Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtre
1.
Yonsei Medical Journal ; : 35-42, 2017.
Article Dans Anglais | WPRIM | ID: wpr-65065

Résumé

PURPOSE: Decitabine, a DNA hypomethylating agent, was recently approved for use in Korea for older adults with acute myeloid leukemia (AML) who are not candidates for standard chemotherapy. This study aimed to evaluate the role of decitabine as a first-line treatment for older adults with AML. MATERIALS AND METHODS: Twenty-four patients with AML who received at least one course of decitabine (20 mg/m²/d intravenously for 5 days every 4 weeks) as a first-line therapy at Severance Hospital were evaluated retrospectively. RESULTS: The median age of the patients was 73.5 years. The longest follow-up duration was 502 days. A total of 113 cycles of treatment were given to 24 patients, and the median number of cycles was four (range, 1–14). Thirteen patients dropped out because of death, no or loss of response, patient refusal, or transfer to another hospital. Twenty-one (87.5%) and 12 (50%) patients completed the second and fourth cycles, respectively, and responses to treatment were evaluated in 17. A complete response (CR) or CR with incomplete blood-count recovery was achieved in six (35.3%) patients, and the estimated median overall survival was 502 days. Ten patients developed grade >2 hematologic or non-hematologic toxicities. In univariate analysis, bone marrow blasts, lactate dehydrogenase, serum ferritin level, and bone marrow iron were significantly associated with response to decitabine. CONCLUSION: Five-day decitabine treatment showed acceptable efficacy in older patients with AML who are unfit for conventional chemotherapy, with a CR rate 35.3% and about a median overall survival of 18 months.


Sujets)
Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Antimétabolites antinéoplasiques/administration et posologie , Azacitidine/analogues et dérivés , Méthylation de l'ADN , Leucémie aigüe myéloïde/traitement médicamenteux , Induction de rémission , République de Corée , Études rétrospectives , Résultat thérapeutique
3.
Braz. j. med. biol. res ; 47(12): 1029-1035, 12/2014. graf
Article Dans Anglais | LILACS | ID: lil-727661

Résumé

DNA methylation is essential in X chromosome inactivation and genomic imprinting, maintaining repression of XIST in the active X chromosome and monoallelic repression of imprinted genes. Disruption of the DNA methyltransferase genes DNMT1 and DNMT3B in the HCT116 cell line (DKO cells) leads to global DNA hypomethylation and biallelic expression of the imprinted gene IGF2 but does not lead to reactivation of XIST expression, suggesting that XIST repression is due to a more stable epigenetic mark than imprinting. To test this hypothesis, we induced acute hypomethylation in HCT116 cells by 5-aza-2′-deoxycytidine (5-aza-CdR) treatment (HCT116-5-aza-CdR) and compared that to DKO cells, evaluating DNA methylation by microarray and monitoring the expression of XIST and imprinted genes IGF2, H19, and PEG10. Whereas imprinted genes showed biallelic expression in HCT116-5-aza-CdR and DKO cells, the XIST locus was hypomethylated and weakly expressed only under acute hypomethylation conditions, indicating the importance of XIST repression in the active X to cell survival. Given that DNMT3A is the only active DNMT in DKO cells, it may be responsible for ensuring the repression of XIST in those cells. Taken together, our data suggest that XIST repression is more tightly controlled than genomic imprinting and, at least in part, is due to DNMT3A.


Sujets)
Humains , Méthylation de l'ADN/génétique , Répression épigénétique/génétique , Génome humain , Génome/génétique , Empreinte génomique/génétique , Facteur de croissance IGF-II/génétique , ARN long non codant/génétique , Azacitidine/administration et posologie , Azacitidine/analogues et dérivés , /génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Techniques de knock-out de gènes , Génome humain/effets des médicaments et des substances chimiques , Hybridation fluorescente in situ/méthodes , Analyse sur microréseau , Polymorphisme de nucléotide simple , Protéines/métabolisme , ARN long non codant/métabolisme , Réaction de polymérisation en chaine en temps réel/méthodes , RT-PCR/méthodes
4.
Yonsei Medical Journal ; : 1656-1663, 2014.
Article Dans Anglais | WPRIM | ID: wpr-180229

Résumé

PURPOSE: Both genetic and epigenetic alterations can lead to abnormal expression of metastasis-regulating genes in tumor cells. Recent studies suggest that aberrant epigenetic alterations, followed by differential gene expression, leads to an aggressive cancer cell phenotype. We examined epigenetically regulated genes that are involved in ovarian cancer metastasis. MATERIALS AND METHODS: We developed SK-OV-3 human ovarian carcinoma cell xenografts in mice. We compared the mRNA expression and DNA methylation profiles of metastatic tissues to those of the original SK-OV-3 cell line. RESULTS: Metastatic implants showed increased mRNA expression of the carbonic anhydrase 9 (CA9) gene and hypomethylation at CpG sites in the CA9 promoter. Treatment of wild-type SK-OV-3 cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine reduced methylation of the CA9 promoter and increased CA9 mRNA expression. Eight CpGs, which were located at positions -197, -74, -19, -6, +4, +13, +40, and +86, relative to the transcription start site, were hypomethylated in metastatic tumor implants, compared to that of wild-type SK-OV-3. Overexpression of CA9 induced an aggressive phenotype, including increased invasiveness and migration, in SK-OV-3 cells. CONCLUSION: Alterations in the DNA methylation profile of the CA9 promoter were correlated with a more aggressive phenotype in ovarian cancer cells.


Sujets)
Animaux , Femelle , Humains , Souris , Azacitidine/analogues et dérivés , Carbonic anhydrases/métabolisme , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Invasion tumorale/génétique , Métastase tumorale/génétique , Tumeurs expérimentales , Tumeurs épithéliales épidermoïdes et glandulaires/génétique , Tumeurs de l'ovaire/génétique , Phénotype , Régions promotrices (génétique) , ARN messager/métabolisme
5.
IJRM-Iranian Journal of Reproductive Medicine. 2012; 10 (3): 193-200
Dans Anglais | IMEMR | ID: emr-144278

Résumé

Epigenetic reprogramming of differentiated cells can modify somatic cells into pluripotential state. Pluripotency can be induced in somatic cells by several approaches. One of the easy ways to induce pluripotency is the exposure of the somatic cells to the embryonic stem cell [ESC] extract. The objective of this study was to increase the efficiency of reprogramming of granulosa cell as a differentiated cell into pluripotential state by using epigenetic modifier agents and extract. The human granulosa cells were cultured in the medium containing 5-Aza-Deoxycytidine and trichostatin A. Then, the cells were exposed to mouse ESCs extract and co-cultured with mouse embryonic fibroblast in the presence of leukemia inhibitory factor [LIF]. Alkaline phosphatase test and also immunohistochemistery staining for Oct4, Sox2 and Nanog were performed after 24 and 72 hours and 1 week. The granulosa cells showed the alkaline phosphatase activity after 24 hours and the enzyme activity maintained for 72 hours. They also expressed Oct4 after 24 hours. The cells also expressed Sox2 and Nanog, 72 hours after exposure to the ESCs extract. The expression of the pluripotency markers decreased after 1 week. It seems that the extract can induce dedifferentiation in granulosa cells and they can express the stem cell markers. It seems that the inhibitors of the methyl transferase [5-Aza-Deoxycytidine] and histone deacetylase [trichostatin A] could delete the epigenetic markers and prepare the cells for reprogramming by administration of the extract


Sujets)
Humains , Cellules de la granulosa , Reprogrammation cellulaire , Azacitidine/analogues et dérivés , Acides hydroxamiques , Épigenèse génétique
6.
The Korean Journal of Internal Medicine ; : 407-410, 2012.
Article Dans Anglais | WPRIM | ID: wpr-168866

Résumé

BACKGROUND/AIMS: The relationship between Runt-related transcription factor 3 (RUNX3) gene inactivation and various solid tumors has been reported; however, little information is available about RUNX3 in thyroid cancers. METHODS: We evaluated the DNA methylation of RUNX3 in 13 papillary thyroid cancer tissues and four thyroid cancer cell lines. Additionally, using reverse transcriptase-polymerase chain reaction, we analyzed RUNX3 gene expression in several thyroid cancer cell lines after treating with the demethylating agent 5-aza-2'-deoxycytidine (DAC). RESULTS: RUNX3 was hypermethylated in many thyroid cancer cell lines and in 10 of the 12 papillary thyroid cancer tissues. Treatment with DAC increased the expression of RUNX3 in some thyroid cancer cell lines. CONCLUSIONS: We suggest that RUNX3 is associated with thyroid carcinogenesis, and RUNX3 methylation is a potentially useful diagnostic marker for papillary thyroid cancer.


Sujets)
Humains , Azacitidine/analogues et dérivés , Carcinomes/génétique , Lignée cellulaire tumorale , Sous-unité alpha 3 du facteur CBF/génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Expression des gènes/effets des médicaments et des substances chimiques , Tumeurs de la thyroïde/génétique , Marqueurs biologiques tumoraux/génétique
7.
Journal of Korean Medical Science ; : 594-604, 2012.
Article Dans Anglais | WPRIM | ID: wpr-202341

Résumé

DNA methylation is one of the main epigenetic mechanisms and hypermethylation of CpG islands at tumor suppressor genes switches off these genes. To find novel DNA methylation markers in hepatocellular carcinoma (HCC), we performed pharmacological unmasking (treatment with 5-aza-2'-deoxycytidine or trichostatin A) followed by microarray analysis in HCC cell lines. Of the 239 promoter CpG island loci hypermethylated in HCC cell lines (as revealed by methylation-specific PCR), 221 loci were found to be hypermethylated in HCC or nonneoplastic liver tissues. Thirty-three loci showed a 20% higher methylation frequency in tumors than in adjacent nonneoplastic tissues. Correlation of individual cancer-related methylation markers with clinicopathological features of HCC patients (n = 95) revealed that the number of hypermethylated genes in HCC tumors was higher in older than in younger patients. Univariate and multivariate survival analysis revealed that the HIST1H2AE methylation status is closely correlated with the patient's overall survival (P = 0.022 and P = 0.010, respectively). In conclusion, we identified 221 novel DNA methylation markers for HCC. One promising prognostic marker, HIST1H2AE, should be further validated in the prognostication of HCC patients.


Sujets)
Femelle , Humains , Mâle , Adulte d'âge moyen , Azacitidine/analogues et dérivés , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Ilots CpG , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Régulation négative , Cellules HepG2 , Acides hydroxamiques/pharmacologie , Foie/métabolisme , Tumeurs du foie/génétique , Séquençage par oligonucléotides en batterie , Régions promotrices (génétique) , Analyse de survie , Marqueurs biologiques tumoraux/génétique
8.
IJFS-International Journal of Fertility and Sterility. 2011; 4 (4): 148-155
Dans Anglais | IMEMR | ID: emr-109861

Résumé

Reconstructed embryos from terminally differentiated somatic cells have revealed high levels of genomic methylation which results in inappropriate expression patterns of imprinted and non-imprinted genes. These aberrant expressions are probably responsible for different abnormalities during the development of clones. Improvement in cloning competency may be achieved through modification of epigenetic markers in donor cells. Our objective was to determine if treatment of donor cells for 72 hours with 5-aza-2'-deoxycytidine [5-aza-dc; 0-0.3 microM], a DNA methyl transferase inhibitor, improved development and expression of Oct-4. In comparison with untreated cells, 0.01 and 0.08 microM 5-aza-dc treated cells insignificantly decreased the blastocyst rate [32.1% vs. 28.6% and 27.2%, respectively] while it was significant for 0.3 microM treated cells [6.5%]. Embryo quality as measured by the total cell number [TCN] decreased in a dose-related fashion, which was significant at 0.08 and 0.3 microM 5-aza-dc treated cells when compared with 0 and 0.01 microM 5-aza-dc treated cells. Although reconstructed embryos from 0.08 and 0.3 microM 5-aza-dc treated cells showed lower levels of DNA methylation and histone H3 acetylation, development to blastocyst stage was decreased. The epigenetic markers of embryos cloned from 0.01 microM 5-aza-dc remained unchanged. These results show that 5-aza-dc is not a suitable choice for modifying nuclear reprogramming. Finally, it was concluded that the wide genomic hypomethylation induced by 5-aza-dc deleteriously impacts the developmental competency of cloned embryo


Sujets)
Animaux , Techniques de transfert nucléaire , Épigenèse génétique , Azacitidine/analogues et dérivés , Bovins
9.
Experimental & Molecular Medicine ; : 401-410, 2011.
Article Dans Anglais | WPRIM | ID: wpr-102683

Résumé

Epstein-Barr virus (EBV) microRNAs (miRNAs) are expressed in EBV-associated tumors and cell lines, but the regulation mechanism of their expression is unclear yet. We investigated whether the expression of EBV miRNAs is epigenetically regulated in EBV-infected B cell lines. The expression of BART miRNAs was inversely related with the methylation level of the BART promoter at both steady-state and following 5-aza-2'-deoxycytidine treatment of the cells. The expression of BHRF1 miRNAs also became detectable with the demethylation of Cp/Wp in latency I EBV-infected cell lines. Furthermore, in vitro methylation of the BART and Cp promoters reduced the promoter-driven transactivation. In contrast, tricostatin A had little effect on the expression of EBV miRNA expression as well as on the BART and Cp/Wp promoters. Our results suggest that promoter methylation, but not histone acetylation, plays a role in regulation of the EBV miRNA expression in EBV-infected B cell lines.


Sujets)
Humains , Azacitidine/analogues et dérivés , Lymphocytes B/métabolisme , Lignée cellulaire , Méthylation de l'ADN , DNA modification methylases/antagonistes et inhibiteurs , Régulation de l'expression des gènes viraux , Extinction de l'expression des gènes , Herpèsvirus humain de type 4/génétique , microARN/génétique , Régions promotrices (génétique) , ARN viral/génétique , Protéines virales/génétique
10.
Journal of Korean Medical Science ; : 1152-1159, 2010.
Article Dans Anglais | WPRIM | ID: wpr-187254

Résumé

Promoter CpG island hypermethylation has become recognized as an important mechanism for inactivating tumor suppressor genes or tumor-related genes in human cancers of various tissues. Gene inactivation in association with promoter CpG island hypermethylation has been reported to be four times more frequent than genetic changes in human colorectal cancers. Hepatocellular carcinoma is also one of the human cancer types in which aberrant promoter CpG island hypermethylation is frequently found. However, the number of genes identified to date as hypermethylated for hepatocellular carcinoma (HCC) is fewer than that for colorectal cancer or gastric cancer, which can be attributed to fewer attempts to perform genome-wide methylation profiling for HCC. In the present study, we used bead-array technology and coupled methylation-specific PCR to identify new genes showing cancer-specific methylation in HCC. Twenty-four new genes have been identified as hypermethylated at their promoter CpG island loci in a cancer-specific manner. Of these, TNFRSF10C, HOXA9, NPY, and IRF5 were frequently hypermethylated in hepatocellular carcinoma tissue samples and their methylation was found to be closely associated with inactivation of gene expression. Further study will be required to elucidate the clinicopathological implications of these newly found DNA methylation markers in hepatocellular carcinoma.


Sujets)
Humains , Antimétabolites antinéoplasiques/usage thérapeutique , Azacitidine/analogues et dérivés , Carcinome hépatocellulaire/traitement médicamenteux , Lignée cellulaire tumorale , Ilots CpG , Méthylation de l'ADN , Protéines liées au GPI/génétique , Analyse de profil d'expression de gènes , Protéines à homéodomaine/génétique , Facteurs de régulation d'interféron/génétique , Tumeurs du foie/traitement médicamenteux , Neuropeptide Y/génétique , Séquençage par oligonucléotides en batterie , Régions promotrices (génétique) , Récepteurs leurres aux facteurs de nécrose tumorale/génétique
11.
Experimental & Molecular Medicine ; : 276-281, 2005.
Article Dans Anglais | WPRIM | ID: wpr-177644

Résumé

Methylation events play a critical role in various cellular processes including regulation of gene transcription and proliferation. Recently, RUNX3 gene, one of TGF-beta-Smads signaling transduction pathway genes, showed strong tumor-suppressor activity by regulation of epithelial proliferation and apoptosis. To elucidate the potential etiological role of the RUNX3 gene in the development of hepatocellular carcinoma (HCC), we have analyzed the methylation status of 5' CpG island of the RUNX3 gene in a series of 73 HCC tissues and 11 liver cell lines. Expectedly, promoter methylation of RUNX3 gene was found in 2 (2.7%) of 73 corresponding normal liver, whereas 30 (41.1%) of 73 HCCs and 4 (40%) of 10 liver cancer cell lines showed hypermethylation of the gene, respectively. There was no significant difference between promoter hypermethylaion and clinicopathologic parameters of primary HCC samples, including histologic grade, microvascular invasion, and clinical stage. Interestingly, demethylating agent 5-aza-2-deoxycytidine induced reactivation and more potent expression of RUNX3 gene in HCC cell lines. Our findings indicate that promoter hypermethylation of RUNX3 gene may occur as an early event in the development of HCC and that methylation may be a major mechanism for inactivation of RUNX3 gene in HCC.


Sujets)
Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Humains , Mâle , Adulte d'âge moyen , Azacitidine/analogues et dérivés , Carcinome hépatocellulaire/génétique , Méthylation de l'ADN , ADN tumoral/effets des médicaments et des substances chimiques , Tumeurs du foie/génétique , Régions promotrices (génétique)
SÉLECTION CITATIONS
Détails de la recherche