivo tumor growth and metastasis in an orthotopic prostate cancer xenograft mouse model. The expression of α-smooth muscle actin in stroma along with interleukin-6 (IL-6) in cancer cells was also decreased in tumors of mice bearing PC-3-cavin-1 tumor cells. To determine whether cavin-1 acts by neutralizing caveolin-1, we expressed cavin-1 in caveolin-1-negative prostate cancer LNCaP and 22Rv1 cells. Caveolin-1 but not cavin-1 expression increased anchorage-independent growth in LNCaP and 22Rv1 cells. Cavin-1 co-expression reversed caveolin-1 effects in caveolin-1-positive LNCaP cells. Taken together, these results suggest that caveolin-1 in advanced prostate cancer is present outside of caveolae, because of the lack of cavin-1 expression. Cavin-1 expression attenuates the effects of non-caveolar caveolin-1 microdomains partly via reduced IL-6 microenvironmental function. With circulating caveolin-1 as a potential biomarker for advanced prostate cancer, identification of the molecular pathways affected by cavin-1 could provide novel therapeutic targets.


Asunto(s)
Caveolina 1/metabolismo , Microdominios de Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas de Unión al ARN/metabolismo , Actinas/metabolismo , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/metabolismo
4.
Pharmacology ; 85(4): 248-58, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20375535

RESUMEN

gamma-Tocotrienol (gammaT3) is known to selectively kill prostate cancer (PCa) cells and to sensitize the cells to docetaxel (DTX)-induced apoptosis. In the present study, the pharmacokinetics of gammaT3 and the in vivo cytotoxic response of androgen-independent prostate cancer (AIPCa) tumor following gammaT3 treatment were investigated. Here, we investigated these antitumor effects for PCa tumors in vivo. The pharmacokinetic and tissue distribution of gammaT3 after exogenous gammaT3 supplementation were examined. Meanwhile, the response of the tumor to gammaT3 alone or in combination with DTX were studied by real-time in vivo bioluminescent imaging and by examination of biomarkers associated with cell proliferation and apoptosis. After intraperitoneal injection, gammaT3 rapidly disappeared from the serum and was selectively deposited in the AIPCa tumor cells. Administration of gammaT3 alone for 2 weeks resulted in a significant shrinkage of the AIPCa tumors. Meanwhile, further inhibition of the AIPCa tumor growth was achieved by combined treatment of gammaT3 and DTX (p < 0.002). The in vivo cytotoxic antitumor effects induced by gammaT3 seem to be associated with a decrease in expression of cell proliferation markers (proliferating cell nuclear antigen, Ki-67 and Id1) and an increase in the rate of cancer cell apoptosis [cleaved caspase 3 and poly(ADP-ribose) polymerase]. Additionally, the combined agents may be more effective at suppressing the invasiveness of AIPCa. Overall, our results indicate that gammaT3, either alone or in combination with DTX, may provide a treatment strategy that can improve therapeutic efficacy against AIPCa while reducing the toxicity often seen in patients treated with DTX.


Asunto(s)
Antineoplásicos/uso terapéutico , Cromanos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Vitamina E/análogos & derivados , Animales , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Cadherinas/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromanos/farmacocinética , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Distribución Tisular , Vitamina E/farmacocinética , Vitamina E/uso terapéutico
5.
Cancer Lett ; 291(2): 187-99, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19926394

RESUMEN

Gamma-tocotrienol has demonstrated anti-proliferative effect on breast cancer (BCa) cells, but mechanisms involved are largely unknown. This study aimed at deciphering the molecular pathways responsible for its activity. Our results showed that treatment of BCa cells with gamma-tocotrienol resulted in induction of apoptosis as evidenced by activation of pro-caspases, accumulation of sub-G1 cells and DNA fragmentations. Examination of the pro-survival genes revealed that the gamma-tocotrienol-induced cell death was associated with suppression of Id1 and NF-kappaB through modulation of their upstream regulators (Src, Smad1/5/8, Fak and LOX). Meanwhile, gamma-tocotrienol treatment also resulted in the induction of JNK signaling pathway and inhibition of JNK activity by specific inhibitor partially blocked the effect of gamma-tocotrienol. Furthermore, synergistic effect was observed when cells were co-treated with gamma-tocotrienol and Docetaxel. Interestingly, in cells that treated with gamma-tocotrienol, alpha-tocopherol or beta-aminoproprionitrile were found to partially restore Id1 expression. Meanwhile, this restoration of Id1 was found to protect the cells from gamma-tocotrienol induced apoptosis. Consistent outcome was observed in cells ectopically transfected with the Id-1 gene. Our results suggested that the anti-proliferative and chemosensitization effect of gamma-tocotrienol on BCa cells may be mediated through downregulation of Id1 protein.


Asunto(s)
Neoplasias de la Mama/patología , Cromanos/farmacología , Proteína 1 Inhibidora de la Diferenciación/genética , Vitamina E/análogos & derivados , Vitamina E/farmacología , Andrógenos/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Muerte Celular/efectos de los fármacos , Diferenciación Celular , División Celular/efectos de los fármacos , Colágeno , Fragmentación del ADN , Regulación hacia Abajo , Combinación de Medicamentos , Estrógenos/fisiología , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Laminina , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteoglicanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas
6.
Nutr Cancer ; 61(3): 357-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19373609

RESUMEN

To date, the most effective cure for metastatic melanoma remains the surgical resection of the primary tumor. Recently, tocotrienol-rich-fraction has shown antiproliferative effect on cancer cells. To elucidate this anticancer property in malignant melanoma, this study aimed, first, to identify the most potent isomer for eliminating melanoma cells and second to decipher the molecular pathway responsible for its activity. Results showed that the inhibitory effect of gamma-tocotrienol was most potent, which resulted in induction of apoptosis as evidenced by activation of procaspases and the accumulation of sub-G1 cell population. Examination of the prosurvival genes revealed that the gamma-tocotrienol-induced cell death was associated with suppression of NF-kappaB, EGF-R, and Id family proteins. Meanwhile, gamma-tocotrienol treatment also resulted in induction of JNK signaling pathway, and inhibition of JNK activity by selective inhibitor was able to partially block the effect of gamma-tocotrienol. Interestingly, gamma-tocotrienol treatment led to suppression of mesenchymal markers and the restoration of E-cadherin and gamma-catenin expression, which was associated with suppression of cell invasion capability. Furthermore, synergistic effect was observed when cells were cotreated with gamma-tocotrienol and chemotherapy drugs. Together, our results demonstrated for the first time the anti-invasion and chemonsensitization effect of gamma-tocotrienol against human malignant melanoma cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cromanos/farmacología , Melanoma/tratamiento farmacológico , Vitamina E/análogos & derivados , Cadherinas/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dacarbazina/farmacología , Docetaxel , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Melanoma/patología , Invasividad Neoplásica , Transducción de Señal , Taxoides/farmacología , Vitamina E/farmacología
7.
Br J Cancer ; 99(11): 1832-41, 2008 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19002171

RESUMEN

Tocotrienol-rich fraction (TRF) has demonstrated antiproliferative effect on prostate cancer (PCa) cells. To elucidate this anticancer property in PCa cells, this study aimed, first, to identify the most potent isomer for eliminating PCa cells; and second, to decipher the molecular pathway responsible for its activity. Results showed that the inhibitory effect of gamma-tocotrienol was most potent, which resulted in induction of apoptosis as evidenced by activation of pro-caspases and the presence of sub-G(1) cell population. Examination of the pro-survival genes revealed that the gamma-tocotrienol-induced cell death was associated with suppression of NF-kappaB, EGF-R and Id family proteins (Id1 and Id3). Meanwhile, gamma-tocotrienol treatment also resulted in the induction of JNK-signalling pathway and inhibition of JNK activity by a specific inhibitor (SP600125) was able to partially block the effect of gamma-tocotrienol. Interestingly, gamma-tocotrienol treatment led to suppression of mesenchymal markers and the restoration of E-cadherin and gamma-catenin expression, which was associated with suppression of cell invasion capability. Furthermore, a synergistic effect was observed when cells were co-treated with gamma-tocotrienol and Docetaxel. Our results suggested that the antiproliferative effect of gamma-tocotrienol act through multiple-signalling pathways, and demonstrated for the first time the anti-invasion and chemosensitisation effect of gamma-tocotrienol against PCa cells.


Asunto(s)
Antineoplásicos/farmacología , Cromanos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Vitamina E/análogos & derivados , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Masculino , Invasividad Neoplásica , Vitamina E/farmacología
8.
Oncogene ; 27(32): 4456-66, 2008 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-18372912

RESUMEN

Id-1 (Inhibitor of DNA binding/differential-1) plays a positive role in tumorigenesis through regulation of multiple signaling pathways. Recently, it is suggested that upregulation of Id-1 in cancer cells promotes chromosomal instability. However, the underlying molecular mechanism is not known. In this study, we report a novel function of Id-1 in regulation of mitosis through physical interaction with Cdc20 (cell division cycle protein 20) and Cdh1 (Cdc20 homolog 1). During early mitosis, Id-1 interacts with Cdc20 and RASSF1A (Ras association domain family 1A), leading to enhanced APC(Cdc20) activity, which in turn promotes cyclin B1/securin degradation and premature mitosis. During late mitosis, Id-1 binds to Cdh1 and disrupts the interaction between Cdh1 and APC, resulting in suppression of APC(Cdh1) activity. On the other hand, overexpression of Cdh1 leads to Id-1 protein degradation, suggesting that Id-1 may also act as a substrate of APC(Cdh1). The negative effect of Id-1 on APC(Cdh1) results in suppression of APC(Cdh1)-induced Aurora A and Cdc20 degradation, leading to failure in cytokinesis. As a result, overexpression of Id-1 in human prostate epithelial cells leads to polyploidy in response to microtubule disruption, and this effect is abolished when Id-1 expression is suppressed using antisense technology. These results demonstrate a novel function of Id-1 in promoting chromosomal instability through modification of APC/C activity during mitosis and provide a novel molecular mechanism accounted for the function of Id-1 as an oncogene.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Inestabilidad Cromosómica , Proteína 1 Inhibidora de la Diferenciación/fisiología , Microtúbulos/fisiología , Mitosis , Complejos de Ubiquitina-Proteína Ligasa/fisiología , Ciclosoma-Complejo Promotor de la Anafase , Aurora Quinasas , Proteínas Cdc20 , Línea Celular , Ciclina B/metabolismo , Ciclina B1 , Fase G1 , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/fisiología , Ubiquitina/metabolismo
9.
Oncogene ; 27(3): 347-57, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-17621272

RESUMEN

The mitotic arrest deficient 2 (MAD2) is suggested to play a key role in a functional mitotic checkpoint because of its inhibitory effect on anaphase-promoting complex/cyclosome (APC/C) during mitosis. The binding of MAD2 to mitotic checkpoint regulators MAD1 and Cdc20 is thought to be crucial for its function and loss of which leads to functional inactivation of the MAD2 protein. However, little is known about the biological significance of this MAD2 mutant in human cells. In this study, we stably transfected a C-terminal-deleted MAD2 gene (MAD2DeltaC) into a human prostate epithelial cell line, Hpr-1 and studied its effect on chromosomal instability, cell proliferation, mitotic checkpoint control and soft agar colony-forming ability. We found that MAD2DeltaC was able to induce aneuploidy through promoting chromosomal duplication, which was a result of an impaired mitotic checkpoint and cytokinesis, suggesting a crucial role of MAD2-mediated mitotic checkpoint in chromosome stability in human cells. In addition, the MAD2DeltaC-transfected cells displayed anchorage-independent growth in soft agar after challenged by 7,12-dimethylbenz[A]anthracene (DMBA), demonstrating a cancer-promoting effect of a defective mitotic checkpoint in human cells. Furthermore, the DMBA-induced transformation was accompanied by a complete loss of DNA damage-induced p53 response and activation of the MAPK pathway in MAD2DeltaC cells. These results indicate that a defective mitotic checkpoint alone is not a direct cause of tumorigenesis, but it may predispose human cells to carcinogen-induced malignant transformation. The evidence presented here provides a link between MAD2 inactivation and malignant transformation of epithelial cells.


Asunto(s)
Aneuploidia , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Carcinógenos/toxicidad , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Humanos , Proteínas Mad2 , Masculino , Mitosis , Próstata/patología , Neoplasias de la Próstata/inducido químicamente , Neoplasias de la Próstata/patología , Eliminación de Secuencia , Proteína p53 Supresora de Tumor/metabolismo
10.
Oncogene ; 27(23): 3313-28, 2008 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-18071305

RESUMEN

Recent studies indicate that estrogen-related receptors (ERRs) are involved in similar estrogen receptor (ER) regulatory pathways and play roles in energy and lipid metabolism. Here, we analysed the functional role of ERRbeta in prostate cancer cell growth regulation in an androgen-sensitive and androgen-insensitive prostate cancer cell lines. ERRbeta was expressed in normal human prostates, but exhibited a reduced expression in prostate cancer lesions. Stable ERRbeta expression suppressed significantly cell proliferation and tumorigenicity of LNCaP and DU145 cells, accompanied by an S-phase suppression and increased p21 expression. Reporter and chromatin immunoprecipitation assays showed that ERRbeta could directly transactivate p21 gene promoter, which could be further enhanced by peroxisome proliferator-activated receptor-gamma coactivator-1alpha. Truncation analysis showed that ERRbeta-mediated p21 transactivation and prostate cancer cell growth inhibition required intact DNA-binding domain and AF2 domains in ERRbeta. Interestingly, ERRbeta displayed a cell cycle associated downregulated expression pattern in ERRbeta-transduced and non-transduced cells. Finally, we showed that ERRbeta-mediated growth inhibition could be potentiated by an ERRbeta/gamma agonist DY131. Knockdown of ERRbeta by RNA interference could reduce the DY131-induced growth inhibition in prostate cancer cells. Taken together, our findings indicate that ERRbeta performs a tumor suppressing function in prostate cancer cells, and targeting ERRbeta could be a potential therapeutic strategy for prostate cancer.


Asunto(s)
Carcinoma/genética , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Sistemas de Liberación de Medicamentos , Neoplasias de la Próstata/genética , Receptores de Estrógenos/fisiología , Animales , Antineoplásicos/uso terapéutico , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Células HeLa , Proteínas de Choque Térmico/fisiología , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Estructura Terciaria de Proteína/fisiología , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Factores de Transcripción/fisiología , Transfección , Trasplante Heterólogo , Regulación hacia Arriba
11.
Br J Cancer ; 95(4): 475-84, 2006 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-16880791

RESUMEN

Testicular germ cell tumour (TGCT) is the most common malignancy in young males. Although most TGCTs are sensitive to cisplatin-based chemotherapy, significant numbers of TGCT patients still relapse and die each year because of the development of resistance to cisplatin. Previously, we first reported that a key regulator of the mitotic checkpoint, mitotic arrest deficient-2 (MAD2), was a mediator of cisplatin sensitivity in human cancer cells. In this study, we investigated whether MAD2 played a role in cellular sensitivity to cisplatin in TGCT cells and the underlying molecular mechanisms responsible. Using 10 TGCT cell lines, we found that increased MAD2 expression was correlated with cellular sensitivity to cisplatin, which was associated with activation of the MEK pathway. Treatment of cells expressing high levels of MAD2 with an MEK inhibitor, U0126, led to cellular protection against cisplatin-induced apoptosis. Inactivation of MAD2 by transfecting a dominant-negative construct in TGCT cells with high levels of MAD2 resulted in the suppression of MEK pathway and resistance to cisplatin-induced cell death. These results support previous suggestion on the involvement of mitotic checkpoint in DNA damage response in human cancer cells and demonstrate a possible molecular mechanism responsible for the MAD2-mediated sensitivity to cisplatin in TGCT cells. Our results also suggest that downregulation of MAD2 may be an indicator for identification of TGCT cancer cells that are potentially resistant to cisplatin-based therapy.


Asunto(s)
Proteínas de Unión al Calcio/farmacología , Proteínas de Ciclo Celular/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos , MAP Quinasa Quinasa 1/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Represoras/farmacología , Neoplasias Testiculares/tratamiento farmacológico , Butadienos/farmacología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Proteínas Mad2 , Masculino , Modelos Biológicos , Neoplasias de Células Germinales y Embrionarias , Nitrilos/farmacología , Transducción de Señal , Transfección
12.
Prostate Cancer Prostatic Dis ; 9(3): 293-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16683014

RESUMEN

Benign prostate hyperplasia (BPH) is a common disease in elderly men. Although it is a non-malignant disease, it has a significant detrimental impact on the quality of life in patients with late-stage disease. Owing to the lack of specific markers, diagnosis of early-stage BPH has been proven unsuccessful. Recently, using two-dimensional electrophoresis, we identified a group of prostatic secretory proteins that are specifically produced by BPH cells (Xu et al., Electrophoresis 2003; 24: 1311). In this study, we investigated the potential diagnostic value of one of the secretory proteins, alphas1-Casein, in BPH by inmmunohistological staining of normal, BPH and prostate cancer tissues. We found that 90% (20 out of 22) of BPH tissues showed moderate to strong alphas1-Casein protein expression whereas none of the normal tissues (0 out of 10) and less than 10% of the prostate cancer tissues (3 out of 30) showed similar staining intensity. Our results suggest that alphas1-Casein may be a potential biomarker for early identification of BPH patients.


Asunto(s)
Biomarcadores de Tumor/aislamiento & purificación , Caseínas/aislamiento & purificación , Proteínas de la Leche/aislamiento & purificación , Hiperplasia Prostática/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Autopsia , Biomarcadores de Tumor/metabolismo , Caseínas/metabolismo , Diagnóstico Diferencial , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Próstata/metabolismo , Próstata/patología , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas de Secreción Prostática/aislamiento & purificación , Proteínas de Secreción Prostática/metabolismo , Regulación hacia Arriba
13.
Br J Cancer ; 91(12): 2042-7, 2004 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-15599381

RESUMEN

Increased EGFR (epidermal growth factor receptor) expression has been reported in many types of human cancer and its levels are positively associated with advanced cancers. Recently, upregulation of Id-1 (inhibitor of differentiation or DNA binding) protein was found in over 70% of ovarian cancer samples and correlated with poor survival of ovarian cancer patients. However, the molecular mechanisms responsible for the role of Id-1 in ovarian cancer are not clear. The aim of this study was to investigate the effect of Id-1 on ovarian cancer proliferation and its association with the EGFR pathway. To achieve this, we transfected an Id-1 expression vector into three ovarian cancer cell lines and examined cell proliferation rate by flow cytometry and bromodeoxyuridine staining. We found that ectopic Id-1 expression led to increased cell proliferation demonstrated by increased BrdU incorporation rate and S-phase fraction. The Id-1-induced cell growth was associated with upregulation of EGFR at both transcriptional and protein levels. In contrast, inactivation of Id-1 through transfection of an Id-1 antisense vector resulted in downregulation of EGFR. Our results indicate that increased Id-1 in ovarian cancer cells may promote cancer cell proliferation through upregulation of EGFR. Our findings also implicate that Id-1 may be a potential target for the development of novel strategies in the treatment of ovarian cancer.


Asunto(s)
Proliferación Celular , Receptores ErbB/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Proteína 1 Inhibidora de la Diferenciación
14.
BMC Cancer ; 4: 72, 2004 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-15469618

RESUMEN

BACKGROUND: Precise classification of cancer types is critically important for early cancer diagnosis and treatment. Numerous efforts have been made to use gene expression profiles to improve precision of tumor classification. However, reliable cancer-related signals are generally lacking. METHOD: Using recent datasets on colon and prostate cancer, a data transformation procedure from single gene expression to pair-wise gene expression ratio is proposed. Making use of the internal consistency of each expression profiling dataset this transformation improves the signal to noise ratio of the dataset and uncovers new relevant cancer-related signals (features). The efficiency in using the transformed dataset to perform normal/tumor classification was investigated using feature partitioning with informative features (gene annotation) as discriminating axes (single gene expression or pair-wise gene expression ratio). Classification results were compared to the original datasets for up to 10-feature model classifiers. RESULTS: 82 and 262 genes that have high correlation to tissue phenotype were selected from the colon and prostate datasets respectively. Remarkably, data transformation of the highly noisy expression data successfully led to lower the coefficient of variation (CV) for the within-class samples as well as improved the correlation with tissue phenotypes. The transformed dataset exhibited lower CV when compared to that of single gene expression. In the colon cancer set, the minimum CV decreased from 45.3% to 16.5%. In prostate cancer, comparable CV was achieved with and without transformation. This improvement in CV, coupled with the improved correlation between the pair-wise gene expression ratio and tissue phenotypes, yielded higher classification efficiency, especially with the colon dataset - from 87.1% to 93.5%. Over 90% of the top ten discriminating axes in both datasets showed significant improvement after data transformation. The high classification efficiency achieved suggested that there exist some cancer-related signals in the form of pair-wise gene expression ratio. CONCLUSION: The results from this study indicated that: 1) in the case when the pair-wise expression ratio transformation achieves lower CV and higher correlation to tissue phenotypes, a better classification of tissue type will follow. 2) the comparable classification accuracy achieved after data transformation suggested that pair-wise gene expression ratio between some pairs of genes can identify reliable markers for cancer.


Asunto(s)
Neoplasias del Colon/genética , Perfilación de la Expresión Génica/métodos , Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias de la Próstata/genética , Colon/citología , Neoplasias del Colon/patología , Sondas de ADN , Femenino , Humanos , Masculino , Fenotipo , Próstata/citología , Neoplasias de la Próstata/patología
15.
Apoptosis ; 9(3): 279-89, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15258459

RESUMEN

The Id (inhibitor of differentiation or DNA binding) helix-loop-helix (HLH) proteins are a group of dominant negative regulators of basic HLH transcriptional factors which promote cell differentiation. Recent evidence has revealed that Id proteins, especially Id-1, are also able to promote cell proliferation and cell cycle progression through inactivation of tumour suppressor and activation of growth promoting pathways in mammalian cells. In addition, upregulation of Id-1 has been found in many types of human cancer and its expression levels are also associated with advanced tumour stage. Furthermore, ectopic expression of Id-1 in human cancer cells is able to induce cell proliferation under sub-optimal conditions and protect the cells against apoptosis. These lines of evidence strongly indicate Id-1 as a positive regulator of cell growth and its expression may be a key factor required for tumour cell proliferation. This review will discuss recent evidence on the role of Id-1 in cell proliferation and survival, and its significance in malignant transformation. In addition, we will highlight the recent development in the understanding of the molecular mechanisms responsible for the action of Id-1 in promoting cell survival and tumourigenesis. Finally, the therapeutic implications through inactivation of Id-1 in the treatment of human cancer will also be addressed.


Asunto(s)
Ciclo Celular , Fenómenos Fisiológicos Celulares , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , Diferenciación Celular , División Celular , Supervivencia Celular , Transformación Celular Neoplásica , Proteínas de Unión al ADN/genética , Secuencias Hélice-Asa-Hélice , Humanos , Proteína 1 Inhibidora de la Diferenciación , Modelos Biológicos , Estadificación de Neoplasias , Proteínas Represoras/química , Factores de Transcripción/química , Factores de Transcripción/genética , Regulación hacia Arriba
16.
Oncogene ; 23(25): 4488-94, 2004 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15064751

RESUMEN

Nasopharyngeal carcinoma is closely associated with Epstein-Barr virus (EBV) infection. The EBV-encoded LMP1 has cell transformation property. It suppresses cellular senescence and enhances cell survival in various cell types. Many of the downstream events of LMP1 expression are mediated through its ability to activate NF-kappaB. In this study, we report a novel function of LMP1 to induce Id1 expression in nasopharyngeal epithelial cells (NP69) and human embryonal kidney cells (HEK293). The Id1 is a basic helix-loop-helix (bHLH) protein and a negative transcriptional regulator of p16(INK4a). Expression of Id1 facilitates cellular immortalization and stimulates cell proliferation. With the combination of both specific chemical inhibitors and genetic inhibitors of cell signaling, we showed that induction of Id1 by LMP1 was dependent on its NF-kappaB activation domain at the carboxy-terminal region, CTAR1 and CTAR2. Induction of Id1 by LMP1 may facilitate clonal expansion of premalignant nasopharyngeal epithelial cells infected with EBV and may promote their malignant transformation.


Asunto(s)
Células Epiteliales/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/fisiología , Nasofaringe/citología , Proteínas Represoras , Factores de Transcripción/fisiología , Proteínas de la Matriz Viral/fisiología , Carcinoma/epidemiología , Carcinoma/etiología , Carcinoma/virología , Células Clonales/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/genética , Genes p16 , Hong Kong/epidemiología , Humanos , Proteína 1 Inhibidora de la Diferenciación , FN-kappa B/fisiología , Neoplasias Nasofaríngeas/epidemiología , Neoplasias Nasofaríngeas/etiología , Neoplasias Nasofaríngeas/virología , Estructura Terciaria de Proteína , Eliminación de Secuencia , Transducción de Señal , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética , Proteínas de la Matriz Viral/química
17.
Int Rev Cytol ; 227: 65-130, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14518550

RESUMEN

The process involved in the development and carcinogenesis of the prostate gland is complex. During early prostate development, the androgenic hormone from embryonic testicles is required for ductal formation, growth, and branching morphogenesis of the prostate gland. From this early stage, interactions between the epithelium and mesenchyme become firmly established through paracrine influence (i.e., growth factors) from mesenchyme (stroma), in response to testosterone, acting on epithelium to stimulate its proliferation, morphogenetic differentiation, and function. In return, the epithelium also exerts its paracrine effects on mesenchyme by regulating the differentiation and specific organizational pattern of its stromal smooth muscle. In a normal adult prostate, the maintenance of normal glandular structure and function is dependent not only on the constant presence of testosterone, but also on a normal intact and stable stroma. This chapter will concentrate first on factors involved in the normal development of the prostate gland and then on the aberrant changes in the homeostatic balance arising either from within (i.e., mutations) or outside (i.e., changes in hormonal balance) that result in derangements of the prostate gland. Finally, environmental and genetic factors that lead to prostate carcinogenesis including activation of oncogenes and mutations of tumor suppressor genes are also discussed.


Asunto(s)
Próstata/embriología , Próstata/crecimiento & desarrollo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/fisiología , Humanos , Masculino , Mesodermo/citología , Mesodermo/fisiología , Comunicación Paracrina
18.
Biochim Biophys Acta ; 1570(3): 145-52, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12020803

RESUMEN

Transforming growth factor beta1 (TGF beta 1) plays important roles in the regulation of cell growth and differentiation in both normal and malignant prostate epithelial cells. Although certain pathways have been suggested, the mechanisms responsible for the action of TGF beta 1 are not well understood. In the present study, using a human papilloma virus 16 E6/E7 immortalized prostate epithelial cell line, HPr-1, we report that TGF beta 1 was able to suppress the expression of Id-1, a helix-loop-helix (HLH) protein, which plays important roles in the inhibition of cell differentiation and growth arrest. In addition, a decrease at both Id-1 mRNA and protein expression levels was associated with TGF beta 1-induced growth arrest and differentiation, indicating that Id-1 may be involved in TGF beta 1 signaling pathway. The fact that up-regulation of p21(WAF1), one of the downstream effectors of Id-1, was observed after exposure to TGF beta 1 further indicates the involvement of Id-1 in the TGF beta 1-induced growth arrest in HPr-1 cells. However, increased expression of p27(KIP1) was also observed in the TGF beta 1-treated cells, suggesting that in addition to down-regulation of Id-1, other factors may be involved in the TGF beta 1-induced cell growth arrest and differentiation in prostate epithelial cells. Our results provide evidence for the first time that TGF beta 1 may be one of the upstream regulators of Id-1.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo , Secuencias Hélice-Asa-Hélice/fisiología , Proteínas Represoras , Factores de Transcripción/biosíntesis , Factor de Crecimiento Transformador beta/fisiología , Western Blotting , Proteínas de Ciclo Celular/biosíntesis , Diferenciación Celular , División Celular , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Proteínas de Unión al ADN/fisiología , Células Epiteliales/metabolismo , Citometría de Flujo , Humanos , Proteína 1 Inhibidora de la Diferenciación , Masculino , Próstata/metabolismo , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta1 , Proteínas Supresoras de Tumor/biosíntesis , Regulación hacia Arriba
19.
J Endocrinol ; 170(1): 287-96, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11431162

RESUMEN

Androgen signaling is crucial for the growth and development, as well as for tumorigenesis of the prostate. However, many of the prostate epithelial cell lines developed previously, either normal or tumorigenic, do not express androgen receptor (AR) or respond to androgen. In order to advance our understanding on how androgen signaling regulates the growth and the differentiation status, and affects tumorigenicity of the epithelial cell, we performed experiments on HPr-1, a prostate cell line recently immortalized from normal human prostate epithelial cells. In the present study, AR was stably transfected into HPr-1 cells by replication-defective retrovirus. Treatment of HPr-1AR cells with androgen resulted in cell differentiation and growth retardation accompanied with up-regulation of cytokeratins K8 and K18, prostate specific antigen, p21 and p27, and down-regulation of c-myc, bcl-2 and telomerase activity. Our results suggest that androgen promotes the process of differentiation in a human papillomavirus 16 E6/E7 immortalized prostate epithelial cell line which may reflect the normal effects of androgen on prostate cells.


Asunto(s)
Andrógenos/farmacología , Línea Celular Transformada/efectos de los fármacos , Proteínas Musculares , Papillomaviridae , Próstata/efectos de los fármacos , Receptores Androgénicos/genética , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Queratinas/metabolismo , Masculino , Proteínas de Microfilamentos/metabolismo , Próstata/citología , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/terapia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Estimulación Química , Telomerasa/metabolismo , Transfección/métodos , Proteína X Asociada a bcl-2
20.
Gynecol Oncol ; 78(3 Pt 1): 293-301, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10985883

RESUMEN

OBJECTIVE: In cervical cancer, high-risk human papillomavirus (HPV) genes are expressed solely in cancerous cells and have been proposed to be the most important etiological factors for cervical cancer, thus making them suitable targets for gene therapy. In this study, we aim to inactivate the HPV16 E7 in CaSki cells and test the possibility of reducing the tumorigenicity of these cells. METHODS: The full-length HPV16 E7 cDNA was cloned in the pBabe-puro or pWZL-Hygro retrovirus vector in reverse orientation and was stably transfected into CaSki cells by replication-defective retrovirus infection giving rise to CaSki-E7AS and CaSki-E7AS2X cells. Immunoprecipitation/Western analysis and real-time RT-PCR were performed to document the levels of HPV16 E7 gene product. Flow cytometry was performed to study changes in the cell cycle in response to reduced E7 protein. The expression of bcl-2, RB, and E2F-1 was studied using Western blot analysis. Tumorigenicity of CaSki, CaSki-E7AS, and CaSki-E7AS2X cells was assayed with subepidermal tumor growth in nude mice. RESULTS: We have documented that the delivery of the antisense gene construct resulted in the reduction of HPV16 E7 protein expression and cell proliferation in CaSki cells. Furthermore, we demonstrated that these changes were accompanied by cell cycle arrest, up-regulation of RB, and down-regulation of E2F-1 and bcl-2 proteins. More importantly, dose-dependent transduction of the antisense HPV16E7 construct was able to inhibit and/or retard the tumorigenicity of CaSki cells in vivo. CONCLUSIONS: Down-regulation of HPV16 E7 with antisense RNA is beneficial in reducing the tumorigenicity of CaSki cells and can potentially be useful for HPV-associated malignancy gene therapy.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , ARN sin Sentido/administración & dosificación , Neoplasias del Cuello Uterino/virología , Animales , Diferenciación Celular , División Celular , Regulación hacia Abajo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Oncogénicas Virales/biosíntesis , Proteínas E7 de Papillomavirus , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN sin Sentido/genética , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/metabolismo , Proteína 1 de Unión a Retinoblastoma , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción DP1 , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...