Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 24(18): 4602-4611, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29898989

ABSTRACT

Purpose:MLH1 is a major tumor suppressor gene involved in the pathogenesis of Lynch syndrome and various sporadic cancers. Despite their potential pathogenic importance, genomic regions capable of regulating MLH1 expression over long distances have yet to be identified.Experimental Design: Here, we use chromosome conformation capture (3C) to screen a 650-kb region flanking the MLH1 locus to identify interactions between the MLH1 promoter and distal regions in MLH1-expressing and nonexpressing cells. Putative enhancers were functionally validated using luciferase reporter assays, chromatin immunoprecipitation, and CRISPR-Cas9-mediated deletion of endogenous regions. To evaluate whether germline variants in the enhancer might contribute to impaired MLH1 expression in patients with suspected Lynch syndrome, we also screened germline DNA from a cohort of 74 patients with no known coding mutations or epimutations at the MLH1 promoter.Results: A 1.8-kb DNA fragment, 35 kb upstream of the MLH1 transcription start site enhances MLH1 gene expression in colorectal cells. The enhancer was bound by CTCF and CRISPR-Cas9-mediated deletion of a core binding region impairs endogenous MLH1 expression. A total of 5.4% of suspected Lynch syndrome patients have a rare single-nucleotide variant (G > A; rs143969848; 2.5% in gnomAD European, non-Finnish) within a highly conserved CTCF-binding motif, which disrupts enhancer activity in SW620 colorectal carcinoma cells.Conclusions: A CTCF-bound region within the MLH1-35 enhancer regulates MLH1 expression in colorectal cells and is worthy of scrutiny in future genetic screening strategies for suspected Lynch syndrome associated with loss of MLH1 expression. Clin Cancer Res; 24(18); 4602-11. ©2018 AACR.


Subject(s)
CCCTC-Binding Factor/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms/genetics , MutL Protein Homolog 1/genetics , Adult , Aged , Aged, 80 and over , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Methylation/genetics , DNA Mismatch Repair/genetics , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic , Germ-Line Mutation/genetics , Humans , Male , Middle Aged , Promoter Regions, Genetic/genetics
2.
Cancer Genet ; 209(11): 497-500, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27886675

ABSTRACT

Lynch syndrome is a hereditary cancer syndrome caused by the autosomal dominant inheritance of loss-of-function mutations in DNA mismatch repair (MMR) genes. Approximately one quarter of clinically suspected cases have no identifiable germline mutation in any MMR gene, a condition known as Lynch-like syndrome (LLS). MCM9 was recently identified as the DNA helicase in the mammalian MMR complex and loss of helicase activity results in microsatellite instability. We hypothesized that pathogenic variants in MCM9 may account for LLS. The 5'UTR and coding region of MCM9 were sequenced in germline DNA of 109 Australian patients with LLS and variants were cross-referenced with three population-based databases (dbSNP144, 1000 Genomes, ExAC). The functional effect of variants was assessed in silico with PolyPhen-2, SIFT and CONDEL. Fifteen variants that included six common SNPs and nine variants of unknown significance (VUS) were identified. We conclude that VUS occur in MCM9 in a small proportion of LLS patients and MCM9 mutations are unlikely to explain most LLS cases.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Germ-Line Mutation , Minichromosome Maintenance Proteins/genetics , Sequence Analysis, DNA/methods , 5' Untranslated Regions , Adult , Aged , Aged, 80 and over , Australia , Computational Biology , Computer Simulation , Databases, Genetic , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Open Reading Frames
3.
Mol Cancer Res ; 14(12): 1217-1228, 2016 12.
Article in English | MEDLINE | ID: mdl-27671336

ABSTRACT

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.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Gene Regulatory Networks , Genomics/methods , Computational Biology/methods , DNA Methylation , Epigenesis, Genetic , Female , Gene Dosage , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study/methods , Humans , Mutation , Sequence Analysis, RNA/methods
4.
J Pathol Clin Res ; 2(3): 127-37, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27499922

ABSTRACT

Serrated polyposis is a rare disorder characterised by the presence of multiple serrated polyps in the large intestine, and an increased personal and familial risk of colorectal cancer. Knowledge of the molecular characteristics of colonic lesions which develop in this syndrome is fragmented, making it difficult to understand the underlying genetic basis of this condition. We conducted a systematic review and meta-analysis of all studies which evaluated the molecular characteristics of colorectal neoplasms found in individuals with serrated polyposis. We identified 4561 potentially relevant studies, but due to a lack of consensus in the reporting of findings, only fourteen studies were able to be included in the meta-analysis. BRAF mutation was found in 73% (95% CI 65-80%) of serrated polyps, 0% (95% CI 0-3%) of conventional adenomas and 49% (95%CI 33-64%) of colorectal cancers. In contrast, KRAS mutation was present in 8% (95% CI 5-11%) of serrated polyps, 3% (95% CI 0-13%) of conventional adenomas and 6% (95% CI 0-13%) of colorectal cancers. Absence of MLH1 immunostaining was found in 3% (95% CI 0-10%) of serrated polyps and 53% (95% CI 36-71%) of colorectal cancers. Overall, microsatellite instability was found in 40% (95% CI 18-64%) of colorectal cancers arising in the setting of serrated polyposis. Our results indicate that diverse molecular pathways are likely to contribute to the increased predisposition for colorectal cancer in individuals with serrated polyposis. We also propose a set of minimum standards for the reporting of future research in serrated polyposis as this is a rare syndrome and collation of research findings from different centres will be essential to identify the molecular mechanisms involved in the pathogenesis of this condition.

5.
Clin Epigenetics ; 8: 39, 2016.
Article in English | MEDLINE | ID: mdl-27096027

ABSTRACT

In the January 2016 issue of Clinical Epigenetics, Quiñonez-Silva et al. (Clin Epigenetics 8:1, 2016) described a possible constitutional epimutation of the RB1 gene as a cause of hereditary predisposition to retinoblastoma. The term constitutional epimutation describes an epigenetic aberration in normal tissues that predisposes to disease. The data presented by Quiñonez-Silva et al. are interesting, but further analysis is required to demonstrate a constitutional epimutation in this family. Here, we define the criteria and describe the experimental approach necessary to identify an epigenetic aberration as a constitutional epimutation.


Subject(s)
DNA Methylation , Mutation , Epigenesis, Genetic , Genetic Predisposition to Disease , Humans
6.
Hum Mutat ; 37(5): 417-26, 2016 May.
Article in English | MEDLINE | ID: mdl-26888055

ABSTRACT

Lynch syndrome is the most common familial cancer condition that mainly predisposes to tumors of the colon and endometrium. Cancer susceptibility is caused by the autosomal dominant inheritance of a loss-of-function mutation or epimutation in one of the DNA mismatch repair (MMR) genes. Cancer risk assessment is often possible with nonsynonymous coding region mutations, but in many cases patients present with DNA sequence changes within noncoding regions, including the promoters, of MMR genes. The pathogenic role of promoter variants, and hence clinical significance, is unclear and this hinders the clinical management of carriers. In this review, we provide an overview of the classification of MMR gene variants, outline the laboratory assays and online resources that can be used to assess the causality of promoter variants in Lynch syndrome, and highlight some of the practical challenges of demonstrating the pathogenicity of these variants. In conclusion, we propose a guide that could be integrated into the current InSiGHT classification scheme to help determine if a MMR gene promoter variant is pathogenic.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair , Genetic Variation , Promoter Regions, Genetic , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics
7.
Carcinogenesis ; 37(1): 10-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26498247

ABSTRACT

Lynch syndrome is an autosomal dominant disorder that predisposes carriers of DNA mismatch repair (MMR) gene mutations to early-onset cancer. Germline testing screens exons and splice sites for mutations, but does not examine introns or RNA transcripts for alterations. Pathogenic mutations have not been detected in ~30% of suspected Lynch syndrome cases with standard screening practices. We present a 38-year-old male with a clinicopathological and family history consistent with Lynch syndrome, including loss of MSH2 expression in his tumor. Germline testing revealed normal MSH2 coding sequence, splice sites and exon copy number, however, cDNA sequencing identified an aberrant MSH2 transcript lacking exons 2-6. An inversion PCR on germline DNA identified an ~18kb unbalanced, paracentric inversion within MSH2, with breakpoints in a long terminal repeat in intron 1 and an Alu repeat in intron 6. The 3' end of the inversion had a 1.2 kb deletion and an 8 bp insertion at the junction with intron 6. Screening of 55 additional Australian patients presenting with MSH2-deficient tumors who were negative in germline genetic tests for MSH2 mutations identified another inversion-positive patient. We propose an Alu-mediated recombination model to explain the origin of the inversion. Our study illustrates the potential value of cDNA screening to identify patients with cryptic MMR gene rearrangements, clarifies why standard testing may not detect some pathogenic alterations, and provides a genetic test for screening individuals with suspected Lynch syndrome that present with unexplained MSH2-deficient tumors.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Exons , MutS Homolog 2 Protein/genetics , Sequence Inversion , Adult , Amino Acid Sequence , Base Sequence , Colorectal Neoplasms, Hereditary Nonpolyposis/blood , DNA Mutational Analysis/methods , DNA, Complementary/blood , DNA, Complementary/genetics , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Gene Rearrangement , Germ-Line Mutation , Humans , Male , Molecular Sequence Data , Pedigree
8.
Oncotarget ; 6(32): 32509-25, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26356674

ABSTRACT

With the advent of high-throughput and relatively inexpensive whole-genome sequencing technology, the focus of cancer research has begun to shift toward analyses of somatic mutations in non-coding cis-regulatory elements of the cancer genome. Cis-regulatory elements play an important role in gene regulation, with mutations in these elements potentially resulting in changes to the expression of linked genes. The recent discoveries of recurrent TERT promoter mutations in melanoma, and recurrent mutations that create a super-enhancer regulating TAL1 expression in T-cell acute lymphoblastic leukaemia (T-ALL), have sparked significant interest in the search for other somatic cis-regulatory mutations driving cancer development. In this review, we look more closely at the TERT promoter and TAL1 enhancer alterations and use these examples to ask whether other cis-regulatory mutations may play a role in cancer susceptibility. In doing so, we make observations from the data emerging from recent research in this field, and describe the experimental and analytical approaches which could be adopted in the hope of better uncovering the true functional significance of somatic cis-regulatory mutations in cancer.


Subject(s)
Biomarkers, Tumor/genetics , Genome, Human , Mutation , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Animals , Biomarkers, Tumor/metabolism , DNA Mutational Analysis/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/metabolism , Neoplasms/pathology , Phenotype
9.
JAMA Oncol ; 1(7): 953-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26181641

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Epigenesis, Genetic , Mosaicism , Nuclear Proteins/genetics , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Colorectal Neoplasms/pathology , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genetic Testing , Heredity , Humans , Male , MutL Protein Homolog 1 , Pedigree , Phenotype , Risk Assessment , Risk Factors , Young Adult
10.
Hum Mutat ; 36(6): 622-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25762362

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Allelic Imbalance , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Gene Expression , Genetic Variation , Nuclear Proteins/genetics , Promoter Regions, Genetic , 5' Untranslated Regions , Age of Onset , CpG Islands , DNA Methylation , Epigenesis, Genetic , Female , Genetic Association Studies , Genetic Loci , High-Throughput Nucleotide Sequencing , Humans , Male , Microsatellite Repeats , MutL Protein Homolog 1 , Mutation , Pedigree
11.
Epigenetics ; 9(10): 1422-30, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25437056

ABSTRACT

Gene silencing in cancer frequently involves hypermethylation and dense nucleosome occupancy across promoter regions. How a promoter transitions to this silent state is unclear. Using colorectal adenomas, we investigated nucleosome positioning, DNA methylation, and gene expression in the early stages of gene silencing. Genome-wide gene expression correlated with highly positioned nucleosomes upstream and downstream of a nucleosome-depleted transcription start site (TSS). Hypermethylated promoters displayed increased nucleosome occupancy, specifically at the TSS. We investigated 2 genes, CDH1 and CDKN2B, which were silenced in adenomas but lacked promoter hypermethylation. Instead, silencing correlated with loss of nucleosomes from the -2 position upstream of the TSS relative to normal mucosa. In contrast, permanent CDH1 silencing in carcinoma cells was characterized by promoter hypermethylation and dense nucleosome occupancy. Our findings suggest that silenced genes transition through an intermediary stage involving altered promoter nucleosome positioning, before permanent silencing by hypermethylation and dense nucleosome occupancy.


Subject(s)
DNA Methylation , Gene Silencing , Nucleosomes/genetics , Promoter Regions, Genetic , Transcription Initiation Site , Adenoma/genetics , Aged , Antigens, CD , Cadherins/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Epigenesis, Genetic , Female , Humans , Male , Middle Aged
12.
Clin Epigenetics ; 6(1): 32, 2014.
Article in English | MEDLINE | ID: mdl-25530820

ABSTRACT

BACKGROUND: Splicing is more efficient when coupled with transcription and it has been proposed that nucleosomes enriched in exons are important for splice site recognition. Lynch syndrome is a familial cancer syndrome that can be caused by the autosomal dominant inheritance of splice site mutations in the MutL homolog 1 (MLH1) gene. To better understand the role of nucleosomes in splicing, we used MLH1 splice site mutations in Lynch syndrome cases as a model to investigate if abnormal splicing was associated with altered nucleosome positioning at exon-intron boundaries. FINDINGS: Nucleosome Occupancy and Methylome sequencing (NOMe-seq) was used to determine the allele-specific positioning of nucleosomes around heterozygous splice site mutations in lymphoblastoid cells lines (LCLs) derived from six Lynch syndrome patients. These mutations were previously shown to cause exon skipping in five of the six patients. Allele-specific high-resolution nucleosome mapping across exons and exon-intron boundaries revealed high levels of nucleosomes across all regions examined. Alleles containing donor or acceptor splice site mutations showed no consistent alteration in nucleosome positioning or occupancy. CONCLUSION: Nucleosomes were enriched at MLH1 exons in LCLs derived from Lynch syndrome patients, and in this model system the positioning of nucleosomes was unaltered at exon-intron boundaries containing splice site mutations. Thus, these splice site mutations alone do not significantly change the local organisation of nucleosomes.

13.
Epigenetics ; 9(8): 1092-100, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24837038

ABSTRACT

In mouse models, loss of the candidate tumor suppressor gene Ubiquitin Specific Protease 44 (USP44) is associated with aneuploidy and cancer. USP44 is also transcriptionally silenced in human cancers. Here we investigated the molecular mechanism of USP44 silencing and whether this correlated with aneuploidy in colorectal adenomas. DNA methylation at the USP44 CpG island (CGI) promoter was measured using combined bisulfite restriction analysis (COBRA) in colorectal cancer (CRC) cell lines (n = 18), and with COBRA and bisulfite sequencing in colorectal adenomas (n = 89) and matched normal colonic mucosa (n = 51). The USP44 CGI was hypermethylated in all CRC cell lines, in most colorectal adenomas (79 of 89, 89%) but rarely in normal mucosa samples (3 of 51, 6%). USP44 expression was also compared between normal mucosa and paired hypermethylated adenomas in six patients using qRT-PCR. Hypermethylation of the USP44 CGI in adenomas was associated with a 1.8 to 5.5-fold reduction in expression compared with paired normal mucosa. Treatment of CRC cell lines with the DNA hypomethylating agent decitabine resulted in a 14 to 270-fold increase in USP44 expression. Whole genome SNP array data showed that gain or loss of individual chromosomes occurred in adenomas, but hypermethylation did not correlate with more aneuploidy. In summary, our data shows that USP44 is epigenetically inactivated in colorectal adenomas, but this alone is not sufficient to cause aneuploidy in colorectal neoplasia.


Subject(s)
Adenoma/genetics , Aneuploidy , Colorectal Neoplasms/genetics , Epigenesis, Genetic , Ubiquitin-Specific Proteases/genetics , Adenoma/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Colon/metabolism , Colorectal Neoplasms/metabolism , CpG Islands , Decitabine , Gene Expression , Genes, Tumor Suppressor , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Rectum/metabolism , Ubiquitin Thiolesterase , Young Adult
14.
PLoS Genet ; 9(7): e1003636, 2013.
Article in English | MEDLINE | ID: mdl-23935509

ABSTRACT

Hypomethylating agents reactivate tumor suppressor genes that are epigenetically silenced in cancer. Inevitably these genes are resilenced, leading to drug resistance. Using the MLH1 tumor suppressor gene as a model, we showed that decitabine-induced re-expression was dependent upon demethylation and eviction of promoter nucleosomes. Following decitabine withdrawal, MLH1 was rapidly resilenced despite persistent promoter demethylation. Single molecule analysis at multiple time points showed that gene resilencing was initiated by nucleosome reassembly on demethylated DNA and only then was followed by remethylation and stable silencing. Taken together, these data establish the importance of nucleosome positioning in mediating resilencing of drug-induced gene reactivation and suggest a role for therapeutic targeting of nucleosome assembly as a mechanism to overcome drug resistance.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA Methylation/genetics , Drug Resistance, Neoplasm/genetics , Nuclear Proteins/genetics , Nucleosomes/genetics , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , CpG Islands/genetics , DNA Methylation/drug effects , Decitabine , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , MutL Protein Homolog 1 , Nucleosomes/metabolism , Promoter Regions, Genetic/drug effects
15.
Genome Res ; 19(2): 221-33, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047520

ABSTRACT

In mammals, genome-wide chromatin maps and immunofluorescence studies show that broad domains of repressive histone modifications are present on pericentromeric and telomeric repeats and on the inactive X chromosome. However, only a few autosomal loci such as silent Hox gene clusters have been shown to lie in broad domains of repressive histone modifications. Here we present a ChIP-chip analysis of the repressive H3K27me3 histone modification along chr 17 in mouse embryonic fibroblast cells using an algorithm named broad local enrichments (BLOCs), which allows the identification of broad regions of histone modifications. Our results, confirmed by BLOC analysis of a whole genome ChIP-seq data set, show that the majority of H3K27me3 modifications form BLOCs rather than focal peaks. H3K27me3 BLOCs modify silent genes of all types, plus flanking intergenic regions and their distribution indicates a negative correlation between H3K27me3 and transcription. However, we also found that some nontranscribed gene-poor regions lack H3K27me3. We therefore performed a low-resolution analysis of whole mouse chr 17, which revealed that H3K27me3 is enriched in mega-base-pair-sized domains that are also enriched for genes, short interspersed elements (SINEs) and active histone modifications. These genic H3K27me3 domains alternate with similar-sized gene-poor domains. These are deficient in active histone modifications, as well as H3K27me3, but are enriched for long interspersed elements (LINEs) and long-terminal repeat (LTR) transposons and H3K9me3 and H4K20me3. Thus, an autosome can be seen to contain alternating chromatin bands that predominantly separate genes from one retrotransposon class, which could offer unique domains for the specific regulation of genes or the silencing of autonomous retrotransposons.


Subject(s)
Chromosomes, Mammalian/metabolism , DNA, Intergenic/metabolism , Gene Silencing/physiology , Histones/metabolism , Protein Multimerization/physiology , Algorithms , Animals , Chromatin Immunoprecipitation/methods , Chromosome Banding/methods , Chromosomes, Mammalian/chemistry , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Lysine/metabolism , Methylation , Mice , Models, Biological , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational/physiology , Substrate Specificity
16.
EMBO J ; 27(23): 3116-28, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-19008856

ABSTRACT

The Airn macro ncRNA is the master regulator of imprinted expression in the Igf2r imprinted gene cluster where it silences three flanking genes in cis. Airn transcription shows unusual features normally viewed as promoter specific, such as impaired post-transcriptional processing and a macro size. The Airn transcript is 108 kb long, predominantly unspliced and nuclear localized, with only a minority being variably spliced and exported. Here, we show by deletion of the Airn ncRNA promoter and replacement with a constitutive strong or weak promoter that splicing suppression and termination, as well as silencing activity, are maintained by strong Airn expression from an exogenous promoter. This indicates that all functional regions are located within the Airn transcript. DNA methylation of the maternal imprint control element (ICE) restricts Airn expression to the paternal allele and we also show that a strong active promoter is required to maintain the unmethylated state of the paternal ICE. Thus, Airn expression not only induces silencing of flanking mRNA genes but also protects the paternal copy of the ICE from de novo methylation.


Subject(s)
Gene Silencing , Promoter Regions, Genetic , RNA, Untranslated/genetics , Transcription, Genetic , Cells, Cultured , DNA Methylation , Gene Expression , Humans , Multigene Family , Sequence Deletion
17.
Mol Cell ; 27(3): 353-66, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17679087

ABSTRACT

The Igf2r imprinted cluster is an epigenetic silencing model in which expression of a ncRNA silences multiple genes in cis. Here, we map a 250 kb region in mouse embryonic fibroblast cells to show that histone modifications associated with expressed and silent genes are mutually exclusive and localized to discrete regions. Expressed genes were modified at promoter regions by H3K4me3 + H3K4me2 + H3K9Ac and on putative regulatory elements flanking active promoters by H3K4me2 + H3K9Ac. Silent genes showed two types of nonoverlapping profile. One type spread over large domains of tissue-specific silent genes and contained H3K27me3 alone. A second type formed localized foci on silent imprinted gene promoters and a nonexpressed pseudogene and contained H3K9me3 + H4K20me3 +/- HP1. Thus, mammalian chromosome arms contain active chromatin interspersed with repressive chromatin resembling the type of heterochromatin previously considered a feature of centromeres, telomeres, and the inactive X chromosome.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Genome , Genomic Imprinting , Histones/metabolism , Animals , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Chromosomes , Embryo, Mammalian , Fibroblasts/metabolism , Gene Silencing , Methylation , Mice , Physical Chromosome Mapping
18.
Genet Res ; 79(3): 203-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12220127

ABSTRACT

We used polymorphic microsatellite markers to look for recombination during parthenogenetic oogenesis between the X chromosomes of aphids of the tribe Macrosiphini. We examined the X chromosome because it comprises approximately 25 % of the genome and previous cytological observations of chromosome pairing and nucleolar organizer (NOR) heteromorphism suggest recombination, although the same is not true for autosomes. A total of 564 parthenogenetic females of Myzus clones with three distinct reproductive modes (cyclical parthenogenesis, obligate parthenogenesis and obligate parthenogenesis with male production) were genotyped at three informative X-linked loci. Also, parthenogenetically produced males from clones encompassing the full range of male-producing reproductive strategies were genotyped. These included 391 Myzus persicae males that were genotyped at three X-linked loci and 538 males from Sitobion clones that were genotyped at five informative X-linked loci. Our results show no departure from clonality in parthenogenetic generations of aphids of the tribe Macrosiphini: no recombinant genotypes were observed in parthenogenetically produced males or females.


Subject(s)
Aphids/physiology , Parthenogenesis/genetics , Recombination, Genetic , X Chromosome , Animals , Female , Genetic Linkage , Heterochromatin/genetics , Male , Microsatellite Repeats
19.
Genet Res ; 79(2): 119-27, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12073550

ABSTRACT

Most aphids are cyclic parthenogens, so are ideal models in studies of the mechanisms and consequences of sex and recombination. However, owing to a shortage of physical and genetic markers, there have been few studies of the most fundamental genetic processes in these organisms. For example, it is not known whether autosomal segregation during male spermatogenesis is in Mendelian proportions: we address that question here. The aphid Myzus persicae has a typical karyotype of 2n = 12 in females (XX), while males are XO (2n = 11). During male meiosis, only the spermatocytes with an X chromosome are viable. We hypothesized that assortment of autosomes might be non-random because chromosomal imprinting leading to elimination of the paternal autosomes is seen in the closely related coccoids. In other aphid models, we have observed segregation distortions at single microsatellite loci (Wilson, 2000). Such distortions may have nothing to do with 'selfish' behaviour, but may be caused by mutation accumulation causing fitness differentials. Thus single-locus distortions might be predicted to be more likely to be detected via the male lines of clones that have lost the ability to reproduce sexually (male-producing obligate parthenogenesis (androcyclic)). Using microsatellites we show that genetic imprinting or selfish autosome behaviour does not occur in male M. persicae. Generally, loci segregated in Mendelian proportions in both sexes of cyclically parthenogenetic (holocyclic) clones. However, in androcyclic clones, segregation distortions consistently involved the same two autosomes. This is consistent with linkage of markers to deleterious mutations associated with a loss of sexual reproduction.


Subject(s)
Aphids/genetics , Chromosome Segregation/genetics , Spermatogenesis/genetics , Animals , Chromosome Aberrations , Crosses, Genetic , Female , Genetic Linkage , Genotype , Male , Microsatellite Repeats
SELECTION OF CITATIONS
SEARCH DETAIL
...