Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Genes Dev ; 37(13-14): 605-620, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37536952

ABSTRACT

The transcription factor RUNX1 is mutated in familial platelet disorder with associated myeloid malignancy (FPDMM) and in sporadic myelodysplastic syndrome and leukemia. RUNX1 was shown to regulate inflammation in multiple cell types. Here we show that RUNX1 is required in granulocyte-monocyte progenitors (GMPs) to epigenetically repress two inflammatory signaling pathways in neutrophils: Toll-like receptor 4 (TLR4) and type I interferon (IFN) signaling. RUNX1 loss in GMPs augments neutrophils' inflammatory response to the TLR4 ligand lipopolysaccharide through increased expression of the TLR4 coreceptor CD14. RUNX1 binds Cd14 and other genes encoding proteins in the TLR4 and type I IFN signaling pathways whose chromatin accessibility increases when RUNX1 is deleted. Transcription factor footprints for the effectors of type I IFN signaling-the signal transducer and activator of transcription (STAT1::STAT2) and interferon regulatory factors (IRFs)-were enriched in chromatin that gained accessibility in both GMPs and neutrophils when RUNX1 was lost. STAT1::STAT2 and IRF motifs were also enriched in the chromatin of retrotransposons that were derepressed in RUNX1-deficient GMPs and neutrophils. We conclude that a major direct effect of RUNX1 loss in GMPs is the derepression of type I IFN and TLR4 signaling, resulting in a state of fixed maladaptive innate immunity.


Subject(s)
Neutrophils , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Monocytes/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Cytokines/metabolism , Chromatin/metabolism , STAT1 Transcription Factor/metabolism
2.
Proc Natl Acad Sci U S A ; 117(38): 23626-23635, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32883883

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) formation and lineage differentiation involve gene expression programs orchestrated by transcription factors and epigenetic regulators. Genetic disruption of the chromatin remodeler chromodomain-helicase-DNA-binding protein 7 (CHD7) expanded phenotypic HSPCs, erythroid, and myeloid lineages in zebrafish and mouse embryos. CHD7 acts to suppress hematopoietic differentiation. Binding motifs for RUNX and other hematopoietic transcription factors are enriched at sites occupied by CHD7, and decreased RUNX1 occupancy correlated with loss of CHD7 localization. CHD7 physically interacts with RUNX1 and suppresses RUNX1-induced expansion of HSPCs during development through modulation of RUNX1 activity. Consequently, the RUNX1:CHD7 axis provides proper timing and function of HSPCs as they emerge during hematopoietic development or mature in adults, representing a distinct and evolutionarily conserved control mechanism to ensure accurate hematopoietic lineage differentiation.


Subject(s)
Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins , Hematopoiesis , Animals , Cell Differentiation , Cell Line , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Hematopoietic Stem Cells , Humans , Male , Mice , Spleen/cytology , Zebrafish
3.
Blood ; 136(21): 2373-2385, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32929473

ABSTRACT

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia, which generates a CBFB-MYH11 fusion gene. It is generally considered that CBFß-SMMHC, the fusion protein encoded by CBFB-MYH11, is a dominant negative repressor of RUNX1. However, recent findings challenge the RUNX1-repression model for CBFß-SMMHC-mediated leukemogenesis. To definitively address the role of Runx1 in CBFB-MYH11-induced leukemia, we crossed conditional Runx1 knockout mice (Runx1f/f) with conditional Cbfb-MYH11 knockin mice (Cbfb+/56M). On Mx1-Cre activation in hematopoietic cells induced by poly (I:C) injection, all Mx1-CreCbfb+/56M mice developed leukemia in 5 months, whereas no leukemia developed in Runx1f/fMx1-CreCbfb+/56M mice, and this effect was cell autonomous. Importantly, the abnormal myeloid progenitors (AMPs), a leukemia-initiating cell population induced by Cbfb-MYH11 in the bone marrow, decreased and disappeared in Runx1f/fMx1-CreCbfb+/56M mice. RNA-seq analysis of AMP cells showed that genes associated with proliferation, differentiation blockage, and leukemia initiation were differentially expressed between Mx1-CreCbfb+/56M and Runx1f/fMx1-CreCbfb+/56M mice. In addition, with the chromatin immunocleavage sequencing assay, we observed a significant enrichment of RUNX1/CBFß-SMMHC target genes in Runx1f/fMx1-CreCbfb+/56M cells, especially among downregulated genes, suggesting that RUNX1 and CBFß-SMMHC mainly function together as activators of gene expression through direct target gene binding. These data indicate that Runx1 is indispensable for Cbfb-MYH11-induced leukemogenesis by working together with CBFß-SMMHC to regulate critical genes associated with the generation of a functional AMP population.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation, Leukemic , Leukemia, Experimental/genetics , Myeloid Cells/metabolism , Neoplasm Proteins/physiology , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/physiology , Transcriptional Activation , Animals , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Knock-In Techniques , Humans , Leukemia, Experimental/etiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Neoplastic Stem Cells/cytology , Oncogene Proteins, Fusion/genetics , Poly I-C/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA-Seq , Single-Cell Analysis
4.
Hum Mol Genet ; 28(24): 4173-4185, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31691804

ABSTRACT

DHX15, a DEAH box containing RNA helicase, is a splicing factor required for the last step of splicing. Recent studies identified a recurrent mutational hotspot, R222G, in DHX15 in ∼ 6% of acute myeloid leukemia (AML) patients that carry the fusion protein RUNX1-RUNX1T1 produced by t (8;21) (q22;q22). Studies using yeast mutants showed that substitution of G for the residue equivalent to R222 leads to loss of its helicase function, suggesting that it is a loss-of-function mutation. To elucidate the role of DHX15 during development, we established the first vertebrate knockout model with CRISPR/Cas9 in zebrafish. Our data showed that dhx15 expression is enriched in the brain, eyes, pectoral fin primordia, liver and intestinal bulb during embryonic development. Dhx15 deficiency leads to pleiotropic morphological phenotypes in homozygous mutant embryos starting at 3 days post fertilization (dpf) that result in lethality by 7 dpf, revealing an essential role during embryonic development. RNA-seq analysis suggested important roles of Dhx15 in chromatin and nucleosome assembly and regulation of the Mdm2-p53 pathway. Interestingly, exons corresponding to the alternate transcriptional start sites for tp53 and mdm2 were preferentially expressed in the mutant embryos, leading to significant upregulation of their alternate isoforms, Δ113p53 (orthologous to Δ133p53 isoform in human) and mdm2-P2 (isoform using distal promoter P2), respectively. We speculate that these alterations in the Mdm2-p53 pathway contribute to the development of AML in patients with t(8;21) and somatically mutated DHX15.


Subject(s)
Proto-Oncogene Proteins c-mdm2/genetics , RNA Helicases/genetics , Tumor Suppressor Protein p53/genetics , Zebrafish Proteins/genetics , Alternative Splicing , Animals , Animals, Genetically Modified , Humans , Promoter Regions, Genetic , Protein Isoforms , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Helicases/metabolism , RNA Splice Sites , RNA Splicing , RNA Splicing Factors/genetics , Transcription Initiation Site , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Zebrafish , Zebrafish Proteins/metabolism
5.
Immunity ; 34(5): 755-68, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21600797

ABSTRACT

Missense mutations in the C-terminal B30.2 domain of pyrin cause familial Mediterranean fever (FMF), the most common Mendelian autoinflammatory disease. However, it remains controversial as to whether FMF is due to the loss of an inhibitor of inflammation or to the activity of a proinflammatory molecule. We generated both pyrin-deficient mice and "knockin" mice harboring mutant human B30.2 domains. Homozygous knockin, but not pyrin-deficient, mice exhibited spontaneous bone marrow-dependent inflammation similar to but more severe than human FMF. Caspase-1 was constitutively activated in knockin macrophages and active IL-1ß was secreted when stimulated with lipopolysaccharide alone, which is also observed in FMF patients. The inflammatory phenotype of knockin mice was completely ablated by crossing with IL-1 receptor-deficient or adaptor molecule ASC-deficient mice, but not NLRP3-deficient mice. Thus, our data provide evidence for an ASC-dependent NLRP3-independent inflammasome in which gain-of-function pyrin mutations cause autoinflammatory disease.


Subject(s)
Autoimmune Diseases/immunology , Carrier Proteins/immunology , Cytoskeletal Proteins/genetics , Mutation , Adaptive Immunity , Animals , Autoimmune Diseases/pathology , Cells, Cultured , Female , Humans , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Pyrin , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/immunology
6.
Blood ; 130(22): 2431-2442, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29018080

ABSTRACT

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia, which generates a CBFB-MYH11 fusion gene. Previous studies showed that the interaction between CBFß-smooth muscle myosin heavy chain (SMMHC; encoded by CBFB-MYH11) and RUNX1 plays a critical role in the pathogenesis of this leukemia. Recently, it was shown that chromodomain helicase DNA-binding protein-7 (CHD7) interacts with RUNX1 and suppresses RUNX1-induced expansion of hematopoietic stem and progenitor cells. These results suggest that CHD7 is also critical for CBFB-MYH11-induced leukemogenesis. To test this hypothesis, we generated Chd7f/fMx1-CreCbfb+/56M mice, which expressed the Cbfb-MYH11 fusion gene and deactivated Chd7 in hematopoietic cells upon inducing Cre with polyinosinic-polycytidylic acid. The Lin-Sca1-c-Kit+ (LK) population was significantly lower in Chd7f/fMx1-CreCbfb+/56M mice than in Mx1-CreCbfb+/56M mice. In addition, there were fewer 5-bromo-2'-deoxyuridine-positive cells in the LK population in Chd7f/fMx1-CreCbfb+/56M mice, and genes associated with cell cycle, cell growth, and proliferation were differentially expressed between Chd7f/fMx1-CreCbfb+/56M and Mx1-CreCbfb+/56M leukemic cells. In vitro studies showed that CHD7 interacted with CBFß-SMMHC through RUNX1 and that CHD7 enhanced transcriptional activity of RUNX1 and CBFß-SMMHC on Csf1r, a RUNX1 target gene. Moreover, RNA sequencing of c-Kit+ cells showed that CHD7 functions mostly through altering the expression of RUNX1 target genes. Most importantly, Chd7 deficiency delayed Cbfb-MYH11-induced leukemia in both primary and transplanted mice. These data indicate that Chd7 is important for Cbfb-MYH11-induced leukemogenesis by facilitating RUNX1 regulation of transcription and cellular proliferation.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cell Cycle , Cell Proliferation , Gene Deletion , Humans , Leukemia/pathology , Mice , Mice, Knockout
7.
Blood ; 124(1): 70-8, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24850758

ABSTRACT

CBFß and RUNX1 form a DNA-binding heterodimer and are both required for hematopoietic stem cell (HSC) generation in mice. However, the exact role of CBFß in the production of HSCs remains unclear. Here, we generated and characterized 2 zebrafish cbfb null mutants. The cbfb(-/-) embryos underwent primitive hematopoiesis and developed transient erythromyeloid progenitors, but they lacked definitive hematopoiesis. Unlike runx1 mutants, in which HSCs are not formed, nascent, runx1(+)/c-myb(+) HSCs were formed in cbfb(-/-) embryos. However, the nascent HSCs were not released from the aorta-gonad-mesonephros (AGM) region, as evidenced by the accumulation of runx1(+) cells in the AGM that could not enter circulation. Moreover, wild-type embryos treated with an inhibitor of RUNX1-CBFß interaction, Ro5-3335, phenocopied the hematopoietic defects in cbfb(-/-) mutants, rather than those in runx1(-/-) mutants. Finally, we found that cbfb was downstream of the Notch pathway during HSC development. Our data suggest that runx1 and cbfb are required at 2 different steps during early HSC development. CBFß is not required for nascent HSC emergence but is required for the release of HSCs from AGM into circulation. Our results also indicate that RUNX1 can drive the emergence of nascent HSCs in the AGM without its heterodimeric partner CBFß.


Subject(s)
CCAAT-Binding Factor/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Zebrafish Proteins/genetics , Animals , CCAAT-Binding Factor/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Knockout Techniques , In Situ Hybridization , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish , Zebrafish Proteins/metabolism
8.
Blood ; 124(12): 1926-30, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25114263

ABSTRACT

Familial platelet disorder with predisposition to acute myeloid leukemia (FPD/AML) is an autosomal dominant disease of the hematopoietic system that is caused by heterozygous mutations in RUNX1. FPD/AML patients have a bleeding disorder characterized by thrombocytopenia with reduced platelet numbers and functions, and a tendency to develop AML. No suitable animal models exist for FPD/AML, as Runx11/2 mice and zebra fish do not develop bleeding disorders or leukemia. Here we derived induced pluripotent stem cells (iPSCs) from 2 patients in a family with FPD/AML, and found that the FPD iPSCs display defects in megakaryocytic differentiation in vitro. We corrected the RUNX1 mutation in 1 FPD iPSC line through gene targeting, which led to normalization of megakaryopoiesis of the iPSCs in culture. Our results demonstrate successful in vitro modeling of FPD with patient-specific iPSCs and confirm that RUNX1 mutations are responsible for megakaryopoietic defects in FPD patients.


Subject(s)
Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/therapy , Blood Platelet Disorders/genetics , Blood Platelet Disorders/therapy , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mutation, Missense , Targeted Gene Repair/methods , Animals , Blood Coagulation Disorders, Inherited/pathology , Blood Platelet Disorders/pathology , Core Binding Factor Alpha 2 Subunit/chemistry , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/transplantation , Leukemia, Myeloid, Acute/pathology , Mice , Thrombopoiesis/genetics
9.
Blood ; 121(4): 638-42, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23152542

ABSTRACT

The C-terminus of CBFß-SMMHC, the fusion protein produced by a chromosome 16 inversion in acute myeloid leukemia subtype M4Eo, contains domains for self-multimerization and transcriptional repression, both of which have been proposed to be important for leukemogenesis by CBFß-SMMHC. To test the role of the fusion protein's C-terminus in vivo, we generated knock-in mice expressing a C-terminally truncated CBFß-SMMHC (CBFß-SMMHCΔC95). Embryos with a single copy of CBFß-SMMHCΔC95 were viable and showed no defects in hematopoiesis, whereas embryos homozygous for the CBFß-SMMHCΔC95 allele had hematopoietic defects and died in mid-gestation, similar to embryos with a single-copy of the full-length CBFß-SMMHC. Importantly, unlike mice expressing full-length CBFß-SMMHC, none of the mice expressing CBFß-SMMHCΔC95 developed leukemia, even after treatment with a mutagen, although some of the older mice developed a nontransplantable myeloproliferative disease. Our data indicate that the CBFß-SMMHC's C-terminus is essential to induce embryonic hematopoietic defects and leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Hematopoiesis/genetics , Leukemia/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Gene Knock-In Techniques , Humans , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism
10.
Blood ; 119(22): 5239-49, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22493295

ABSTRACT

Proper cell fate choice in myelopoiesis is essential for generating correct numbers of distinct myeloid subsets manifesting a wide spectrum of subset-specific activities during development and adulthood. Studies have suggested that myeloid fate choice is primarily regulated by transcription factors; however, new intrinsic regulators and their underlying mechanisms remain to be elucidated. Zebrafish embryonic myelopoiesis gives rise to neutrophils and macrophages and represents a promising system to derive new regulatory mechanisms for myeloid fate decision in vertebrates. Here we present an in vivo study of cell fate specification during zebrafish embryonic myelopoiesis through characterization of the embryos with altered Pu.1, Runx1 activity alone, or their combinations. Genetic analysis shows that low and high Pu.1 activities determine embryonic neutrophilic granulocyte and macrophage fate, respectively. Inactivation and overexpression of Runx1 in zebrafish uncover Runx1 as a key embryonic myeloid fate determinant that favors neutrophil over macrophage fate. Runx1 is induced by high Pu.1 level and in turn transrepresses pu.1 expression, thus constituting a negative feedback loop that fashions a favorable Pu.1 level required for balanced fate commitment to neutrophils versus macrophages. Our findings define a Pu.1-Runx1 regulatory loop that governs the equilibrium between distinct myeloid fates by assuring an appropriate Pu.1 dosage.


Subject(s)
Core Binding Factor Alpha 2 Subunit/biosynthesis , Gene Expression Regulation, Developmental/physiology , Myelopoiesis/physiology , Proto-Oncogene Proteins/biosynthesis , Trans-Activators/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Macrophages/cytology , Macrophages/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
11.
Blood ; 119(6): 1511-21, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22160378

ABSTRACT

KIT mutations are the most common secondary mutations in inv(16) acute myeloid leukemia (AML) patients and are associated with poor prognosis. It is therefore important to verify that KIT mutations cooperate with CBFB-MYH11, the fusion gene generated by inv(16), for leukemogenesis. Here, we transduced wild-type and conditional Cbfb-MYH11 knockin (KI) mouse bone marrow (BM) cells with KIT D816V/Y mutations. KIT transduction caused massive BM Lin(-) cell death and fewer colonies in culture that were less severe in the KI cells. D816Y KIT but not wild-type KIT enhanced proliferation in Lin(-) cells and led to more mixed lineage colonies from transduced KI BM cells. Importantly, 60% and 80% of mice transplanted with KI BM cells expressing D816V or D816Y KIT, respectively, died from leukemia within 9 months, whereas no control mice died. Results from limiting dilution transplantations indicate higher frequencies of leukemia-initiating cells in the leukemia expressing mutated KIT. Signaling pathway analysis revealed that p44/42 MAPK and Stat3, but not AKT and Stat5, were strongly phosphorylated in the leukemia cells. Finally, leukemia cells carrying KIT D816 mutations were sensitive to the kinase inhibitor PKC412. Our data provide clear evidence for cooperation between mutated KIT and CBFB-MYH11 during leukemogenesis.


Subject(s)
Leukemia/genetics , Mutation , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Blotting, Western , Bone Marrow Transplantation , Cell Survival/drug effects , Cells, Cultured , Disease Progression , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukemia/metabolism , Leukemia/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
12.
Nat Genet ; 32(4): 639-44, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12434156

ABSTRACT

Runx2 (runt-related transcription factor 2, also known as Cbfa1, Osf2 and AML3) is essential for bone development in mice, and mutations in RUNX2 are found in 65-80% of individuals with cleidocranial dysplasia. Although all Runx family members can interact with Cbfbeta (core-binding factor b, encoded by Cbfb), a role for Cbfbeta in bone development has not been demonstrated owing to lethality in Cbfb(-/-) mouse embryos at 12.5 days post coitum (d.p.c.) from hemorrhages and lack of definitive hematopoiesis. Using a 'knock-in' strategy, we generated mouse embryonic stem (ES) cells that express Cbfb fused in-frame to a cDNA encoding green fluorescent protein (GFP). Cbfb(+/GFP) mice had normal life spans and appeared normal, but Cbfb(GFP/GFP) pups died within the first day after birth. The Cbfb(GFP/GFP) mice exhibited a delay in endochondral and intramembranous ossification as well as in chondrocyte differentiation, similar to but less severe than delays observed in Runx2(-/-) mice. We demonstrate that Cbfbeta is expressed in developing bone and forms a functional interaction with Runx2, and that Cbfb(GFP) is a hypomorphic allele. The fusion allele maintains sufficient function in hematopoietic cells to bypass the early embryonic lethality, and identifies a new role for Cbfb in bone development. Our findings raise the possibility that mutations in CBFB may be responsible for some cases of cleidocranial dysplasia that are not linked to mutations in RUNX2.


Subject(s)
DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Neoplasm Proteins , Osteogenesis , Transcription Factors/metabolism , Transcription Factors/physiology , Alleles , Animals , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/physiology , Core Binding Factor Alpha 1 Subunit , Core Binding Factor alpha Subunits , Core Binding Factor beta Subunit , Core Binding Factors , DNA-Binding Proteins/genetics , Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental , Genes, Lethal , Green Fluorescent Proteins , HeLa Cells , Homozygote , Humans , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Phenotype , Pluripotent Stem Cells/physiology , Recombinant Fusion Proteins/physiology , Transcription Factor AP-2 , Transcription Factors/genetics , Tumor Cells, Cultured
13.
Dev Dyn ; 241(5): 879-89, 2012 May.
Article in English | MEDLINE | ID: mdl-22411201

ABSTRACT

BACKGROUND: Vertebrate hematopoiesis is a complex developmental process that is controlled by genes in diverse pathways. To identify novel genes involved in early hematopoiesis, we conducted an ENU (N-ethyl-N-nitrosourea) mutagenesis screen in zebrafish. The mummy (mmy) line was investigated because of its multiple hematopoietic defects. RESULTS: Homozygous mmy embryos lacked circulating blood cell types and were dead by 30 hr post-fertilization (hpf). The mmy mutants did not express myeloid markers and had significantly decreased expression of progenitor and erythroid markers in primitive hematopoiesis. Through positional cloning, we identified a truncation mutation in dhx8 in the mmy fish. dhx8 is the zebrafish ortholog of the yeast splicing factor prp22, which is a DEAH-box RNA helicase. mmy mutants had splicing defects in many genes, including several hematopoietic genes. mmy embryos also showed cell division defects as characterized by disorganized mitotic spindles and formation of multiple spindle poles in mitotic cells. These cell division defects were confirmed by DHX8 knockdown in HeLa cells. CONCLUSIONS: Together, our results confirm that dhx8 is involved in mRNA splicing and suggest that it is also important for cell division during mitosis. This is the first vertebrate model for dhx8, whose function is essential for primitive hematopoiesis in developing embryos.


Subject(s)
Cell Division/genetics , DEAD-box RNA Helicases/genetics , Hematopoiesis/genetics , RNA Splicing/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Cell Differentiation/genetics , DEAD-box RNA Helicases/metabolism , Embryo, Nonmammalian/metabolism , Hematopoietic System/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
14.
Blood Adv ; 7(23): 7304-7318, 2023 12 12.
Article in English | MEDLINE | ID: mdl-37756546

ABSTRACT

Germ line mutations in the RUNX1 gene cause familial platelet disorder (FPD), an inherited disease associated with lifetime risk to hematopoietic malignancies (HM). Patients with FPD frequently show clonal expansion of premalignant cells preceding HM onset. Despite the extensive studies on the role of RUNX1 in hematopoiesis, its function in the premalignant bone marrow (BM) is not well-understood. Here, we characterized the hematopoietic progenitor compartments using a mouse strain carrying an FPD-associated mutation, Runx1R188Q. Immunophenotypic analysis showed an increase in the number of hematopoietic stem and progenitor cells (HSPCs) in the Runx1R188Q/+ mice. However, the comparison of Sca-1 and CD86 markers suggested that Sca-1 expression may result from systemic inflammation. Cytokine profiling confirmed the dysregulation of interferon-response cytokines in the BM. Furthermore, the expression of CD48, another inflammation-response protein, was also increased in Runx1R188Q/+ HSPCs. The DNA-damage response activity of Runx1R188Q/+ hematopoietic progenitor cells was defective in vitro, suggesting that Runx1R188Q may promote genomic instability. The differentiation of long-term repopulating HSCs was reduced in Runx1R188Q/+ recipient mice. Furthermore, we found that Runx1R188Q/+ HSPCs outcompete their wild-type counterparts in bidirectional repopulation assays, and that the genetic makeup of recipient mice did not significantly affect the clonal dynamics under this setting. Finally, we demonstrate that Runx1R188Q predisposes to HM in cooperation with somatic mutations found in FPDHM, using 3 mouse models. These studies establish a novel murine FPDHM model and demonstrate that germ line Runx1 mutations induce a premalignant phenotype marked by BM inflammation, selective expansion capacity, defective DNA-damage response, and predisposition to HM.


Subject(s)
Blood Platelet Disorders , Hematologic Neoplasms , Animals , Mice , Humans , Germ-Line Mutation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Disease Susceptibility , Blood Platelet Disorders/genetics , Inflammation/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/complications , DNA
15.
bioRxiv ; 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36747636

ABSTRACT

The transcription factor RUNX1 is mutated in familial platelet disorder with associated myeloid malignancies (FPDMM) and in sporadic myelodysplastic syndrome and leukemia. RUNX1 regulates inflammation in multiple cell types. Here we show that RUNX1 is required in granulocyte-monocyte progenitors (GMPs) to restrict the inflammatory response of neutrophils to toll-like receptor 4 (TLR4) signaling. Loss of RUNX1 in GMPs increased the TLR4 coreceptor CD14 on neutrophils, which contributed to neutrophils’ increased inflammatory cytokine production in response to the TLR4 ligand lipopolysaccharide. RUNX1 loss increased the chromatin accessibility of retrotransposons in GMPs and neutrophils and induced a type I interferon signature characterized by enriched footprints for signal transducer and activator of transcription (STAT1::STAT2) and interferon regulatory factors (IRF) in opened chromatin, and increased expression of interferon-stimulated genes. The overproduction of inflammatory cytokines by neutrophils was reversed by inhibitors of type I IFN signaling. We conclude that RUNX1 restrains the chromatin accessibility of retrotransposons in GMPs and neutrophils, and that loss of RUNX1 increases proinflammatory cytokine production by elevating tonic type I interferon signaling.

16.
Blood ; 115(7): 1433-43, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20007544

ABSTRACT

It is known that CBFB-MYH11, the fusion gene generated by inversion of chromosome 16 in human acute myeloid leukemia, is causative for oncogenic transformation. However, the mechanism by which CBFB-MYH11 initiates leukemogenesis is not clear. Previously published reports showed that CBFB-MYH11 dominantly inhibits RUNX1 and CBFB, and such inhibition has been suggested as the mechanism for leukemogenesis. Here we show that Cbfb-MYH11 caused Cbfb/Runx1 repression-independent defects in both primitive and definitive hematopoiesis. During primitive hematopoiesis, Cbfb-MYH11 delayed differentiation characterized by sustained expression of Gata2, Il1rl1, and Csf2rb, a phenotype not found in Cbfb and Runx1 knockout mice. Expression of Cbfb-MYH11 in the bone marrow induced the accumulation of abnormal progenitor-like cells expressing Csf2rb in preleukemic mice. The expression of all 3 genes was detected in most human and murine CBFB-MYH11(+) leukemia samples. Interestingly, Cbfb-MYH11(+) preleukemic progenitors and leukemia-initiating cells did not express Csf2rb, although the majority of leukemia cells in our Cbfb-MYH11 knockin mice were Csf2rb(+). Therefore Csf2rb can be used as a negative selection marker to enrich preleukemic progenitor cells and leukemia-initiating cells from Cbfb-MYH11 mice. These results suggest that Cbfb/Runx1 repression-independent activities contribute to leukemogenesis by Cbfb-MYH11.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Cytokine Receptor Common beta Subunit/metabolism , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Apoptosis/physiology , Biomarkers , Cell Differentiation/physiology , Cell Division/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Cytokine Receptor Common beta Subunit/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Leukemic , Hematopoiesis/physiology , Hematopoietic Stem Cells/pathology , Humans , Interleukin-1 Receptor-Like 1 Protein , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins, Fusion/genetics , Phenotype , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Up-Regulation/physiology
17.
Blood ; 115(14): 2806-9, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20154212

ABSTRACT

Runx1 is required for the emergence of hematopoietic stem cells (HSCs) from hemogenic endothelium during embryogenesis. However, its role in the generation and maintenance of HSCs during adult hematopoiesis remains uncertain. Here, we present analysis of a zebrafish mutant line carrying a truncation mutation, W84X, in runx1. The runx1(W84X/W84X) embryos showed blockage in the initiation of definitive hematopoiesis, but some embryos were able to recover from a larval "bloodless" phase and develop to fertile adults with multilineage hematopoiesis. Using cd41-green fluorescent protein transgenic zebrafish and lineage tracing, we demonstrated that the runx1(W84X/W84X) embryos developed cd41(+) HSCs in the aorta-gonad-mesonephros region, which later migrated to the kidney, the site of adult hematopoiesis. Overall, our data suggest that in zebrafish adult HSCs can be formed without an intact runx1.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Mutation , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Core Binding Factor Alpha 2 Subunit/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Hematopoietic Stem Cells/cytology , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Platelet Membrane Glycoprotein IIb/biosynthesis , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
19.
Blood ; 114(25): 5162-72, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19843882

ABSTRACT

The transcription factor Gata1 is required for the development of erythrocytes and megakaryocytes. Previous studies with a complementation rescue approach showed that the zinc finger domains are required for both primitive and definitive hematopoiesis. Here we report a novel zebrafish gata1 mutant with an N-ethyl-N-nitrosourea-induced point mutation in the C-finger (gata1(T301K)). The Gata1 protein with this mutation bound to its DNA target sequence with reduced affinity and transactivated inefficiently in a reporter assay. gata1(T301K/T301K) fish had a decreased number of erythrocytes during primitive hematopoiesis but normal adult hematopoiesis. We crossed the gata1(T301K/T301K) fish with those carrying the R339X mutation, also known as vlad tepes (vlt), which abolishes DNA binding and transactivation activities. As we reported previously, gata1(vlt/vlt) embryos were "bloodless" and died approximately 11 to 15 days after fertilization. Interestingly, the gata1(T301K/vlt) fish had nearly a complete block of primitive hematopoiesis, but they resumed hematopoiesis between 7 and 14 days after fertilization and grew to phenotypically normal fish with normal adult hematopoiesis. Our findings suggest that the impact of Gata1 on hematopoiesis correlates with its DNA-binding ability and that primitive hematopoiesis is more sensitive to reduction in Gata1 function than definitive hematopoiesis.


Subject(s)
DNA/metabolism , GATA1 Transcription Factor/metabolism , Hematopoiesis , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , DNA/chemistry , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Flow Cytometry , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental , In Situ Hybridization , Male , Models, Molecular , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcriptional Activation , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
20.
J Cell Biochem ; 110(5): 1039-45, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20589720

ABSTRACT

The core binding factor (CBF) acute myeloid leukemias (AMLs) are a prognostically distinct subgroup that includes patients with the inv(16) and t(8:21) chromosomal rearrangements. Both of these rearrangements result in the formation of fusion proteins, CBFB-MYH11 and AML1-ETO, respectively, that involve members of the CBF family of transcription factors. It has been proposed that both of these fusion proteins function primarily by dominantly repressing normal CBF transcription. However, recent reports have indicted that additional, CBF-repression independent activities may be equally important during leukemogenesis. This article will focus on these recent advances.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Leukemia/genetics , Oncogene Proteins, Fusion/genetics , Acute Disease , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid/genetics , Models, Genetic , RUNX1 Translocation Partner 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL