Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BMC Cancer ; 17(1): 260, 2017 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-28403857

RESUMO

BACKGROUND: Breast carcinogenesis is a multistep process involving genetic and epigenetic changes. Tumor tissues are frequently characterized by gene-specific hypermethylation and global DNA hypomethylation. Aberrant DNA methylation levels have, however, not only been found in tumors, but also in tumor-surrounding tissue appearing histologically normal. This phenomenon is called field cancerization. Knowledge of the existence of a cancer field and its spread are of clinical relevance. If the tissue showing pre-neoplastic lesions is not removed by surgery, it may develop into invasive carcinoma. METHODS: We investigated the prevalence of gene-specific and global DNA methylation changes in tumor-adjacent and tumor-distant tissues in comparison to tumor tissues from the same breast cancer patients (n = 18) and normal breast tissues from healthy women (n = 4). Methylation-sensitive high resolution melting (MS-HRM) analysis was applied to determine methylation levels in the promoters of APC, BRCA1, CDKN2A (p16), ESR1, HER2/neu and PTEN, in CDKN2A exon 2 and in LINE-1, as indicator for the global DNA methylation extent. The methylation status of the ESR2 promoter was determined by pyrosequencing. RESULTS: Tumor-adjacent and tumor-distant tissues frequently showed pre-neoplastic gene-specific and global DNA methylation changes. The APC promoter (p = 0.003) and exon 2 of CDKN2A (p < 0.001) were significantly higher methylated in tumors than in normal breast tissues from healthy women. For both regions, significant differences were also found between tumor and tumor-adjacent tissues (p = 0.001 and p < 0.001, respectively) and tumor and tumor-distant tissues (p = 0.001 and p < 0.001, respectively) from breast cancer patients. In addition, tumor-adjacent (p = 0.002) and tumor-distant tissues (p = 0.005) showed significantly higher methylation levels of CDKN2A exon 2 than normal breast tissues serving as control. Significant correlations were found between the proliferative activity and the methylation status of CDKN2A exon 2 in tumor (r = -0.485, p = 0.041) and tumor-distant tissues (r = -0.498, p = 0.036). CONCLUSIONS: From our results we can conclude that methylation changes in CDKN2A exon 2 are associated with breast carcinogenesis. Further investigations are, however, necessary to confirm that hypermethylation of CDKN2A exon 2 is associated with tumor proliferative activity.


Assuntos
Neoplasias da Mama/genética , Inibidor de Quinase Dependente de Ciclina p18/genética , Metilação de DNA , Receptor beta de Estrogênio/genética , Análise de Sequência de DNA/métodos , Adulto , Idoso , Linhagem Celular Tumoral , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina , Epigênese Genética , Éxons , Feminino , Humanos , Células MCF-7 , Pessoa de Meia-Idade , Regiões Promotoras Genéticas
2.
Amino Acids ; 49(1): 117-127, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27714514

RESUMO

AMPA receptors mediate most fast excitatory synaptic transmission in the brain. Highly dynamic AMPA receptors are subjected to trafficking, recycling, and/or degradation and replacement. Changes in AMPA receptor abundance is an important mechanism involved in learning and memory formation. Results obtained with the Morris water maze (MWM), a paradigm for testing spatial memory in rodent, correlate with hippocampal synaptic plasticity and NMDA function. Different phases of spatial learning like acquisition and retrieval involve AMPA receptors. Long-term memory formation requires dynamic changes in gene transcription and protein synthesis. It is, however, not known so far if epigenetic marks such as DNA methylation and mRNA levels participate in regulation of AMPA receptors in hippocampus during memory retrieval. In the present study, rats were trained or untrained in the MWM. Steady state levels of hippocampal GluA1-4 mRNA were determined by RT-PCR and promoter methylation levels of GluA1-4 by in-house developed bisulfite pyrosequencing methods. GluA1-4 protein levels were determined in parallel in a membrane fraction by SDS-PAGE followed by Western blotting. Our results indicate that changes of hippocampal membrane AMPA receptors were modulated at the protein level, while no changes were observed at the mRNA and at the promoter methylation level of hippocampal GluA1-4. Training in the MWM at retrieval may, therefore, involve GluA2 and GluA4 subunits that may be regulated by protein stability or trafficking as protein determinations were carried out in a hippocampal membrane fraction.


Assuntos
Epigênese Genética , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Isoformas de Proteínas/genética , Receptores de AMPA/genética , Animais , Metilação de DNA , Masculino , Plasticidade Neuronal , Regiões Promotoras Genéticas , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Análise de Sequência de DNA , Transmissão Sináptica
3.
Breast Cancer Res ; 17: 125, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26370119

RESUMO

INTRODUCTION: It has been shown in some articles that genetic and epigenetic abnormalities cannot only be found in tumor tissues but also in adjacent regions that appear histologically normal. This phenomenon is metaphorically called field cancerization or field defect. Field cancerization is regarded as clinically significant because it is assumed to be an important factor in local recurrence of cancer. As the field showing these molecular abnormalities may not be removed completely by surgery, these changes might lead to neoplasms and subsequent transformation to a tumor. We aimed to investigate the applicability of the methylation status of six tumor suppressor genes as biomarkers for detecting field cancerization in breast cancer. METHODS: The promoter methylation status of CCND2, DAPK1, GSTP1, HIN-1, MGMT and RASSF1A was determined by methylation-sensitive high-resolution melting (MS-HRM) analysis. MS-HRM methods for CCND2, MGMT and RASSF1A were developed in-house, primer sequences for DAPK1, GSTP1 and HIN-1 have already been published. Biopsy samples were taken from tumor, tumor-adjacent and tumor-distant tissue from 17 breast cancer patients. Normal breast tissues of four healthy women served as controls. RESULTS: All MS-HRM methods proved to be very sensitive. LODs were in the range from 0.1 to 1.5 %, LOQs ranged from 0.3 to 5.3 %. A total of 94 %, 82 % and 65 % of the tumors showed methylation of RASSF1A, HIN-1 and MGMT promoters, respectively. The methylation status of these promoters was significantly lower in tumor-distant tissues than in tumor tissues. Tumor-adjacent tissues showed higher methylation status of RASSF1A, HIN-1 and MGMT promoters than tumor-distant tissues, indicating field cancerization. The methylation status of the HIN-1 promoter in tumor-adjacent tissues was found to correlate strongly with that in the corresponding tumors (r = 0.785, p < 0.001), but not with that in the corresponding tumor-distant tissues (r = 0.312, p = 0.239). CONCLUSIONS: Among the gene promoters investigated, the methylation status of the HIN-1 promoter can be considered the best suitable biomarker for detecting field cancerization. Further investigation is needed to test whether it can be used for defining surgical margins in order to prevent future recurrence of breast cancer.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Citocinas/genética , Metilação de DNA/genética , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Regiões Promotoras Genéticas/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Biópsia , Neoplasias da Mama/patologia , Ciclina D2/genética , Proteínas Quinases Associadas com Morte Celular/genética , Epigênese Genética/genética , Feminino , Glutationa S-Transferase pi/genética , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia
4.
Oncotarget ; 7(45): 73347-73369, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27689338

RESUMO

Overexpression of ABCB1, ABCC1 and ABCG2 in tumor tissues is considered a major cause of limited efficacy of anticancer drugs. Gene expression of ABC transporters is regulated by multiple mechanisms, including changes in the DNA methylation status. Most of the studies published so far only report promoter methylation levels for either ABCB1 or ABCG2, and data on the methylation status for ABCC1 are scarce. Thus, we determined the promoter methylation patterns of ABCB1, ABCC1 and ABCG2 in 19 human cancer cell lines. In order to contribute to the elucidation of the role of DNA methylation changes in acquisition of a multidrug resistant (MDR) phenotype, we also analyzed the promoter methylation patterns in drug-resistant sublines of the cancer cell lines GLC-4, SW1573, KB-3-1 and HL-60. In addition, we investigated if aberrant promoter methylation levels of ABCB1, ABCC1 and ABCG2 occur in tumor and tumor-surrounding tissues from breast cancer patients.Our data indicates that hypomethylation of the ABCC1 promoter is not cancer type-specific but occurs in cancer cell lines of different origins. Promoter methylation was found to be an important mechanism in gene regulation of ABCB1 in parental cancer cell lines and their drug-resistant sublines. Overexpression of ABCC1 in MDR cell models turned out to be mediated by gene amplification, not by changes in the promoter methylation status of ABCC1. In contrast to the promoters of ABCC1 and ABCG2, the promoter of ABCB1 was significantly higher methylated in tumor tissues than in tumor-adjacent and tumor-distant tissues from breast cancer patients.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Neoplasias da Mama/genética , Metilação de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Adulto , Idoso , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ilhas de CpG , Variações do Número de Cópias de DNA , Resistência a Múltiplos Medicamentos/genética , Feminino , Expressão Gênica , Humanos , Pessoa de Meia-Idade , Variantes Farmacogenômicos , Reprodutibilidade dos Testes , Análise de Sequência de DNA
5.
Oncotarget ; 7(19): 27379-93, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27036042

RESUMO

As an environmental poison, arsenic is responsible for many cancer deaths. Paradoxically, arsenic trioxide (ATO) presents also a powerful therapy used to treat refractory acute promyelocytic leukemia (APL) and is intensively investigated for treatment of other cancer types. Noteworthy, cancer therapy is frequently hampered by drug resistance, which is also often associated with enhancement of tumor aggressiveness. In this study, we analyzed ATO-selected cancer cells (A2780ATO) for the mechanisms underlying their enhanced tumorigenicity and aggressiveness. These cells were characterized by enhanced proliferation and spheroid growth as well as increased tumorigenicity of xenografts in SCID mice. Noteworthy, subsequent studies revealed that overexpression of Met receptor was the underlying oncogenic driver of these effects, as A2780ATO cells were characterized by collateral sensitivity against Met inhibitors. This finding was also confirmed by array comparative genomic hybridization (array CGH) and whole genome gene expression arrays, which revealed that Met overexpression by chronic ATO exposure was based on the transcriptional regulation via activation of AP-1. Finally, it was shown that treatment with the Met inhibitor crizotinib was also effective against A2780ATO cell xenografts in vivo, indicating that targeting of Met presents a promising strategy for the treatment of Met-overexpressing tumors after either arsenic exposure or failure to ATO treatment.


Assuntos
Arsenicais/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias/genética , Óxidos/farmacologia , Proteínas Proto-Oncogênicas c-met/genética , Animais , Trióxido de Arsênio , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Crizotinibe , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
Oncotarget ; 7(31): 50161-50179, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27367030

RESUMO

Genomically amplified fibroblast growth factor receptor 1 (FGFR1) is an oncogenic driver in defined lung cancer subgroups and predicts sensibility against FGFR1 inhibitors in this patient cohort. The FGFR inhibitor nintedanib has recently been approved for treatment of lung adenocarcinoma and is currently evaluated for small cell lung cancer (SCLC). However, tumor recurrence due to development of nintedanib resistance might occur. Hence, we aimed at characterizing the molecular mechanisms underlying acquired nintedanib resistance in FGFR1-driven lung cancer. Chronic nintedanib exposure of the FGFR1-driven SCLC cell line DMS114 (DMS114/NIN) but not of two NSCLC cell lines induced massive overexpression of the multidrug-resistance transporter ABCB1. Indeed, we proved nintedanib to be both substrate and modulator of ABCB1-mediated efflux. Importantly, the oncogenic FGFR1 signaling axis remained active in DMS114/NIN cells while bioinformatic analyses suggested hyperactivation of the endothelin-A receptor (ETAR) signaling axis. Indeed, ETAR inhibition resensitized DMS114/NIN cells against nintedanib by downregulation of ABCB1 expression. PKC and downstream NFκB were identified as major downstream players in ETAR-mediated ABCB1 hyperactivation. Summarizing, ABCB1 needs to be considered as a factor underlying nintedanib resistance. Combination approaches with ETAR antagonists or switching to non-ABCB1 substrate FGFR inhibitors represent innovative strategies to manage nintedanib resistance in lung cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Indóis/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Endotelina/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Adenocarcinoma de Pulmão , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Separação Celular , Sobrevivência Celular , Hibridização Genômica Comparativa , Metilação de DNA , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Hibridização in Situ Fluorescente , Recidiva Local de Neoplasia , Fenótipo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA