Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
3.
Oncogene ; 35(49): 6330-6340, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27270442

RESUMEN

MicroRNA-101, a tumor suppressor microRNA (miR), is often downregulated in cancer and is known to target multiple oncogenes. Some of the genes that are negatively regulated by miR-101 expression include histone methyltransferase EZH2 (enhancer of zeste homolog 2), COX2 (cyclooxygenase-2), POMP (proteasome maturation protein), CERS6, STMN1, MCL-1 and ROCK2, among others. In the present study, we show that miR-101 targets transcriptional coactivator SUB1 homolog (Saccharomyces cerevisiae)/PC4 (positive cofactor 4) and regulates its expression. SUB1 is known to have diverse role in vital cell processes such as DNA replication, repair and heterochromatinization. SUB1 is known to modulate transcription and acts as a mediator between the upstream activators and general transcription machinery. Expression profiling in several cancers revealed SUB1 overexpression, suggesting a potential role in tumorigenesis. However, detailed regulation and function of SUB1 has not been elucidated. In this study, we show elevated expression of SUB1 in aggressive prostate cancer. Knockdown of SUB1 in prostate cancer cells resulted in reduced cell proliferation, invasion and migration in vitro, and tumor growth and metastasis in vivo. Gene expression analyses coupled with chromatin immunoprecipitation revealed that SUB1 binds to the promoter regions of several oncogenes such as PLK1 (Polo-like kinase 1), C-MYC, serine-threonine kinase BUB1B and regulates their expression. Additionally, we observed SUB1 downregulated CDKN1B expression. PLK1 knockdown or use of PLK1 inhibitor can mitigate oncogenic function of SUB1 in benign prostate cancer cells. Thus, our study suggests that miR-101 loss results in increased SUB1 expression and subsequent activation of known oncogenes driving prostate cancer progression and metastasis. This study therefore demonstrates functional role of SUB1 in prostate cancer, and identifies its regulation and potential downstream therapeutic targets of SUB1 in prostate cancer.


Asunto(s)
Proteínas de Unión al ADN/genética , MicroARNs/genética , Neoplasias de la Próstata/genética , Factores de Transcripción/genética , Animales , Proliferación Celular/genética , Proteínas de Unión al ADN/biosíntesis , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Masculino , Ratones , MicroARNs/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factores de Transcripción/biosíntesis
4.
Ann Oncol ; 27(9): 1706-12, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27217541

RESUMEN

BACKGROUND: Despite aggressive multimodal therapy, locally advanced and/or metastatic penile squamous cell carcinoma (SqCC) is associated with significant morbidity and mortality, indicating a need for new therapeutic options. Given the emerging clinical utility of immunotherapeutics, we sought to assess the incidence and potential clinical significance of PD-L1 expression in penile SqCC. PATIENTS AND METHODS: Using an anti-PD-L1 primary antibody (clone 5H1), immunohistochemistry was carried out on whole tumor sections from 37 patients with penile SqCC treated at our institution between 2005 and 2013. PD-L1-positive tumors were defined as those with membranous staining in ≥5% of tumor cells. Association between PD-L1 expression and clinicopathologic parameters was examined using Fisher's exact test. Correlation between PD-L1 expression in primary tumors and matched metastases was assessed using the Spearman rank correlation coefficient (ρ). The difference in cancer-specific mortality between PD-L1-positive and -negative groups was examined using the log-rank test. RESULTS: Twenty-three (62.2%) of 37 primary tumors were positive for PD-L1 expression, and there was strong positive correlation of PD-L1 expression in primary and metastatic samples (ρ = 0.72; 0.032 < P < 0.036). Primary tumor PD-L1 expression was significantly associated with usual type histology (P = 0.040) and regional lymph node metastasis (P = 0.024), as well as decreased cancer-specific survival (P = 0.011). CONCLUSIONS: The majority of primary penile SqCC tumors express PD-L1, which is associated with high-risk clinicopathologic features and poor clinical outcome. These data provide a rational basis for further investigation of anti-PD-1 and anti-PD-L1 immunotherapeutics in patients with advanced penile SqCC.


Asunto(s)
Antígeno B7-H1/genética , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Neoplasias del Pene/genética , Anciano , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Inmunoterapia , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Neoplasias del Pene/inmunología , Neoplasias del Pene/patología , Neoplasias del Pene/terapia , Factores de Riesgo
5.
Ann Oncol ; 26(8): 1589-604, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26041764

RESUMEN

The first St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) Expert Panel identified and reviewed the available evidence for the ten most important areas of controversy in advanced prostate cancer (APC) management. The successful registration of several drugs for castration-resistant prostate cancer and the recent studies of chemo-hormonal therapy in men with castration-naïve prostate cancer have led to considerable uncertainty as to the best treatment choices, sequence of treatment options and appropriate patient selection. Management recommendations based on expert opinion, and not based on a critical review of the available evidence, are presented. The various recommendations carried differing degrees of support, as reflected in the wording of the article text and in the detailed voting results recorded in supplementary Material, available at Annals of Oncology online. Detailed decisions on treatment as always will involve consideration of disease extent and location, prior treatments, host factors, patient preferences as well as logistical and economic constraints. Inclusion of men with APC in clinical trials should be encouraged.


Asunto(s)
Adenocarcinoma/terapia , Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/terapia , Neoplasias de la Próstata/terapia , Taxoides/uso terapéutico , Adenocarcinoma/patología , Antineoplásicos/uso terapéutico , Docetaxel , Humanos , Masculino , Orquiectomía , Guías de Práctica Clínica como Asunto , Prostatectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/patología , Radioterapia Adyuvante
6.
Oncogene ; 32(42): 5026-37, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23160375

RESUMEN

Despite the dismal prognosis for patients with squamous cell carcinoma of the head and neck (SCCHN), there have been no novel treatments in over 40 years. Identification of novel tumor antigens in SCCHN will facilitate the identification of potential novel treatment targets. Tumor antigens are proteins selectively expressed by tumor cells and recognized by the host immune system. Phage-displayed tumor antigens were enriched by biopanning with normal and then SCCHN-specific serum. Ninety-six phage clones were sequenced for identification, and 21 clones were validated using Luminex. One of these proteins, L23, a novel tumor antigen in SCCHN, was validated as an oncogene. L23 is upregulated in SCCHN compared with normal keratinocytes. Knockdown of L23 inhibited proliferation, invasion and cell survival. Overexpression of L23 had the reverse effect. Overexpression of L23 in non malignant cells led to transformation. Injection of SCCHN cells with knockdown of L23 in mice, induced tumors that were significantly smaller than control tumors. In conclusion, the immunomic screen yielded a panel of antigens specific to SCCHN; one of these proteins, L23, is a novel oncogene in SCCHN.


Asunto(s)
Antígenos de Neoplasias/genética , Autoanticuerpos/inmunología , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Proteínas Ribosómicas/genética , Animales , Apoptosis/genética , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/inmunología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Técnicas de Visualización de Superficie Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Queratinocitos/inmunología , Ratones , Ratones Desnudos , Células 3T3 NIH , Oncogenes , Valores de Referencia , Reproducibilidad de los Resultados , Proteínas Ribosómicas/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello
7.
Oncogene ; 30(42): 4339-49, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-21532618

RESUMEN

Rap1GAP is a critical tumor suppressor gene that is downregulated in multiple aggressive cancers, such as head and neck squamous cell carcinoma, melanoma and pancreatic cancer. However, the mechanistic basis of rap1GAP downregulation in cancers is poorly understood. By employing an integrative approach, we demonstrate polycomb-mediated repression of rap1GAP that involves Enhancer of Zeste Homolog 2 (EZH2), a histone methyltransferase in head and neck cancers. We further demonstrate that the loss of miR-101 expression correlates with EZH2 upregulation, and the concomitant downregulation of rap1GAP in head and neck cancers. EZH2 represses rap1GAP by facilitating the trimethylation of histone 3 at lysine 27, a mark of gene repression, and also hypermethylation of rap1GAP promoter. These results provide a conceptual framework involving a microRNA-oncogene-tumor suppressor axis to understand head and neck cancer progression.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Silenciador del Gen , Neoplasias de Cabeza y Cuello/metabolismo , MicroARNs/metabolismo , Factores de Transcripción/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2 , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Neoplasias de Cabeza y Cuello/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas
8.
Oncogene ; 29(39): 5370-80, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20622896

RESUMEN

The neuronal repellent SLIT2 is repressed in a number of cancer types primarily through promoter hypermethylation. SLIT2, however, has not been studied in prostate cancer. Through genome-wide location analysis we identified SLIT2 as a target of polycomb group (PcG) protein EZH2. The EZH2-containing polycomb repressive complexes bound to the SLIT2 promoter inhibiting its expression. SLIT2 was downregulated in a majority of metastatic prostate tumors, showing a negative correlation with EZH2. This repressed expression could be restored by methylation inhibitors or EZH2-suppressing compounds. In addition, a low level of SLIT2 expression was associated with aggressive prostate, breast and lung cancers. Functional assays showed that SLIT2 inhibited prostate cancer cell proliferation and invasion. Thus, this study showed for the first time the epigenetic silencing of SLIT2 in prostate tumors, and supported SLIT2 as a potential biomarker for aggressive solid tumors. Importantly, PcG-mediated repression may serve as a precursor for the silencing of SLIT2 by DNA methylation in cancer.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Represoras/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Neoplasias Pulmonares/genética , Masculino , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Represoras/genética , Factores de Transcripción/metabolismo
9.
Oncogene ; 27(58): 7274-84, 2008 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18806826

RESUMEN

Enhancer of zeste homolog 2 (EZH2) is a critical component of the polycomb-repressive complex 2 (PRC2), which is involved in gene silencing and histone H3 lysine 27 methylation. EZH2 has a master regulatory function in controlling such processes as stem cell differentiation, cell proliferation, early embryogenesis and X chromosome inactivation. Although benign epithelial cells express very low levels of EZH2, increased levels of EZH2 have been observed in aggressive solid tumors such as those of the prostate, breast and bladder. The mechanism by which EZH2 mediates tumor aggressiveness is unclear. Here, we demonstrate that EZH2 mediates transcriptional silencing of the tumor suppressor gene E-cadherin by trimethylation of H3 lysine 27. Histone deacetylase inhibitors can prevent EZH2-mediated repression of E-cadherin and attenuate cell invasion, suggesting a possible mechanism that may be useful for the development of therapeutic treatments. Taken together, these observations provide a novel mechanism of E-cadherin regulation and establish a functional link between dysregulation of EZH2 and repression of E-cadherin during cancer progression.


Asunto(s)
Cadherinas/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/genética , Línea Celular Transformada , Línea Celular Tumoral , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Silenciador del Gen , Histonas/metabolismo , Humanos , Masculino , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/patología , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Represoras/genética , Factores de Transcripción/genética
11.
J Pathol ; 212(1): 91-101, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17385188

RESUMEN

The TMPRSS2-ETS fusion prostate cancers comprise 50-70% of the prostate-specific antigen (PSA)-screened hospital-based prostate cancers examined to date, making it perhaps the most common genetic rearrangement in human cancer. The most common variant involves androgen-regulated TMPRSS2 and ERG, both located on chromosome 21. Emerging data from our group and others suggests that TMPRSS2-ERG fusion prostate cancer is associated with higher tumour stage and prostate cancer-specific death. The goal of this study was to determine if this common somatic alteration is associated with a morphological phenotype. We assessed 253 prostate cancer cases for TMPRSS2-ERG fusion status using an ERG break-apart FISH assay. Blinded to gene fusion status, two reviewers assessed each tumour for presence or absence of eight morphological features. Statistical analysis was performed to look for significant associations between morphological features and TMPRSS2-ERG fusion status. Five morphological features were associated with TMPRSS2-ERG fusion prostate cancer: blue-tinged mucin, cribriform growth pattern, macronucleoli, intraductal tumour spread, and signet-ring cell features, all with p-values < 0.05. Only 24% (n=30/125) of tumours without any of these features displayed the TMPRSS2-ERG fusion. By comparison, 55% (n=38/69) of cases with one feature (RR=3.88), 86% (n=38/44) of cases with two features (RR=20.06), and 93% (n=14/15) of cases with three or more features (RR=44.33) were fusion positive (p<0.001). To our knowledge, this is the first study that demonstrates a significant link between a molecular alteration in prostate cancer and distinct phenotypic features. The strength of these findings is similar to microsatellite unstable colon cancer and breast cancer involving BRCA1 and BRCA2 mutations. The biological effect of TMPRSS2-ERG overexpression may drive pathways that favour these common morphological features that pathologists observe daily. These features may also be helpful in diagnosing TMPRSS2-ERG fusion prostate cancer, which may have both prognostic and therapeutic implications.


Asunto(s)
Proteínas de Unión al ADN/genética , Fusión Génica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Serina Endopeptidasas/genética , Transactivadores/genética , Biomarcadores de Tumor/análisis , Biopsia con Aguja , Humanos , Hibridación Fluorescente in Situ , Interfase , Masculino , Persona de Mediana Edad , Mucina-1/análisis , Mucinas/análisis , Estadificación de Neoplasias , Fenotipo , Neoplasias de la Próstata/diagnóstico , Regulador Transcripcional ERG
12.
Oncogene ; 26(31): 4596-9, 2007 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-17237811

RESUMEN

The identification of the TMPRSS2:ERG fusion in prostate cancer suggests that distinct molecular subtypes may define risk for disease progression. In surgical series, TMPRSS2:ERG fusion was identified in 50% of the tumors. Here, we report on a population-based cohort of men with localized prostate cancers followed by expectant (watchful waiting) therapy with 15% (17/111) TMPRSS2:ERG fusion. We identified a statistically significant association between TMPRSS2:ERG fusion and prostate cancer specific death (cumulative incidence ratio=2.7, P<0.01, 95% confidence interval=1.3-5.8). Quantitative reverse-transcription-polymerase chain reaction demonstrated high ets-related [corrected] gene (ERG) expression to be associated with TMPRSS2:ERG fusion (P<0.005). These data suggest that TMPRSS2:ERG fusion prostate cancers may have a more aggressive phenotype, possibly mediated through increased ERG expression.


Asunto(s)
Proteínas de Fusión Oncogénica/metabolismo , Neoplasias de la Próstata/genética , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Fusión Génica , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa/métodos
13.
Oncogene ; 26(23): 3431-9, 2007 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-17146438

RESUMEN

We have recently reported that treatment with gemcitabine, a potent chemotherapeutic agent and radiation sensitizer, stimulates phosphorylation of the epidermal growth factor receptor (EGFR). Because phosphorylation of EGFR is known to precede receptor degradation, we hypothesized that gemcitabine treatment may also result in EGFR degradation. In two human head and neck cancer cell lines, UMSCC-1 and UMSCC-6, we demonstrated an approximately 80% decrease in total EGFR levels at 72 h after a 2-h treatment with 1 muM gemcitabine. Neither cisplatin nor 5-fluorouracil, which are used to treat head and neck cancer, caused EGFR degradation. EGFR downregulation did not occur at the level of transcription, as assessed by reverse transcription-polymerase chain reaction (RT-PCR), but instead occurred via phosphorylation and ubiquitination of the receptor along a proteosome/lysosome-mediated pathway. Inhibition of EGFR degradation, by either pretreatment with the EGFR tyrosine kinase inhibitor gefitinib or by exposure to the proteosome/lysosome inhibitor MG132, significantly reduced gemcitabine-induced cell death. These results suggest that EGFR degradation may be a novel mechanism for gemcitabine-mediated cell death. These findings also indicate that caution should be exercised when combining gemcitabine with agents that may prevent EGFR degradation, such as EGFR tyrosine kinase inhibitors administered in a suboptimal sequence or proteosome inhibitors.


Asunto(s)
Citotoxinas/toxicidad , Desoxicitidina/análogos & derivados , Receptores ErbB/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Desoxicitidina/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Lisosomas/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , Transcripción Genética/genética , Ubiquitina/metabolismo , Gemcitabina
14.
Br J Cancer ; 95(4): 425-30, 2006 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-16880794

RESUMEN

Molecular alterations in the prostate cancer proteome mediate the functional and phenotypic transformation from clinically localised to metastatic cancer, a transition that drives patient's mortality and challenges therapeutic intervention. A first approximation of differential proteomic alterations stratified by disease stage has yielded repertoires of potential diagnostic and prognostic markers, multiplex signatures of predictive value, and yield fundamental insight into molecular commonalities in cancer progression. Deciphering these causative proteomic alterations from the molecular noise will continue to mature our understanding of tumour biology and drive new computational and integrative approaches to model a system's view that accommodates the heterogeneity of prostate cancer progression.


Asunto(s)
Perfilación de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Biomarcadores de Tumor/análisis , Progresión de la Enfermedad , Humanos , Masculino , Modelos Biológicos , Metástasis de la Neoplasia/diagnóstico
15.
Histopathology ; 47(6): 587-96, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16324196

RESUMEN

AIM: Studies using a monoclonal (P504S) and a polyclonal antibody (p-AMACR) to alpha-methylacyl-CoA racemase (AMACR) have shown variable expression in prostate cancer (PCa). The goal is to compare the sensitivity of both antibodies in PCa and evaluate their utility in the work-up of atypical prostate needle biopsies (NBXs). METHODS AND RESULTS: A tissue microarray (TMA) with 248 samples of benign prostate, high-grade prostatic intraepithelial neoplasia (HGPIN) and PCa samples, 20 NBXs with minute PCa and 32 NBXs with 'atypical' foci were stained with P504S and p-AMACR. Ninety percent of PCa (76/76 TMA, 16/20 NBXs) showed predominantly strong p-AMACR expression while 87% (65/69 TMA, 16/20 NBXs) showed variable P504S expression (sensitivity 90% versus 87%, P = 0.10). In HGPIN, P504S and p-AMACR were positive in 77% and 91% of samples, respectively. In the 'atypical' NBXs group, 53% were classified as PCa, 12% benign and 35% atypical, suspicious for PCa, after review of the basal marker. Of atypical, suspicious for PCa, P504S/p-AMACR helped convert the diagnosis to PCa in 5/11 (45%) cases, where, despite negative basal cell markers, morphology was less than optimal. CONCLUSIONS: Differences between P504S and p-AMACR appear marginal and clinically insignificant. AMACR is negative in a subset of unequivocal minute PCa with both antibodies. However, when utilized in proper context, AMACR may offer significant advantage in converting an 'atypical' diagnosis to PCa where morphology and basal markers are less than optimal in resolving the diagnosis.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Anticuerpos/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Racemasas y Epimerasas/metabolismo , Biopsia con Aguja , Humanos , Inmunohistoquímica , Masculino , Hiperplasia Prostática/enzimología , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/cirugía , Neoplasia Intraepitelial Prostática/enzimología , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Neoplasia Intraepitelial Prostática/cirugía , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/cirugía , Sensibilidad y Especificidad
16.
Am J Pathol ; 159(4): 1199-209, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11583946

RESUMEN

During sepsis the host's system-wide response to microbial invasion seems dysregulated. Here we explore the diverse multiorgan transcriptional programs activated during systemic inflammation in a cecal ligation/puncture model of sepsis in rats. Using DNA microarrays representing 7398 genes, we examined the temporal sequence of sepsis-induced gene expression patterns in major organ systems including lung, liver, kidney, thymus, spleen, and brain. Although genes known to be associated with systemic inflammation were identified by our global transcript analysis, many genes and expressed sequence tags not previously linked to the septic response were also elucidated. Taken together, our results suggest activation of a highly complex transcriptional response in individual organs of the septic animal. Several overlying themes emerged from our genome-scale analysis that includes 1) the sepsis response elicited gene expression profiles that were either organ-specific, common to more than one organ, or distinctly opposite in some organs; 2) the brain is protected from sepsis-induced gene activation relative to other organs; 3) the thymus and spleen have an interesting cohort of genes with opposing gene expression patterns; 4) genes with proinflammatory effects were often balanced by genes with anti-inflammatory effects (eg, interleukin-1beta/decoy receptor, xanthine oxidase/superoxide dismutase, Ca2+-dependent PLA2/Ca2+-independent PLA2); and 5) differential gene expression was observed in proteins responsible for preventing tissue injury and promoting homeostasis including anti-proteases (TIMP-1, Cpi-26), oxidant neutralizing enzymes (metallothionein), cytokine decoy receptors (interleukin-1RII), and tissue/vascular permeability factors (aquaporin 5, vascular endothelial growth factor). This global perspective of the sepsis response should provide a molecular framework for future research into the pathophysiology of systemic inflammation. Understanding, on a genome scale, how an organism responds to infection, may facilitate the development of enhanced detection and treatment modalities for sepsis.


Asunto(s)
Perfilación de la Expresión Génica , Infecciones/genética , Animales , ADN Complementario/genética , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Long-Evans
17.
Cancer Res ; 61(20): 7585-93, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11606398

RESUMEN

The advent of DNA microarray technology will likely have a major impact on the molecular classification and understanding of human cancer. Obtaining a global perspective of proteins expressed in cancer cells is considerably more challenging. Here we describe a microarray-based platform that can be used to measure protein levels and activities in a complex biological milieu such as a cellular lysate. Using a protein microarray made up of 1920 elements (146 distinct antibodies) we were able to monitor alterations of protein levels in LoVo colon carcinoma cells treated with ionizing radiation. The protein microarray approach revealed radiation-induced up-regulation of apoptotic regulators including p53, DNA fragmentation factor 40/caspase activated DNase, DNA fragmentation factor 45/inhibitor of caspase activated DNase, tumor necrosis factor-related apoptosis-inducing ligand, death receptor 5, decoy receptor 2, FLICE-like inhibitory protein, signal transducers and activators of transcription 1alpha, and uncoupling protein 2, among others. Consistent with this observation, an increased percentage of apoptosis was observed in irradiated LoVo cells. Interestingly, we also observed radiation-induced down-regulation of carcinoembryonic antigen, a prototypic cancer biomarker. Selected proteins assessed by microarray were validated by traditional immunoblotting. Taken together, our work suggests that protein/antibody microarrays will facilitate high-throughput proteomic studies of human cancer and carcinogenesis.


Asunto(s)
Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Proteínas de Neoplasias/biosíntesis , Anticuerpos Antineoplásicos , Antígenos de Neoplasias , Proteínas Reguladoras de la Apoptosis , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/genética , Neoplasias del Colon/genética , Desoxirribonucleasas/biosíntesis , Desoxirribonucleasas/genética , Regulación hacia Abajo/efectos de la radiación , Humanos , Proteínas de Neoplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/biosíntesis , Receptores del Factor de Necrosis Tumoral/genética , Reproducibilidad de los Resultados , Células Tumorales Cultivadas/efectos de la radiación , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
18.
Nature ; 412(6849): 822-6, 2001 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-11518967

RESUMEN

Prostate cancer is the most frequently diagnosed cancer in American men. Screening for prostate-specific antigen (PSA) has led to earlier detection of prostate cancer, but elevated serum PSA levels may be present in non-malignant conditions such as benign prostatic hyperlasia (BPH). Characterization of gene-expression profiles that molecularly distinguish prostatic neoplasms may identify genes involved in prostate carcinogenesis, elucidate clinical biomarkers, and lead to an improved classification of prostate cancer. Using microarrays of complementary DNA, we examined gene-expression profiles of more than 50 normal and neoplastic prostate specimens and three common prostate-cancer cell lines. Signature expression profiles of normal adjacent prostate (NAP), BPH, localized prostate cancer, and metastatic, hormone-refractory prostate cancer were determined. Here we establish many associations between genes and prostate cancer. We assessed two of these genes-hepsin, a transmembrane serine protease, and pim-1, a serine/threonine kinase-at the protein level using tissue microarrays consisting of over 700 clinically stratified prostate-cancer specimens. Expression of hepsin and pim-1 proteins was significantly correlated with measures of clinical outcome. Thus, the integration of cDNA microarray, high-density tissue microarray, and linked clinical and pathology data is a powerful approach to molecular profiling of human cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Serina Endopeptidasas/metabolismo , Biomarcadores de Tumor/genética , ADN Complementario , ADN de Neoplasias , Humanos , Inmunohistoquímica , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-pim-1 , Valores de Referencia , Serina Endopeptidasas/genética
19.
Proc Natl Acad Sci U S A ; 97(4): 1754-9, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10677530

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent endogenous activator of the cell death pathway and functions by activating the cell surface death receptors 4 and 5 (DR4 and DR5). TRAIL is nontoxic in vivo and preferentially kills neoplastically transformed cells over normal cells by an undefined mechanism. Radiotherapy is a common treatment for breast cancer as well as many other cancers. Here we demonstrate that ionizing radiation can sensitize breast carcinoma cells to TRAIL-induced apoptosis. This synergistic effect is p53-dependent and may be the result of radiation-induced up-regulation of the TRAIL-receptor DR5. Importantly, TRAIL and ionizing radiation have a synergistic effect in the regression of established breast cancer xenografts. Changes in tumor cellularity and extracellular space were monitored in vivo by diffusion-weighted magnetic resonance imaging (diffusion MRI), a noninvasive technique to produce quantitative images of the apparent mobility of water within a tissue. Increased water mobility was observed in combined TRAIL- and radiation-treated tumors but not in tumors treated with TRAIL or radiation alone. Histological analysis confirmed the loss of cellularity and increased numbers of apoptotic cells in TRAIL- and radiation-treated tumors. Taken together, our results provide support for combining radiation with TRAIL to improve tumor eradication and suggest that efficacy of apoptosis-inducing cancer therapies may be monitored noninvasively, using diffusion MRI.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Glicoproteínas de Membrana/uso terapéutico , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/efectos de la radiación , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Imagen por Resonancia Magnética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos , Trasplante de Neoplasias , Radiación Ionizante , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
20.
Neoplasia ; 1(1): 5-15, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10935465

RESUMEN

Apoptosis is a fundamental biologic process by which metazoan cells orchestrate their own self-demise. Genetic analyses of the nematode C elegans identified three core components of the suicide apparatus which include CED-3, CED-4, and CED-9. An analogous set of core constituents exists in mammalian cells and includes caspase-9, Apaf-1, and bcl-2/xL, respectively. CED-3 and CED-4, along with their mammalian counterparts, function to kill cells, whereas CED-9 and its mammalian equivalents protect cells from death. These central components biochemically intermingle in a ternary complex recently dubbed the "apoptosome." The C elegans protein EGL-1 and its mammalian counterparts, pro-apoptotic members of the bcl-2 family, induce cell death by disrupting apoptosome interactions. Thus, EGL-1 may represent a primordial signal integrator for the apoptosome. Various biochemical processes including oligomerization, adenosine triphosphate ATP/dATP binding, and cytochrome c interaction play a role in regulating the ternary death complex. Recent studies suggest that cell death receptors, such as CD95, may amplify their suicide signal by activating the apoptosome. These mutual associations by core components of the suicide apparatus provide a molecular framework in which diverse death signals likely interface. Understanding the apoptosome and its cellular connections will facilitate the design of novel therapeutic strategies for cancer and other disease states in which apoptosis plays a pivotal role.


Asunto(s)
Apoptosis , Proteínas de Caenorhabditis elegans , Adenosina Trifosfato/metabolismo , Animales , Proteínas de Unión al Calcio/fisiología , Caspasa 8 , Caspasa 9 , Caspasas/fisiología , Cisteína Endopeptidasas/fisiología , Proteínas del Helminto/fisiología , Humanos , Proteínas Represoras/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA