Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nucleic Acids Res ; 48(12): 6513-6529, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32449925

RESUMO

Monocytes and macrophages are essential components of the innate immune system. Herein, we report that intron retention (IR) plays an important role in the development and function of these cells. Using Illumina mRNA sequencing, Nanopore direct cDNA sequencing and proteomics analysis, we identify IR events that affect the expression of key genes/proteins involved in macrophage development and function. We demonstrate that decreased IR in nuclear-detained mRNA is coupled with increased expression of genes encoding regulators of macrophage transcription, phagocytosis and inflammatory signalling, including ID2, IRF7, ENG and LAT. We further show that this dynamic IR program persists during the polarisation of resting macrophages into activated macrophages. In the presence of proinflammatory stimuli, intron-retaining CXCL2 and NFKBIZ transcripts are rapidly spliced, enabling timely expression of these key inflammatory regulators by macrophages. Our study provides novel insights into the molecular factors controlling vital regulators of the innate immune response.


Assuntos
Ativação de Macrófagos , Macrófagos/imunologia , Splicing de RNA , RNA Mensageiro/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Cultivadas , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Endoglina/genética , Endoglina/metabolismo , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Íntrons , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , RNA Mensageiro/metabolismo , Células THP-1
2.
Nat Commun ; 8: 15134, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28480880

RESUMO

While intron retention (IR) is considered a widely conserved and distinct mechanism of gene expression control, its regulation is poorly understood. Here we show that DNA methylation directly regulates IR. We also find reduced occupancy of MeCP2 near the splice junctions of retained introns, mirroring the reduced DNA methylation at these sites. Accordingly, MeCP2 depletion in tissues and cells enhances IR. By analysing the MeCP2 interactome using mass spectrometry and RNA co-precipitation, we demonstrate that decreased MeCP2 binding near splice junctions facilitates IR via reduced recruitment of splicing factors, including Tra2b, and increased RNA polymerase II stalling. These results suggest an association between IR and a slower rate of transcription elongation, which reflects inefficient splicing factor recruitment. In summary, our results reinforce the interdependency between alternative splicing involving IR and epigenetic controls of gene expression.


Assuntos
Processamento Alternativo , Íntrons , Proteína 2 de Ligação a Metil-CpG/metabolismo , Fatores de Processamento de RNA/metabolismo , Animais , Células Cultivadas , Metilação de DNA , Células Precursoras de Granulócitos/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Ligação Proteica , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Sítios de Splice de RNA , Fatores de Processamento de RNA/genética
4.
J Hematol Oncol ; 10(1): 39, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28153030

RESUMO

Methylation of N6 adenosine (m6A) is known to be important for diverse biological processes including gene expression control, translation of protein, and messenger RNA (mRNA) splicing. However, its role in the development of human cancers is poorly understood. By analyzing datasets from the Cancer Genome Atlas Research Network (TCGA) acute myeloid leukemia (AML) study, we discover that mutations and/or copy number variations of m6A regulatory genes are strongly associated with the presence of TP53 mutations in AML patients. Further, our analyses reveal that alterations in m6A regulatory genes confer a worse survival in AML. Our work indicates that genetic alterations of m6A regulatory genes may cooperate with TP53 and/or its regulator/downstream targets in the pathogenesis and/or maintenance of AML.


Assuntos
Adenosina/análogos & derivados , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , RNA Mensageiro/metabolismo , Adenosina/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Feminino , Amplificação de Genes , Genes p53 , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidade , Masculino , Metilação , Pessoa de Meia-Idade , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Modelos de Riscos Proporcionais , Deleção de Sequência , Adulto Jovem
5.
Mol Cancer Res ; 14(12): 1217-1228, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27671336

RESUMO

Laterally spreading tumors (LST) are colorectal adenomas that develop into extremely large lesions with predominantly slow progression to cancer, depending on lesion subtype. Comparing and contrasting the molecular profiles of LSTs and colorectal cancers offers an opportunity to delineate key molecular alterations that drive malignant transformation in the colorectum. In a discovery cohort of 11 LSTs and paired normal mucosa, we performed a comprehensive and unbiased screen of the genome, epigenome, and transcriptome followed by bioinformatics integration of these data and validation in an additional 84 large, benign colorectal lesions. Mutation rates in LSTs were comparable with microsatellite-stable colorectal cancers (2.4 vs. 2.6 mutations per megabase); however, copy number alterations were infrequent (averaging only 1.5 per LST). Frequent genetic, epigenetic, and transcriptional alterations were identified in genes not previously implicated in colorectal neoplasia (ANO5, MED12L, EPB41L4A, RGMB, SLITRK1, SLITRK5, NRXN1, ANK2). Alterations to pathways commonly mutated in colorectal cancers, namely, the p53, PI3K, and TGFß pathways, were rare. Instead, LST-altered genes converged on axonal guidance, Wnt, and actin cytoskeleton signaling. These integrated omics data identify molecular features associated with noncancerous LSTs and highlight that mutation load, which is relatively high in LSTs, is a poor predictor of invasive potential. IMPLICATIONS: The novel genetic, epigenetic, and transcriptional changes associated with LST development reveal important insights into why some adenomas do not progress to cancer. The finding that LSTs exhibit a mutational load similar to colorectal carcinomas has implications for the validity of molecular biomarkers for assessing cancer risk. Mol Cancer Res; 14(12); 1217-28. ©2016 AACR.


Assuntos
Adenoma/genética , Neoplasias Colorretais/genética , Redes Reguladoras de Genes , Genômica/métodos , Biologia Computacional/métodos , Metilação de DNA , Epigênese Genética , Feminino , Dosagem de Genes , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla/métodos , Humanos , Mutação , Análise de Sequência de RNA/métodos
6.
Nucleic Acids Res ; 44(6): 2888-97, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26825461

RESUMO

Fever is commonly used to diagnose disease and is consistently associated with increased mortality in critically ill patients. However, the molecular controls of elevated body temperature are poorly understood. We discovered that the expression of RNA-binding motif protein 3 (RBM3), known to respond to cold stress and to modulate microRNA (miRNA) expression, was reduced in 30 patients with fever, and in THP-1-derived macrophages maintained at a fever-like temperature (40 °C). Notably, RBM3 expression is reduced during fever whether or not infection is demonstrable. Reduced RBM3 expression resulted in increased expression of RBM3-targeted temperature-sensitive miRNAs, we termed thermomiRs. ThermomiRs such as miR-142-5p and miR-143 in turn target endogenous pyrogens including IL-6, IL6ST, TLR2, PGE2 and TNF to complete a negative feedback mechanism, which may be crucial to prevent pathological hyperthermia. Using normal PBMCs that were exogenously exposed to fever-like temperature (40 °C), we further demonstrate the trend by which decreased levels of RBM3 were associated with increased levels of miR-142-5p and miR-143 and vice versa over a 24 h time course. Collectively, our results indicate the existence of a negative feedback loop that regulates fever via reduced RBM3 levels and increased expression of miR-142-5p and miR-143.


Assuntos
Retroalimentação Fisiológica , Febre/genética , Leucócitos Mononucleares/imunologia , MicroRNAs/genética , Proteínas de Ligação a RNA/genética , Temperatura Corporal , Regulação da Temperatura Corporal/genética , Linhagem Celular , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/imunologia , Febre/imunologia , Febre/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , MicroRNAs/imunologia , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/imunologia , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/imunologia , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
7.
JAMA Oncol ; 1(7): 953-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26181641

RESUMO

IMPORTANCE: Constitutional hypermethylation of 1 allele throughout the soma (constitutional epimutation) is an accepted mechanism of cancer predisposition. Understanding the origin and inheritance of epimutations is important for assessing cancer risk in affected families. OBSERVATIONS: We report a 29-year-old man with early-onset colorectal cancer who showed a constitutional MLH1 epimutation (approximately 50% of alleles methylated and allele-specific loss of MLH1 expression) that was stable over a 16-year period. The epimutation was inherited without a genetic alteration from his asymptomatic mother. She showed methylation on the same allele but in less than 5% of her somatic cells. CONCLUSIONS AND RELEVANCE: These findings indicate that low-level somatic mosaicism for an epimutation in an asymptomatic parent can produce a nonmosaic constitutional epimutation in a child. Asymptomatic low-level methylation in some individuals may be associated with substantial cancer risk to their offspring.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/genética , Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Epigênese Genética , Mosaicismo , Proteínas Nucleares/genética , Adenocarcinoma/patologia , Adolescente , Adulto , Idoso , Neoplasias Colorretais/patologia , Metilação de DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Testes Genéticos , Hereditariedade , Humanos , Masculino , Proteína 1 Homóloga a MutL , Linhagem , Fenótipo , Medição de Risco , Fatores de Risco , Adulto Jovem
8.
Methods Mol Biol ; 1315: 153-71, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26103898

RESUMO

Pyrosequencing(®) is able to quantitate the level of a nucleotide at a designated germ-line or somatic variant, including single nucleotide polymorphisms (SNPs). SNPs within a gene of interest may be used to distinguish between the two genetic alleles and study their behavior in heterozygous individuals. With regard to cancer etiology and development, identification of alleles and the detection of allelic imbalances, such as transcriptional loss from one allele or loss-of-heterozygosity (due to deletion of one allele), within a tumor are particularly useful. Lynch syndrome, the most common form of hereditary bowel and uterine cancer, is caused by heterozygous germ-line mutations within the DNA mismatch repair genes and tumors develop following inactivation of the remaining functional allele within somatic tissues, usually by acquired loss-of-heterozygosity. MLH1 is the most frequently mutated gene in Lynch syndrome; however, some cases whose tumors display immunohistochemical loss of the MLH1 protein have no apparent mutation within the coding region of MLH1. Allelic loss of expression or reduced function of MLH1 can also result in the propensity to develop Lynch syndrome associated cancers. In this chapter we describe allele quantification Pyrosequencing assays designed at a common benign SNP within the MLH1 coding region for application to either DNA or mRNA (cDNA) templates, which enabled us to detect pathological allelic imbalances in such cases with suspected Lynch syndrome. Our allele quantification Pyrosequencing assays at the MLH1 c.655A > G (rs1799977) exonic SNP were applied to clinical specimens and detected both constitutional allelic expression loss and tumor loss-of-heterozygosity in some cases, facilitating the identification of the mechanistic cause underlying their cancer development. We provide detailed protocols for implementing these Pyrosequencing assays and illustrative examples of their application in patients.


Assuntos
Alelos , Análise Mutacional de DNA/métodos , Perda de Heterozigosidade/genética , Polimorfismo de Nucleotídeo Único/genética , Biotinilação , DNA/química , DNA/genética , Reparo de Erro de Pareamento de DNA , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Sefarose/química , Software
9.
Hum Mutat ; 36(6): 622-30, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25762362

RESUMO

Lynch syndrome is a hereditary cancer syndrome caused by a constitutional mutation in one of the mismatch repair genes. The implementation of predictive testing and targeted preventative surveillance is hindered by the frequent finding of sequence variants of uncertain significance in these genes. We aimed to determine the pathogenicity of previously reported variants (c.-28A>G and c.-7C>T) within the MLH1 5'untranslated region (UTR) in two individuals from unrelated suspected Lynch syndrome families. We investigated whether these variants were associated with other pathogenic alterations using targeted high-throughput sequencing of the MLH1 locus. We also determined their relationship to gene expression and epigenetic alterations at the promoter. Sequencing revealed that the c.-28A>G and c.-7C>T variants were the only potentially pathogenic alterations within the MLH1 gene. In both individuals, the levels of transcription from the variant allele were reduced to 50% compared with the wild-type allele. Partial loss of expression occurred in the absence of constitutional epigenetic alterations within the MLH1 promoter. We propose that these variants may be pathogenic due to constitutional partial loss of MLH1 expression, and that this may be associated with intermediate penetrance of a Lynch syndrome phenotype. Our findings provide further evidence of the potential importance of noncoding variants in the MLH1 5'UTR in the pathogenesis of Lynch syndrome.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Desequilíbrio Alélico , Neoplasias Colorretais Hereditárias sem Polipose/genética , Expressão Gênica , Variação Genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Regiões 5' não Traduzidas , Idade de Início , Ilhas de CpG , Metilação de DNA , Epigênese Genética , Feminino , Estudos de Associação Genética , Loci Gênicos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Repetições de Microssatélites , Proteína 1 Homóloga a MutL , Mutação , Linhagem
10.
Eur J Hum Genet ; 22(5): 617-24, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24084575

RESUMO

Germline mutations of the DNA mismatch repair genes MLH1, MSH2, MSH6 or PMS2, and deletions affecting the EPCAM gene adjacent to MSH2, underlie Lynch syndrome by predisposing to early-onset colorectal, endometrial and other cancers. An alternative but rare cause of Lynch syndrome is constitutional epimutation of MLH1, whereby promoter methylation and transcriptional silencing of one allele occurs throughout normal tissues. A dominantly transmitted constitutional MLH1 epimutation has been linked to an MLH1 haplotype bearing two single-nucleotide variants, NM_000249.2: c.-27C>A and c.85G>T, in a Caucasian family with Lynch syndrome from Western Australia. Subsequently, a second seemingly unrelated Caucasian Australian case with the same MLH1 haplotype and concomitant epimutation was reported. We now describe three additional, ostensibly unrelated, cancer-affected families of European heritage with this MLH1 haplotype in association with constitutional epimutation, bringing the number of index cases reported to five. Array-based genotyping in four of these families revealed shared haplotypes between individual families that extended across ≤2.6-≤6.4 megabase regions of chromosome 3p, indicating common ancestry. A minimal ≤2.6 megabase founder haplotype common to all four families was identified, which encompassed MLH1 and additional flanking genes and segregated with the MLH1 epimutation in each family. Our findings indicate that the MLH1 c.-27C>A and c.85G>T variants are borne on a European ancestral haplotype and provide conclusive evidence for its pathogenicity via a mechanism of epigenetic silencing of MLH1 within normal tissues. Additional descendants bearing this founder haplotype may exist who are also at high risk of developing Lynch syndrome-related cancers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Epigênese Genética , Genes Dominantes , Haplótipos , Proteínas Nucleares/genética , Mutação Puntual , População Branca/genética , Alelos , Estudos de Casos e Controles , Cromossomos Humanos Par 3 , Neoplasias Colorretais Hereditárias sem Polipose/genética , Variações do Número de Cópias de DNA , Metilação de DNA , Reparo de Erro de Pareamento de DNA , Análise Mutacional de DNA , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Proteína 1 Homóloga a MutL , Linhagem , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Transcrição Gênica
11.
Dis Model Mech ; 6(4): 993-1000, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23611895

RESUMO

Development of asthma in childhood is linked to viral infections of the lower respiratory tract in early life, with subsequent chronic exposure to allergens. Progression to persistent asthma is associated with a Th2-biased immunological response and structural remodelling of the airways. The underlying mechanisms are unclear, but could involve epigenetic changes. To investigate this, we employed a recently developed mouse model in which self-limited neonatal infection with a pneumovirus, followed by sensitisation to ovalbumin via the respiratory tract and low-level chronic challenge with aerosolised antigen, leads to development of an asthmatic phenotype. We assessed expression of microRNA by cells in the proximal airways, comparing changes over the period of disease progression, and used target prediction databases to identify genes likely to be up- or downregulated as a consequence of altered regulation of microRNA. In parallel, we assessed DNA methylation in pulmonary CD4(+) T cells. We found that a limited number of microRNAs exhibited marked up- or downregulation following early-life infection and sensitisation, for many of which the levels of expression were further changed following chronic challenge with the sensitizing antigen. Targets of these microRNAs included genes involved in immune or inflammatory responses (e.g. Gata3, Kitl) and in tissue remodelling (e.g. Igf1, Tgfbr1), as well as genes for various transcription factors and signalling proteins. In pulmonary CD4(+) T cells, there was significant demethylation at promoter sites for interleukin-4 and interferon-γ, the latter increasing following chronic challenge. We conclude that, in this model, progression to an asthmatic phenotype is linked to epigenetic regulation of genes associated with inflammation and structural remodelling, and with T-cell commitment to a Th2 immunological response. Epigenetic changes associated with this pattern of gene activation might play a role in the development of childhood asthma.


Assuntos
Asma/genética , Asma/patologia , Progressão da Doença , Epigênese Genética , Animais , Asma/virologia , Linfócitos T CD4-Positivos/metabolismo , Criança , Ilhas de CpG/genética , Metilação de DNA/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Feminino , Humanos , Interferon gama/genética , Interleucina-4/genética , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/metabolismo , Vírus da Pneumonia Murina/fisiologia , Regiões Promotoras Genéticas/genética , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/genética
12.
Cancer Cell ; 20(2): 200-13, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21840485

RESUMO

Constitutional epimutations of tumor suppressor genes manifest as promoter methylation and transcriptional silencing of a single allele in normal somatic tissues, thereby predisposing to cancer. Constitutional MLH1 epimutations occur in individuals with young-onset cancer and demonstrate non-Mendelian inheritance through their reversal in the germline. We report a cancer-affected family showing dominant transmission of soma-wide highly mosaic MLH1 methylation and transcriptional repression linked to a particular genetic haplotype. The epimutation was erased in spermatozoa but reinstated in the somatic cells of the next generation. The affected haplotype harbored two single nucleotide substitutions in tandem; c.-27C > A located near the transcription initiation site and c.85G > T. The c.-27C > A variant significantly reduced transcriptional activity in reporter assays and is the probable cause of this epimutation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Epigênese Genética , Inativação Gênica , Genes Dominantes , Proteínas Nucleares/genética , Regiões 5' não Traduzidas , Metilação de DNA , Ligação Genética , Humanos , Proteína 1 Homóloga a MutL
13.
Int J Cancer ; 128(4): 869-78, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20473912

RESUMO

Lynch syndrome is an autosomal dominant cancer predisposition syndrome classically caused by germline mutations of the mismatch repair genes, MLH1, MSH2, MSH6 and PMS2. Constitutional epimutations of the MLH1 gene, characterized by soma-wide methylation of a single allele of the promoter and allelic transcriptional silencing, have been identified in a subset of Lynch syndrome cases lacking a sequence mutation in MLH1. We report two individuals with no family history of colorectal cancer who developed that disease at age 18 and 20 years. In both cases, cancer had arisen because of the de novo occurrence of a constitutional MLH1 epimutation and somatic loss-of-heterozygosity of the functional allele in the tumors. We show for the first time that the epimutation in one case arose on the paternally inherited allele. Analysis of 13 tumors from seven individuals with constitutional MLH1 epimutations showed eight tumors had lost the second MLH1 allele, two tumors had a novel pathogenic missense mutation and three had retained heterozygosity. Only 1 of 12 tumors demonstrated the BRAF V600E mutation and 3 of 11 tumors harbored a mutation in KRAS. The finding that epimutations can originate on the paternal allele provides important new insights into the mechanism of origin of epimutations. It is clear that the second hit in MLH1 epimutation-associated tumors typically has a genetic not epigenetic basis. Individuals with mismatch repair-deficient cancers without the BRAF V600E mutation are candidates for germline screening for sequence or methylation changes in MLH1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais/genética , Epigenômica , Mutação em Linhagem Germinativa/genética , Proteínas Nucleares/genética , Adolescente , Adulto , Idade de Início , Idoso , Alelos , DNA de Neoplasias/genética , Feminino , Predisposição Genética para Doença , Haplótipos/genética , Humanos , Perda de Heterozigosidade , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Linhagem , Reação em Cadeia da Polimerase , Prognóstico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras) , Adulto Jovem , Proteínas ras/genética
14.
Fam Cancer ; 9(3): 345-56, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20063070

RESUMO

Lynch syndrome is an autosomal dominant cancer susceptibility syndrome characterized by the early development of microsatellite unstable colorectal, endometrial and other cancers. Lynch syndrome is caused by germline heterozygous loss-of-function sequence mutations within the mismatch repair genes MLH1, MSH2, MSH6 or PMS2. Some individuals with Lynch syndrome have constitutional epimutations, characterized by promoter methylation and transcriptional inactivation of a single allele in normal somatic tissues, while others lack identifiable pathogenic changes in the germline. We hypothesized that analysis of the relative levels of allelic expression of MLH1 would assist in the identification of cryptic pathogenic defects of MLH1 in five presumed Lynch syndrome cases whose tumours demonstrated MLH1 loss, but whose causative mutation remained unidentified. We exploited the common benign c.655A>G SNP (rs1799977) within MLH1 exon 8 to distinguish between the two genetic alleles in heterozygous individuals and to study their transcriptional activity, using quantitative pyrosequencing assays. In one of the five patients we detected loss of expression of one allele and deletion of the other allele in the tumour, prompting renewed germline screening. A novel intronic splice mutation was subsequently identified, which resulted in loss of an entire exon from the transcript. This pyrosequencing assay also proved useful in demonstrating the gradual reversal of a constitutional MLH1 epimutation during lymphoblastoid cell culture, suggesting this defect may not be stably maintained in immortalized cells. Our findings illustrate that the study of allelic behaviour can complement conventional molecular analyses by providing new insight into the genetic or epigenetic mechanisms underlying disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Perfilação da Expressão Gênica/métodos , Proteínas Nucleares/genética , Desequilíbrio Alélico , Sequência de Bases , Metilação de DNA , Mutação em Linhagem Germinativa , Humanos , Perda de Heterozigosidade , Dados de Sequência Molecular , Proteína 1 Homóloga a MutL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Mod Pathol ; 22(12): 1588-99, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19734844

RESUMO

O(6)-methylguanine DNA methyltransferase (MGMT) is a DNA repair protein that restores mutagenic O(6)-methylguanine to guanine. MGMT methylation is frequently observed in sporadic colorectal cancer and was recently correlated with the C>T allele at SNP rs16906252, within the transcriptional enhancer element of the promoter. MGMT methylation has also been associated with KRAS mutations, particularly G>A transitions. We studied 1123 colorectal carcinoma to define the molecular and clinicopathological profiles associated with MGMT methylation. Furthermore, we assessed factors contributing to MGMT methylation in the development of colorectal cancer by studying the allelic pattern of MGMT methylation using SNP rs16906252, and the methylation status of neighbouring genes within 10q26 in selected tumours and matched normal colonic mucosa. MGMT methylation was detected by combined bisulphite restriction analysis in 28% of tumours and was associated with a number of characteristics, including CDKN2A methylation, absent lymphovascular space invasion and KRAS mutations (but not specifically with KRAS G>A transitions). In a multivariate analysis adjusted for age and sex, MGMT methylation was associated with the T allele of SNP rs16906252 (P<0.0001, OR 5.5, 95% CI 3.8-7.9). Low-level methylation was detected by quantitative methylation-specific PCR in the normal colonic mucosa of cases, particularly those with a correspondingly methylated tumour, as well as controls without neoplasia, and this was also associated with the C>T SNP. We show that the T allele at SNP rs16906252 is a key determinant in the onset of MGMT methylation in colorectal cancer, whereas the association of methylation at MGMT and CDKN2A suggests that these loci may be targets of a common mechanism of epigenetic dysregulation.


Assuntos
Carcinoma/genética , Cromossomos Humanos Par 10 , Neoplasias Colorretais/genética , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Mutação em Linhagem Germinativa , Mucosa Intestinal/enzimologia , Polimorfismo de Nucleotídeo Único , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Austrália , Carcinoma/enzimologia , Carcinoma/patologia , Carcinoma/cirurgia , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Epigênese Genética , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Frequência do Gene , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Razão de Chances , Fenótipo , Reação em Cadeia da Polimerase , Estudos Prospectivos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Proteínas Supressoras de Tumor/metabolismo , Proteínas ras/genética
16.
Cancer Res ; 67(19): 9107-16, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909015

RESUMO

Biallelic promoter methylation and transcriptional silencing of the MLH1 gene occurs in the majority of sporadic colorectal cancers exhibiting microsatellite instability due to defective DNA mismatch repair. Long-range epigenetic silencing of contiguous genes has been found on chromosome 2q14 in colorectal cancer. We hypothesized that epigenetic silencing of MLH1 could occur on a regional scale affecting additional genes within 3p22, rather than as a focal event. We studied the levels of CpG island methylation and expression of multiple contiguous genes across a 4 Mb segment of 3p22 including MLH1 in microsatellite-unstable and -stable cancers, and their paired normal colonic mucosa. We found concordant CpG island hypermethylation, H3-K9 dimethylation and transcriptional silencing of MLH1 and multiple flanking genes spanning up to 2.4 Mb in microsatellite-unstable colorectal cancers. This region was interspersed with unmethylated genes, which were also transcriptionally repressed. Expression of both methylated and unmethylated genes was reactivated by methyltransferase and histone deacetylase inhibitors in a microsatellite-unstable colorectal carcinoma cell line. Two genes at the telomeric end of the region were also hypermethylated in microsatellite-stable cancers, adenomas, and at low levels in normal colonic mucosa from older individuals. Thus, the cluster of genes flanking MLH1 that was specifically methylated in the microsatellite-unstable group of cancers extended across 1.1 Mb. Our results show that coordinate epigenetic silencing extends across a large chromosomal region encompassing MLH1 in microsatellite-unstable colorectal cancers. Simultaneous epigenetic silencing of this cluster of 3p22 genes may contribute to the development or progression of this type of cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Instabilidade de Microssatélites , Proteínas Nucleares/genética , Alelos , Linhagem Celular Tumoral , Cromatina/genética , Cromossomos Humanos Par 3 , Metilação de DNA , Inibidores de Histona Desacetilases , Humanos , Metiltransferases/antagonistas & inibidores , Família Multigênica , Proteína 1 Homóloga a MutL , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...