Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(10): e2317147121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38422019

ABSTRACT

Mutations in genes encoding transcription factors inactivate or generate ectopic activities to instigate pathogenesis. By disrupting hematopoietic stem/progenitor cells, GATA2 germline variants create a bone marrow failure and leukemia predisposition, GATA2 deficiency syndrome, yet mechanisms underlying the complex phenotypic constellation are unresolved. We used a GATA2-deficient progenitor rescue system to analyze how genetic variation influences GATA2 functions. Pathogenic variants impaired, without abrogating, GATA2-dependent transcriptional regulation. Variants promoted eosinophil and repressed monocytic differentiation without regulating mast cell and erythroid differentiation. While GATA2 and T354M required the DNA-binding C-terminal zinc finger, T354M disproportionately required the N-terminal finger and N terminus. GATA2 and T354M activated a CCAAT/Enhancer Binding Protein-ε (C/EBPε) enhancer, creating a feedforward loop operating with the T-cell Acute Lymphocyte Leukemia-1 (TAL1) transcription factor. Elevating C/EBPε partially normalized hematopoietic defects of GATA2-deficient progenitors. Thus, pathogenic germline variation discriminatively spares or compromises transcription factor attributes, and retaining an obligate enhancer mechanism distorts a multilineage differentiation program.


Subject(s)
Leukemia , Regulatory Sequences, Nucleic Acid , Humans , Cell Differentiation/genetics , Genotype , Hematopoietic Stem Cells , GATA2 Transcription Factor/genetics
2.
Curr Opin Hematol ; 30(4): 117-123, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37254854

ABSTRACT

PURPOSE OF REVIEW: Recent discoveries have provided evidence for mechanistic links between the master regulator of hematopoiesis GATA2 and the key component of interferon and innate immunity signaling pathways, interferon-regulatory factor-8 (IRF8). These links have important implications for the control of myeloid differentiation in physiological and pathological states. RECENT FINDINGS: GATA2 deficiency resulting from loss of the Gata2 -77 enhancer in progenitors triggers an alarm that instigates the transcriptional induction of innate immune signaling and distorts a myeloid differentiation program. This pathological alteration renders progenitors hyperresponsive to interferon γ, toll-like receptor and interleukin-6 signaling and impaired in granulocyte-macrophage colony-stimulating factor signaling. IRF8 upregulation in -77-/- progenitors promotes monocyte and dendritic cell differentiation while suppressing granulocytic differentiation. As PU.1 promotes transcription of Irf8 and other myeloid and B-lineage genes, GATA2-mediated repression of these genes opposes the PU.1-dependent activating mechanism. SUMMARY: As GATA2 deficiency syndrome is an immunodeficiency disorder often involving myelodysplastic syndromes and acute myeloid leukemia, elucidating how GATA2 commissions and decommissions genome activity and developmental regulatory programs will unveil mechanisms that go awry when GATA2 levels and/or activities are disrupted.


Subject(s)
GATA2 Deficiency , Humans , Cell Differentiation/genetics , GATA2 Transcription Factor/genetics , Immunity, Innate , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferons/metabolism , Animals
3.
Genome Res ; 30(11): 1655-1666, 2020 11.
Article in English | MEDLINE | ID: mdl-32958497

ABSTRACT

Publicly available RNA-seq data is routinely used for retrospective analysis to elucidate new biology. Novel transcript discovery enabled by joint analysis of large collections of RNA-seq data sets has emerged as one such analysis. Current methods for transcript discovery rely on a '2-Step' approach where the first step encompasses building transcripts from individual data sets, followed by the second step that merges predicted transcripts across data sets. To increase the power of transcript discovery from large collections of RNA-seq data sets, we developed a novel '1-Step' approach named Pooling RNA-seq and Assembling Models (PRAM) that builds transcript models from pooled RNA-seq data sets. We demonstrate in a computational benchmark that 1-Step outperforms 2-Step approaches in predicting overall transcript structures and individual splice junctions, while performing competitively in detecting exonic nucleotides. Applying PRAM to 30 human ENCODE RNA-seq data sets identified unannotated transcripts with epigenetic and RAMPAGE signatures similar to those of recently annotated transcripts. In a case study, we discovered and experimentally validated new transcripts through the application of PRAM to mouse hematopoietic RNA-seq data sets. We uncovered new transcripts that share a differential expression pattern with a neighboring gene Pik3cg implicated in human hematopoietic phenotypes, and we provided evidence for the conservation of this relationship in human. PRAM is implemented as an R/Bioconductor package.


Subject(s)
RNA-Seq/methods , Animals , Class Ib Phosphatidylinositol 3-Kinase/genetics , DNA, Intergenic , Genomics , Hematopoietic Stem Cells/metabolism , Humans , Mice , RNA/metabolism , Software
4.
Nat Immunol ; 12(2): 129-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186366

ABSTRACT

The nuclear adaptor Ldb1 functions as a core component of multiprotein transcription complexes that regulate differentiation in diverse cell types. In the hematopoietic lineage, Ldb1 forms a complex with the non-DNA-binding adaptor Lmo2 and the transcription factors E2A, Scl and GATA-1 (or GATA-2). Here we demonstrate a critical and continuous requirement for Ldb1 in the maintenance of both fetal and adult mouse hematopoietic stem cells (HSCs). Deletion of Ldb1 in hematopoietic progenitors resulted in the downregulation of many transcripts required for HSC maintenance. Genome-wide profiling by chromatin immunoprecipitation followed by sequencing (ChIP-Seq) identified Ldb1 complex-binding sites at highly conserved regions in the promoters of genes involved in HSC maintenance. Our results identify a central role for Ldb1 in regulating the transcriptional program responsible for the maintenance of HSCs.


Subject(s)
Adult Stem Cells/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Fetal Development , Hematopoietic Stem Cells/metabolism , Adoptive Transfer , Adult Stem Cells/cytology , Adult Stem Cells/immunology , Adult Stem Cells/transplantation , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/transplantation , Female , Fetal Development/genetics , Fetal Development/immunology , Gene Expression Regulation, Developmental/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , High-Throughput Nucleotide Sequencing , LIM Domain Proteins , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , Protein Binding , Regulatory Elements, Transcriptional/genetics , Regulatory Elements, Transcriptional/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
5.
EMBO Rep ; 22(1): e50535, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33319461

ABSTRACT

Alternative splicing (AS) leads to transcriptome diversity in eukaryotic cells and is one of the key regulators driving cellular differentiation. Although AS is of crucial importance for normal hematopoiesis and hematopoietic malignancies, its role in early hematopoietic development is still largely unknown. Here, by using high-throughput transcriptomic analyses, we show that pervasive and dynamic AS takes place during hematopoietic development of human pluripotent stem cells (hPSCs). We identify a splicing factor switch that occurs during the differentiation of mesodermal cells to endothelial progenitor cells (EPCs). Perturbation of this switch selectively impairs the emergence of EPCs and hemogenic endothelial progenitor cells (HEPs). Mechanistically, an EPC-induced alternative spliced isoform of NUMB dictates EPC specification by controlling NOTCH signaling. Furthermore, we demonstrate that the splicing factor SRSF2 regulates splicing of the EPC-induced NUMB isoform, and the SRSF2-NUMB-NOTCH splicing axis regulates EPC generation. The identification of this splicing factor switch provides a new molecular mechanism to control cell fate and lineage specification.


Subject(s)
Cell Lineage , Pluripotent Stem Cells , Serine-Arginine Splicing Factors/genetics , Cell Differentiation , Cell Lineage/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells , Humans , Membrane Proteins , Nerve Tissue Proteins
6.
Mol Cell ; 59(1): 62-74, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26073540

ABSTRACT

Thousands of cis-elements in genomes are predicted to have vital functions. Although conservation, activity in surrogate assays, polymorphisms, and disease mutations provide functional clues, deletion from endogenous loci constitutes the gold-standard test. A GATA-2-binding, Gata2 intronic cis-element (+9.5) required for hematopoietic stem cell genesis in mice is mutated in a human immunodeficiency syndrome. Because +9.5 is the only cis-element known to mediate stem cell genesis, we devised a strategy to identify functionally comparable enhancers ("+9.5-like") genome-wide. Gene editing revealed +9.5-like activity to mediate GATA-2 occupancy, chromatin opening, and transcriptional activation. A +9.5-like element resided in Samd14, which encodes a protein of unknown function. Samd14 increased hematopoietic progenitor levels/activity and promoted signaling by a pathway vital for hematopoietic stem/progenitor cell regulation (stem cell factor/c-Kit), and c-Kit rescued Samd14 loss-of-function phenotypes. Thus, the hematopoietic stem/progenitor cell cistrome revealed a mediator of a signaling pathway that has broad importance for stem/progenitor cell biology.


Subject(s)
GATA2 Transcription Factor/genetics , Hematopoietic Stem Cells/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-kit/genetics , Transcriptional Activation/genetics , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cell Line , Mice , Molecular Sequence Data , Proteins/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction , Transcription, Genetic/genetics
7.
Nucleic Acids Res ; 49(16): 9007-9025, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34059908

ABSTRACT

Cellular differentiation requires vast remodeling of transcriptomes, and therefore machinery mediating remodeling controls differentiation. Relative to transcriptional mechanisms governing differentiation, post-transcriptional processes are less well understood. As an important post-transcriptional determinant of transcriptomes, the RNA exosome complex (EC) mediates processing and/or degradation of select RNAs. During erythropoiesis, the erythroid transcription factor GATA1 represses EC subunit genes. Depleting EC structural subunits prior to GATA1-mediated repression is deleterious to erythroid progenitor cells. To assess the importance of the EC catalytic subunits Dis3 and Exosc10 in this dynamic process, we asked if these subunits function non-redundantly to control erythropoiesis. Dis3 or Exosc10 depletion in primary murine hematopoietic progenitor cells reduced erythroid progenitors and their progeny, while sparing myeloid cells. Dis3 loss severely compromised erythroid progenitor and erythroblast survival, rendered erythroblasts hypersensitive to apoptosis-inducing stimuli and induced γ-H2AX, indicative of DNA double-stranded breaks. Dis3 loss-of-function phenotypes were more severe than those caused by Exosc10 depletion. We innovated a genetic rescue system to compare human Dis3 with multiple myeloma-associated Dis3 mutants S447R and R750K, and only wild type Dis3 was competent to rescue progenitors. Thus, Dis3 establishes a disease mutation-sensitive, cell type-specific survival mechanism to enable a differentiation program.


Subject(s)
Erythropoiesis , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/metabolism , RNA Processing, Post-Transcriptional , Animals , Apoptosis , Cells, Cultured , DNA Breaks, Double-Stranded , Erythroblasts/cytology , Erythroblasts/metabolism , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/genetics , GATA1 Transcription Factor/metabolism , Humans , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Transcriptome
8.
Nucleic Acids Res ; 49(22): e127, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34581807

ABSTRACT

Single-cell transcriptome sequencing (scRNA-seq) enabled investigations of cellular heterogeneity at exceedingly higher resolutions. Identification of novel cell types or transient developmental stages across multiple experimental conditions is one of its key applications. Linear and non-linear dimensionality reduction for data integration became a foundational tool in inference from scRNA-seq data. We present multilayer graph clustering (MLG) as an integrative approach for combining multiple dimensionality reduction of multi-condition scRNA-seq data. MLG generates a multilayer shared nearest neighbor cell graph with higher signal-to-noise ratio and outperforms current best practices in terms of clustering accuracy across large-scale benchmarking experiments. Application of MLG to a wide variety of datasets from multiple conditions highlights how MLG boosts signal-to-noise ratio for fine-grained sub-population identification. MLG is widely applicable to settings with single cell data integration via dimension reduction.


Subject(s)
RNA-Seq/methods , Single-Cell Analysis/methods , Algorithms , Animals , Cluster Analysis , Hematopoietic Stem Cells/metabolism , Humans , Mice
9.
PLoS Genet ; 16(12): e1009286, 2020 12.
Article in English | MEDLINE | ID: mdl-33370779

ABSTRACT

Developmental-regulatory networks often include large gene families encoding mechanistically-related proteins like G-protein-coupled receptors, zinc finger transcription factors and solute carrier (SLC) transporters. In principle, a common mechanism may confer expression of multiple members integral to a developmental process, or diverse mechanisms may be deployed. Using genetic complementation and enhancer-mutant systems, we analyzed the 456 member SLC family that establishes the small molecule constitution of cells. This analysis identified SLC gene cohorts regulated by GATA1 and/or GATA2 during erythroid differentiation. As >50 SLC genes shared GATA factor regulation, a common mechanism established multiple members of this family. These genes included Slc29a1 encoding an equilibrative nucleoside transporter (Slc29a1/ENT1) that utilizes adenosine as a preferred substrate. Slc29a1 promoted erythroblast survival and differentiation ex vivo. Targeted ablation of murine Slc29a1 in erythroblasts attenuated erythropoiesis and erythrocyte regeneration in response to acute anemia. Our results reveal a GATA factor-regulated SLC ensemble, with a nucleoside transporter component that promotes erythropoiesis and prevents anemia, and establish a mechanistic link between GATA factor and adenosine mechanisms. We propose that integration of the GATA factor-adenosine circuit with other components of the GATA factor-regulated SLC ensemble establishes the small molecule repertoire required for progenitor cells to efficiently generate erythrocytes.


Subject(s)
Equilibrative Nucleoside Transporter 1/metabolism , Erythropoiesis , GATA Transcription Factors/metabolism , Adenosine/metabolism , Animals , Cells, Cultured , Equilibrative Nucleoside Transporter 1/genetics , Mice , Mice, Inbred C57BL
10.
Nucleic Acids Res ; 48(21): 11913-11928, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33119769

ABSTRACT

Given the complexity of intracellular RNA ensembles and vast phenotypic remodeling intrinsic to cellular differentiation, it is instructive to consider the role of RNA regulatory machinery in controlling differentiation. Dynamic post-transcriptional regulation of protein-coding and non-coding transcripts is vital for establishing and maintaining proteomes that enable or oppose differentiation. By contrast to extensively studied transcriptional mechanisms governing differentiation, many questions remain unanswered regarding the involvement of post-transcriptional mechanisms. Through its catalytic activity to selectively process or degrade RNAs, the RNA exosome complex dictates the levels of RNAs comprising multiple RNA classes, thereby regulating chromatin structure, gene expression and differentiation. Although the RNA exosome would be expected to control diverse biological processes, studies to elucidate its biological functions and how it integrates into, or functions in parallel with, cell type-specific transcriptional mechanisms are in their infancy. Mechanistic analyses have demonstrated that the RNA exosome confers expression of a differentiation regulatory receptor tyrosine kinase, downregulates the telomerase RNA component TERC, confers genomic stability and promotes DNA repair, which have considerable physiological and pathological implications. In this review, we address how a broadly operational RNA regulatory complex interfaces with cell type-specific machinery to control cellular differentiation.


Subject(s)
Chromatin/chemistry , Erythrocytes/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Animals , Cell Differentiation , Chromatin/metabolism , Chromatin/ultrastructure , DNA Repair , Erythrocytes/ultrastructure , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/ultrastructure , Genomic Instability , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/ultrastructure , Humans , Mice , Organ Specificity , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
11.
J Biol Chem ; 295(20): 7113-7125, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32241909

ABSTRACT

Hundreds of sterile α-motif (SAM) domains have predicted structural similarities and are reported to bind proteins, lipids, or RNAs. However, the majority of these domains have not been analyzed functionally. Previously, we demonstrated that a SAM domain-containing protein, SAMD14, promotes SCF/proto-oncogene c-Kit (c-Kit) signaling, erythroid progenitor function, and erythrocyte regeneration. Deletion of a Samd14 enhancer (Samd14-Enh), occupied by GATA2 and SCL/TAL1 transcription factors, reduces SAMD14 expression in bone marrow and spleen and is lethal in a hemolytic anemia mouse model. To rigorously establish whether Samd14-Enh deletion reduces anemia-dependent c-Kit signaling by lowering SAMD14 levels, we developed a genetic rescue assay in murine Samd14-Enh-/- primary erythroid precursor cells. SAMD14 expression at endogenous levels rescued c-Kit signaling. The conserved SAM domain was required for SAMD14 to increase colony-forming activity, c-Kit signaling, and progenitor survival. To elucidate the molecular determinants of SAM domain function in SAMD14, we substituted its SAM domain with distinct SAM domains predicted to be structurally similar. The chimeras were less effective than SAMD14 itself in rescuing signaling, survival, and colony-forming activities. Thus, the SAMD14 SAM domain has attributes that are distinct from other SAM domains and underlie SAMD14 function as a regulator of cellular signaling and erythrocyte regeneration.


Subject(s)
Anemia, Hemolytic/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Amino Acid Motifs , Anemia, Hemolytic/genetics , Anemia, Hemolytic/pathology , Animals , Mice , Mice, Mutant Strains , Protein Domains , Proteins/genetics , Proto-Oncogene Proteins c-kit/genetics
12.
Development ; 145(1)2018 01 10.
Article in English | MEDLINE | ID: mdl-29321181

ABSTRACT

Hemoglobin-expressing erythrocytes (red blood cells) act as fundamental metabolic regulators by providing oxygen to cells and tissues throughout the body. Whereas the vital requirement for oxygen to support metabolically active cells and tissues is well established, almost nothing is known regarding how erythrocyte development and function impact regeneration. Furthermore, many questions remain unanswered relating to how insults to hematopoietic stem/progenitor cells and erythrocytes can trigger a massive regenerative process termed 'stress erythropoiesis' to produce billions of erythrocytes. Here, we review the cellular and molecular mechanisms governing erythrocyte development and regeneration, and discuss the potential links between these events and other regenerative processes.


Subject(s)
Cell Differentiation/physiology , Erythrocytes/metabolism , Erythropoiesis/physiology , Hematopoietic Stem Cells/metabolism , Regeneration/physiology , Animals , Biological Transport, Active/physiology , Erythrocytes/cytology , Hematopoietic Stem Cells/cytology , Humans , Oxygen/metabolism
13.
Proc Natl Acad Sci U S A ; 115(43): E10109-E10118, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30301799

ABSTRACT

By inducing the generation and function of hematopoietic stem and progenitor cells, the master regulator of hematopoiesis GATA-2 controls the production of all blood cell types. Heterozygous GATA2 mutations cause immunodeficiency, myelodysplastic syndrome, and acute myeloid leukemia. GATA2 disease mutations commonly disrupt amino acid residues that mediate DNA binding or cis-elements within a vital GATA2 intronic enhancer, suggesting a haploinsufficiency mechanism of pathogenesis. Mutations also occur in GATA2 coding regions distinct from the DNA-binding carboxyl-terminal zinc finger (C-finger), including the amino-terminal zinc finger (N-finger), and N-finger function is not established. Whether distinct mutations differentially impact GATA-2 mechanisms is unknown. Here, we demonstrate that N-finger mutations decreased GATA-2 chromatin occupancy and attenuated target gene regulation. We developed a genetic complementation assay to quantify GATA-2 function in myeloid progenitor cells from Gata2 -77 enhancer-mutant mice. GATA-2 complementation increased erythroid and myeloid differentiation. While GATA-2 disease mutants were not competent to induce erythroid differentiation of Lin-Kit+ myeloid progenitors, unexpectedly, they promoted myeloid differentiation and proliferation. As the myelopoiesis-promoting activity of GATA-2 mutants exceeded that of GATA-2, GATA2 disease mutations are not strictly inhibitory. Thus, we propose that the haploinsufficiency paradigm does not fully explain GATA-2-linked pathogenesis, and an amalgamation of qualitative and quantitative defects instigated by GATA2 mutations underlies the complex phenotypes of GATA-2-dependent pathologies.


Subject(s)
GATA2 Transcription Factor/genetics , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Haploinsufficiency/genetics , Hematopoiesis/genetics , Humans , Mice , Myelodysplastic Syndromes/genetics , Regulatory Sequences, Nucleic Acid/genetics , Stem Cells/metabolism , Zinc Fingers/genetics
14.
Curr Opin Hematol ; 27(3): 163-171, 2020 05.
Article in English | MEDLINE | ID: mdl-32205587

ABSTRACT

PURPOSE OF REVIEW: By establishing mechanisms that deliver oxygen to sustain cells and tissues, fight life-threatening pathogens and harness the immune system to eradicate cancer cells, hematopoietic stem and progenitor cells (HSPCs) are vital in health and disease. The cell biological framework for HSPC generation has been rigorously developed, yet recent single-cell transcriptomic analyses have unveiled permutations of the hematopoietic hierarchy that differ considerably from the traditional roadmap. Deploying mutants that disrupt specific steps in hematopoiesis constitutes a powerful strategy for deconvoluting the complex cell biology. It is striking that a single transcription factor, GATA2, is so crucial for HSPC generation and function, and therefore it is instructive to consider mechanisms governing GATA2 expression and activity. The present review focuses on an essential GATA2 enhancer (+9.5) and how +9.5 mutants inform basic and clinical/translational science. RECENT FINDINGS: +9.5 is essential for HSPC generation and function during development and hematopoietic regeneration. Human +9.5 mutations cause immunodeficiency, myelodysplastic syndrome, and acute myeloid leukemia. Qualitatively and quantitatively distinct contributions of +9.5 cis-regulatory elements confer context-dependent enhancer activity. The discovery of +9.5 and its mutant alleles spawned fundamental insights into hematopoiesis, and given its role to suppress blood disease emergence, clinical centers test for mutations in this sequence to diagnose the cause of enigmatic cytopenias. SUMMARY: Multidisciplinary approaches to discover and understand cis-regulatory elements governing expression of key regulators of hematopoiesis unveil biological and mechanistic insights that provide the logic for innovating clinical applications.


Subject(s)
Enhancer Elements, Genetic , GATA2 Transcription Factor , Hematopoietic Stem Cells , Immunologic Deficiency Syndromes , Leukemia, Myeloid, Acute , Mutation , Myelodysplastic Syndromes , Animals , GATA2 Transcription Factor/biosynthesis , GATA2 Transcription Factor/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Precision Medicine
15.
Genome Res ; 26(8): 1124-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27405803

ABSTRACT

RNA-seq is currently the technology of choice for global measurement of transcript abundances in cells. Despite its successes, isoform-level quantification remains difficult because short RNA-seq reads are often compatible with multiple alternatively spliced isoforms. Existing methods rely heavily on uniquely mapping reads, which are not available for numerous isoforms that lack regions of unique sequence. To improve quantification accuracy in such difficult cases, we developed a novel computational method, prior-enhanced RSEM (pRSEM), which uses a complementary data type in addition to RNA-seq data. We found that ChIP-seq data of RNA polymerase II and histone modifications were particularly informative in this approach. In qRT-PCR validations, pRSEM was shown to be superior than competing methods in estimating relative isoform abundances within or across conditions. Data-driven simulations suggested that pRSEM has a greatly decreased false-positive rate at the expense of a small increase in false-negative rate. In aggregate, our study demonstrates that pRSEM transforms existing capacity to precisely estimate transcript abundances, especially at the isoform level.


Subject(s)
Alternative Splicing/genetics , RNA/genetics , Sequence Analysis, RNA/methods , Algorithms , Computational Biology/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing/methods , Humans , RNA Polymerase II/genetics , Software
16.
Blood ; 129(15): 2092-2102, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28179282

ABSTRACT

The discovery of the GATA binding protein (GATA factor) transcription factor family revolutionized hematology. Studies of GATA proteins have yielded vital contributions to our understanding of how hematopoietic stem and progenitor cells develop from precursors, how progenitors generate red blood cells, how hemoglobin synthesis is regulated, and the molecular underpinnings of nonmalignant and malignant hematologic disorders. This thrilling journey began with mechanistic studies on a ß-globin enhancer- and promoter-binding factor, GATA-1, the founding member of the GATA family. This work ushered in the cloning of related proteins, GATA-2-6, with distinct and/or overlapping expression patterns. Herein, we discuss how the hematopoietic GATA factors (GATA-1-3) function via a battery of mechanistic permutations, which can be GATA factor subtype, cell type, and locus specific. Understanding this intriguing protein family requires consideration of how the mechanistic permutations are amalgamated into circuits to orchestrate processes of interest to the hematologist and more broadly.


Subject(s)
Enhancer Elements, Genetic , GATA Transcription Factors , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms , Hematopoietic Stem Cells/metabolism , beta-Globins , Animals , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , beta-Globins/biosynthesis , beta-Globins/genetics
17.
Blood ; 130(Suppl_1): 7, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-31940664

ABSTRACT

DISCLOSURES: No relevant conflicts of interest to declare.

18.
Blood ; 129(3): 358-370, 2017 01 19.
Article in English | MEDLINE | ID: mdl-27815262

ABSTRACT

Somatic mutations in TP53 and NRAS are associated with transformation of human chronic myeloid diseases to acute myeloid leukemia (AML). Here, we report that concurrent RAS pathway and TP53 mutations are identified in a subset of AML patients and confer an inferior overall survival. To further investigate the genetic interaction between p53 loss and endogenous NrasG12D/+ in AML, we generated conditional NrasG12D/+p53-/- mice. Consistent with the clinical data, recipient mice transplanted with NrasG12D/+p53-/- bone marrow cells rapidly develop a highly penetrant AML. We find that p53-/- cooperates with NrasG12D/+ to promote increased quiescence in megakaryocyte-erythroid progenitors (MEPs). NrasG12D/+p53-/- MEPs are transformed to self-renewing AML-initiating cells and are capable of inducing AML in serially transplanted recipients. RNA sequencing analysis revealed that transformed MEPs gain a partial hematopoietic stem cell signature and largely retain an MEP signature. Their distinct transcriptomes suggests a potential regulation by p53 loss. In addition, we show that during AML development, transformed MEPs acquire overexpression of oncogenic Nras, leading to hyperactivation of ERK1/2 signaling. Our results demonstrate that p53-/- synergizes with enhanced oncogenic Nras signaling to transform MEPs and drive AML development. This model may serve as a platform to test candidate therapeutics in this aggressive subset of AML.


Subject(s)
Cell Transformation, Neoplastic/genetics , GTP Phosphohydrolases/genetics , Leukemia, Myeloid, Acute/pathology , Megakaryocyte-Erythroid Progenitor Cells/pathology , Membrane Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Bone Marrow Transplantation , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System , Mice , Mutation , Signal Transduction , Tumor Suppressor Protein p53/deficiency
19.
EMBO Rep ; 17(2): 249-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26698166

ABSTRACT

Metal ion-containing macromolecules have fundamental roles in essentially all biological processes throughout the evolutionary tree. For example, iron-containing heme is a cofactor in enzyme catalysis and electron transfer and an essential hemoglobin constituent. To meet the intense demand for hemoglobin assembly in red blood cells, the cell type-specific factor GATA-1 activates transcription of Alas2, encoding the rate-limiting enzyme in heme biosynthesis, 5-aminolevulinic acid synthase-2 (ALAS-2). Using genetic editing to unravel mechanisms governing heme biosynthesis, we discovered a GATA factor- and heme-dependent circuit that establishes the erythroid cell transcriptome. CRISPR/Cas9-mediated ablation of two Alas2 intronic cis elements strongly reduces GATA-1-induced Alas2 transcription, heme biosynthesis, and surprisingly, GATA-1 regulation of other vital constituents of the erythroid cell transcriptome. Bypassing ALAS-2 function in Alas2 cis element-mutant cells by providing its catalytic product 5-aminolevulinic acid rescues heme biosynthesis and the GATA-1-dependent genetic network. Heme amplifies GATA-1 function by downregulating the heme-sensing transcriptional repressor Bach1 and via a Bach1-insensitive mechanism. Through this dual mechanism, heme and a master regulator collaborate to orchestrate a cell type-specific transcriptional program that promotes cellular differentiation.


Subject(s)
GATA1 Transcription Factor/metabolism , Gene Regulatory Networks , Hematopoiesis , Heme/metabolism , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Amino Acid Sequence , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , CHO Cells , Cricetinae , Cricetulus , Erythroid Cells/cytology , Erythroid Cells/metabolism , Mice , Molecular Sequence Data , Transcriptome
20.
Proc Natl Acad Sci U S A ; 112(25): 7689-94, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056298

ABSTRACT

Multiple myeloma (MM), a malignancy of plasma cells, is characterized by widespread genomic heterogeneity and, consequently, differences in disease progression and drug response. Although recent large-scale sequencing studies have greatly improved our understanding of MM genomes, our knowledge about genomic structural variation in MM is attenuated due to the limitations of commonly used sequencing approaches. In this study, we present the application of optical mapping, a single-molecule, whole-genome analysis system, to discover new structural variants in a primary MM genome. Through our analysis, we have identified and characterized widespread structural variation in this tumor genome. Additionally, we describe our efforts toward comprehensive characterization of genome structure and variation by integrating our findings from optical mapping with those from DNA sequencing-based genomic analysis. Finally, by studying this MM genome at two time points during tumor progression, we have demonstrated an increase in mutational burden with tumor progression at all length scales of variation.


Subject(s)
DNA Copy Number Variations , Multiple Myeloma/genetics , DNA/genetics , Humans , Loss of Heterozygosity , Multiple Myeloma/pathology , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL