Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Language
Publication year range
1.
Mol Oncol ; 16(3): 630-647, 2022 02.
Article in English | MEDLINE | ID: mdl-34520622

ABSTRACT

Wilms tumour (WT), an embryonal kidney cancer, has been extensively characterised for genetic and epigenetic alterations, but a proportion of WTs still lack identifiable abnormalities. To uncover DNA methylation changes critical for WT pathogenesis, we compared the epigenome of foetal kidney with two WT cell lines, filtering our results to remove common cancer-associated epigenetic changes and to enrich for genes involved in early kidney development. This identified four hypermethylated genes, of which ESRP2 (epithelial splicing regulatory protein 2) was the most promising for further study. ESRP2 was commonly repressed by DNA methylation in WT, and this occurred early in WT development (in nephrogenic rests). ESRP2 expression was reactivated by DNA methyltransferase inhibition in WT cell lines. When ESRP2 was overexpressed in WT cell lines, it inhibited cellular proliferation in vitro, and in vivo it suppressed tumour growth of orthotopic xenografts in nude mice. RNA-seq of the ESRP2-expressing WT cell lines identified several novel splicing targets. We propose a model in which epigenetic inactivation of ESRP2 disrupts the mesenchymal to epithelial transition in early kidney development to generate WT.


Subject(s)
Kidney Neoplasms , Wilms Tumor , Animals , Cell Line, Tumor , DNA/metabolism , DNA Methylation/genetics , Genes, Tumor Suppressor , Humans , Kidney Neoplasms/genetics , Mice , Mice, Nude , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Wilms Tumor/genetics
2.
Sci Rep ; 9(1): 18934, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831790

ABSTRACT

To discover epigenetic changes that may underly neuroblastoma pathogenesis, we identified differentially methylated genes in neuroblastoma cells compared to neural crest cells, the presumptive precursors cells for neuroblastoma, by using genome-wide DNA methylation analysis. We previously described genes that were hypermethylated in neuroblastoma; in this paper we report on 67 hypomethylated genes, which were filtered to select genes that showed transcriptional over-expression and an association with poor prognosis in neuroblastoma, highlighting GATA3 for detailed studies. Specific methylation assays confirmed the hypomethylation of GATA3 in neuroblastoma, which correlated with high expression at both the RNA and protein level. Demethylation with azacytidine in cultured sympathetic ganglia cells led to increased GATA3 expression, suggesting a mechanistic link between GATA3 expression and DNA methylation. Neuroblastomas that had completely absent GATA3 methylation and/or very high levels of protein expression, were associated with poor prognosis. Knock-down of GATA3 in neuroblastoma cells lines inhibited cell proliferation and increased apoptosis but had no effect on cellular differentiation. These results identify GATA3 as an epigenetically regulated component of the neuroblastoma transcriptional control network, that is essential for neuroblastoma proliferation. This suggests that the GATA3 transcriptional network is a promising target for novel neuroblastoma therapies.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Epigenesis, Genetic , GATA3 Transcription Factor/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neuroblastoma/metabolism , Cell Line, Tumor , DNA, Neoplasm/genetics , GATA3 Transcription Factor/genetics , Humans , Neoplasm Proteins/genetics , Neuroblastoma/diagnosis , Neuroblastoma/genetics , Neuroblastoma/pathology , Prognosis
3.
Mol Carcinog ; 56(4): 1290-1301, 2017 04.
Article in English | MEDLINE | ID: mdl-27862318

ABSTRACT

Neuroblastoma is a childhood cancer in which many children still have poor outcomes, emphasising the need to better understand its pathogenesis. Despite recent genome-wide mutation analyses, many primary neuroblastomas do not contain recognizable driver mutations, implicating alternate molecular pathologies such as epigenetic alterations. To discover genes that become epigenetically deregulated during neuroblastoma tumorigenesis, we took the novel approach of comparing neuroblastomas to neural crest precursor cells, using genome-wide DNA methylation analysis. We identified 93 genes that were significantly differentially methylated of which 26 (28%) were hypermethylated and 67 (72%) were hypomethylated. Concentrating on hypermethylated genes to identify candidate tumor suppressor loci, we found the cell engulfment and adhesion factor gene MEGF10 to be epigenetically repressed by DNA hypermethylation or by H3K27/K9 methylation in neuroblastoma cell lines. MEGF10 showed significantly down-regulated expression in neuroblastoma tumor samples; furthermore patients with the lowest-expressing tumors had reduced relapse-free survival. Our functional studies showed that knock-down of MEGF10 expression in neuroblastoma cell lines promoted cell growth, consistent with MEGF10 acting as a clinically relevant, epigenetically deregulated neuroblastoma tumor suppressor gene. © 2016 The Authors. Molecular Carcinogenesis Published by Wiley Periodicals, Inc.


Subject(s)
DNA Methylation , Genes, Tumor Suppressor , Membrane Proteins/genetics , Neuroblastoma/genetics , Cell Line, Tumor , Child , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histone Code , Humans
4.
Mol Carcinog ; 53(5): 413-20, 2014 May.
Article in English | MEDLINE | ID: mdl-23280764

ABSTRACT

Tumor suppressor genes such as RASSF1A are often epigenetically repressed by DNA hypermethylation in neuroblastoma, where the MYCN proto-oncogene is frequently amplified. MYC has been shown to associate with DNA methyltransferases, thereby inducing transcriptional repression of target genes, which suggested that MYCN might play a similar mechanistic role in the hypermethylation of tumor suppressor genes in neuroblastoma. This study tested that hypothesis by using co-immunoprecipitation and ChIP to investigate MYCN-DNA methyltransferase interactions, together with MYCN knock-down and over-expression systems to examine the effect of MYCN expression changes on gene methylation, employing both candidate gene and genome-wide assays. We show that MYCN interacts with DNA methyltransferases and is recruited to the promoter region of RASSF1A. However, using four model systems, we showed that long-term silencing of MYCN induces only a small loss of DNA methylation at the RASSF1A promoter in MYCN amplified neuroblastoma cell lines and over-expression of MYCN does not induce any DNA methylation, suggesting that MYCN is not critical for DNA hypermethylation in neuroblastoma.


Subject(s)
DNA Methylation , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/genetics , Blotting, Western , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Humans , Immunoprecipitation , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/metabolism , Polymerase Chain Reaction , Proto-Oncogene Mas , RNA, Small Interfering/genetics , Tumor Cells, Cultured
5.
Mol Cancer Res ; 6(7): 1114-23, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18644976

ABSTRACT

Epigenetic changes occur frequently in Wilms' tumor (WT), especially loss of imprinting (LOI) of IGF2/H19 at 11p15. Our previous results have identified imprinted transcripts (WT1-AS and AWT1) from the WT1 locus at 11p13 and showed LOI of these in some WTs. In this article, we set out to test the relationship between LOI at 11p13 and 11p15 and their timing in WT progression relative to other genetic changes. We found a higher level (83%) of 11p13 LOI in WT than of 11p15 LOI (71%). There was no correlation between methylation levels at the 11p13 and 11p15 differentially methylated regions or between allelic expression of WT1-AS/AWT1 and IGF2. Interestingly, retention of normal imprinting at 11p13 was associated with a small group of relatively late-onset, high-stage WTs. An examination of genetic and epigenetic alterations in nephrogenic rests, which are premalignant WT precursors, showed that LOI at both 11p13 and 11p15 occurred before either 16q loss of heterozygosity (LOH) or 7p LOH. This suggests that these LOH events are very unlikely to be a cause of LOI but that LOH may act by potentiating the effects of overexpression of IGF2 and/or WT1-AS/AWT1 that result from LOI.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Genomic Imprinting/genetics , Wilms Tumor/genetics , Wilms Tumor/pathology , Adult , DNA Methylation , Disease Progression , Humans , Insulin-Like Growth Factor II/genetics , Loss of Heterozygosity/genetics , Middle Aged , Time Factors
6.
RNA ; 13(12): 2287-99, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17940140

ABSTRACT

Many mammalian genes contain overlapping antisense RNAs, but the functions and mechanisms of action of these transcripts are mostly unknown. WT1 is a well-characterized developmental gene that is mutated in Wilms' tumor (WT) and acute myeloid leukaemia (AML) and has an antisense transcript (WT1-AS), which we have previously found to regulate WT1 protein levels. In this study, we show that WT1-AS is present in multiple spliceoforms that are usually expressed in parallel with WT1 RNA in human and mouse tissues. We demonstrate that the expression of WT1-AS correlates with methylation of the antisense regulatory region (ARR) in WT1 intron 1, displaying imprinted monoallelic expression in normal kidney and loss of imprinting in WT. However, we find no evidence for imprinting of mouse Wt1-as. WT1-AS transcripts are exported into the cytoplasm and form heteroduplexes with WT1 mRNA in the overlapping region in WT1 exon 1. In AML, there is often abnormal splicing of WT1-AS, which may play a role in the development of this malignancy. These results show that WT1 encodes conserved antisense RNAs that may have an important regulatory role in WT1 expression via RNA:RNA interactions, and which can become deregulated by a variety of mechanisms in cancer.


Subject(s)
Alternative Splicing , Neoplasm Proteins/genetics , RNA, Antisense/genetics , RNA, Neoplasm/genetics , Transcription, Genetic , WT1 Proteins/genetics , Animals , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Leukemia, Myeloid, Acute/genetics , Mice , Mutation , Wilms Tumor/genetics
7.
J Gen Virol ; 69(7): 1695-710, July 1988.
Article in English | MedCarib | ID: med-10044

ABSTRACT

We report the first complete nucleotide sequence of an adult T cell leukaemia virus/human T cell leukaemia virus type I (ATLV/HTLV) isolate from a British patient of Caribbean origin. Sequence comparisons of our proviral clone (HS-35) with other molecular clones are shown. We note the strong sequence conservation between isolates of Caribbean and Japanese origin (2.3 percent divergence), but demonstrate the higher homologies existing between isolates originating from similar geographical areas (approximately 1 percent divergence). Implications for the origin, evolution and dissemination of the ATLV/HTLV-I subgroup are discussed. Analysis of defective proviral clones isolated from the same genomic library is also reported,and suggests a pattern of proviral sequence deletions during the biogenesis of defective proviruses. (AU)


Subject(s)
Humans , Genes, Viral , Deltaretrovirus Infections/microbiology , Deltaretrovirus/genetics , Amino Acid Sequence , Base Sequence , England , Deltaretrovirus Infections/ethnology , Deltaretrovirus/classification , Deltaretrovirus/isolation & purification , Molecular Sequence Data , Phylogeny , Proviruses/genetics , Sequence Homology, Nucleic Acid , Viral Envelope Proteins/genetics , West Indies
SELECTION OF CITATIONS
SEARCH DETAIL
...