Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Cell Rep ; 43(4): 114107, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38613785

The production of type 1 conventional dendritic cells (cDC1s) requires high expression of the transcription factor IRF8. Three enhancers at the Irf8 3' region function in a differentiation stage-specific manner. However, whether and how these enhancers interact physically and functionally remains unclear. Here, we show that the Irf8 3' enhancers directly interact with each other and contact the Irf8 gene body during cDC1 differentiation. The +56 kb enhancer, which functions from multipotent progenitor stages, activates the other 3' enhancers through an IRF8-dependent transcription factor program, that is, in trans. Then, the +32 kb enhancer, which operates in cDC1-committed cells, reversely acts in cis on the other 3' enhancers to maintain the high expression of Irf8. Indeed, mice with compound heterozygous deletion of the +56 and +32 kb enhancers are unable to generate cDC1s. These results illustrate how multiple enhancers cooperate to induce a lineage-determining transcription factor gene during cell differentiation.


Cell Differentiation , Dendritic Cells , Enhancer Elements, Genetic , Interferon Regulatory Factors , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Animals , Dendritic Cells/metabolism , Dendritic Cells/cytology , Enhancer Elements, Genetic/genetics , Mice , Mice, Inbred C57BL
2.
Exp Hematol ; 132: 104173, 2024 Apr.
Article En | MEDLINE | ID: mdl-38309573

In the intricate orchestration of the central dogma, pre-mRNA splicing plays a crucial role in the post-transcriptional process that transforms DNA into mature mRNA. Widely acknowledged as a pivotal RNA processing step, it significantly influences gene expression and alters the functionality of gene product proteins. Although U2-dependent spliceosomes efficiently manage the removal of over 99% of introns, a distinct subset of essential genes undergo splicing with a different intron type, denoted as minor introns, using U12-dependent spliceosomes. Mutations in spliceosome component genes are now recognized as prevalent genetic abnormalities in cancer patients, especially those with hematologic malignancies. Despite the relative rarity of minor introns, genes containing them are evolutionarily conserved and play crucial roles in functions such as the RAS-MAPK pathway. Disruptions in U12-type minor intron splicing caused by mutations in snRNA or its regulatory components significantly contribute to cancer progression. Notably, recurrent mutations associated with myelodysplastic syndrome (MDS) in the minor spliceosome component ZRSR2 underscore its significance. Examination of ZRSR2-mutated MDS cells has revealed that only a subset of minor spliceosome-dependent genes, such as LZTR1, consistently exhibit missplicing. Recent technological advancements have uncovered insights into minor introns, raising inquiries beyond current understanding. This review comprehensively explores the importance of minor intron regulation, the molecular implications of minor (U12-type) spliceosomal mutations and cis-regulatory regions, and the evolutionary progress of studies on minor, aiming to provide a sophisticated understanding of their intricate role in cancer biology.


Hematologic Neoplasms , Myelodysplastic Syndromes , Humans , Introns , Spliceosomes/genetics , Spliceosomes/metabolism , RNA Splicing , RNA, Messenger/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Transcription Factors/genetics
3.
Nat Aging ; 4(3): 319-335, 2024 Mar.
Article En | MEDLINE | ID: mdl-38388781

Plasma membrane damage (PMD) occurs in all cell types due to environmental perturbation and cell-autonomous activities. However, cellular outcomes of PMD remain largely unknown except for recovery or death. In this study, using budding yeast and normal human fibroblasts, we found that cellular senescence-stable cell cycle arrest contributing to organismal aging-is the long-term outcome of PMD. Our genetic screening using budding yeast unexpectedly identified a close genetic association between PMD response and replicative lifespan regulations. Furthermore, PMD limits replicative lifespan in budding yeast; upregulation of membrane repair factors ESCRT-III (SNF7) and AAA-ATPase (VPS4) extends it. In normal human fibroblasts, PMD induces premature senescence via the Ca2+-p53 axis but not the major senescence pathway, DNA damage response pathway. Transient upregulation of ESCRT-III (CHMP4B) suppressed PMD-dependent senescence. Together with mRNA sequencing results, our study highlights an underappreciated but ubiquitous senescent cell subtype: PMD-dependent senescent cells.


Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Saccharomyces cerevisiae/genetics , Longevity , Tumor Suppressor Protein p53/genetics , Fibroblasts , Cell Membrane/metabolism , Cellular Senescence/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Adenosine Triphosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism
4.
Nat Commun ; 14(1): 8372, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-38102116

ATP-dependent chromatin remodeling SWI/SNF complexes exist in three subcomplexes: canonical BAF (cBAF), polybromo BAF (PBAF), and a newly described non-canonical BAF (ncBAF). While cBAF and PBAF regulate fates of multiple cell types, roles for ncBAF in hematopoietic stem cells (HSCs) have not been investigated. Motivated by recent discovery of disrupted expression of BRD9, an essential component of ncBAF, in multiple cancers, including clonal hematopoietic disorders, we evaluate here the role of BRD9 in normal and malignant HSCs. BRD9 loss enhances chromatin accessibility, promoting myeloid lineage skewing while impairing B cell development. BRD9 significantly colocalizes with CTCF, whose chromatin recruitment is augmented by BRD9 loss, leading to altered chromatin state and expression of myeloid-related genes within intact topologically associating domains. These data uncover ncBAF as critical for cell fate specification in HSCs via three-dimensional regulation of gene expression and illuminate roles for ncBAF in normal and malignant hematopoiesis.


Chromatin , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin Assembly and Disassembly , Cell Differentiation , Hematopoietic Stem Cells/metabolism
5.
Nat Cancer ; 4(12): 1675-1692, 2023 Dec.
Article En | MEDLINE | ID: mdl-37872381

Despite recent advances in the treatment of acute myeloid leukemia (AML), there has been limited success in targeting surface antigens in AML, in part due to shared expression across malignant and normal cells. Here, high-density immunophenotyping of AML coupled with proteogenomics identified unique expression of a variety of antigens, including the RNA helicase U5 snRNP200, on the surface of AML cells but not on normal hematopoietic precursors and skewed Fc receptor distribution in the AML immune microenvironment. Cell membrane localization of U5 snRNP200 was linked to surface expression of the Fcγ receptor IIIA (FcγIIIA, also known as CD32A) and correlated with expression of interferon-regulated immune response genes. Anti-U5 snRNP200 antibodies engaging activating Fcγ receptors were efficacious across immunocompetent AML models and were augmented by combination with azacitidine. These data provide a roadmap of AML-associated antigens with Fc receptor distribution in AML and highlight the potential for targeting the AML cell surface using Fc-optimized therapeutics.


Leukemia, Myeloid, Acute , Receptors, IgG , Humans , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antigens, Surface , Leukemia, Myeloid, Acute/drug therapy , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Ribonucleoproteins, Small Nuclear , Tumor Microenvironment
6.
Leukemia ; 37(9): 1802-1811, 2023 09.
Article En | MEDLINE | ID: mdl-37464069

SETBP1 is a potential epigenetic regulator whose hotspot mutations preventing proteasomal degradation are recurrently detected in myeloid malignancies with poor prognosis. It is believed that the mutant SETBP1 exerts amplified effects of wild-type SETBP1 rather than neomorphic functions. This indicates that dysregulated quantitative control of SETBP1 would result in the transformation of hematopoietic cells. However, little is known about the roles of endogenous SETBP1 in malignant and normal hematopoiesis. Thus, we integrated the analyses of primary AML and healthy samples, cancer cell lines, and a newly generated murine model, Vav1-iCre;Setbp1fl/fl. Despite the expression in long-term hematopoietic stem cells, SETBP1 depletion in normal hematopoiesis minimally alters self-renewal, differentiation, or reconstitution in vivo. Indeed, its loss does not profoundly alter transcription or chromatin accessibilities. Furthermore, although AML with high SETBP1 mRNA is associated with genetic and clinical characteristics for dismal outcomes, SETBP1 is dispensable for the development or maintenance of AML. Contrary to the evidence that SETBP1 mutations are restricted to myeloid malignancies, dependency on SETBP1 mRNA expression is not observed in AML. These unexpected results shed light on the unrecognized idea that a physiologically nonessential gene can act as an oncogene when the machinery of protein degradation is damaged.


Hematopoiesis , Leukemia, Myeloid, Acute , Animals , Humans , Mice , Carrier Proteins/genetics , Cell Differentiation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/pathology , Mutation , Nuclear Proteins/genetics
7.
Int J Hematol ; 117(6): 821-829, 2023 Jun.
Article En | MEDLINE | ID: mdl-37041345

Hematopoiesis is maintained and regulated by a bone marrow-specific microenvironment called a niche. In hematological malignancies, tumor cells induce niche remodeling, and the reconstructed niche is closely linked to disease pathogenesis. Recent studies have suggested that extracellular vesicles (EVs) secreted from tumor cells play a principal role in niche remodeling in hematological malignancies. Although EVs are emerging as potential therapeutic targets, the underlying mechanism of action remains unclear, and selective inhibition remains a challenge. This review summarizes remodeling of the bone marrow microenvironment in hematological malignancies and its contribution to pathogenesis, as well as roles of tumor-derived EVs, and provides a perspective on future research in this field.


Extracellular Vesicles , Hematologic Neoplasms , Myeloproliferative Disorders , Humans , Bone Marrow/pathology , Extracellular Vesicles/pathology , Myeloproliferative Disorders/pathology , Hematologic Neoplasms/pathology , Hematopoiesis , Tumor Microenvironment
8.
Cell Rep ; 42(3): 112165, 2023 03 28.
Article En | MEDLINE | ID: mdl-36862552

Inflammatory stimuli cause a state of emergency myelopoiesis leading to neutrophil-like monocyte expansion. However, their function, the committed precursors, or growth factors remain elusive. In this study we find that Ym1+Ly6Chi monocytes, an immunoregulatory entity of neutrophil-like monocytes, arise from progenitors of neutrophil 1 (proNeu1). Granulocyte-colony stimulating factor (G-CSF) favors the production of neutrophil-like monocytes through previously unknown CD81+CX3CR1lo monocyte precursors. GFI1 promotes the differentiation of proNeu2 from proNeu1 at the cost of producing neutrophil-like monocytes. The human counterpart of neutrophil-like monocytes that also expands in response to G-CSF is found in CD14+CD16- monocyte fraction. The human neutrophil-like monocytes are discriminated from CD14+CD16- classical monocytes by CXCR1 expression and the capacity to suppress T cell proliferation. Collectively, our findings suggest that the aberrant expansion of neutrophil-like monocytes under inflammatory conditions is a process conserved between mouse and human, which may be beneficial for the resolution of inflammation.


Monocytes , Neutrophils , Mice , Animals , Humans , Monocytes/physiology , Myelopoiesis , Cell Differentiation , Granulocyte Colony-Stimulating Factor
9.
Cancer Sci ; 113(9): 2934-2942, 2022 Sep.
Article En | MEDLINE | ID: mdl-35766428

Pre-mRNA splicing is now widely recognized as a cotranscriptional and post-transcriptional mechanism essential for regulating gene expression and modifying gene product function. Mutations in genes encoding core spliceosomal proteins and accessory regulatory splicing factors are now considered among the most recurrent genetic abnormalities in patients with cancer, particularly hematologic malignancies. These include mutations in the major (U2-type) and minor (U12-type) spliceosomes, which remove >99% and ~0.35% of introns, respectively. Growing evidence indicates that aberrant splicing of evolutionarily conserved U12-type minor introns plays a crucial role in cancer as the minor spliceosome component, ZRSR2, is subject to recurrent, leukemia-associated mutations, and intronic mutations have been shown to disrupt the splicing of minor introns. Here, we review the importance of minor intron regulation, the molecular effects of the minor (U12-type) spliceosomal mutations and cis-regulatory regions, and the development of minor intron studies for better understanding of cancer biology.


Neoplasms , RNA Splicing , Humans , Introns/genetics , Mutation , Neoplasms/genetics , Neoplasms/metabolism , RNA Splicing/genetics , Spliceosomes/genetics , Spliceosomes/metabolism
10.
Genome Res ; 30(8): 1119-1130, 2020 08.
Article En | MEDLINE | ID: mdl-32747411

Polycomb group proteins are important for maintaining gene expression patterns and cell identity in metazoans. The mammalian Polycomb repressive deubiquitinase (PR-DUB) complexes catalyze removal of monoubiquitination on lysine 119 of histone H2A (H2AK119ub1) through a multiprotein core comprised of BAP1, HCFC1, FOXK1/2, and OGT in combination with either of ASXL1, 2, or 3. Mutations in PR-DUB components are frequent in cancer. However, mechanistic understanding of PR-DUB function in gene regulation is limited. Here, we show that BAP1 is dependent on the ASXL proteins and FOXK1/2 in facilitating gene activation across the genome. Although PR-DUB was previously shown to cooperate with PRC2, we observed minimal overlap and functional interaction between BAP1 and PRC2 in embryonic stem cells. Collectively, these results demonstrate that PR-DUB, by counteracting accumulation of H2AK119ub1, maintains chromatin in an optimal configuration ensuring expression of genes important for general functions such as cell metabolism and homeostasis.


Chromatin/metabolism , Deubiquitinating Enzymes/metabolism , Gene Expression Regulation/genetics , Histones/metabolism , Polycomb-Group Proteins/metabolism , Animals , Cell Proliferation/genetics , Cells, Cultured , Chromatin/genetics , Deubiquitinating Enzymes/genetics , Forkhead Transcription Factors/metabolism , Gene Knockout Techniques , Mice , Mice, Inbred BALB C , Mice, Knockout , Mouse Embryonic Stem Cells , Polycomb-Group Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
11.
Blood ; 134(14): 1154-1158, 2019 10 03.
Article En | MEDLINE | ID: mdl-31434704

KDM4/JMJD2 are H3K9- and H3K36-specific demethylases, which are considered promising therapeutic targets for the treatment of acute myeloid leukemia (AML) harboring MLL translocations. Here, we investigate the long-term effects of depleting KDM4 activity on normal hematopoiesis to probe potential side effects of continuous inhibition of these enzymes. Utilizing conditional Kdm4a/Kdm4b/Kdm4c triple-knockout mice, we show that KDM4 activity is required for hematopoietic stem cell (HSC) maintenance in vivo. The knockout of the KDM4 demethylases leads to accumulation of H3K9me3 on transcription start sites and the corresponding downregulation of expression of several genes in HSCs. We show that 2 of these genes, Taf1b and Nom1, are essential for the maintenance of hematopoietic cells. Taken together, our results show that the KDM4 demethylases are required for the expression of genes essential for the long-term maintenance of normal hematopoiesis.


Hematopoietic Stem Cells/cytology , Histone Demethylases/genetics , Animals , Cell Survival , Cells, Cultured , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Mice, Inbred C57BL , Mice, Knockout , Transcription Initiation Site
12.
Nat Commun ; 10(1): 981, 2019 02 28.
Article En | MEDLINE | ID: mdl-30816115

Animal cells undergo rapid rounding during mitosis, ensuring proper chromosome segregation, during which an outward rounding force abruptly increases upon prometaphase entry and is maintained at a constant level during metaphase. Initial cortical tension is generated by the actomyosin system to which both myosin motors and actin network architecture contribute. However, how cortical tension is maintained and its physiological significance remain unknown. We demonstrate here that Cdk1-mediated phosphorylation of DIAPH1 stably maintains cortical tension after rounding and inactivates the spindle assembly checkpoint (SAC). Cdk1 phosphorylates DIAPH1, preventing profilin1 binding to maintain cortical tension. Mutation of DIAPH1 phosphorylation sites promotes cortical F-actin accumulation, increases cortical tension, and delays anaphase onset due to SAC activation. Measurement of the intra-kinetochore length suggests that Cdk1-mediated cortex relaxation is indispensable for kinetochore stretching. We thus uncovered a previously unknown mechanism by which Cdk1 coordinates cortical tension maintenance and SAC inactivation at anaphase onset.


Adaptor Proteins, Signal Transducing/metabolism , CDC2 Protein Kinase/metabolism , Chromosome Segregation/physiology , M Phase Cell Cycle Checkpoints/physiology , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Anaphase/physiology , Cyclin B1/metabolism , Formins , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Kinetochores/metabolism , Metaphase/physiology , Phosphorylation , Profilins/chemistry , Profilins/genetics , Profilins/metabolism , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Genome Res ; 29(4): 564-575, 2019 04.
Article En | MEDLINE | ID: mdl-30796038

The epigenetic regulator TET2 is frequently mutated in hematological diseases. Mutations have been shown to arise in hematopoietic stem cells early in disease development and lead to altered DNA methylation landscapes and an increased risk of hematopoietic malignancy. Here, we show by genome-wide mapping of TET2 binding sites in different cell types that TET2 localizes to regions of open chromatin and cell-type-specific enhancers. We find that deletion of Tet2 in native hematopoiesis as well as fully transformed acute myeloid leukemia (AML) results in changes in transcription factor (TF) activity within these regions, and we provide evidence that loss of TET2 leads to attenuation of chromatin binding of members of the basic helix-loop-helix (bHLH) TF family. Together, these findings demonstrate that TET2 activity shapes the local chromatin environment at enhancers to facilitate TF binding and provides an example of how epigenetic dysregulation can affect gene expression patterns and drive disease development.


Chromatin/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cells, Cultured , Dioxygenases , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/metabolism , Mice , Protein Binding
14.
J Biochem ; 159(1): 17-25, 2016 Jan.
Article En | MEDLINE | ID: mdl-26590301

Recent progress in high-speed sequencing technology has revealed that tumors harbor novel mutations in a variety of genes including those for molecules involved in epigenetics and splicing, some of which were not categorized to previously thought malignancy-related genes. However, despite thorough identification of mutations in solid tumors and hematological malignancies, how these mutations induce cell transformation still remains elusive. In addition, each tumor usually contains multiple mutations or sometimes consists of multiple clones, which makes functional analysis difficult. Fifteen years ago, it was proposed that combination of two types of mutations induce acute leukemia; Class I mutations induce cell growth or inhibit apoptosis while class II mutations block differentiation, co-operating in inducing acute leukemia. This notion has been proven using a variety of mouse models, however most of recently found mutations are not typical class I/II mutations. Although some novel mutations have been found to functionally work as class I or II mutation in leukemogenesis, the classical class I/II theory seems to be too simple to explain the whole story. We here overview the molecular basis of hematological malignancies based on clinical and experimental results, and propose a new working hypothesis for leukemogenesis.


Carcinogenesis/genetics , Hematologic Neoplasms/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Translocation, Genetic , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Transgenic , Mutation , Oncogene Proteins, Fusion/genetics , Phenotype
15.
Exp Hematol ; 43(4): 300-8.e1, 2015 Apr.
Article En | MEDLINE | ID: mdl-25534203

Two types of CCAAT-enhancer-binding protein α (C/EBPα) mutants are found in acute myeloid leukemia (AML) patients: N-terminal frame-shift mutants (C/EBPα-N(m)) generating p30 as a dominant form and C-terminal basic leucine zipper domain mutants (C/EBPα-C(m)). We have previously shown that C/EBPα-K304_R323dup belonging to C/EBPα-C(m), but not C/EBPα-T60fsX159 belonging to C/EBPα-N(m), alone induced AML in mouse bone marrow transplantation (BMT) models. Here we show that various C/EBPα-C(m) mutations have a similar, but not identical, potential in myeloid leukemogenesis. Notably, like C/EBPα-K304_R323dup, any type of C/EBPα-C(m) tested (C/EBPα-S299_K304dup, K313dup, or N321D) by itself induced AML, albeit with different latencies after BMT; C/EBPα-N321D induced AML with the shortest latency. By analyzing the gene expression profiles of C/EBPα-N321D- and mock-transduced c-kit(+)Sca-1(+)Lin(-) cells, we identified Csf1r as a gene downregulated by C/EBPα-N321D. In addition, leukemic cells expressing C/EBPα-C(m) exhibited low levels of colony stimulating factor 1 receptor in mice. On the other hand, transduction with C/EBPα-N(m) did not influence Csf1r expression in c-kit(+)Sca-1(+)Lin(-) cells, implying a unique role for C/EBPα-C(m) in downregulating Csf1r. Importantly, Csf1r overexpression collaborated with C/EBPα-N321D to induce fulminant AML with leukocytosis in mouse BMT models to a greater extent than did C/EBPα-N321D alone. Collectively, these results suggest that C/EBPα-C(m)-mediated downregulation of Csf1r has a negative, rather than a positive, impact on the progression of AML involving C/EBPα-C(m), which might possibly be accelerated by additional genetic and/or epigenetic alterations inducing Csf1r upregulation.


CCAAT-Enhancer-Binding Protein-alpha/physiology , Down-Regulation/physiology , Leukemia, Myeloid, Acute/pathology , Mutation , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Base Sequence , CCAAT-Enhancer-Binding Protein-alpha/genetics , DNA Primers , Disease Progression , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/physiopathology , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Sci Rep ; 4: 4012, 2014 Feb 06.
Article En | MEDLINE | ID: mdl-24500246

The quiescent (G0) phase of the cell cycle is the reversible phase from which the cells exit from the cell cycle. Due to the difficulty of defining the G0 phase, quiescent cells have not been well characterized. In this study, a fusion protein consisting of mVenus and a defective mutant of CDK inhibitor, p27 (p27K(-)) was shown to be able to identify and isolate a population of quiescent cells and to effectively visualize the G0 to G1 transition. By comparing the expression profiles of the G0 and G1 cells defined by mVenus-p27K(-), we have identified molecular features of quiescent cells. Quiescence is also an important feature of many types of stem cells, and mVenus-p27K(-)-transgenic mice enabled the detection of the quiescent cells with muscle stem cell markers in muscle in vivo. The mVenus-p27K(-) probe could be useful in investigating stem cells as well as quiescent cells.


Bacterial Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , G1 Phase/genetics , Luminescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Resting Phase, Cell Cycle/genetics , 3T3 Cells , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Gene Expression , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Selenium-Binding Proteins/biosynthesis , Selenium-Binding Proteins/genetics , Stem Cells/cytology , Tumor Suppressor Protein p53/biosynthesis
17.
Exp Hematol ; 42(5): 369-379.e3, 2014 May.
Article En | MEDLINE | ID: mdl-24486648

We have previously shown that elevated expression of Hairy enhancer of split 1 (Hes1) contributes to blast crisis transition in Bcr-Abl-positive chronic myelogenous leukemia. Here we investigate whether Hes1 is involved in the development of other myeloid neoplasms. Notably, Hes1 expression was elevated in only a few cases of 65 samples with different types of myeloid neoplasms. Interestingly, elevated expression of Hes1 was found in two of five samples of Fip1-like1 platelet-derived growth factor receptor-α (FIP1L1-PDGFA)-positive myeloid neoplasms associated with eosinophilia. Whereas FIP1L1-PDGFRα alone induced acute T-cell leukemia or myeloproliferative neoplasms in mouse bone marrow transplantation models, mice transplanted with bone marrow cells expressing both Hes1 and FIP1L1-PDGFRα developed acute leukemia characterized by an expansion of myeloid blasts and leukemic cells without eosinophilic granules. FIP1L1-PDGFRα conferred cytokine-independent growth to Hes1-transduced common myeloid progenitors, interleukin-3-dependent cells. Imatinib inhibited the growth of common myeloid progenitors expressing Hes1 with FIP1L1-PDGFRα, but not with imatinib-resistant FIP1L1-PDGFRα mutants harboring T674I or D842V. In contrast, ponatinib efficiently eradicated leukemic cells expressing Hes1 and the imatinib-resistant FLP1L1-PDGFRΑ mutant in vitro and in vivo. Thus, we have established mouse models of FIP1L1-PDGFRA-positive leukemia in myeloid blast crisis, which will help elucidate the pathogenesis of the disease and develop a new treatment for it.


Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Blast Crisis/metabolism , Gene Expression Regulation, Leukemic , Homeodomain Proteins/biosynthesis , Leukemia, Myeloid, Acute/mortality , Neoplasms, Experimental/metabolism , Oncogene Proteins, Fusion/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Substitution , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Benzamides/pharmacology , Blast Crisis/genetics , Blast Crisis/pathology , Female , Homeodomain Proteins/genetics , Humans , Imatinib Mesylate , Interleukin-3/biosynthesis , Interleukin-3/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred BALB C , Mutation, Missense , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oncogene Proteins, Fusion/genetics , Piperazines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transcription Factor HES-1 , mRNA Cleavage and Polyadenylation Factors/genetics
18.
J Clin Invest ; 123(11): 4627-40, 2013 Nov.
Article En | MEDLINE | ID: mdl-24216483

Recurrent mutations in the gene encoding additional sex combs-like 1 (ASXL1) are found in various hematologic malignancies and associated with poor prognosis. In particular, ASXL1 mutations are common in patients with hematologic malignancies associated with myelodysplasia, including myelodysplastic syndromes (MDSs), and chronic myelomonocytic leukemia. Although loss-of-function ASXL1 mutations promote myeloid transformation, a large subset of ASXL1 mutations is thought to result in stable truncation of ASXL1. Here we demonstrate that C-terminal­truncating Asxl1 mutations (ASXL1-MTs) inhibited myeloid differentiation and induced MDS-like disease in mice. ASXL1-MT mice displayed features of human-associated MDS, including multi-lineage myelodysplasia, pancytopenia, and occasional progression to overt leukemia. ASXL1-MT resulted in derepression of homeobox A9 (Hoxa9) and microRNA-125a (miR-125a) expression through inhibition of polycomb repressive complex 2­mediated (PRC2-mediated) methylation of histone H3K27. miR-125a reduced expression of C-type lectin domain family 5, member a (Clec5a), which is involved in myeloid differentiation. In addition, HOXA9 expression was high in MDS patients with ASXL1-MT, while CLEC5A expression was generally low. Thus, ASXL1-MT­induced MDS-like disease in mice is associated with derepression of Hoxa9 and miR-125a and with Clec5a dysregulation. Our data provide evidence for an axis of MDS pathogenesis that implicates both ASXL1 mutations and miR-125a as therapeutic targets in MDS.


Mutant Proteins/genetics , Mutation , Myelodysplastic Syndromes/genetics , Repressor Proteins/genetics , Animals , Cell Line , Disease Models, Animal , Histones/metabolism , Homeodomain Proteins/genetics , Humans , Lectins, C-Type/genetics , Methylation , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Myelopoiesis/genetics , Peptide Fragments/genetics , Receptors, Cell Surface/genetics
19.
PLoS One ; 8(5): e63001, 2013.
Article En | MEDLINE | ID: mdl-23696789

BACKGROUND: Male germ cell RacGTPase activating protein (MgcRacGAP) is an important regulator of the Rho family GTPases--RhoA, Rac1, and Cdc42--and is indispensable in cytokinesis and cell cycle progression. Inactivation of RhoA by phosphorylated MgcRacGAP is an essential step in cytokinesis. MgcRacGAP is also involved in G1-S transition and nuclear transport of signal transducer and activator of transcription 3/5 (STAT3/5). Expression of MgcRacGAP is strictly controlled in a cell cycle-dependent manner. However, the underlying mechanisms have not been elucidated. METHODOLOGY/PRINCIPAL FINDINGS: Using MgcRacGAP deletion mutants and the fusion proteins of full-length or partial fragments of MgcRacGAP to mVenus fluorescent protein, we demonstrated that MgcRacGAP is degraded by the ubiquitin-proteasome pathway in the late M to G1 phase via APC(CDH1). We also identified the critical region for destruction located in the C-terminus of MgcRacGAP, AA537-570, which is necessary and sufficient for CDH1-mediated MgcRacGAP destruction. In addition, we identified a PEST domain-like structure with charged residues in MgcRacGAP and implicate it in effective ubiquitination of MgcRacGAP. CONCLUSIONS/SIGNIFICANCE: Our findings not only reveal a novel mechanism for controlling the expression level of MgcRacGAP but also identify a new target of APC(CDH1). Moreover our results identify a C-terminal region AA537-570 of MgcRacGAP as its degron.


Cadherins/metabolism , Cdh1 Proteins/metabolism , GTPase-Activating Proteins/metabolism , Animals , Antigens, CD , Blotting, Western , Cadherins/genetics , Cdh1 Proteins/genetics , Cell Cycle , Cell Line , Flow Cytometry , G1 Phase/genetics , G1 Phase/physiology , GTPase-Activating Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Mice , NIH 3T3 Cells , Protein Binding , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/physiology
20.
Int J Hematol ; 96(3): 334-41, 2012 Sep.
Article En | MEDLINE | ID: mdl-22843432

MicroRNA-125b-1 (miR-125b-1) is a target of the chromosomal translocations t(11;14)(q24;q32) and t(2;11)(p21;q23), which are found in human B-lymphoid and myeloid malignancies, respectively. These translocations result in overexpression of mature miR-125b, consisting of 22 nucleotides. To analyze the role of miR-125b-1 in leukemogenesis, we created a bone marrow transplantation model using a retrovirus vector containing GFP expression elements. Sole transduction of miR-125b-1 into bone marrow cells resulted in expansion of hematopoietic cells expressing GFP. Compared with cells lacking GFP expression, we observed that GFP(+)/CD11b(+) or GFP(+)/Gr(-)1(+) cells were increased in the bone marrow and spleen. Although previous studies reported sole induction of miR-125b-induced leukemia, we did not find leukemic transformation in our model. Transduction of miR-125b-1 did accelerate myeloid tumors induced by a C-terminal mutant of CAAT-enhancer binding protein (C/EBPα-C(m)), a class II-like mutation. As miR-125b has been shown to hasten the development of leukemia in a BCR/ABL-transduced animal model, our present results support the conclusion that overexpression of miR-125b cooperates with other genetic alterations in the pathogenesis of myeloid malignancies.


CCAAT-Enhancer-Binding Protein-alpha/genetics , Leukemia, Myeloid/genetics , MicroRNAs/genetics , Mutation , Animals , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Disease Progression , Gene Expression , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Mice , Myeloid Cells/pathology , Transduction, Genetic
...