Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(12): e82516, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24340035

RESUMEN

In humans, adipose tissue is distributed in subcutaneous abdominal and subcutaneous gluteal depots that comprise a variety of functional differences. Whereas energy storage in gluteal adipose tissue has been shown to mediate a protective effect, an increase of abdominal adipose tissue is associated with metabolic disorders. However, the molecular basis of depot-specific characteristics is not completely understood yet. Using array-based analyses of transcription profiles, we identified a specific set of genes that was differentially expressed between subcutaneous abdominal and gluteal adipose tissue. To investigate the role of epigenetic regulation in depot-specific gene expression, we additionally analyzed genome-wide DNA methylation patterns in abdominal and gluteal depots. By combining both data sets, we identified a highly significant set of depot-specifically expressed genes that appear to be epigenetically regulated. Interestingly, the majority of these genes form part of the homeobox gene family. Moreover, genes involved in fatty acid metabolism were also differentially expressed. Therefore we suppose that changes in gene expression profiles might account for depot-specific differences in lipid composition. Indeed, triglycerides and fatty acids of abdominal adipose tissue were more saturated compared to triglycerides and fatty acids in gluteal adipose tissue. Taken together, our results uncover clear differences between abdominal and gluteal adipose tissue on the gene expression and DNA methylation level as well as in fatty acid composition. Therefore, a detailed molecular characterization of adipose tissue depots will be essential to develop new treatment strategies for metabolic syndrome associated complications.


Asunto(s)
Metilación de ADN/fisiología , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/fisiología , Grasa Subcutánea Abdominal/metabolismo , Triglicéridos/metabolismo , Adulto , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Síndrome Metabólico/mortalidad , Especificidad de Órganos/fisiología
2.
PLoS One ; 7(5): e36125, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563479

RESUMEN

Silencing of genes by hypermethylation contributes to cancer progression and has been shown to occur with increased frequency at specific genomic loci. However, the precise mechanisms underlying the establishment and maintenance of aberrant methylation marks are still elusive. The de novo DNA methyltransferase 3B (DNMT3B) has been suggested to play an important role in the generation of cancer-specific methylation patterns. Previous studies have shown that a reduction of DNMT3B protein levels induces antiproliferative effects in cancer cells that were attributed to the demethylation and reactivation of tumor suppressor genes. However, methylation changes have not been analyzed in detail yet. Using RNA interference we reduced DNMT3B protein levels in colon cancer cell lines. Our results confirm that depletion of DNMT3B specifically reduced the proliferation rate of DNMT3B-overexpressing colon cancer cell lines. However, genome-scale DNA methylation profiling failed to reveal methylation changes at putative DNMT3B target genes, even in the complete absence of DNMT3B. These results show that DNMT3B is dispensable for the maintenance of aberrant DNA methylation patterns in human colon cancer cells and they have important implications for the development of targeted DNA methyltransferase inhibitors as epigenetic cancer drugs.


Asunto(s)
Proliferación Celular , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Interferencia de ARN , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Immunoblotting , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ADN Metiltransferasa 3B
4.
PLoS One ; 6(3): e17388, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21408221

RESUMEN

The DNA methyltransferase inhibitors azacytidine and decitabine represent archetypal drugs for epigenetic cancer therapy. To characterize the demethylating activity of azacytidine and decitabine we treated colon cancer and leukemic cells with both drugs and used array-based DNA methylation analysis of more than 14,000 gene promoters. Additionally, drug-induced demethylation was compared to methylation patterns of isogenic colon cancer cells lacking both DNA methyltransferase 1 (DNMT1) and DNMT3B. We show that drug-induced demethylation patterns are highly specific, non-random and reproducible, indicating targeted remethylation of specific loci after replication. Correspondingly, we found that CG dinucleotides within CG islands became preferentially remethylated, indicating a role for DNA sequence context. We also identified a subset of genes that were never demethylated by drug treatment, either in colon cancer or in leukemic cell lines. These demethylation-resistant genes were enriched for Polycomb Repressive Complex 2 components in embryonic stem cells and for transcription factor binding motifs not present in demethylated genes. Our results provide detailed insights into the DNA methylation patterns induced by azacytidine and decitabine and suggest the involvement of complex regulatory mechanisms in drug-induced DNA demethylation.


Asunto(s)
Azacitidina/análogos & derivados , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Genes Relacionados con las Neoplasias/genética , Cromatina/metabolismo , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Decitabina , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Genoma Humano/genética , Células HCT116 , Células HL-60 , Humanos , Leucemia Mieloide/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , ADN Metiltransferasa 3B
5.
Mol Cancer Ther ; 9(5): 1256-64, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20442313

RESUMEN

Azacytidine is an established nucleoside drug that is well known for its ability to modulate epigenetic gene regulation by inhibition of DNA methylation. Despite recent advances in the clinical development of azacytidine, the use of the drug is limited by its low bioavailability and dependency on variably expressed nucleoside transporters for cellular uptake. We show here that CP-4200, an elaidic acid derivative of azacytidine, has strong epigenetic modulatory potency in human cancer cell lines, as evidenced by efficient depletion of DNA methyltransferase protein, genome-wide DNA demethylation, and robust reactivation of epigenetically silenced tumor suppressor genes. Importantly, however, the cellular uptake of CP-4200 was substantially less dependent on the nucleoside transporters that are known to be involved in azacytidine uptake. In agreement with this notion, CP-4200 showed a significantly higher antitumoral activity than azacytidine in an orthotopic mouse tumor model for acute lymphocytic leukemia. Together, these data represent a detailed characterization of the CP-4200 mode of action and suggest that elaidic acid modification improves the therapeutic efficacy of azacytidine.


Asunto(s)
Azacitidina/administración & dosificación , Neoplasias/tratamiento farmacológico , Ácido Oléico/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Azacitidina/análogos & derivados , Azacitidina/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Epigénesis Genética/efectos de los fármacos , Esterificación , Ésteres/administración & dosificación , Ésteres/química , Ésteres/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HL-60 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos , Neoplasias/genética , Neoplasias/patología , Ácido Oléico/química , Ácido Oléico/metabolismo , Ácidos Oléicos , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell Cycle ; 6(9): 1001-5, 2007 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-17457051

RESUMEN

MicroRNAs (miRNAs) are small RNA molecules that control gene expression by inhibition of protein translation or degradation of cognate target mRNAs. Eventhough strict Even though strict developmental and tissue-specific regulation appears to be critical for miRNA function, very little is known about the mechanisms governing miRNA gene expression. Several recent studies have shown that miRNA genes can regulated DNA methylation and other epigenetic mechanisms. The observation of altered miRNA gene methylation patterns in human cancers also suggested that miRNA gene methylation is functional relevant for tumorigenesis. We have now performed a comprehensive analysis of miRNA genes and found that about half of these genes are associated with CpG islands and thus represent candidate targets of the DNA methylation machinery An expanded analysis of several miRNA-associated CpG islands in five cell lines indicated that miRNA gene methylation is detectable at high frequencies, both in normal and malignant cells. Possible explanations for this phenomenon include the specific structure of miRNA genes and/or their requirement for strict expression regulation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias/genética , Animales , Células Cultivadas , Islas de CpG , Metilación de ADN , Regulación de la Expresión Génica , Humanos , Modelos Biológicos , Células Tumorales Cultivadas
7.
Cancer J ; 13(1): 17-22, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17464242

RESUMEN

Aberrant DNA methylation patterns, including hypermethylation of tumor suppressor genes, have been described in many human cancers. These epigenetic mutations can be reversed by DNA methyltransferase inhibitors, which provide novel opportunities for cancer therapy. Clinical concepts for epigenetic therapies are currently being developed by using azanucleosides for the treatment of leukemias and other tumors. These trials will greatly benefit from the inclusion of molecular markers for monitoring epigenetic changes in patients and for maximizing biologic responses. In addition, novel inhibitors need to be developed that result in a direct and specific inhibition of DNA methyltransferase activity. Several recent developments indicate that rational design of small molecule DNA methyltransferase inhibitors is feasible and that this approach can result in the establishment of novel drug candidates. The use of novel DNA methyltransferase inhibitors in clinical trials that allow monitoring of drug-induced DNA methylation changes should provide the foundation for improved epigenetic cancer therapies.


Asunto(s)
Metilasas de Modificación del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Azacitidina/uso terapéutico , Metilación de ADN/efectos de los fármacos , Decitabina , Epigénesis Genética/efectos de los fármacos , Humanos , Neoplasias/genética
8.
Cancer Res ; 67(4): 1419-23, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308078

RESUMEN

MicroRNAs (miRNAs) are small noncoding RNAs that repress their target mRNAs by complementary base pairing and induction of the RNA interference pathway. It has been shown that miRNA expression can be regulated by DNA methylation and it has been suggested that altered miRNA gene methylation might contribute to human tumorigenesis. In this study, we show that the human let-7a-3 gene on chromosome 22q13.31 is associated with a CpG island. Let-7a-3 belongs to the archetypal let-7 miRNA gene family and was found to be methylated by the DNA methyltransferases DNMT1 and DNMT3B. The gene was heavily methylated in normal human tissues but hypomethylated in some lung adenocarcinomas. Let-7a-3 hypomethylation facilitated epigenetic reactivation of the gene and elevated expression of let-7a-3 in a human lung cancer cell line resulted in enhanced tumor phenotypes and oncogenic changes in transcription profiles. Our results thus identify let-7a-3 as an epigenetically regulated miRNA gene with oncogenic function and suggest that aberrant miRNA gene methylation might contribute to the human cancer epigenome.


Asunto(s)
Metilación de ADN , MicroARNs/genética , Neoplasias/genética , Oncogenes , Adenocarcinoma/genética , Adenoma/genética , Neoplasias Colorrectales/genética , Células HCT116 , Humanos , Neoplasias Pulmonares/genética , MicroARNs/metabolismo , Transfección
9.
Bioconjug Chem ; 17(2): 261-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16536454

RESUMEN

Small-molecule inhibitors of DNA methyltransferases such as RG108 represent promising candidates for cancer drug development. We report the synthesis and in vitro analysis of a biotinylated RG108 conjugate, 2-(1,3-dioxo-1,3-dihydro-isoindol-2-yl)-3-(5-[3-[5-(2-oxo-hexahydro-thieno[3,4-d]imidazol-4-yl)pentanoylamino]propoxy]-1H-indol-3-yl)propionic acid (bio-RG108), for the evaluation of interactions with DNA methyltransferase enzymes. The structural design of the chemically modified inhibitor was aided by molecular modeling, which suggested the possibility for extensive chemical modifications at the 5-position of the tryptophan moiety in RG108. The inhibitory activity of the corresponding derivative was confirmed in a cell-free biochemical assay, where bio-RG108 showed an undiminished inhibition of DNA methyltransferase activity (IC50 = 40 nM). Bio-RG108 therefore represents a suitable bioconjugate for the elucidation of inhibitory mechanisms and for the affinity purification of RG108-associated proteins.


Asunto(s)
Biotina/química , Metilasas de Modificación del ADN/antagonistas & inhibidores , Indoles/química , Propionatos/química , Biotina/metabolismo , Sistema Libre de Células , Metilasas de Modificación del ADN/metabolismo , Humanos , Indoles/metabolismo , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Ftalimidas , Propionatos/metabolismo , Unión Proteica , Triptófano/análogos & derivados
10.
Cancer Res ; 66(5): 2794-800, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510601

RESUMEN

DNA methyltransferase inhibitors represent promising new drugs for cancer therapies. The first of these compounds (5-azacytidine, Vidaza) has recently been approved as an antitumor agent, and others are presently in various stages of their preclinical or clinical development. Most of the archetypal inhibitors have been established and characterized in different experimental systems, which has thus far precluded their direct comparison. We have now established defined experimental conditions that allowed a comparative analysis of the six most widely known DNA methyltransferase inhibitors: 5-azacytidine (5-aza-CR), 5-aza-2'-deoxycytidine (5-aza-CdR), zebularine, procaine, (-)-epigallocatechin-3-gallate (EGCG), and RG108. Of these, 5-aza-CR, 5-aza-CdR, zebularine, and EGCG were found to exhibit significant cytotoxicity in human cancer cell lines. 5-aza-CdR and EGCG were also found to be genotoxic, as evidenced by the induction of micronuclei. In addition, 5-aza-CR, 5-aza-CdR, zebularine, and RG108 caused concentration-dependent demethylation of genomic DNA, whereas procaine and EGCG failed to induce significant effects. Finally, the experiments in cancer cell lines were complemented by a cell-free in vitro assay with purified recombinant DNA methyltransferase, which indicated that RG108 is the only drug capable of direct enzyme inhibition. These results show a substantial diversity in the molecular activities of DNA methyltransferase inhibitors and provide valuable insights into the developmental potential of individual drugs.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Catequina/análogos & derivados , Catequina/farmacología , Línea Celular Tumoral , Citidina/análogos & derivados , Citidina/farmacología , Metilación de ADN/efectos de los fármacos , Decitabina , Humanos , Indoles/farmacología , Células Jurkat , Ftalimidas , Procaína/farmacología , Propionatos/farmacología , Triptófano/análogos & derivados
11.
J Med Chem ; 49(2): 678-83, 2006 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-16420053

RESUMEN

DNA methyltransferases are promising targets for cancer therapy. In many cancer cells promoters of tumor suppressor genes are hypermethylated, which results in gene inactivation. It has been shown that DNA methyltransferase inhibitors can suppress tumor growth and have significant therapeutic value. However, the established inhibitors are limited in their application due to their substantial cytotoxicity. To discover novel compounds for the inhibition of human DNA methyltransferases, we have screened a set of small molecules available from the NCI database. Using a 3-dimensional model of the human DNA methyltransferase 1 and a modified docking and scoring procedure, we have identified a small list of molecules with high affinities for the active site of the enzyme. The two highest scoring structures were found to inhibit DNA methyltransferase activity in vitro and in vivo. The newly discovered inhibitors validate our screening procedure and also provide a useful basis for further rational drug development.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/química , Metilación de ADN , Indoles/química , Modelos Moleculares , Ftalimidas/química , Piridinas/química , Triazoles/química , Dominio Catalítico , ADN (Citosina-5-)-Metiltransferasa 1 , Humanos
12.
Epigenetics ; 1(1): 7-13, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17998812

RESUMEN

Hypermethylation of tumor suppressor genes is one of the most consistent hallmarks of human cancers. This epigenetic alteration has been associated with gene silencing and thus represents an important pathway for generating loss-of-function mutations. In this review, we survey the available literature on systematic, genome-wide approaches aimed at the identification of epigenetically silenced loci. These studies uncovered a variety of diverse genes, but a common signature for epigenetic reactivation has not been identified. Nevertheless, DNA methyltransferase inhibitors have shown significant clinical benefits, mostly in the therapy of leukemias. Recent analyses revealed substantial drug-induced methylation changes that can now be used as endpoints for the further refinement of clinical treatment schedules. Further optimization of epigenetic cancer therapies should be feasible through the use of novel DNA methyltransferase inhibitors with improved specificity. Rational design of epigenetic inhibitors might provide the foundation for a broader use of these drugs in the treatment of cancer.


Asunto(s)
Metilasas de Modificación del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Animales , Metilación de ADN/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Silenciador del Gen/efectos de los fármacos , Humanos
13.
Cancer Res ; 65(14): 6305-11, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024632

RESUMEN

DNA methylation regulates gene expression in normal and malignant cells. The possibility to reactivate epigenetically silenced genes has generated considerable interest in the development of DNA methyltransferase inhibitors. Here, we provide a detailed characterization of RG108, a novel small molecule that effectively blocked DNA methyltransferases in vitro and did not cause covalent enzyme trapping in human cell lines. Incubation of cells with low micromolar concentrations of the compound resulted in significant demethylation of genomic DNA without any detectable toxicity. Intriguingly, RG108 caused demethylation and reactivation of tumor suppressor genes, but it did not affect the methylation of centromeric satellite sequences. These results establish RG108 as a DNA methyltransferase inhibitor with fundamentally novel characteristics that will be particularly useful for the experimental modulation of epigenetic gene regulation.


Asunto(s)
Metilasas de Modificación del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor/efectos de los fármacos , Indoles/farmacología , Propionatos/farmacología , Sitios de Unión , Metilación de ADN/efectos de los fármacos , Células HCT116 , Humanos , Ftalimidas , Triptófano/análogos & derivados
14.
Trends Pharmacol Sci ; 25(11): 551-4, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15491775

RESUMEN

The pharmacological inhibition of DNA methyltransferases provides novel opportunities for the therapy of human cancers. Several Phase III trials of DNA methyltransferase inhibitors have been completed successfully, but the efficacy of these compounds appears to be compromised by their lack of specificity. To address this issue, much work has been focused on defining the functional characteristics of human DNA methyltransferases. This knowledge will be essential for the design of a new generation of pharmacological inhibitors that target epigenetic processes.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Animales , Metilación de ADN/efectos de los fármacos , Epigénesis Genética , Humanos , Neoplasias/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...