Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
iScience ; 27(7): 110386, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39055911

ABSTRACT

[This corrects the article DOI: 10.1016/j.isci.2023.106444.].

2.
iScience ; 26(4): 106444, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37096048

ABSTRACT

P53 is a master transcriptional regulator and effector of the DNA damage response (DDR) that localizes to DNA damage sites, in part, via an interaction with PARP1. However, the mechanisms that regulate p53 abundance and activity at PARP1-decorated DNA damage sites remain undefined. The PARP9 (BAL1) macrodomain-containing protein and its partner DTX3L (BBAP) E3 ligase are rapidly recruited to PARP1-PARylated DNA damage sites. During an initial DDR, we found that DTX3L rapidly colocalized with p53, polyubiquitylated its lysine-rich C-terminal domain, and targeted p53 for proteasomal degradation. DTX3L knockout significantly increased and prolonged p53 retention at PARP-decorated DNA damage sites. These findings reveal a non-redundant, PARP- and PARylation-dependent role for DTX3L in the spatiotemporal regulation of p53 during an initial DDR. Our studies suggest that targeted inhibition of DTX3L may augment the efficacy of certain DNA-damaging agents by increasing p53 abundance and activity.

3.
J Med Chem ; 63(23): 14522-14529, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33034194

ABSTRACT

Histone demethylase LSDl (KDMlA) belongs to the flavin adenine dinucleotide (FAD) dependent family of monoamine oxidases and is vital in regulation of mammalian biology. Dysregulation and overexpression of LSD1 are hallmarks of a number of human diseases, particularly cancers that are characterized as morphologically poorly differentiated. As such, inhibitors of LSD1 have potential to be beneficial as a cancer therapy. The most clinically advanced inhibitors of LSDl are covalent inhibitors derived from tranylcypromine (TCP). Herein, we report the discovery of a novel series of reversible and selective LSDl inhibitors. Exploration of structure-activity relationships (SARs) and optimization of ADME properties resulted in the identification of clinical candidate CC-90011. CC-90011 exhibits potent on-target induction of cellular differentiation in acute myeloid leukemia (AML) and small cell lung cancer (SCLC) cell lines, and antitumor efficacy in patient-derived xenograft (PDX) SCLC models. CC-90011 is currently in phase 2 trials in patients with first line, extensive stage SCLC (ClinicalTrials.gov identifier: NCT03850067).


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Organic Chemicals/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Organic Chemicals/chemistry , Structure-Activity Relationship
4.
Nat Med ; 26(9): 1468-1479, 2020 09.
Article in English | MEDLINE | ID: mdl-32778827

ABSTRACT

PD-1 blockade is highly effective in classical Hodgkin lymphomas (cHLs), which exhibit frequent copy-number gains of CD274 (PD-L1) and PDC1LG2 (PD-L2) on chromosome 9p24.1. However, in this largely MHC-class-I-negative tumor, the mechanism of action of anti-PD-1 therapy remains undefined. We utilized the complementary approaches of T cell receptor (TCR) sequencing and cytometry by time-of-flight analysis to obtain a peripheral immune signature of responsiveness to PD-1 blockade in 56 patients treated in the CheckMate 205 phase II clinical trial (NCT02181738). Anti-PD-1 therapy was most effective in patients with a diverse baseline TCR repertoire and an associated expansion of singleton clones during treatment. CD4+, but not CD8+, TCR diversity significantly increased during therapy, most strikingly in patients who had achieved complete responses. Additionally, patients who responded to therapy had an increased abundance of activated natural killer cells and a newly identified CD3-CD68+CD4+GrB+ subset. These studies highlight the roles of recently expanded, clonally diverse CD4+ T cells and innate effectors in the efficacy of PD-1 blockade in cHL.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hodgkin Disease/drug therapy , Killer Cells, Natural/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antineoplastic Agents, Immunological/therapeutic use , CD4-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/classification , Humans , Lymphocyte Activation/immunology , Nivolumab/therapeutic use , Receptors, Antigen, T-Cell/genetics , Tumor Microenvironment/immunology
5.
Haematologica ; 105(5): 1361-1368, 2020 05.
Article in English | MEDLINE | ID: mdl-31471373

ABSTRACT

B-cell receptor (BCR) signaling pathway components represent promising treatment targets in multiple B-cell malignancies including diffuse large B-cell lymphoma (DLBCL). In in vitro and in vivo model systems, a subset of DLBCLs depend upon BCR survival signals and respond to proximal BCR/phosphoinositide 3 kinase (PI3K) blockade. However, single-agent BCR pathway inhibitors have had more limited activity in patients with DLBCL, underscoring the need for indicators of sensitivity to BCR blockade and insights into potential resistance mechanisms. Here, we report highly significant transcriptional upregulation of C-X-C chemokine receptor 4 (CXCR4) in BCR-dependent DLBCL cell lines and primary tumors following chemical spleen tyrosine kinase (SYK) inhibition, molecular SYK depletion or chemical PI3K blockade. SYK or PI3K inhibition also selectively upregulated cell surface CXCR4 protein expression in BCR-dependent DLBCLs. CXCR4 expression was directly modulated by fork-head box O1 via the PI3K/protein kinase B/forkhead box O1 signaling axis. Following chemical SYK inhibition, all BCR-dependent DLBCLs exhibited significantly increased stromal cell-derived factor-1α (SDF-1α) induced chemotaxis, consistent with the role of CXCR4 signaling in B-cell migration. Select PI3K isoform inhibitors also augmented SDF-1α induced chemotaxis. These data define CXCR4 upregulation as an indicator of sensitivity to BCR/PI3K blockade and identify CXCR4 signaling as a potential resistance mechanism in BCR-dependent DLBCLs.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Phosphatidylinositol 3-Kinases , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Phosphatidylinositol 3-Kinase , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Up-Regulation
6.
Blood Adv ; 3(23): 4065-4080, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31816062

ABSTRACT

Classical Hodgkin lymphoma (cHL) is composed of rare malignant Hodgkin Reed-Sternberg (HRS) cells within an extensive, but ineffective, inflammatory/immune cell infiltrate. HRS cells exhibit near-universal somatic copy gains of chromosome 9p/9p24.1, which increase expression of the programmed cell death protein 1 (PD-1) ligands. To define genetic mechanisms of response and resistance to PD-1 blockade and identify complementary treatment targets, we performed whole-exome sequencing of flow cytometry-sorted HRS cells from 23 excisional biopsies of newly diagnosed cHLs, including 8 Epstein-Barr virus-positive (EBV+) tumors. We identified significantly mutated cancer candidate genes (CCGs) as well as somatic copy number alterations and structural variations and characterized their contribution to disease-defining immune evasion mechanisms and nuclear factor κB (NF-κB), JAK/STAT, and PI3K signaling pathways. EBV- cHLs had a higher prevalence of genetic alterations in the NF-κB and major histocompatibility complex class I antigen presentation pathways. In this young cHL cohort (median age, 26 years), we identified a predominant mutational signature of spontaneous deamination of cytosine- phosphate-guanines ("Aging"), in addition to apolipoprotein B mRNA editing catalytic polypeptide-like, activation-induced cytidine deaminase, and microsatellite instability (MSI)-associated hypermutation. In particular, the mutational burden in EBV- cHLs was among the highest reported, similar to that of carcinogen-induced tumors. Together, the overall high mutational burden, MSI-associated hypermutation, and newly identified genetic alterations represent additional potential bases for the efficacy of PD-1 blockade in cHL. Of note, recurrent cHL alterations, including B2M, TNFAIP3, STAT6, GNA13, and XPO1 mutations and 2p/2p15, 6p21.32, 6q23.3, and 9p/9p24.1 copy number alterations, were also identified in >20% of primary mediastinal B-cell lymphomas, highlighting shared pathogenetic mechanisms in these diseases.


Subject(s)
Genomics/methods , Reed-Sternberg Cells/immunology , Adult , Humans , Immune Evasion
7.
Nat Commun ; 10(1): 5622, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819055

ABSTRACT

A heritable polymorphism within regulatory sequences of the LMO1 gene is associated with its elevated expression and increased susceptibility to develop neuroblastoma, but the oncogenic pathways downstream of the LMO1 transcriptional co-regulatory protein are unknown. Our ChIP-seq and RNA-seq analyses reveal that a key gene directly regulated by LMO1 and MYCN is ASCL1, which encodes a basic helix-loop-helix transcription factor. Regulatory elements controlling ASCL1 expression are bound by LMO1, MYCN and the transcription factors GATA3, HAND2, PHOX2B, TBX2 and ISL1-all members of the adrenergic (ADRN) neuroblastoma core regulatory circuitry (CRC). ASCL1 is required for neuroblastoma cell growth and arrest of differentiation. ASCL1 and LMO1 directly regulate the expression of CRC genes, indicating that ASCL1 is a member and LMO1 is a coregulator of the ADRN neuroblastoma CRC.


Subject(s)
Adrenergic Agents/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , LIM Domain Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Transcription Factors/metabolism , Cell Differentiation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/genetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Survival Analysis
8.
Blood ; 133(1): 70-80, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30322870

ABSTRACT

Inhibition of the B-cell receptor (BCR) signaling pathway is a promising treatment strategy in multiple B-cell malignancies. However, the role of BCR blockade in diffuse large B-cell lymphoma (DLBCL) remains undefined. We recently characterized primary DLBCL subsets with distinct genetic bases for perturbed BCR/phosphoinositide 3-kinase (PI3K) signaling and dysregulated B-cell lymphoma 2 (BCL-2) expression. Herein, we explore the activity of PI3K inhibitors and BCL-2 blockade in a panel of functionally and genetically characterized DLBCL cell line models. A PI3K inhibitor with predominant α/δ activity, copanlisib, exhibited the highest cytotoxicity in all BCR-dependent DLBCLs. The proapoptotic effect of copanlisib was associated with DLBCL subtype-specific dysregulated expression of BCL-2 family members including harakiri (HRK) and its antiapoptotic partner BCL extra large (BCL-xL), BCL2 related protein A1, myeloid cell leukemia 1 (MCL-1), and BCL2 interacting mediator of cell death. Using functional BH3 profiling, we found that the cytotoxic activity of copanlisib was primarily mediated through BCL-xL and MCL-1-dependent mechanisms that might complement BCL-2 blockade. For these reasons, we evaluated single-agent activity of venetoclax in the DLBCLs and identified a subset with limited sensitivity to BCL-2 blockade despite having genetic bases of BCL-2 dysregulation. As these were largely BCR-dependent DLBCLs, we hypothesized that combined inhibition of PI3Kα/δ and BCL-2 would perturb BCR-dependent and BCL-2-mediated survival pathways. Indeed, we observed synergistic activity of copanlisib/venetoclax in BCR-dependent DLBCLs with genetic bases for BCL-2 dysregulation in vitro and confirmed these findings in a xenograft model. These results provide preclinical evidence for the rational combination of PI3Kα/δ and BCL-2 blockade in genetically defined DLBCLs.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Synergism , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrimidines/pharmacology , Quinazolines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Female , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Genes Dev ; 31(23-24): 2343-2360, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29326336

ABSTRACT

The oncogenic transcription factor TAL1/SCL induces an aberrant transcriptional program in T-cell acute lymphoblastic leukemia (T-ALL) cells. However, the critical factors that are directly activated by TAL1 and contribute to T-ALL pathogenesis are largely unknown. Here, we identified AT-rich interactive domain 5B (ARID5B) as a collaborating oncogenic factor involved in the transcriptional program in T-ALL. ARID5B expression is down-regulated at the double-negative 2-4 stages in normal thymocytes, while it is induced by the TAL1 complex in human T-ALL cells. The enhancer located 135 kb upstream of the ARID5B gene locus is activated under a superenhancer in T-ALL cells but not in normal T cells. Notably, ARID5B-bound regions are associated predominantly with active transcription. ARID5B and TAL1 frequently co-occupy target genes and coordinately control their expression. ARID5B positively regulates the expression of TAL1 and its regulatory partners. ARID5B also activates the expression of the oncogene MYC Importantly, ARID5B is required for the survival and growth of T-ALL cells, and forced expression of ARID5B in immature thymocytes results in thymus retention, differentiation arrest, radioresistance, and tumor formation in zebrafish. Our results indicate that ARID5B reinforces the oncogenic transcriptional program by positively regulating the TAL1-induced regulatory circuit and MYC in T-ALL, thereby contributing to T-cell leukemogenesis.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, myc/genetics , HEK293 Cells , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Protein Binding , Protein Domains/genetics , Thymocytes/metabolism , Thymus Gland/growth & development , Transcription Factors/genetics , Transcriptional Activation/genetics , Zebrafish
10.
Genes Dev ; 29(5): 483-8, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25737277

ABSTRACT

We performed a genome-scale shRNA screen for modulators of B-cell leukemia progression in vivo. Results from this work revealed dramatic distinctions between the relative effects of shRNAs on the growth of tumor cells in culture versus in their native microenvironment. Specifically, we identified many "context-specific" regulators of leukemia development. These included the gene encoding the zinc finger protein Phf6. While inactivating mutations in PHF6 are commonly observed in human myeloid and T-cell malignancies, we found that Phf6 suppression in B-cell malignancies impairs tumor progression. Thus, Phf6 is a "lineage-specific" cancer gene that plays opposing roles in developmentally distinct hematopoietic malignancies.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Leukemia/genetics , Cell Lineage , Cell Proliferation/genetics , Genome, Human/genetics , Humans , Leukemia/physiopathology , Mutation/genetics , RNA, Small Interfering/genetics , Repressor Proteins
11.
J Exp Med ; 210(8): 1545-57, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23857984

ABSTRACT

The oncogenic transcription factor TAL1/SCL is aberrantly expressed in 60% of cases of human T cell acute lymphoblastic leukemia (T-ALL) and initiates T-ALL in mouse models. By performing global microRNA (miRNA) expression profiling after depletion of TAL1, together with genome-wide analysis of TAL1 occupancy by chromatin immunoprecipitation coupled to massively parallel DNA sequencing, we identified the miRNA genes directly controlled by TAL1 and its regulatory partners HEB, E2A, LMO1/2, GATA3, and RUNX1. The most dynamically regulated miRNA was miR-223, which is bound at its promoter and up-regulated by the TAL1 complex. miR-223 expression mirrors TAL1 levels during thymic development, with high expression in early thymocytes and marked down-regulation after the double-negative-2 stage of maturation. We demonstrate that aberrant miR-223 up-regulation by TAL1 is important for optimal growth of TAL1-positive T-ALL cells and that sustained expression of miR-223 partially rescues T-ALL cells after TAL1 knockdown. Overexpression of miR-223 also leads to marked down-regulation of FBXW7 protein expression, whereas knockdown of TAL1 leads to up-regulation of FBXW7 protein levels, with a marked reduction of its substrates MYC, MYB, NOTCH1, and CYCLIN E. We conclude that TAL1-mediated up-regulation of miR-223 promotes the malignant phenotype in T-ALL through repression of the FBXW7 tumor suppressor.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins/metabolism , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Humans , Mice , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Thymus Gland/metabolism , Ubiquitin-Protein Ligases/genetics
12.
Cancer Cell ; 22(2): 209-21, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22897851

ABSTRACT

The oncogenic transcription factor TAL1/SCL is aberrantly expressed in over 40% of cases of human T cell acute lymphoblastic leukemia (T-ALL), emphasizing its importance in the molecular pathogenesis of T-ALL. Here we identify the core transcriptional regulatory circuit controlled by TAL1 and its regulatory partners HEB, E2A, LMO1/2, GATA3, and RUNX1. We show that TAL1 forms a positive interconnected autoregulatory loop with GATA3 and RUNX1 and that the TAL1 complex directly activates the MYB oncogene, forming a positive feed-forward regulatory loop that reinforces and stabilizes the TAL1-regulated oncogenic program. One of the critical downstream targets in this circuitry is the TRIB2 gene, which is oppositely regulated by TAL1 and E2A/HEB and is essential for the survival of T-ALL cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Regulatory Networks/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Leukemic , Genes, Neoplasm/genetics , Genome, Human/genetics , Homeostasis/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/classification , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myb/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
13.
Cell ; 147(3): 577-89, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036566

ABSTRACT

BMP and Wnt signaling pathways control essential cellular responses through activation of the transcription factors SMAD (BMP) and TCF (Wnt). Here, we show that regeneration of hematopoietic lineages following acute injury depends on the activation of each of these signaling pathways to induce expression of key blood genes. Both SMAD1 and TCF7L2 co-occupy sites with master regulators adjacent to hematopoietic genes. In addition, both SMAD1 and TCF7L2 follow the binding of the predominant lineage regulator during differentiation from multipotent hematopoietic progenitor cells to erythroid cells. Furthermore, induction of the myeloid lineage regulator C/EBPα in erythroid cells shifts binding of SMAD1 to sites newly occupied by C/EBPα, whereas expression of the erythroid regulator GATA1 directs SMAD1 loss on nonerythroid targets. We conclude that the regenerative response mediated by BMP and Wnt signaling pathways is coupled with the lineage master regulators to control the gene programs defining cellular identity.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Hematopoiesis , Signal Transduction , Wnt Signaling Pathway , Animals , DNA-Binding Proteins/metabolism , Humans , Regeneration , Smad1 Protein/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Zebrafish
14.
Cell Stem Cell ; 9(3): 219-32, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21885018

ABSTRACT

Using mouse skin, where bountiful reservoirs of synchronized hair follicle stem cells (HF-SCs) fuel cycles of regeneration, we explore how adult SCs remodel chromatin in response to activating cues. By profiling global mRNA and chromatin changes in quiescent and activated HF-SCs and their committed, transit-amplifying (TA) progeny, we show that polycomb-group (PcG)-mediated H3K27-trimethylation features prominently in HF-lineage progression by mechanisms distinct from embryonic-SCs. In HF-SCs, PcG represses nonskin lineages and HF differentiation. In TA progeny, nonskin regulators remain PcG-repressed, HF-SC regulators acquire H3K27me3-marks, and HF-lineage regulators lose them. Interestingly, genes poised in embryonic stem cells, active in HF-SCs, and PcG-repressed in TA progeny encode not only key transcription factors, but also signaling regulators. We document their importance in balancing HF-SC quiescence, underscoring the power of chromatin mapping in dissecting SC behavior. Our findings explain how HF-SCs cycle through quiescent and activated states without losing stemness and define roles for PcG-mediated repression in governing a fate switch irreversibly.


Subject(s)
Adult Stem Cells/metabolism , Hair Follicle/metabolism , Histones/metabolism , Repressor Proteins/metabolism , Adult Stem Cells/pathology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin Assembly and Disassembly , Genome-Wide Association Study , Guided Tissue Regeneration , Hair Follicle/pathology , Mice , Polycomb-Group Proteins , Protein Processing, Post-Translational , Signal Transduction/genetics , Skin/pathology
15.
Cell ; 144(2): 296-309, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241896

ABSTRACT

Though many individual transcription factors are known to regulate hematopoietic differentiation, major aspects of the global architecture of hematopoiesis remain unknown. Here, we profiled gene expression in 38 distinct purified populations of human hematopoietic cells and used probabilistic models of gene expression and analysis of cis-elements in gene promoters to decipher the general organization of their regulatory circuitry. We identified modules of highly coexpressed genes, some of which are restricted to a single lineage but most of which are expressed at variable levels across multiple lineages. We found densely interconnected cis-regulatory circuits and a large number of transcription factors that are differentially expressed across hematopoietic states. These findings suggest a more complex regulatory system for hematopoiesis than previously assumed.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Hematopoiesis , Transcription Factors/metabolism , Gene Expression Profiling , Humans
16.
Genes Dev ; 22(9): 1174-89, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451107

ABSTRACT

The ability of somatic stem cells to self-renew and differentiate into downstream lineages is dependent on specialized chromatin environments that keep stem cell-specific genes active and key differentiation factors repressed but poised for activation. The epigenetic factors that provide this type of regulation remain ill-defined. Here we provide the first evidence that the SNF2-like ATPase Mi-2beta of the Nucleosome Remodeling Deacetylase (NuRD) complex is required for maintenance of and multilineage differentiation in the early hematopoietic hierarchy. Shortly after conditional inactivation of Mi-2beta, there is an increase in cycling and a decrease in quiescence in an HSC (hematopoietic stem cell)-enriched bone marrow population. These cycling mutant cells readily differentiate into the erythroid lineage but not into the myeloid and lymphoid lineages. Together, these effects result in an initial expansion of mutant HSC and erythroid progenitors that are later depleted as more differentiated proerythroblasts accumulate at hematopoietic sites exhibiting features of erythroid leukemia. Examination of gene expression in the mutant HSC reveals changes in the expression of genes associated with self-renewal and lineage priming and a pivotal role of Mi-2beta in their regulation. Thus, Mi-2beta provides the hematopoietic system with immune cell capabilities as well as with an extensive regenerative capacity.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Differentiation/physiology , Chromatin/metabolism , Hematopoietic Stem Cells/metabolism , Adenosine Triphosphatases/genetics , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Apoptosis , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Cycle , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation , Cells, Cultured , DNA Helicases , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Flow Cytometry/methods , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis/methods , Receptors, Transferrin/analysis
17.
Genes Dev ; 22(24): 3403-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19141473

ABSTRACT

Mixed-lineage leukemia (MLL) fusion proteins are potent inducers of leukemia, but how these proteins generate aberrant gene expression programs is poorly understood. Here we show that the MLL-AF4 fusion protein occupies developmental regulatory genes important for hematopoietic stem cell identity and self-renewal in human leukemia cells. These MLL-AF4-bound regions have grossly altered chromatin structure, with histone modifications catalyzed by trithorax group proteins and DOT1 extending across large domains. Our results define direct targets of the MLL fusion protein, reveal the global role of epigenetic misregulation in leukemia, and identify new targets for therapeutic intervention in cancer.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/physiology , Leukemia/genetics , Cell Line , Hematopoietic Stem Cells/cytology , Humans , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL