Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Oncotarget ; 11(46): 4201-4223, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33245731

RESUMEN

COVID-19 affects vulnerable populations including elderly individuals and patients with cancer. Natural Killer (NK) cells and innate-immune TRAIL suppress transformed and virally-infected cells. ACE2, and TMPRSS2 protease promote SARS-CoV-2 infectivity, while inflammatory cytokines IL-6, or G-CSF worsen COVID-19 severity. We show MEK inhibitors (MEKi) VS-6766, trametinib and selumetinib reduce ACE2 expression in human cells. In some human cells, remdesivir increases ACE2-promoter luciferase-reporter expression, ACE2 mRNA and protein, and ACE2 expression is attenuated by MEKi. In serum-deprived and stimulated cells treated with remdesivir and MEKi we observed correlations between pRB, pERK, and ACE2 expression further supporting role of proliferative state and MAPK pathway in ACE2 regulation. We show elevated cytokines in COVID-19-(+) patient plasma (N = 9) versus control (N = 11). TMPRSS2, inflammatory cytokines G-CSF, M-CSF, IL-1α, IL-6 and MCP-1 are suppressed by MEKi alone or with remdesivir. We observed MEKi stimulation of NK-cell killing of target-cells, without suppressing TRAIL-mediated cytotoxicity. Pseudotyped SARS-CoV-2 virus with a lentiviral core and SARS-CoV-2 D614 or G614 SPIKE (S) protein on its envelope infected human bronchial epithelial cells, small airway epithelial cells, or lung cancer cells and MEKi suppressed infectivity of the pseudovirus. We show a drug class-effect with MEKi to stimulate NK cells, inhibit inflammatory cytokines and block host-factors for SARS-CoV-2 infection leading also to suppression of SARS-CoV-2-S pseudovirus infection of human cells. MEKi may attenuate SARS-CoV-2 infection to allow immune responses and antiviral agents to control disease progression.

2.
bioRxiv ; 2020 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-32793908

RESUMEN

COVID-19 affects vulnerable populations including elderly individuals and patients with cancer. Natural Killer (NK) cells and innate-immune TRAIL suppress transformed and virally-infected cells. ACE2, and TMPRSS2 protease promote SARS-CoV-2 infectivity, while inflammatory cytokines IL-6, or G-CSF worsen COVID-19 severity. We show MEK inhibitors (MEKi) VS-6766, trametinib and selumetinib reduce ACE2 expression in human cells. In some human cells, remdesivir increases ACE2-promoter luciferase-reporter expression, ACE2 mRNA and protein, and ACE2 expression is attenuated by MEKi. In serum-deprived and stimulated cells treated with remdesivir and MEKi we observed correlations between pRB, pERK, and ACE2 expression further supporting role of proliferative state and MAPK pathway in ACE2 regulation. We show elevated cytokines in COVID-19-(+) patient plasma (N=9) versus control (N=11). TMPRSS2, inflammatory cytokines G-CSF, M-CSF, IL-1α, IL-6 and MCP-1 are suppressed by MEKi alone or with remdesivir. We observed MEKi stimulation of NK-cell killing of target-cells, without suppressing TRAIL-mediated cytotoxicity. Pseudotyped SARS-CoV-2 virus with a lentiviral core and SARS-CoV-2 D614 or G614 SPIKE (S) protein on its envelope infected human bronchial epithelial cells, small airway epithelial cells, or lung cancer cells and MEKi suppressed infectivity of the pseudovirus. We show a drug class-effect with MEKi to stimulate NK cells, inhibit inflammatory cytokines and block host-factors for SARS-CoV-2 infection leading also to suppression of SARS-CoV-2-S pseudovirus infection of human cells. MEKi may attenuate SARS-CoV-2 infection to allow immune responses and antiviral agents to control disease progression.

3.
Clin Cancer Res ; 13(14): 4225-32, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17634552

RESUMEN

PURPOSE: 1-beta-D-Arabinofuranosylcytosine (cytarabine; ara-C) is the most active agent in myeloid leukemia. 5-Aza-2'-deoxycytidine (DAC) is a cytosine analogue that inhibits DNA methylation and also has activity in myeloid leukemia. Therefore, we investigated combining these two drugs in human leukemia cell lines in vitro. EXPERIMENTAL DESIGN: We initially examined the effects of ara-C and DAC on human leukemia cell lines HL60, ML-1, RAji, and Jurkat. We measured IC(50) of DAC and ara-C in these cell lines and calculated a combination index of these two drugs given either simultaneously or sequentially. In searching for mechanisms relative to epigenetic regulation for this effect, we examined DNA methylation of LINE and Alu repetitive elements as a surrogate for global genomic DNA methylation. In addition, we sorted Annexin V positive and negative cells and measured differences in LINE methylation between them. RESULTS: The combination of DAC and ara-C showed additive induction of cell death in ML-1 and synergistic induction in HL60, Raji, and Jurkat. Sequentially, DAC followed by ara-C was a synergistic combination in all cell lines. Low-dose DAC induced more hypomethylation than high doses of the drug, whereas ara-C had no effects on methylation. The combination of ara-C with DAC either together or DAC followed by ara-C resulted in inhibition of LINE demethylation in HL60. The RIL gene, which is silenced by DNA hypermethylation, was activated by DAC, but the addition of ara-C to DAC reduced RIL gene activation. DAC treatment increased H3 Lys(9) acetylation of Alu elements, whereas ara-C had no effect, and the addition of ara-C to DAC inhibited this effect. Finally, we showed that after DAC exposure, Annexin V positive cells were more hypomethylated than Annexin V negative cells. CONCLUSION: The combination of DAC and ara-C showed additive or synergistic effects on cell death in four human leukemia cell lines in vitro, but antagonism in terms of epigenetic effects. One possible explanation for these paradoxical observations is that hypomethylated cells are sensitized to cell killing by ara-C. These data suggest that DAC used in combination with ara-C has clinical potential in the treatment of acute myeloid leukemia.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Citarabina/farmacología , Células HL-60/efectos de los fármacos , Anexina A5/metabolismo , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , Decitabina , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Cancer Biol Ther ; 5(11): 1494-501, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17369752

RESUMEN

The epidermal growth factor receptor (EGFR) is a member of the HER/ERB-B family of transmembrane receptor kinases. Overexpression of EGFR confers advantages in cell proliferation, survival, and migration and correlates with decreased survival in multiple solid tumors. However, a proportion of these malignancies have little or no expression of EGFR. CpG island hypermethylation and associated transcriptional silencing are common in solid tumors. The methylation status of the EGFR CpG island was examined in a series of cell lines and tissues. Dense EGFR methylation (90%) was found in the breast cancer cell line CAMA1, and a moderate degree of methylation (30-50%) was observed in the breast cancer cell lines MB435 and MB453. Transcriptional silencing of EGFR in these cell lines closely correlated with methylation. By contrast, no methylation of the HER-2/ neu CpG island was detected. EGFR hypermethylation was also found in a subset of unselected primary breast (20%), head and neck squamous cell carcinoma (35%), and lung tumors (11%). Treatment with decitabine resulted in the reexpression of EGFR in CAMA1 and MB453. Both cell lines are relatively resistant to killing by the EGFR inhibitor gefitinib. However, after cotreatment with decitabine and gefitinib, a significant effect on the induction of apoptosis was observed. In conclusion, EGFR is hypermethylated and silenced in a subset of solid tumor cell lines and primary tumor specimens, and cotreatment with decitabine and gefitinib has an additive effect only in EGFR methylated breast cancer cell lines.


Asunto(s)
Línea Celular Tumoral , Metilación de ADN , Fosfatos de Dinucleósidos/genética , Epigénesis Genética , Receptores ErbB/genética , Terapia Genética/métodos , Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Azacitidina/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular , Cartilla de ADN , Decitabina , Femenino , Gefitinib , Humanos , Especificidad de Órganos , Quinazolinas/uso terapéutico , Mapeo Restrictivo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Cancer Res ; 64(7): 2411-7, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15059893

RESUMEN

A variety of tumor suppressor genes are down-regulated by hypermethylation during carcinogenesis. Using methylated CpG amplification-representation difference analysis, we identified a DNA fragment corresponding to the Tazarotene-induced gene 1 (TIG1) promoter-associated CpG island as one of the genes hypermethylated in the leukemia cell line K562. Because TIG1 has been proposed to act as a tumor suppressor, we tested the hypothesis that cytosine methylation of the TIG1 promoter suppresses its expression and causes a loss of responsiveness to retinoic acid in some neoplastic cells. We examined TIG1 methylation and expression status in 53 human cancer cell lines and 74 primary tumors, including leukemia and head and neck, breast, colon, skin, brain, lung, and prostate cancer. Loss of TIG1 expression was strongly associated with TIG1 promoter hypermethylation (P < 0.001). There was no correlation between TIG1 promoter methylation and that of retinoid acid receptor beta2 (RARbeta2), another retinoic-induced putative tumor suppressor gene (P = 0.78). Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine for 5 days restored TIG1 expression in all eight silenced cell lines tested. TIG1 expression was also inducible by treatment with 1 micro M all-trans-retinoic acid for 3 days except in densely methylated cell lines. Treatment of the K562 leukemia cells with demethylating agent combined with all-trans-retinoic acid induced apoptosis. These findings indicate that silencing of TIG1 promoter by hypermethylation is common in human cancers and may contribute to the loss of retinoic acid responsiveness in some neoplastic cells.


Asunto(s)
Azacitidina/análogos & derivados , Metilación de ADN , Silenciador del Gen , Neoplasias/genética , Proteínas/genética , Antineoplásicos/farmacología , Azacitidina/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Decitabina , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Proteínas de la Membrana , Neoplasias/metabolismo , Neoplasias/patología , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Receptores de Ácido Retinoico/genética , Tretinoina/farmacología
6.
Clin Cancer Res ; 8(12): 3824-31, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12473596

RESUMEN

PURPOSE: Premature death associated with colorectal adenocarcinoma occurs in PIK3CG(-/-) mice and overexpression of PIK3CG in colon cancer cells suppresses cell proliferation. We examined expression levels of PIK3CG, a catalytic subunit of phosphatidylinositide 3-OH kinase (PI3K), in colon cancer cells to investigate the hypothesis that PIK3CG might contribute to the growth and progression of colorectal cancers. EXPERIMENTAL DESIGN: The effects of LY294002, a PI3K inhibitor, on cell growth were examined to elucidate the role of the PI3K-Akt/protein kinase B (PKB) pathway in colon cancer. We used reverse transcription-PCR, Western blotting, and immunohistochemical analyses to examine PIK3CG mRNA and protein expression levels in colon cancer cells and primary colorectal cancers. To clarify the mechanism responsible for the silencing of this gene in colon cancers, we performed methylation-sensitive PCR analysis of DNA digested with HpaII and MspI and analyzed PI3KCG expression in DLD-1 and LoVo cells treated with the demethylating agent 5-aza-2'-deoxycytidine (5-Aza). RESULTS: LY294002 suppressed growth and decreased expression of Akt (Ser(473)) expression in cancer cells. Three (60%) of 5 colon cancer cell lines did not express PIK3CG, but hypermethylation at CpG sites of the promoter regions of this gene was detected. However, 5-Aza-treated DLD-1 and LoVo cells did express PIK3CG. Reduction of PIK3CG expression was detected immunohistochemically in 85% of human colorectal cancers and was closely associated with invasion, metastasis, and poor differentiation. Down-regulation of PIK3CG expression and hypermethylation of promoter regions were also detected in primary colon cancers. CONCLUSIONS: Our findings suggest that the silencing of the PIK3CG gene plays an important role in inhibiting the PI3K-Akt/PKB signaling system responsible for tumorigenesis and the progression of colorectal cancers.


Asunto(s)
Neoplasias Colorrectales/enzimología , Islas de CpG/genética , Metilación de ADN , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Azacitidina/farmacología , Cromonas/farmacología , Cromosomas Humanos Par 7/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN de Neoplasias/metabolismo , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Subunidades de Proteína , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología , Células Tumorales Cultivadas/patología
7.
Clin Cancer Res ; 10(5): 1733-42, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15014026

RESUMEN

PURPOSE: Retinoic acid receptor-beta(2) (RAR-beta(2)) expression is suppressed in oral premalignant lesions and head and neck squamous cell carcinomas (HNSCCs). This study was conducted to determine whether RAR-beta(2) gene expression in such lesions can be silenced by promoter methylation. EXPERIMENTAL DESIGN: RAR-beta(2) methylation was analyzed in DNA samples from 22 pairs of primary HNSCC and adjacent normal epithelium, 124 samples of oral leukoplakia, and 18 HNSCC cell lines using methylation-specific PCR. RAR-beta(2) promoter was methylated in 67, 56, and 53% of HNSCC tumors, HNSCC cell lines, and microdissected oral leukoplakia specimens, respectively. RAR-beta(2) hypermethylation was confirmed by sodium bisulfite-PCR combined with restriction enzyme digestion analysis and by random cloning and sequencing of bisulfite-treated DNA isolates. RESULTS: Significantly higher RAR-beta(2) hypermethylation levels were found in tumor tissue compared with adjacent normal tissue (P = 0.002). RAR-beta(2) methylation in the cell lines was correlated with loss of RAR-beta(2) expression (P = 0.013) and inversely related to the presence of mutated p53 (P = 0.025). The demethylating agent 5-aza-2'-deoxycytidine (5-aza-CdR) restored RAR-beta(2) inducibility by all-trans-retinoic acid (ATRA) in some of the cell lines, which posses a methylated RAR-beta(2) promoter. In some cell lines, this effect was associated with increased growth inhibition after combined treatment with 5-aza-CdR and ATRA. CONCLUSIONS: RAR-beta(2) silencing by methylation is an early event in head and neck carcinogenesis; 5-Aza-CdR can restore RAR-beta(2) inducibility by ATRA in most cell lines, and the combination of 5-aza-CdR and ATRA is more effective in growth inhibition than single agents.


Asunto(s)
ADN de Neoplasias/genética , Neoplasias de Cabeza y Cuello/genética , Receptores de Ácido Retinoico/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , División Celular , Línea Celular Tumoral , Clonación Molecular , Metilación de ADN , Cartilla de ADN , Femenino , Silenciador del Gen , Neoplasias de Cabeza y Cuello/patología , Humanos , Leucoplasia Bucal/genética , Leucoplasia Bucal/patología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Reacción en Cadena de la Polimerasa , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Regiones Promotoras Genéticas/genética , Receptores de Ácido Retinoico/efectos de los fármacos , Tretinoina/farmacología
8.
Cancer Biol Ther ; 3(1): 82-6, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14726683

RESUMEN

Tumor suppressor genes can become inactivated in cancer via hypermethylation of their promoter. The retinoic acid receptor beta (RARbeta) gene is expressed from two distinct promoters, both of which have CpG islands. RARbeta1 is expressed primarily during embryogenesis, whereas RARbeta2 is expressed in adult tissues and hypermethylated in a number of cancer cells. We used combined bisulfite restriction analysis to evaluate their methylation in colorectal mucosa and tumors. Methylation of RARbeta1 was detected, with a mean of 2% in normal colon tissues in young subjects (< 32 years), and 16% in older subjects (> 75 years) (P < 0.001). Using paired normal/tumor tissue samples, we found higher mean methylation rate in tumors than in adjacent normal tissue (mean, 46% versus 16%; P < 0.001) and hypermethylation of RARbeta1 in all eight cell lines examined. By RT-PCR, RARbeta1 was not expressed in normal adult colon tissues and its expression could not be efficiently activated in most cell lines by the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR). RARbeta2 methylation was also observed in normal colon tissues and was lower in young individuals than in older ones (mean, 11% versus 23%; P < 0.05). Among paired samples, RARbeta2 methylation was higher in tumor tissue than in normal tissue in 14 cases, vice versa in 7 cases, and equal in 6 cases. All eight cell lines were hypermethylated and did not express RARbeta2, but RARbeta2 expression could be reactivated easily by 5-Aza-CdR. We suggest that the embryonic RARbeta1 isoform is readily hypermethylated in aging colon mucosa and all colorectal cancers because of its lack of expression in normal tissues. The adult RARbeta2 isoform also shows age-related methylation in normal tissues but more variable methylation in colorectal cancer, perhaps because its expression offers continued protection against methylation or its silencing does not provide a selective advantage in the early stages of the disease.


Asunto(s)
Neoplasias del Colon/genética , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica/genética , Receptores de Ácido Retinoico/genética , Secuencia de Bases , División Celular/genética , Línea Celular Tumoral , Metilación de ADN , Cartilla de ADN , ADN de Neoplasias/aislamiento & purificación , Humanos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética
9.
Cancer Biol Ther ; 3(10): 1002-6, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467435

RESUMEN

Retinoids have shown significant activities in cancer prevention and therapy. Many of their effects are mediated by nuclear retinoid receptors including retinoic acid receptors (RARs alpha, beta and gamma) and retinoid X receptors (RXRs alpha, beta and gamma). Human retinoic acid receptor beta (RARbeta) has three different isoforms: beta1, beta2 and beta4. The tumor suppressive characteristics of RARbeta2, its silencing by promoter hypermethylation, and its reexpression following demethylation have been reported. In contrast, RARbeta1, an embryonic isoform with restricted expression in adult tissues has been linked to carcinogenesis. However, factors regulating RARbeta1 expression have not yet been clarified. During studies on the head and neck squamous cell carcinoma cells, we found that the expression of RARbeta increased in cells grown to high density. Real-time reverse-transcriptase polymerase chain reaction revealed that the isoform increased in these cells was RARbeta1. Epigenetic modifications of this isoform were tested using combined bisulfite restriction analysis and chromatin immunoprecipitation assays. The UMSCC38 cell line showed significant RARbeta1 expression (p < 0.001), which was dependent on cell density and culture duration. The increased expression of RARbeta1 was not due to demethylation of its promoter. However, higher cell densities were associated with increased acetylation of histone 3 at lysine 9 in RARbeta1 but not in RARbeta2. These findings reveal that the expression of RARbeta1 is regulated by cell density through changes in histone acetylation.


Asunto(s)
Carcinoma de Células Escamosas/genética , Ensamble y Desensamble de Cromatina/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Receptores de Ácido Retinoico/genética , Acetilación , Recuento de Células , Inmunoprecipitación de Cromatina , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA