Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(9): 4857-4871, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38647050

RESUMEN

CpG islands near promoters are normally unmethylated despite being surrounded by densely methylated regions. Aberrant hypermethylation of these CpG islands has been associated with the development of various human diseases. Although local genetic elements have been speculated to play a role in protecting promoters from methylation, only a limited number of methylation barriers have been identified. In this study, we conducted an integrated computational and experimental investigation of colorectal cancer methylomes. Our study revealed 610 genes with disrupted methylation barriers. Genomic sequences of these barriers shared a common 41-bp sequence motif (MB-41) that displayed homology to the chicken HS4 methylation barrier. Using the CDKN2A (P16) tumor suppressor gene promoter, we validated the protective function of MB-41 and showed that loss of such protection led to aberrant hypermethylation. Our findings highlight a novel sequence signature of cis-acting methylation barriers in the human genome that safeguard promoters from silencing.


Asunto(s)
Neoplasias Colorrectales , Metilación de ADN , Regiones Promotoras Genéticas , Animales , Humanos , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Islas de CpG , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Genoma Humano , Motivos de Nucleótidos , Pollos , Estudio de Asociación del Genoma Completo
2.
Artículo en Inglés | MEDLINE | ID: mdl-32247558

RESUMEN

Cadmium oxide nanoparticles (CdO NPs) are among some of the most studied and industrially used metal oxide NPs. They have been widely used for industrial application, such as paint pigments and electronic devices, and medical therapeutics. With increasing use of CdO NPs and concerns for their potential adverse effects on the environment and public health, evaluation of the cytotoxicity and genotoxicity of CdO NPs becomes very important. To date, there is a limited understanding of the potential hazard brought by CdO NPs and a lack of information and research, particularly on the genotoxicity assessment of these NPs. In this study, 10 nm CdO core-PEG stabilized NPs were synthesized, characterized and used for evaluation of CdO NPs' cytotoxicity and genotoxicity. Release of cadmium ions (Cd+2) from the CdO NPs in cell culture medium, cellular uptake of the NPs, and the endotoxin content of the particles were measured prior to the toxicity assays. Cytotoxicity was evaluated using the MTS assay, ATP content detection assay, and LDH assay. Genotoxicity was assessed using the Ames test, Comet assay, micronucleus assay, and mouse lymphoma assay. The cytotoxicity of cadmium chloride (CdCl2) was also evaluated along with that of the CdO NPs. The results showed that endotoxin levels within the CdO NPs were below the limit of detection. CdO NPs induced concentration-dependent cytotoxicity in TK6 and HepG2 cells with the MTS, ATP and LDH assays. Although the genotoxicity of CdO NPs was negative in the Ames test, positive results were obtained with the micronucleus, Comet, and mouse lymphoma assays. The negative response of CdO NPs with the Ames test may be the result of unsuitability of the assay for measuring NPs, while the positive responses from other genotoxicity assays suggest that CdO NPs can induce chromosomal damage, single or double strand breaks in DNA, and mutations. The toxicity of the CdO NPs results from the NPs themselves and not from the released Cd+2, because the ions released from the NPs were minimal. These results demonstrate that CdO NPs are cytotoxic and genotoxic and provide new insights into risk assessment of CdO NPs for human exposure and environmental protection.


Asunto(s)
Compuestos de Cadmio/toxicidad , Nanopartículas del Metal/toxicidad , Pruebas de Mutagenicidad , Mutágenos/toxicidad , Óxidos/toxicidad , Animales , Compuestos de Cadmio/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN/efectos de los fármacos , Humanos , Nanopartículas del Metal/química , Ratones , Mutágenos/farmacología , Óxidos/farmacología
3.
PLoS One ; 13(7): e0200169, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29979778

RESUMEN

Estimation of remaining capacity is essential for ensuring the safety and reliability of lithium-ion batteries. In actual operation, batteries are seldom fully discharged. For a constant current-constant voltage charging mode, the incomplete discharging process affects not only the initial state but also processed variables of the subsequent charging profile, thereby mainly limiting the applications of many feature-based capacity estimation methods which rely on a whole cycling process. Since the charging information of the constant voltage profile can be completely saved whether the battery is fully discharged or not, a geometrical feature of the constant voltage charging profile is extracted to be a new aging feature of lithium-ion batteries under the incomplete discharging situation in this work. By introducing the quantum computing theory into the classical machine learning technique, an integrated quantum particle swarm optimization-based support vector regression estimation framework, as well as its application to characterize the relationship between extracted feature and battery remaining capacity, are presented and illustrated in detail. With the lithium-ion battery data provided by NASA, experiment and comparison results demonstrate the effectiveness, accuracy, and superiority of the proposed battery capacity estimation framework for the not entirely discharged condition.


Asunto(s)
Suministros de Energía Eléctrica , Litio , Suministros de Energía Eléctrica/estadística & datos numéricos , Electricidad , Electroquímica , Iones , Reproducibilidad de los Resultados , Máquina de Vectores de Soporte
4.
Arch Toxicol ; 91(1): 509-519, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27180073

RESUMEN

In spite of many reports on the toxicity of silver nanoparticles (AgNPs), the mechanisms underlying the toxicity are far from clear. A key question is whether the observed toxicity comes from the silver ions (Ag+) released from the AgNPs or from the nanoparticles themselves. In this study, we explored the genotoxicity and the genotoxicity mechanisms of Ag+ and AgNPs. Human TK6 cells were treated with 5 nM AgNPs or silver nitrate (AgNO3) to evaluate their genotoxicity and induction of oxidative stress. AgNPs and AgNO3 induced cytotoxicity and genotoxicity in a similar range of concentrations (1.00-1.75 µg/ml) when evaluated using the micronucleus assay, and both induced oxidative stress by measuring the gene expression and reactive oxygen species in the treated cells. Addition of N-acetylcysteine (NAC, an Ag+ chelator) to the treatments significantly decreased genotoxicity of Ag+, but not AgNPs, while addition of Trolox (a free radical scavenger) to the treatment efficiently decreased the genotoxicity of both agents. In addition, the Ag+ released from the highest concentration of AgNPs used for the treatment was measured. Only 0.5 % of the AgNPs were ionized in the culture medium and the released silver ions were neither cytotoxic nor genotoxic at this concentration. Further analysis using electron spin resonance demonstrated that AgNPs produced hydroxyl radicals directly, while AgNO3 did not. These results indicated that although both AgNPs and Ag+ can cause genotoxicity via oxidative stress, the mechanisms are different, and the nanoparticles, but not the released ions, mainly contribute to the genotoxicity of AgNPs.


Asunto(s)
Linfocitos/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Mutágenos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Plata/toxicidad , Acetilcisteína/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quelantes/farmacología , Cromanos/farmacología , Depuradores de Radicales Libres/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Linfocitos/enzimología , Linfocitos/metabolismo , Pruebas de Micronúcleos , Mutágenos/análisis , Mutágenos/química , Tamaño de la Partícula , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Plata/análisis , Plata/química , Nitrato de Plata/antagonistas & inhibidores , Nitrato de Plata/toxicidad , Solubilidad , Propiedades de Superficie
5.
Epigenetics ; 10(11): 1006-13, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26440216

RESUMEN

Oxidation of 5-methylcytosine by TET family proteins can induce DNA replication-dependent (passive) DNA demethylation and base excision repair (BER)-based (active) DNA demethylation. The balance of active vs. passive TET-induced demethylation remains incompletely determined. In the context of large scale DNA demethylation, active demethylation may require massive induction of the DNA repair machinery and thus compromise genome stability. To study this issue, we constructed a tetracycline-controlled TET-induced global DNA demethylation system in HEK293T cells. Upon TET overexpression, we observed induction of DNA damage and activation of a DNA damage response; however, BER genes are not upregulated to promote DNA repair. Depletion of TDG (thymine DNA glycosylase) or APEX1 (apurinic/apyrimidinic endonuclease 1), two key BER enzymes, enhances rather than impairs global DNA demethylation, which can be explained by stimulated proliferation. By contrast, growth arrest dramatically blocks TET-induced global DNA demethylation. Thus, in the context of TET-induction in HEK293T cells, the DNA replication-dependent passive mechanism functions as the predominant pathway for global DNA demethylation. In the same context, BER-based active demethylation is markedly restricted by limited BER upregulation, thus potentially preventing a disastrous DNA damage response to extensive active DNA demethylation.


Asunto(s)
Metilación de ADN , Reparación del ADN , Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proliferación Celular , Daño del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/deficiencia , Células HEK293 , Humanos , Timina ADN Glicosilasa/deficiencia
6.
Clin Epigenetics ; 7: 97, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26366234

RESUMEN

BACKGROUND: Aberrant epigenetic silencing of tumor suppressor genes has been recognized as a driving force in cancer. Epigenetic drugs such as the DNA methylation inhibitor decitabine reactivate genes and are effective in myeloid leukemia, but resistance often develops and efficacy in solid tumors is limited. To improve their clinical efficacy, we searched among approved anti-cancer drugs for an epigenetic synergistic combination with decitabine. RESULTS: We used the YB5 cell line, a clonal derivative of the SW48 colon cancer cell line that contains a single copy of a hypermethylated cytomegalovirus (CMV) promoter driving green fluorescent protein (GFP) to screen for drug-induced gene reactivation and synergy with decitabine. None of the 16 anti-cancer drugs tested had effects on their own. However, in combination with decitabine, platinum compounds showed striking synergy in activating GFP. This was dose dependent, observed both in concurrent and sequential combinations, and also seen with other alkylating agents. Clinically achievable concentrations of carboplatin at (25 µM) and decitabine reactivated GFP in 28 % of the YB5 cells as compared to 15 % with decitabine alone. Epigenetic synergy was also seen at endogenously hypermethylated tumor suppressor genes such as MLH1 and PDLIM4. Genome-wide studies showed that reactivation of hypermethylated genes by the combination was significantly better than that induced by decitabine alone or carboplatin alone. Platinum compounds did not enhance decitabine-induced hypomethylation. Rather, we found significantly inhibited HP1α expression by carboplatin and the combination. This was accompanied by increased histone H3 lysine 4 (H3K4) trimethylation and histone H3 lysine 9 (H3K9) acetylation at reactivated genes (P < 0.0001) and reduced occupancy by methyl-binding proteins including MeCP2 and methyl-CpG-binding domain protein 2 (MBD2) (P < 0.0001). CONCLUSIONS: Our results suggest that the combination of decitabine with platinum analogs shows epigenetic synergy that might be exploited in the treatment of different cancers.

7.
BMC Genomics ; 16: 365, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25952319

RESUMEN

BACKGROUND: Aristolochic Acid (AA), a natural component of Aristolochia plants that is found in a variety of herbal remedies and health supplements, is classified as a Group 1 carcinogen by the International Agency for Research on Cancer. Given that microRNAs (miRNAs) are involved in cancer initiation and progression and their role remains unknown in AA-induced carcinogenesis, we examined genome-wide AA-induced dysregulation of miRNAs as well as the regulation of miRNAs on their target gene expression in rat kidney. RESULTS: We treated rats with 10 mg/kg AA and vehicle control for 12 weeks and eight kidney samples (4 for the treatment and 4 for the control) were used for examining miRNA and mRNA expression by deep sequencing, and protein expression by proteomics. AA treatment resulted in significant differential expression of miRNAs, mRNAs and proteins as measured by both principal component analysis (PCA) and hierarchical clustering analysis (HCA). Specially, 63 miRNAs (adjusted p value < 0.05 and fold change > 1.5), 6,794 mRNAs (adjusted p value < 0.05 and fold change > 2.0), and 800 proteins (fold change > 2.0) were significantly altered by AA treatment. The expression of 6 selected miRNAs was validated by quantitative real-time PCR analysis. Ingenuity Pathways Analysis (IPA) showed that cancer is the top network and disease associated with those dysregulated miRNAs. To further investigate the influence of miRNAs on kidney mRNA and protein expression, we combined proteomic and transcriptomic data in conjunction with miRNA target selection as confirmed and reported in miRTarBase. In addition to translational repression and transcriptional destabilization, we also found that miRNAs and their target genes were expressed in the same direction at levels of transcription (169) or translation (227). Furthermore, we identified that up-regulation of 13 oncogenic miRNAs was associated with translational activation of 45 out of 54 cancer-related targets. CONCLUSIONS: Our findings suggest that dysregulated miRNA expression plays an important role in AA-induced carcinogenesis in rat kidney, and that the integrated approach of multiple profiling provides a new insight into a post-transcriptional regulation of miRNAs on their target repression and activation in a genome-wide scale.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Carcinógenos/toxicidad , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Riñón/efectos de los fármacos , ARN Neoplásico/análisis , Animales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Riñón/metabolismo , Neoplasias Renales/etiología , Masculino , MicroARNs/análisis , Datos de Secuencia Molecular , Proteómica , ARN Mensajero/análisis , Ratas , Análisis de Secuencia de ARN
8.
Eur J Clin Invest ; 43(8): 809-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23683178

RESUMEN

BACKGROUND: Melanocytomas of the Central Nervous System (CNS) are rare and benign lesions. These slow-growing tumours can behave aggressively, with local recurrence. Various genetic aberrations occur in malignant melanomas and raise possible new therapeutic options. However, little information is available regarding these characteristic genetic alterations in melanocytomas of the CNS. This study was designed to better understand the clinicopathological and molecular features of melanocytomas. MATERIALS AND METHODS: Twenty cases of melanocytoma were studied by light microscopy, electron microscopy and immunohistochemistry. Clinical characteristics, therapeutic options and prognosis were analysed. BRAF, NRAS and KIT gene mutations were tested by direct DNA sequencing. RESULTS: Fourteen of twenty patients had intracranial tumours including one associated with naevus of Ota and six were spinal. Histologically, these tumours contain fusiform and epithelioid cells with little or no cellular pleomorphism and rare mitoses. Immunohistochemical and ultrastructural findings confirmed the origin of tumour cells as melanocytic. None of the melanocytomas harboured BRAF, NRAS and KIT mutations. Patients with complete resection had no tumour recurrence. Moreover, patients with incomplete tumour resection followed by radiotherapy showed a higher local control (LC) rate than incomplete resection alone (P < 0·05). CONCLUSIONS: BRAF, NRAS and KIT mutations appear to be rare, if not completely absent in melanocytomas of the CNS. The complete resection of the tumour or incomplete resection followed by radiotherapy should be considered as better therapeutic options to reduce the tumour recurrence.


Asunto(s)
Neoplasias del Sistema Nervioso Central/patología , Melanoma/patología , Adolescente , Adulto , Anciano , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/terapia , Análisis Mutacional de ADN , Femenino , GTP Fosfohidrolasas/genética , Humanos , Inmunohistoquímica , Masculino , Melanoma/genética , Melanoma/terapia , Proteínas de la Membrana/genética , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Mutación/genética , Recurrencia Local de Neoplasia/etiología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-kit/genética , Resultado del Tratamiento , Adulto Joven
9.
PLoS One ; 6(8): e23372, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21858090

RESUMEN

PURPOSE: The DNA methylation inhibitor 5-aza-2'-deoxycytidine (DAC) is approved for the treatment of myelodysplastic syndromes (MDS), but resistance to DAC develops during treatment and mechanisms of resistance remain unknown. Therefore, we investigated mechanisms of primary and secondary resistance to DAC in MDS. PATIENTS AND METHODS: We performed Quantitative Real-Time PCR to examine expression of genes related to DAC metabolism prior to therapy in 32 responders and non-responders with MDS as well as 14 patients who achieved a complete remission and subsequently relapsed while on therapy (secondary resistance). We then performed quantitative methylation analyses by bisulfite pyrosequencing of 10 genes as well as Methylated CpG Island Amplification Microarray (MCAM) analysis of global methylation in secondary resistance. RESULTS: Most genes showed no differences by response, but the CDA/DCK ratio was 3 fold higher in non-responders than responders (P<.05), suggesting that this could be a mechanism of primary resistance. There were no significant differences at relapse in DAC metabolism genes, and no DCK mutations were detected. Global methylation measured by the LINE1 assay was lower at relapse than at diagnosis (P<.05). On average, the methylation of 10 genes was lower at relapse (16.1%) compared to diagnosis (18.1%) (P<.05). MCAM analysis showed decreased methylation of an average of 4.5% (range 0.6%-9.7%) of the genes at relapse. By contrast, new cytogenetic changes were found in 20% of patients. CONCLUSION: Pharmacological mechanisms are involved in primary resistance to DAC, whereas hypomethylation does not prevent a relapse for patients with DAC treatment.


Asunto(s)
Azacitidina/análogos & derivados , Resistencia a Medicamentos/genética , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Anciano , Anciano de 80 o más Años , Azacitidina/metabolismo , Azacitidina/uso terapéutico , Aberraciones Cromosómicas/efectos de los fármacos , Islas de CpG/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Metilación de ADN/efectos de los fármacos , Decitabina , Desoxicitidina Quinasa/genética , Desoxicitidina Quinasa/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
10.
Cancer Res ; 70(17): 6968-77, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20713525

RESUMEN

The DNA hypomethylating drug decitabine (DAC) reactivates silenced gene expression in cancer and is approved for the treatment of the myelodysplastic syndrome. Gene reactivation after DAC is variable and incompletely understood. Here, we established a cell line system (YB5) derived from the SW48 colon cancer cell line to study DAC-induced reactivation. YB5 contains a hypermethylated cytomegalovirus promoter driving green fluorescent protein (GFP), and the locus is transcriptionally silent. GFP reexpression can be achieved by DAC treatment, but the expression level of individual cells is heterogeneous. DAC-treated YB5 cells were separated into GFP-positive and GFP-negative subpopulations. By comparing DAC-treated sorted GFP-positive and GFP-negative cells, we found that their methylation levels were similarly decreased but that histone modifications and histone H3 densities were remarkably different. Despite a similar degree of (incomplete) DNA hypomethylation, GFP-positive cells reverted to an active chromatin structure marked by higher H3K9 acetylation, lower H3K27 trimethylation, and lower promoter nucleosome density. GFP-negative cells had histone modifications and promoter nucleosome density, similar to parental cells. On DAC withdrawal, gradual resilencing and remethylation occurred in both GFP-positive and GFP-negative cells, and the resilencing correlated with a gradual increase in nucleosome occupancy in GFP-positive cells. These data show that hypomethylation alone after DAC is insufficient for gene expression induction, and that chromatin resetting to an active state including nucleosome eviction is required for activation of protein expression. Our findings suggest that gene expression is the key in optimizing DAC treatment strategies in the clinic.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Ensamble y Desensamble de Cromatina/genética , Neoplasias del Colon/genética , Metilación de ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Azacitidina/farmacología , Línea Celular Tumoral , Decitabina , Regulación Neoplásica de la Expresión Génica/fisiología , Silenciador del Gen/efectos de los fármacos , Silenciador del Gen/fisiología , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Transfección , Transgenes
11.
Blood ; 113(3): 659-67, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18931345

RESUMEN

5-aza-2'-deoxycytidine (DAC) is approved for the treatment of myelodysplastic syndromes, but resistance to this agent is common. In search for mechanisms of resistance, we measured the half maximal (50%) inhibitory concentration (IC(50)) of DAC and found it differed 1000-fold among a panel of cancer cell lines. The IC(50) was correlated with the doses of DAC that induced the most hypomethylation of long interspersed nuclear elements (LINE; R = 0.94, P < .001), but not with LINE methylation or DNA methyltransferase 1 (DNMT1), 3a, and 3b expression at baseline. Sensitivity to DAC showed a low correlation (R = 0.44, P = .11) to that of 5-azacytidine (AZA), but a good correlation to that of cytarabine (Ara-C; R = 0.89, P < .001). The 5 cell lines most resistant to DAC had a combination of low dCK, hENT1, and 2 transporters, and high cytosine deaminase. In an HL60 clone, resistance to DAC could be rapidly induced by drug exposure and was related to a switch from heterozygous to homozygous mutation of DCK. Transfection of wild-type DCK restored DAC sensitivity. DAC induced DNA breaks as evidenced by H2AX phosphorylation and increased homologous recombination rates by 7- to 10-fold. These results suggest that in vitro resistance to DAC can be explained by insufficient incorporation into DNA.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Azacitidina/análogos & derivados , ADN/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Azacitidina/administración & dosificación , Western Blotting , Línea Celular Tumoral , Daño del ADN , Metilación de ADN , Decitabina , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Pérdida de Heterocigocidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
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
13.
Cancer Res ; 66(10): 5077-84, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707430

RESUMEN

CpG island methylation within promoters is known to silence individual genes in cancer. The involvement of this process in silencing gene pairs controlled by bidirectional promoters is unclear. In a screen for hypermethylated CpG islands in cancer, bidirectional promoters constituted 25.2% of all identified promoters, which matches with the genomic representation of bidirectional promoters. From the screen, we selected three bidirectional gene pairs for detailed analysis, WNT9A/CD558500, CTDSPL/BC040563, and KCNK15/BF195580. Levels of mRNA of all three pairs of genes were inversely correlated with the degree of promoter methylation in multiple cancer cell lines. Hypomethylation of these promoters induced by 5-aza-2'-deoxycytidine treatment reactivated or enhanced gene expression bidirectionally. The bidirectional nature of the WNT9A/CD558500 promoter was confirmed by luciferase assays, and hypermethylation down-regulated expression of both genes in the pair. Methylation of WNT9A/CD558500 and CTDSPL/BC040563 promoters occurs frequently in primary colon cancers and acute lymphoid leukemias (ALL), respectively, and methylation was correlated with decreased gene expression in ALL patient samples. Our study shows that hypermethylation of bidirectional promoter-associated CpG island silences two genes simultaneously, a property that should be taken into account when studying the functional consequences of hypermethylation in cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Silenciador del Gen , Neoplasias/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Islas de CpG , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Neoplasias/patología , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...