Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Blood ; 142(1): 90-105, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37146239

ABSTRACT

RNA-binding proteins (RBPs) form a large and diverse class of factors, many members of which are overexpressed in hematologic malignancies. RBPs participate in various processes of messenger RNA (mRNA) metabolism and prevent harmful DNA:RNA hybrids or R-loops. Here, we report that PIWIL4, a germ stem cell-associated RBP belonging to the RNase H-like superfamily, is overexpressed in patients with acute myeloid leukemia (AML) and is essential for leukemic stem cell function and AML growth, but dispensable for healthy human hematopoietic stem cells. In AML cells, PIWIL4 binds to a small number of known piwi-interacting RNA. Instead, it largely interacts with mRNA annotated to protein-coding genic regions and enhancers that are enriched for genes associated with cancer and human myeloid progenitor gene signatures. PIWIL4 depletion in AML cells downregulates the human myeloid progenitor signature and leukemia stem cell (LSC)-associated genes and upregulates DNA damage signaling. We demonstrate that PIWIL4 is an R-loop resolving enzyme that prevents R-loop accumulation on a subset of AML and LSC-associated genes and maintains their expression. It also prevents DNA damage, replication stress, and activation of the ATR pathway in AML cells. PIWIL4 depletion potentiates sensitivity to pharmacological inhibition of the ATR pathway and creates a pharmacologically actionable dependency in AML cells.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/pathology , Hematopoietic Stem Cells/metabolism , Cell Proliferation , Genomics , RNA, Messenger/metabolism , Neoplastic Stem Cells/pathology
2.
Nature ; 588(7836): 157-163, 2020 12.
Article in English | MEDLINE | ID: mdl-33239784

ABSTRACT

Janus kinases (JAKs) mediate responses to cytokines, hormones and growth factors in haematopoietic cells1,2. The JAK gene JAK2 is frequently mutated in the ageing haematopoietic system3,4 and in haematopoietic cancers5. JAK2 mutations constitutively activate downstream signalling and are drivers of myeloproliferative neoplasm (MPN). In clinical use, JAK inhibitors have mixed effects on the overall disease burden of JAK2-mutated clones6,7, prompting us to investigate the mechanism underlying disease persistence. Here, by in-depth phosphoproteome profiling, we identify proteins involved in mRNA processing as targets of mutant JAK2. We found that inactivation of YBX1, a post-translationally modified target of JAK2, sensitizes cells that persist despite treatment with JAK inhibitors to apoptosis and results in RNA mis-splicing, enrichment for retained introns and disruption of the transcriptional control of extracellular signal-regulated kinase (ERK) signalling. In combination with pharmacological JAK inhibition, YBX1 inactivation induces apoptosis in JAK2-dependent mouse and primary human cells, causing regression of the malignant clones in vivo, and inducing molecular remission. This identifies and validates a cell-intrinsic mechanism whereby differential protein phosphorylation causes splicing-dependent alterations of JAK2-ERK signalling and the maintenance of JAK2V617F malignant clones. Therapeutic targeting of YBX1-dependent ERK signalling in combination with JAK2 inhibition could thus eradicate cells harbouring mutations in JAK2.


Subject(s)
Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Neoplasms/genetics , Neoplasms/pathology , Y-Box-Binding Protein 1/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cells, Cultured , Clone Cells/metabolism , Clone Cells/pathology , Female , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Introns/genetics , Janus Kinase 2/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Mice , Mutation , Neoplasm Transplantation , Neoplasms/drug therapy , Phosphoproteins/analysis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome/analysis , Proteomics , RNA Splicing/genetics , Remission Induction , Y-Box-Binding Protein 1/antagonists & inhibitors , Y-Box-Binding Protein 1/chemistry
3.
J Clin Invest ; 126(4): 1438-50, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26927674

ABSTRACT

Meningioma-1 (MN1) overexpression is frequently observed in patients with acute myeloid leukemia (AML) and is predictive of poor prognosis. In murine models, forced expression of MN1 in hematopoietic progenitors induces an aggressive myeloid leukemia that is strictly dependent on a defined gene expression program in the cell of origin, which includes the homeobox genes Hoxa9 and Meis1 as key components. Here, we have shown that this program is controlled by two histone methyltransferases, MLL1 and DOT1L, as deletion of either Mll1 or Dot1l in MN1-expressing cells abrogated the cell of origin-derived gene expression program, including the expression of Hoxa cluster genes. In murine models, genetic inactivation of either Mll1 or Dot1l impaired MN1-mediated leukemogenesis. We determined that HOXA9 and MEIS1 are coexpressed with MN1 in a subset of clinical MN1hi leukemia, and human MN1hi/HOXA9hi leukemias were sensitive to pharmacologic inhibition of DOT1L. Together, these data point to DOT1L as a potential therapeutic target in MN1hi AML. In addition, our findings suggest that epigenetic modulation of the interplay between an oncogenic lesion and its cooperating developmental program has therapeutic potential in AML.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/metabolism , Methyltransferases/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Female , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Methyltransferases/genetics , Mice , Mice, Knockout , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins/genetics , Trans-Activators , Tumor Suppressor Proteins/genetics
4.
J Clin Invest ; 126(3): 997-1011, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26878175

ABSTRACT

Self-renewal is a hallmark of both hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs); therefore, the identification of mechanisms that are required for LSC, but not HSC, function could provide therapeutic opportunities that are more effective and less toxic than current treatments. Here, we employed an in vivo shRNA screen and identified jumonji domain-containing protein JMJD1C as an important driver of MLL-AF9 leukemia. Using a conditional mouse model, we showed that loss of JMJD1C substantially decreased LSC frequency and caused differentiation of MLL-AF9- and homeobox A9-driven (HOXA9-driven) leukemias. We determined that JMJD1C directly interacts with HOXA9 and modulates a HOXA9-controlled gene-expression program. In contrast, loss of JMJD1C led to only minor defects in blood homeostasis and modest effects on HSC self-renewal. Together, these data establish JMJD1C as an important mediator of MLL-AF9- and HOXA9-driven LSC function that is largely dispensable for HSC function.


Subject(s)
Homeodomain Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/physiology , Oncogene Proteins, Fusion/physiology , Animals , Cell Self Renewal , Gene Expression , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice, Knockout , Neoplasm Transplantation , Tumor Cells, Cultured
5.
Nat Med ; 21(4): 335-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25822366

ABSTRACT

Rearrangements of MLL (encoding lysine-specific methyltransferase 2A and officially known as KMT2A; herein referred to as MLL to denote the gene associated with mixed-lineage leukemia) generate MLL fusion proteins that bind DNA and drive leukemogenic gene expression. This gene expression program is dependent on the disruptor of telomeric silencing 1-like histone 3 lysine 79 (H3K79) methyltransferase DOT1L, and small-molecule DOT1L inhibitors show promise as therapeutics for these leukemias. However, the mechanisms underlying this dependency are unclear. We conducted a genome-scale RNAi screen and found that the histone deacetylase SIRT1 is required for the establishment of a heterochromatin-like state around MLL fusion target genes after DOT1L inhibition. DOT1L inhibits chromatin localization of a repressive complex composed of SIRT1 and the H3K9 methyltransferase SUV39H1, thereby maintaining an open chromatin state with elevated H3K9 acetylation and minimal H3K9 methylation at MLL fusion target genes. Furthermore, the combination of SIRT1 activators and DOT1L inhibitors shows enhanced antiproliferative activity against MLL-rearranged leukemia cells. These results indicate that the dynamic interplay between chromatin regulators controlling the activation and repression of gene expression could provide novel opportunities for combination therapy.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Leukemia/metabolism , Methyltransferases/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Sirtuin 1/metabolism , Alleles , Animals , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , Female , Gene Expression Profiling , Gene Rearrangement , Gene Silencing , Genome , Green Fluorescent Proteins/metabolism , Histones/metabolism , Leukemia/genetics , Mice , Mice, Inbred C57BL , Protein Binding , RNA Interference
6.
Cancer Cell ; 26(6): 896-908, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25464900

ABSTRACT

Homeotic (HOX) genes are dysregulated in multiple malignancies, including several AML subtypes. We demonstrate that H3K79 dimethylation (H3K79me2) is converted to monomethylation (H3K79me1) at HOX loci as hematopoietic cells mature, thus coinciding with a decrease in HOX gene expression. We show that H3K79 methyltransferase activity as well as H3K79me1-to-H3K79me2 conversion is regulated by the DOT1L cofactor AF10. AF10 inactivation reverses leukemia-associated epigenetic profiles, precludes abnormal HOXA gene expression, and impairs the transforming ability of MLL-AF9, MLL-AF6, and NUP98-NSD1 fusions-mechanistically distinct HOX-activating oncogenes. Furthermore, NUP98-NSD1-transformed cells are sensitive to small-molecule inhibition of DOT1L. Our findings demonstrate that pharmacological inhibition of the DOT1L/AF10 complex may provide therapeutic benefits in an array of malignancies with abnormal HOXA gene expression.


Subject(s)
Histones/metabolism , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Methyltransferases/metabolism , Transcription Factors/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Bone Marrow Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Methylation , Methyltransferases/antagonists & inhibitors , Mice , Mice, Transgenic , Molecular Sequence Data , Neoplasms, Experimental , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Phenylurea Compounds/pharmacology
7.
Nat Commun ; 5: 3469, 2014 Mar 24.
Article in English | MEDLINE | ID: mdl-24662245

ABSTRACT

Relapsed paediatric acute lymphoblastic leukaemia (ALL) has high rates of treatment failure. Epigenetic regulators have been proposed as modulators of chemoresistance, here, we sequence genes encoding epigenetic regulators in matched diagnosis-remission-relapse ALL samples. We find significant enrichment of mutations in epigenetic regulators at relapse with recurrent somatic mutations in SETD2, CREBBP, MSH6, KDM6A and MLL2, mutations in signalling factors are not enriched. Somatic alterations in SETD2, including frameshift and nonsense mutations, are present at 12% in a large de novo ALL patient cohort. We conclude that the enrichment of mutations in epigenetic regulators at relapse is consistent with a role in mediating therapy resistance.


Subject(s)
Epigenesis, Genetic/genetics , Histone-Lysine N-Methyltransferase/genetics , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Treatment Failure , Base Sequence , DNA Primers/genetics , Exons/genetics , Humans , Molecular Sequence Data , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence , Sequence Analysis, DNA
8.
Blood ; 121(13): 2533-41, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23361907

ABSTRACT

The t(6;11)(q27;q23) is a recurrent chromosomal rearrangement that encodes the MLLAF6 fusion oncoprotein and is observed in patients with diverse hematologic malignancies. The presence of the t(6;11)(q27;q23) has been linked to poor overall survival in patients with AML. In this study, we demonstrate that MLL-AF6 requires continued activity of the histone-methyltransferase DOT1L to maintain expression of the MLL-AF6-driven oncogenic gene-expression program. Using gene-expression analysis and genome-wide chromatin immunoprecipitation studies followed by next generation sequencing, we found that MLL-fusion target genes display markedly high levels of histone 3 at lysine 79 (H3K79) dimethylation in murine MLL-AF6 leukemias as well as in ML2, a human myelomonocytic leukemia cell line bearing the t(6;11)(q27;q23) translocation. Targeted disruption of Dot1l using a conditional knockout mouse model inhibited leukemogenesis mediated by the MLL-AF6 fusion oncogene. Moreover, both murine MLL-AF6-transformed cells as well as the human MLL-AF6-positive ML2 leukemia cell line displayed specific sensitivity to EPZ0004777, a recently described, selective, small-molecule inhibitor of Dot1l. Dot1l inhibition resulted in significantly decreased proliferation, decreased expression of MLL-AF6 target genes, and cell cycle arrest of MLL-AF6-transformed cells. These results indicate that patients bearing the t(6;11)(q27;q23) translocation may benefit from therapeutic agents targeting aberrant H3K79 methylation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/genetics , Kinesins/genetics , Methyltransferases/physiology , Myeloid-Lymphoid Leukemia Protein/genetics , Myosins/genetics , Oncogene Proteins, Fusion/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Lysine/metabolism , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phenylurea Compounds/pharmacology
9.
Cell Stem Cell ; 11(3): 429-39, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22958934

ABSTRACT

The mechanistic target of rapamycin (mTOR) pathway serves as a key sensor of cellular-energetic state and functions to maintain tissue homeostasis. Hyperactivation of the mTOR pathway impairs hematopoietic stem cell (HSC) function and is associated with leukemogenesis. However, the roles of the unique mTOR complexes (mTORCs) in hematopoiesis and leukemogenesis have not been adequately elucidated. We deleted the mTORC1 component, regulatory-associated protein of mTOR (Raptor), in mouse HSCs and its loss causes a nonlethal phenotype characterized by pancytopenia, splenomegaly, and the accumulation of monocytoid cells. Furthermore, Raptor is required for HSC regeneration, and plays largely nonredundant roles with rapamycin-insensitive companion of mTOR (Rictor) in these processes. Ablation of Raptor also significantly extends survival of mice in models of leukemogenesis evoked by Pten deficiency. These data delineate critical roles for mTORC1 in hematopoietic function and leukemogenesis and inform clinical strategies based on chronic mTORC1 inhibition.


Subject(s)
Cell Transformation, Neoplastic/pathology , Hematopoiesis , Leukemia/enzymology , Leukemia/pathology , Multiprotein Complexes/metabolism , PTEN Phosphohydrolase/deficiency , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins , Cell Cycle/genetics , Cell Differentiation , Cell Lineage , Disease Models, Animal , Gene Expression Regulation, Leukemic , Hematopoiesis/genetics , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Homeostasis , Mechanistic Target of Rapamycin Complex 1 , Mice , PTEN Phosphohydrolase/metabolism , Regulatory-Associated Protein of mTOR , Survival Analysis
10.
PLoS One ; 7(2): e31690, 2012.
Article in English | MEDLINE | ID: mdl-22393367

ABSTRACT

Increasing use of high throughput genomic scale assays requires effective visualization and analysis techniques to facilitate data interpretation. Moreover, existing tools often require programming skills, which discourages bench scientists from examining their own data. We have created iCanPlot, a compelling platform for visual data exploration based on the latest technologies. Using the recently adopted HTML5 Canvas element, we have developed a highly interactive tool to visualize tabular data and identify interesting patterns in an intuitive fashion without the need of any specialized computing skills. A module for geneset overlap analysis has been implemented on the Google App Engine platform: when the user selects a region of interest in the plot, the genes in the region are analyzed on the fly. The visualization and analysis are amalgamated for a seamless experience. Further, users can easily upload their data for analysis--which also makes it simple to share the analysis with collaborators. We illustrate the power of iCanPlot by showing an example of how it can be used to interpret histone modifications in the context of gene expression.


Subject(s)
Computational Biology/methods , Genomics , Algorithms , Computer Graphics , Computers , Database Management Systems , Databases, Genetic , Gene Expression , Gene Expression Profiling , Genome , Hematopoietic Stem Cells/cytology , Histones/metabolism , Humans , Internet , Software , User-Computer Interface
11.
Nature ; 483(7391): 598-602, 2012 Mar 04.
Article in English | MEDLINE | ID: mdl-22388813

ABSTRACT

Generation of induced pluripotent stem cells (iPSCs) by somatic cell reprogramming involves global epigenetic remodelling. Whereas several proteins are known to regulate chromatin marks associated with the distinct epigenetic states of cells before and after reprogramming, the role of specific chromatin-modifying enzymes in reprogramming remains to be determined. To address how chromatin-modifying proteins influence reprogramming, we used short hairpin RNAs (shRNAs) to target genes in DNA and histone methylation pathways, and identified positive and negative modulators of iPSC generation. Whereas inhibition of the core components of the polycomb repressive complex 1 and 2, including the histone 3 lysine 27 methyltransferase EZH2, reduced reprogramming efficiency, suppression of SUV39H1, YY1 and DOT1L enhanced reprogramming. Specifically, inhibition of the H3K79 histone methyltransferase DOT1L by shRNA or a small molecule accelerated reprogramming, significantly increased the yield of iPSC colonies, and substituted for KLF4 and c-Myc (also known as MYC). Inhibition of DOT1L early in the reprogramming process is associated with a marked increase in two alternative factors, NANOG and LIN28, which play essential functional roles in the enhancement of reprogramming. Genome-wide analysis of H3K79me2 distribution revealed that fibroblast-specific genes associated with the epithelial to mesenchymal transition lose H3K79me2 in the initial phases of reprogramming. DOT1L inhibition facilitates the loss of this mark from genes that are fated to be repressed in the pluripotent state. These findings implicate specific chromatin-modifying enzymes as barriers to or facilitators of reprogramming, and demonstrate how modulation of chromatin-modifying enzymes can be exploited to more efficiently generate iPSCs with fewer exogenous transcription factors.


Subject(s)
Cellular Reprogramming , Chromatin/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cellular Reprogramming/genetics , Chromatin/genetics , DNA Methylation/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Fibroblasts/cytology , Fibroblasts/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Methylation , Methyltransferases/antagonists & inhibitors , Methyltransferases/biosynthesis , Methyltransferases/genetics , Methyltransferases/metabolism , Nanog Homeobox Protein , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering , RNA-Binding Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , YY1 Transcription Factor/antagonists & inhibitors , YY1 Transcription Factor/metabolism
12.
Proc Natl Acad Sci U S A ; 109(13): 5028-33, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22396593

ABSTRACT

A growing body of data suggests the importance of epigenetic mechanisms in cancer. Polycomb repressive complex 2 (PRC2) has been implicated in self-renewal and cancer progression, and its components are overexpressed in many cancers. However, its role in cancer development and progression remains unclear. We used conditional alleles for the PRC2 components enhancer of zeste 2 (Ezh2) and embryonic ectoderm development (Eed) to characterize the role of PRC2 function in leukemia development and progression. Compared with wild-type leukemia, Ezh2-null MLL-AF9-mediated acute myeloid leukemia (AML) failed to accelerate upon secondary transplantation. However, Ezh2-null leukemias maintained self-renewal up to the third round of transplantation, indicating that Ezh2 is not strictly required for MLL-AF9 AML, but plays a role in leukemia progression. Genome-wide analyses of PRC2-mediated trimethylation of histone 3 demonstrated locus-specific persistence of H3K27me3 despite inactivation of Ezh2, suggesting partial compensation by Ezh1. In contrast, inactivation of the essential PRC2 gene, Eed, led to complete ablation of PRC2 function, which was incompatible with leukemia growth. Gene expression array analyses indicated more profound gene expression changes in Eed-null compared with Ezh2-null leukemic cells, including down-regulation of Myc target genes and up-regulation of PRC2 targets. Manipulating PRC2 function may be of therapeutic benefit in AML.


Subject(s)
Leukemia/pathology , Oncogene Proteins, Fusion/metabolism , Repressor Proteins/metabolism , Animals , Cell Proliferation , Chromatin Immunoprecipitation , Cytoprotection , Disease Progression , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Gene Silencing , Genes, Neoplasm/genetics , Genetic Loci/genetics , Genome/genetics , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Leukemia/genetics , Methylation , Mice , Mice, Inbred C57BL , Phenotype , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proto-Oncogene Proteins c-myc/metabolism
13.
Genes Dev ; 26(4): 344-9, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22345515

ABSTRACT

Epigenetic mechanisms regulating leukemia stem cells (LSCs) are an attractive target for therapy of blood cancers. Here, we report that conditional knockout of the DNA methyltransferase Dnmt1 blocked development of leukemia, and haploinsufficiency of Dnmt1 was sufficient to delay progression of leukemogenesis and impair LSC self-renewal without altering normal hematopoiesis. Haploinsufficiency of Dnmt1 resulted in tumor suppressor gene derepression associated with reduced DNA methylation and bivalent chromatin marks. These results suggest that LSCs depend on not only active expression of leukemogenic programs, but also DNA methylation-mediated silencing of bivalent domains to enforce transcriptional repression.


Subject(s)
Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Neoplastic , Haploinsufficiency , Leukemia/enzymology , Neoplastic Stem Cells/enzymology , Animals , DNA Methylation , Gene Knockout Techniques , Kaplan-Meier Estimate , Leukemia/pathology , Mice , Neoplastic Stem Cells/cytology , Tumor Suppressor Proteins/genetics
14.
BMC Bioinformatics ; 12: 452, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-22103807

ABSTRACT

BACKGROUND: Genome-wide experiments are routinely conducted to measure gene expression, DNA-protein interactions and epigenetic status. Structured metadata for these experiments is imperative for a complete understanding of experimental conditions, to enable consistent data processing and to allow retrieval, comparison, and integration of experimental results. Even though several repositories have been developed for genomics data, only a few provide annotation of samples and assays using controlled vocabularies. Moreover, many of them are tailored for a single type of technology or measurement and do not support the integration of multiple data types. RESULTS: We have developed eXframe - a reusable web-based framework for genomics experiments that provides 1) the ability to publish structured data compliant with accepted standards 2) support for multiple data types including microarrays and next generation sequencing 3) query, analysis and visualization integration tools (enabled by consistent processing of the raw data and annotation of samples) and is available as open-source software. We present two case studies where this software is currently being used to build repositories of genomics experiments - one contains data from hematopoietic stem cells and another from Parkinson's disease patients. CONCLUSION: The web-based framework eXframe offers structured annotation of experiments as well as uniform processing and storage of molecular data from microarray and next generation sequencing platforms. The framework allows users to query and integrate information across species, technologies, measurement types and experimental conditions. Our framework is reusable and freely modifiable - other groups or institutions can deploy their own custom web-based repositories based on this software. It is interoperable with the most important data formats in this domain. We hope that other groups will not only use eXframe, but also contribute their own useful modifications.


Subject(s)
Genomics/methods , Hematopoietic Stem Cells/metabolism , Parkinson Disease/genetics , Software , Aged , Aged, 80 and over , Female , Gene Expression Profiling/methods , Hematopoietic Stem Cells/cytology , Humans , Internet , Middle Aged , Parkinson Disease/pathology , User-Computer Interface
15.
Cancer Cell ; 20(1): 66-78, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21741597

ABSTRACT

The histone 3 lysine 79 (H3K79) methyltransferase Dot1l has been implicated in the development of leukemias bearing translocations of the Mixed Lineage Leukemia (MLL) gene. We identified the MLL-fusion targets in an MLL-AF9 leukemia model, and conducted epigenetic profiling for H3K79me2, H3K4me3, H3K27me3, and H3K36me3 in hematopoietic progenitor and leukemia stem cells (LSCs). We found abnormal profiles only for H3K79me2 on MLL-AF9 fusion target loci in LSCs. Inactivation of Dot1l led to downregulation of direct MLL-AF9 targets and an MLL translocation-associated gene expression signature, whereas global gene expression remained largely unaffected. Suppression of MLL translocation-associated gene expression corresponded with dependence of MLL-AF9 leukemia on Dot1l in vivo. These data point to DOT1L as a potential therapeutic target in MLL-rearranged leukemia.


Subject(s)
Gene Rearrangement/genetics , Histones/metabolism , Lysine/metabolism , Methyltransferases/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Genetic Loci/genetics , Hematopoiesis , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/metabolism , Humans , Methylation , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Processing, Post-Translational
16.
Science ; 327(5973): 1650-3, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20339075

ABSTRACT

Leukemia stem cells (LSCs) are capable of limitless self-renewal and are responsible for the maintenance of leukemia. Because selective eradication of LSCs could offer substantial therapeutic benefit, there is interest in identifying the signaling pathways that control their development. We studied LSCs in mouse models of acute myelogenous leukemia (AML) induced either by coexpression of the Hoxa9 and Meis1a oncogenes or by the fusion oncoprotein MLL-AF9. We show that the Wnt/beta-catenin signaling pathway is required for self-renewal of LSCs that are derived from either hematopoietic stem cells (HSC) or more differentiated granulocyte-macrophage progenitors (GMP). Because the Wnt/beta-catenin pathway is normally active in HSCs but not in GMP, these results suggest that reactivation of beta-catenin signaling is required for the transformation of progenitor cells by certain oncogenes. beta-catenin is not absolutely required for self-renewal of adult HSCs; thus, targeting the Wnt/beta-catenin pathway may represent a new therapeutic opportunity in AML.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Genes, Homeobox , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/genetics , Indomethacin/pharmacology , Mice , Mice, Inbred C57BL , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Transduction, Genetic
17.
Clin Cancer Res ; 16(7): 2122-30, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20332322

ABSTRACT

PURPOSE: Patients with mixed lineage leukemia (MLL)-rearranged B-lymphoblastic leukemias (B-ALL) have an unfavorable prognosis and require intensified treatment. Multiple MLL fusion partners have been identified, complicating the diagnostic evaluation of MLL rearrangements. We analyzed molecular markers of MLL rearrangement for use in rapid diagnostic assays and found the immunomodulatory protein, Galectin-1 (Gal-1), to be selectively expressed in MLL-rearranged B-ALL. EXPERIMENTAL DESIGN: Transcriptional profiling of ALL subtypes revealed selective overexpression of Gal-1 in MLL-rearranged ALLs. For this reason, we analyzed Gal-1 protein expression in MLL-germline and MLL-rearranged adult and infant pediatric B-ALLs and cell lines by immunoblotting, immunohistochemistry, and intracellular flow cytometry of viable tumor cell suspensions. Because deregulated gene expression in MLL-rearranged leukemias may be related to the altered histone methyltransferase activity of the MLL fusion protein complex, we also analyzed histone H3 lysine 79 (H3K79) dimethylation in the LGALS1 promoter region using chromatin immunoprecipitation. RESULTS: Gal-1 transcripts were significantly more abundant in MLL-rearranged B-ALLs. All 32 primary MLL-rearranged B-ALLs exhibited abundant Gal-1 immunostaining, regardless of the translocation partner, whereas only 2 of 81 germline-MLL B-ALLs expressed Gal-1. In addition, Gal-1 was selectively detected in newly diagnosed MLL-rearranged B-ALLs by intracellular flow cytometry. The LGALS1 promoter H3K79 was significantly hypermethylated in MLL-rearranged B-ALLs compared with MLL-germline B-ALLs and normal pre-B cells. CONCLUSION: In B-ALL, Gal-1 is a highly sensitive and specific biomarker of MLL rearrangement that is likely induced by a MLL-dependent epigenetic modification.


Subject(s)
Galectin 1/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adult , Cell Line, Tumor , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 4 , Chromosomes, Human, Pair 9 , Galectin 1/metabolism , Gene Expression Regulation, Leukemic , Gene Rearrangement/genetics , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Immunologic Factors/genetics , Immunologic Factors/metabolism , Infant , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Translocation, Genetic/physiology
18.
Cancer Cell ; 14(5): 355-68, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18977325

ABSTRACT

We created a mouse model wherein conditional expression of an Mll-AF4 fusion oncogene induces B precursor acute lymphoblastic (ALL) or acute myeloid leukemias (AML). Gene expression profile analysis of the ALL cells demonstrated significant overlap with human MLL-rearranged ALL. ChIP-chip analysis demonstrated histone H3 lysine 79 (H3K79) methylation profiles that correlated with Mll-AF4-associated gene expression profiles in murine ALLs and in human MLL-rearranged leukemias. Human MLL-rearranged ALLs could be distinguished from other ALLs by their H3K79 profiles, and suppression of the H3K79 methyltransferase DOT1L inhibited expression of critical MLL-AF4 target genes. We thus demonstrate that ectopic H3K79 methylation is a distinguishing feature of murine and human MLL-AF4 ALLs and is important for maintenance of MLL-AF4-driven gene expression.


Subject(s)
Gene Expression Regulation, Leukemic , Histones/metabolism , Leukemia, Myeloid, Acute/genetics , Methylation , Myeloid-Lymphoid Leukemia Protein/physiology , Oncogene Proteins, Fusion/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation , Female , Flow Cytometry , Gene Expression Profiling , Gene Rearrangement , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Histones/chemistry , Histones/genetics , Homeodomain Proteins/metabolism , Humans , Immunophenotyping , Integrases/metabolism , Leukemia, Myeloid, Acute/pathology , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Male , Methyltransferases/antagonists & inhibitors , Methyltransferases/physiology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Principal Component Analysis , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , RNA, Small Interfering/pharmacology , Transcription, Genetic
19.
BMC Genomics ; 9: 88, 2008 Feb 23.
Article in English | MEDLINE | ID: mdl-18294391

ABSTRACT

BACKGROUND: The master regulator p53 tumor-suppressor protein through coordination of several downstream target genes and upstream transcription factors controls many pathways important for tumor suppression. While it has been reported that some of the p53's functions are microRNA-mediated, it is not known as to how many other microRNAs might contribute to the p53-mediated tumorigenesis. RESULTS: Here, we use bioinformatics-based integrative approach to identify and prioritize putative p53-regulated miRNAs, and unravel the miRNA-based microregulation of the p53 master regulatory network. Specifically, we identify putative microRNA regulators of a) transcription factors that are upstream or downstream to p53 and b) p53 interactants. The putative p53-miRs and their targets are prioritized using current knowledge of cancer biology and literature-reported cancer-miRNAs. CONCLUSION: Our predicted p53-miRNA-gene networks strongly suggest that coordinated transcriptional and p53-miR mediated networks could be integral to tumorigenesis and the underlying processes and pathways.


Subject(s)
Computational Biology/methods , MicroRNAs/genetics , Tumor Suppressor Protein p53/genetics , Gene Expression Regulation , Genome, Human , Genomics/methods , Humans , MicroRNAs/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
20.
Pac Symp Biocomput ; : 37-48, 2008.
Article in English | MEDLINE | ID: mdl-18229675

ABSTRACT

Identifying syntenic regions and quantifying evolutionary relatedness between genomes by interrogating genome rearrangement events is one of the central goals of comparative genomics. However, identification of synteny blocks and the resulting assessment of genome rearrangements are dependent on the choice of conserved markers, the definition of conserved segments, and the choice of various parameters that are used to construct such segments for two genomes. In this work, we performed an extended sensitivity analysis of synteny block generation using alternative sets of markers in multiple genomes. A simple approach to synteny block aggregation is used, which depends on two principle parameters: the maximum gap (max gap) between adjacent blocks to be merged, and the minimum length (min len) of synteny blocks. In particular, the dependence on the choice of conserved markers and max gap/min len aggregation parameters is assessed for two important quantities that can be used to characterize evolutionary relationships between genomes, namely the reversal distance and breakpoint reuse. We observe that the number of synteny blocks depends on both parameters, while the reversal distance depends mostly on min len. On the other hand, we observe that relative reversal distances between mammalian genomes, which are defined as ratios of distances between different pairs of genomes, are nearly constant for both parameters. Similarly, the breakpoint reuse rate was found to be almost constant for different data sets and a wide range of parameters. Breakpoint reuse is also strongly correlated with evolutionary distances, increasing for pairs of more divergent genomes. Finally, we demonstrate that the role of parameters may be further reduced by using a multi-way analysis that involves markers conserved in multiple genomes, which opens a way to guide the choice of a correct parameterization.


Subject(s)
Genomics/statistics & numerical data , Algorithms , Animals , Chromosome Breakage , Computational Biology , Databases, Genetic , Evolution, Molecular , Gene Rearrangement , Humans , Mammals/genetics , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...