Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Publication year range
1.
Nat Rev Genet ; 25(8): 533, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38684857
2.
Genome Res ; 30(9): 1217-1227, 2020 09.
Article in English | MEDLINE | ID: mdl-32820006

ABSTRACT

Multiple myeloma (MM) is a plasma cell neoplasm associated with a broad variety of genetic lesions. In spite of this genetic heterogeneity, MMs share a characteristic malignant phenotype whose underlying molecular basis remains poorly characterized. In the present study, we examined plasma cells from MM using a multi-epigenomics approach and demonstrated that, when compared to normal B cells, malignant plasma cells showed an extensive activation of regulatory elements, in part affecting coregulated adjacent genes. Among target genes up-regulated by this process, we found members of the NOTCH, NF-kB, MTOR signaling, and TP53 signaling pathways. Other activated genes included sets involved in osteoblast differentiation and response to oxidative stress, all of which have been shown to be associated with the MM phenotype and clinical behavior. We functionally characterized MM-specific active distant enhancers controlling the expression of thioredoxin (TXN), a major regulator of cellular redox status and, in addition, identified PRDM5 as a novel essential gene for MM. Collectively, our data indicate that aberrant chromatin activation is a unifying feature underlying the malignant plasma cell phenotype.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Plasma Cells/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Humans , NF-kappa B/metabolism , Osteogenesis/genetics , Receptors, Notch/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Thioredoxins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation
3.
Nature ; 526(7574): 519-24, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26200345

ABSTRACT

Chronic lymphocytic leukaemia (CLL) is a frequent disease in which the genetic alterations determining the clinicobiological behaviour are not fully understood. Here we describe a comprehensive evaluation of the genomic landscape of 452 CLL cases and 54 patients with monoclonal B-lymphocytosis, a precursor disorder. We extend the number of CLL driver alterations, including changes in ZNF292, ZMYM3, ARID1A and PTPN11. We also identify novel recurrent mutations in non-coding regions, including the 3' region of NOTCH1, which cause aberrant splicing events, increase NOTCH1 activity and result in a more aggressive disease. In addition, mutations in an enhancer located on chromosome 9p13 result in reduced expression of the B-cell-specific transcription factor PAX5. The accumulative number of driver alterations (0 to ≥4) discriminated between patients with differences in clinical behaviour. This study provides an integrated portrait of the CLL genomic landscape, identifies new recurrent driver mutations of the disease, and suggests clinical interventions that may improve the management of this neoplasia.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation/genetics , 3' Untranslated Regions/genetics , Alternative Splicing/genetics , B-Lymphocytes/metabolism , Carrier Proteins/genetics , Chromosomes, Human, Pair 9/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA-Binding Proteins , Enhancer Elements, Genetic/genetics , Genomics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , PAX5 Transcription Factor/biosynthesis , PAX5 Transcription Factor/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Transcription Factors/genetics
5.
Curr Opin Hematol ; 25(4): 299-306, 2018 07.
Article in English | MEDLINE | ID: mdl-29738333

ABSTRACT

PURPOSE OF REVIEW: SOX11 has emerged as a key transcription factor in the pathogenesis of mantle cell lymphoma (MCL) whereas it is not expressed in normal B cells or virtually in any other mature B-cell neoplasm. This review will examine the role of SOX11 as a biomarker in MCL, the new information on its transcriptional targets, and the mechanisms regulating its expression in MCL. RECENT FINDINGS: SOX11 is highly expressed in conventional MCL, including cyclin D1-negative cases, but it is not expressed in the indolent leukemic nonnodal MCL subtype. These two MCL subtypes also differ in their cell-of-origin, IGHV mutational status and genomic instability. SOX11 promotes tumor growth of MCL cells in vivo and regulates a broad transcriptional program that includes B-cell differentiation pathways and tumor-microenvironment interactions, among others. The mechanisms upregulating SOX11 in MCL are not well understood but are mediated in part by the three-dimensional reconfiguration of the DNA, bringing together a distant enhancer region and the SOX11 promoter. SUMMARY: SOX11 is a relevant element in the pathogenesis of MCL and has been instrumental to identify two distinct clinicobiological subtypes of this tumor. Further studies should clarify the mechanisms mediating its oncogenic potential and leading to its intriguing expression in these tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Mantle-Cell/metabolism , Neoplasm Proteins/metabolism , SOXC Transcription Factors/metabolism , Tumor Microenvironment , Animals , Biomarkers, Tumor/genetics , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Neoplasm Proteins/genetics , SOXC Transcription Factors/genetics
6.
Genome Res ; 25(4): 478-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644835

ABSTRACT

While analyzing the DNA methylome of multiple myeloma (MM), a plasma cell neoplasm, by whole-genome bisulfite sequencing and high-density arrays, we observed a highly heterogeneous pattern globally characterized by regional DNA hypermethylation embedded in extensive hypomethylation. In contrast to the widely reported DNA hypermethylation of promoter-associated CpG islands (CGIs) in cancer, hypermethylated sites in MM, as opposed to normal plasma cells, were located outside CpG islands and were unexpectedly associated with intronic enhancer regions defined in normal B cells and plasma cells. Both RNA-seq and in vitro reporter assays indicated that enhancer hypermethylation is globally associated with down-regulation of its host genes. ChIP-seq and DNase-seq further revealed that DNA hypermethylation in these regions is related to enhancer decommissioning. Hypermethylated enhancer regions overlapped with binding sites of B cell-specific transcription factors (TFs) and the degree of enhancer methylation inversely correlated with expression levels of these TFs in MM. Furthermore, hypermethylated regions in MM were methylated in stem cells and gradually became demethylated during normal B-cell differentiation, suggesting that MM cells either reacquire epigenetic features of undifferentiated cells or maintain an epigenetic signature of a putative myeloma stem cell progenitor. Overall, we have identified DNA hypermethylation of developmentally regulated enhancers as a new type of epigenetic modification associated with the pathogenesis of MM.


Subject(s)
DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Multiple Myeloma/genetics , Neoplastic Stem Cells/cytology , Plasma Cells/cytology , Cell Differentiation/genetics , Cell Line, Tumor , CpG Islands/genetics , DNA, Neoplasm/genetics , Down-Regulation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Promoter Regions, Genetic , Transcription Factors/biosynthesis , Transcription Factors/genetics
7.
Blood ; 123(14): 2229-37, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24523240

ABSTRACT

Severe congenital neutropenia (CN) is a preleukemic bone marrow failure syndrome with a 20% risk of evolving into leukemia or myelodysplastic syndrome (MDS). Patterns of acquisition of leukemia-associated mutations were investigated using next-generation deep-sequencing in 31 CN patients who developed leukemia or MDS. Twenty (64.5%) of the 31 patients had mutations in RUNX1. A majority of patients with RUNX1 mutations (80.5%) also had acquired CSF3R mutations. In contrast to their high frequency in CN patients who developed leukemia or MDS, RUNX1 mutations were found in only 9 of 307 (2.9%) patients with de novo pediatric acute myeloid leukemia. A sequential analysis at stages prior to overt leukemia revealed RUNX1 mutations to be late events in leukemic transformation. Single-cell analyses in 2 patients showed that RUNX1 and CSF3R mutations were present in the same malignant clone. Functional studies demonstrated elevated granulocyte colony-stimulating factor (G-CSF)-induced proliferation with diminished myeloid differentiation of hematopoietic CD34(+) cells coexpressing mutated forms of RUNX1 and CSF3R. The high frequency of cooperating RUNX1 and CSF3R mutations in CN patients suggests a novel molecular pathway of leukemogenesis: mutations in the hematopoietic cytokine receptor (G-CSFR) in combination with the second mutations in the downstream hematopoietic transcription fator (RUNX1). The detection of both RUNX1 and CSF3R mutations could be used as a marker for identifying CN patients with a high risk of progressing to leukemia or MDS.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid/genetics , Mutation , Neutropenia/congenital , Receptors, Colony-Stimulating Factor/genetics , Adolescent , Adult , Child , Child, Preschool , Congenital Bone Marrow Failure Syndromes , Cytogenetic Analysis , Female , Humans , Male , Neutropenia/genetics , Neutropenia/pathology , Signal Transduction/genetics , Young Adult
8.
Biochim Biophys Acta ; 1829(11): 1161-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23938249

ABSTRACT

Ever since the discovery of DNA methylation at cytosine residues, the role of this so called fifth base has been extensively studied and debated. Until recently, the majority of DNA methylation studies focused on the analysis of CpG islands associated to promoter regions. However, with the upcoming possibilities to study DNA methylation in a genome-wide context, this epigenetic mark can now be studied in an unbiased manner. As a result, recent studies have shown that not only promoters but also intragenic and intergenic regions are widely modulated during physiological processes and disease. In particular, it is becoming increasingly clear that DNA methylation in the gene body is not just a passive witness of gene transcription but it seems to be actively involved in multiple gene regulation processes. In this review we discuss the potential role of intragenic DNA methylation in alternative promoter usage, regulation of short and long non-coding RNAs, alternative RNA processing, as well as enhancer activity. Furthermore, we summarize how the intragenic DNA methylome is modified both during normal cell differentiation and neoplastic transformation.


Subject(s)
Cell Differentiation/genetics , DNA Methylation , Gene Expression Regulation , Neoplasms/genetics , Transcription, Genetic , DNA Transposable Elements , Enhancer Elements, Genetic , Humans , Neoplasms/pathology , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional
10.
Blood ; 119(22): 5071-7, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22371884

ABSTRACT

Severe congenital neutropenia (SCN) is a BM failure syndrome with a high risk of progression to acute myeloid leukemia (AML). The underlying genetic changes involved in SCN evolution to AML are largely unknown. We obtained serial hematopoietic samples from an SCN patient who developed AML 17 years after the initiation of G-CSF treatment. Next- generation sequencing was performed to identify mutations during disease progression. In the AML phase, we found 12 acquired nonsynonymous mutations. Three of these, in CSF3R, LLGL2, and ZC3H18, co-occurred in a subpopulation of progenitor cells already in the early SCN phase. This population expanded over time, whereas clones harboring only CSF3R mutations disappeared from the BM. The other 9 mutations were only apparent in the AML cells and affected known AML-associated genes (RUNX1 and ASXL1) and chromatin remodelers (SUZ12 and EP300). In addition, a novel CSF3R mutation that conferred autonomous proliferation to myeloid progenitors was found. We conclude that progression from SCN to AML is a multistep process, with distinct mutations arising early during the SCN phase and others later in AML development. The sequential gain of 2 CSF3R mutations implicates abnormal G-CSF signaling as a driver of leukemic transformation in this case of SCN.


Subject(s)
Cell Transformation, Neoplastic/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Neoplasm Proteins/genetics , Neutropenia/genetics , Adult , Bone Marrow/metabolism , Cell Transformation, Neoplastic/metabolism , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Male , Neoplasm Proteins/metabolism , Neutropenia/complications , Neutropenia/congenital , Neutropenia/drug therapy , Neutropenia/metabolism , Stem Cells/metabolism
11.
Blood ; 128(19): 2279-2280, 2016 11 10.
Article in English | MEDLINE | ID: mdl-28829750
12.
Blood ; 115(25): 5131-6, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20237318

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) has been used in the clinic for more than 2 decades to treat congenital and acquired neutropenias and to reduce febrile neutropenia before or during courses of intensive cytoreductive therapy. In addition, healthy stem cell donors receive short-term treatment with G-CSF for mobilization of hematopoietic stem cells. G-CSF has also been applied in priming strategies designed to enhance the sensitivity of leukemia stem cells to cytotoxic agents, in protocols aimed to induce their differentiation and accompanying growth arrest and cell death, and in severe aplastic anemia and myelodysplastic syndrome (MDS) to alleviate anemia. The potential adverse effects of G-CSF administration, particularly the risk of malignant transformation, have fueled ongoing debates, some of which can only be settled in follow-up studies extending over several decades. This specifically applies to children with severe congenital neutropenia who receive lifelong treatment with G-CSF and in which the high susceptibility to develop MDS and acute myeloid leukemia (AML) has now become a major clinical concern. Here, we will highlight some of the controversies and challenges regarding the clinical application of G-CSF and discuss a possible role of G-CSF in malignant transformation, particularly in patients with neutropenia harboring mutations in the gene encoding the G-CSF receptor.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Granulocyte Colony-Stimulating Factor/adverse effects , Leukemia, Myeloid, Acute/chemically induced , Myelodysplastic Syndromes/chemically induced , Neutropenia/drug therapy , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Anemia, Aplastic/pathology , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , Child, Preschool , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Living Donors , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neutropenia/congenital , Neutropenia/metabolism , Neutropenia/pathology , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Proteins , Risk Factors
13.
Genome Biol ; 23(1): 229, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36307828

ABSTRACT

Single-cell DNA methylation profiling currently suffers from excessive noise and/or limited cellular throughput. We developed scTAM-seq, a targeted bisulfite-free method for profiling up to 650 CpGs in up to 10,000 cells per experiment, with a dropout rate as low as 7%. We demonstrate that scTAM-seq can resolve DNA methylation dynamics across B-cell differentiation in blood and bone marrow, identifying intermediate differentiation states that were previously masked. scTAM-seq additionally queries surface-protein expression, thus enabling integration of single-cell DNA methylation information with cell atlas data. In summary, scTAM-seq is a high-throughput, high-confidence method for analyzing DNA methylation at single-CpG resolution across thousands of single cells.


Subject(s)
DNA Methylation , High-Throughput Nucleotide Sequencing , CpG Islands , Sequence Analysis, DNA/methods , High-Throughput Nucleotide Sequencing/methods
14.
Nat Commun ; 12(1): 651, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510161

ABSTRACT

To investigate the three-dimensional (3D) genome architecture across normal B cell differentiation and in neoplastic cells from different subtypes of chronic lymphocytic leukemia and mantle cell lymphoma patients, here we integrate in situ Hi-C and nine additional omics layers. Beyond conventional active (A) and inactive (B) compartments, we uncover a highly-dynamic intermediate compartment enriched in poised and polycomb-repressed chromatin. During B cell development, 28% of the compartments change, mostly involving a widespread chromatin activation from naive to germinal center B cells and a reversal to the naive state upon further maturation into memory B cells. B cell neoplasms are characterized by both entity and subtype-specific alterations in 3D genome organization, including large chromatin blocks spanning key disease-specific genes. This study indicates that 3D genome interactions are extensively modulated during normal B cell differentiation and that the genome of B cell neoplasias acquires a tumor-specific 3D genome architecture.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Genome, Human/genetics , B-Lymphocytes/cytology , Gene Expression Regulation, Neoplastic , Genomics/methods , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology
15.
Leukemia ; 35(7): 2002-2016, 2021 07.
Article in English | MEDLINE | ID: mdl-33953289

ABSTRACT

B cells have the unique property to somatically alter their immunoglobulin (IG) genes by V(D)J recombination, somatic hypermutation (SHM) and class-switch recombination (CSR). Aberrant targeting of these mechanisms is implicated in lymphomagenesis, but the mutational processes are poorly understood. By performing whole genome and transcriptome sequencing of 181 germinal center derived B-cell lymphomas (gcBCL) we identified distinct mutational signatures linked to SHM and CSR. We show that not only SHM, but presumably also CSR causes off-target mutations in non-IG genes. Kataegis clusters with high mutational density mainly affected early replicating regions and were enriched for SHM- and CSR-mediated off-target mutations. Moreover, they often co-occurred in loci physically interacting in the nucleus, suggesting that mutation hotspots promote increased mutation targeting of spatially co-localized loci (termed hypermutation by proxy). Only around 1% of somatic small variants were in protein coding sequences, but in about half of the driver genes, a contribution of B-cell specific mutational processes to their mutations was found. The B-cell-specific mutational processes contribute to both lymphoma initiation and intratumoral heterogeneity. Overall, we demonstrate that mutational processes involved in the development of gcBCL are more complex than previously appreciated, and that B cell-specific mutational processes contribute via diverse mechanisms to lymphomagenesis.


Subject(s)
Genome/genetics , Germinal Center/metabolism , Lymphoma, B-Cell/genetics , Mutation/genetics , Adult , B-Lymphocytes/metabolism , Cell Line , Cell Line, Tumor , Genes, Immunoglobulin/genetics , HeLa Cells , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunoglobulin Class Switching/genetics , K562 Cells , MCF-7 Cells , Somatic Hypermutation, Immunoglobulin/genetics , V(D)J Recombination/genetics
16.
BMC Bioinformatics ; 11: 275, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20492700

ABSTRACT

BACKGROUND: Tiling-arrays are applicable to multiple types of biological research questions. Due to its advantages (high sensitivity, resolution, unbiased), the technology is often employed in genome-wide investigations. A major challenge in the analysis of tiling-array data is to define regions-of-interest, i.e., contiguous probes with increased signal intensity (as a result of hybridization of labeled DNA) in a region. Currently, no standard criteria are available to define these regions-of-interest as there is no single probe intensity cut-off level, different regions-of-interest can contain various numbers of probes, and can vary in genomic width. Furthermore, the chromosomal distance between neighboring probes can vary across the genome among different arrays. RESULTS: We have developed Hypergeometric Analysis of Tiling-arrays (HAT), and first evaluated its performance for tiling-array datasets from a Chromatin Immunoprecipitation study on chip (ChIP-on-chip) for the identification of genome-wide DNA binding profiles of transcription factor Cebpa (used for method comparison). Using this assay, we can refine the detection of regions-of-interest by illustrating that regions detected by HAT are more highly enriched for expected motifs in comparison with an alternative detection method (MAT). Subsequently, data from a retroviral insertional mutagenesis screen were used to examine the performance of HAT among different applications of tiling-array datasets. In both studies, detected regions-of-interest have been validated with (q)PCR. CONCLUSIONS: We demonstrate that HAT has increased specificity for analysis of tiling-array data in comparison with the alternative method, and that it accurately detects regions-of-interest in two different applications of tiling-arrays. HAT has several advantages over previous methods: i) as there is no single cut-off level for probe-intensity, HAT can detect regions-of-interest at various thresholds, ii) it can detect regions-of-interest of any size, iii) it is independent of probe-resolution across the genome, and across tiling-array platforms and iv) it employs a single user defined parameter: the significance level. Regions-of-interest are detected by computing the hypergeometric-probability, while controlling the Family Wise Error. Furthermore, the method does not require experimental replicates, common regions-of-interest are indicated, a sequence-of-interest can be examined for every detected region-of-interest, and flanking genes can be reported.


Subject(s)
Chromatin Immunoprecipitation/methods , Genomics/methods , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Software , Genome
17.
Nat Cancer ; 1(11): 1066-1081, 2020 11.
Article in English | MEDLINE | ID: mdl-34079956

ABSTRACT

We report a systematic analysis of the DNA methylation variability in 1,595 samples of normal cell subpopulations and 14 tumor subtypes spanning the entire human B-cell lineage. Differential methylation among tumor entities relates to differences in cellular origin and to de novo epigenetic alterations, which allowed us to build an accurate machine learning-based diagnostic algorithm. We identify extensive patient-specific methylation variability in silenced chromatin associated with the proliferative history of normal and neoplastic B cells. Mitotic activity generally leaves both hyper- and hypomethylation imprints, but some B-cell neoplasms preferentially gain or lose DNA methylation. Subsequently, we construct a DNA methylation-based mitotic clock called epiCMIT, whose lapse magnitude represents a strong independent prognostic variable in B-cell tumors and is associated with particular driver genetic alterations. Our findings reveal DNA methylation as a holistic tracer of B-cell tumor developmental history, with implications in the differential diagnosis and prediction of clinical outcome.


Subject(s)
Epigenome , Neoplasms , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenome/genetics , Gene Expression Regulation, Neoplastic , Humans
18.
Nat Commun ; 10(1): 3615, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31399598

ABSTRACT

Genome-wide association studies have provided evidence for inherited genetic predisposition to chronic lymphocytic leukemia (CLL). To gain insight into the mechanisms underlying CLL risk we analyze chromatin accessibility, active regulatory elements marked by H3K27ac, and DNA methylation at 42 risk loci in up to 486 primary CLLs. We identify that risk loci are significantly enriched for active chromatin in CLL with evidence of being CLL-specific or differentially regulated in normal B-cell development. We then use in situ promoter capture Hi-C, in conjunction with gene expression data to reveal likely target genes of the risk loci. Candidate target genes are enriched for pathways related to B-cell development such as MYC and BCL2 signalling. At 14 loci the analysis highlights 63 variants as the probable functional basis of CLL risk. By integrating genetic and epigenetic information our analysis reveals novel insights into the relationship between inherited predisposition and the regulatory chromatin landscape of CLL.


Subject(s)
Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Epigenomics , Genetic Predisposition to Disease/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , B-Lymphocytes/metabolism , Base Sequence , Chromatin/metabolism , DNA Methylation , Gene Expression Regulation, Leukemic , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors
19.
Biochemistry ; 47(24): 6418-26, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18500823

ABSTRACT

The ecotropic viral integration site 1 ( Evi1) gene encodes a putative transcription regulator, which is aberrantly expressed in acute myeloid leukemias (AML) with chromosomal abnormalities involving the 3q26 locus. Repression and activation of transcriptional control have been reported, but it is currently unclear how Evi1 may evoke these opposing effects. Using a yeast two-hybrid screen, we identified a novel binding partner of Evi1, i.e., methyl binding domain 3b (Mbd3b) protein, a member of the Mi-2/NuRD histone deacetylase complex. Applying in vitro and in vivo assays, we found that Evi1 interacts with Mbd3b but not with other MBD family members Mbd1, -2, and -4 or MeCP2. We show that interaction of Evi1 with Mbd3 requires 40 amino acids that are adjacent and downstream of the methyl binding domain (MBD). We further demonstrate that the first three zinc fingers of Evi1 are needed for Mbd3 interaction. Evi1 acts as a transcriptional repressor when recruited to an active promoter, yet when present in the Mi-2/NuRD complex through Mbd3 interaction, it inhibits the histone deacetylation function of this multiprotein structure. Our data may in part explain how Evi1 could act as a repressor as well as an activator of transcription.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/physiology , Histone Deacetylase Inhibitors , Histones/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogenes/physiology , Repressor Proteins/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology , Acetylation , Adenosine Triphosphatases/physiology , Amino Acid Sequence , Animals , Cell Line , DNA Helicases , DNA-Binding Proteins/metabolism , Down-Regulation/physiology , Histone Deacetylases/physiology , Histones/metabolism , Humans , MDS1 and EVI1 Complex Locus Protein , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Mice , Molecular Sequence Data , Repressor Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL