Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Blood ; 137(19): 2662-2675, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33569577

ABSTRACT

Patients with familial platelet disorder with a predisposition to myeloid malignancy (FPDMM) harbor germline monoallelic mutations in a key hematopoietic transcription factor, RUNX-1. Previous studies of FPDMM have focused on megakaryocyte (Mk) differentiation and platelet production and signaling. However, the effects of RUNX-1 haploinsufficiency on hematopoietic progenitor cells (HPCs) and subsequent megakaryopoiesis remains incomplete. We studied induced pluripotent stem cell (iPSC)-derived HPCs (iHPCs) and Mks (iMks) from both patient-derived lines and a wild-type (WT) line modified to be RUNX-1 haploinsufficient (RUNX-1+/-), each compared with their isogenic WT control. All RUNX-1+/- lines showed decreased iMk yield and depletion of an Mk-biased iHPC subpopulation. To investigate global and local gene expression changes underlying this iHPC shift, single-cell RNA sequencing was performed on sorted FPDMM and control iHPCs. We defined several cell subpopulations in the Mk-biased iHPCs. Analyses of gene sets upregulated in FPDMM iHPCs indicated enrichment for response to stress, regulation of signal transduction, and immune signaling-related gene sets. Immunoblot analyses in FPDMM iMks were consistent with these findings, but also identified augmented baseline c-Jun N-terminal kinase (JNK) phosphorylation, known to be activated by transforming growth factor-ß1 (TGF-ß1) and cellular stressors. These findings were confirmed in adult human CD34+-derived stem and progenitor cells (HSPCs) transduced with lentiviral RUNX1 short hairpin RNA to mimic RUNX-1+/-. In both iHPCs and CD34+-derived HSPCs, targeted inhibitors of JNK and TGF-ß1 pathways corrected the megakaryopoietic defect. We propose that such intervention may correct the thrombocytopenia in patients with FPDMM.


Subject(s)
Core Binding Factor Alpha 2 Subunit/deficiency , Hematopoietic Stem Cells/pathology , Megakaryocytes/pathology , Neoplastic Syndromes, Hereditary/pathology , Adult , Base Sequence , Core Binding Factor Alpha 2 Subunit/genetics , Flow Cytometry , Haploinsufficiency , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , MAP Kinase Signaling System , Neoplastic Syndromes, Hereditary/genetics , Platelet Glycoprotein GPIb-IX Complex/analysis , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Signal Transduction , Single-Cell Analysis , Thrombopoiesis , Transforming Growth Factor beta1/physiology
2.
Blood ; 137(19): 2579-2584, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33821975

ABSTRACT

The core binding factor composed of CBFß and RUNX subunits plays a critical role in most hematopoietic lineages and is deregulated in acute myeloid leukemia (AML). The fusion oncogene CBFß-SMMHC expressed in AML with the chromosome inversion inv(16)(p13q22) acts as a driver oncogene in hematopoietic stem cells and induces AML. This review focuses on novel insights regarding the molecular mechanisms involved in CBFß-SMMHC-driven leukemogenesis and recent advances in therapeutic approaches to target CBFß-SMMHC in inv(16) AML.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosome Inversion , Chromosomes, Human, Pair 16/genetics , Core Binding Factor beta Subunit/genetics , Immunotherapy/methods , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy , Myosin Heavy Chains/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosomes, Human, Pair 16/ultrastructure , Combined Modality Therapy , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/physiology , Forecasting , Gemtuzumab/therapeutic use , Gene Expression Regulation, Leukemic , Gene Knock-In Techniques , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
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.
Circulation ; 137(1): 57-70, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29030345

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is a leading cause of heart failure and death worldwide. Preservation of contractile function and protection against adverse changes in ventricular architecture (cardiac remodeling) are key factors to limiting progression of this condition to heart failure. Consequently, new therapeutic targets are urgently required to achieve this aim. Expression of the Runx1 transcription factor is increased in adult cardiomyocytes after MI; however, the functional role of Runx1 in the heart is unknown. METHODS: To address this question, we have generated a novel tamoxifen-inducible cardiomyocyte-specific Runx1-deficient mouse. Mice were subjected to MI by means of coronary artery ligation. Cardiac remodeling and contractile function were assessed extensively at the whole-heart, cardiomyocyte, and molecular levels. RESULTS: Runx1-deficient mice were protected against adverse cardiac remodeling after MI, maintaining ventricular wall thickness and contractile function. Furthermore, these mice lacked eccentric hypertrophy, and their cardiomyocytes exhibited markedly improved calcium handling. At the mechanistic level, these effects were achieved through increased phosphorylation of phospholamban by protein kinase A and relief of sarco/endoplasmic reticulum Ca2+-ATPase inhibition. Enhanced sarco/endoplasmic reticulum Ca2+-ATPase activity in Runx1-deficient mice increased sarcoplasmic reticulum calcium content and sarcoplasmic reticulum-mediated calcium release, preserving cardiomyocyte contraction after MI. CONCLUSIONS: Our data identified Runx1 as a novel therapeutic target with translational potential to counteract the effects of adverse cardiac remodeling, thereby improving survival and quality of life among patients with MI.


Subject(s)
Core Binding Factor Alpha 2 Subunit/deficiency , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Calcium Signaling , Calcium-Binding Proteins/metabolism , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Phosphorylation , Rabbits , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Time Factors
5.
Am J Respir Cell Mol Biol ; 57(2): 174-183, 2017 08.
Article in English | MEDLINE | ID: mdl-28314106

ABSTRACT

Runt-related transcription factor 1 (RUNX1), a transcription factor expressed in multiple organs, plays important roles in embryonic development and hematopoiesis. Although RUNX1 is highly expressed in pulmonary tissues, its roles in lung function and homeostasis are unknown. We sought to assess the role of RUNX1 in lung development and inflammation after LPS challenge. Expression of RUNX1 was assessed in the developing and postnatal lung. RUNX1 was conditionally deleted in pulmonary epithelial cells. Pulmonary maturation was evaluated in the developing and postnatal lung, and lung inflammation was investigated in adult mice after LPS challenge. Interactions between RUNX1 and inflammatory signaling via NF-κB-IkB kinase ß were assessed in vitro. RUNX1 was expressed in both mesenchymal and epithelial compartments of the developing and postnatal lung. The RUNX1 gene was efficiently deleted from respiratory epithelial cells producing Runx1∆/∆ mice. Although lung maturation was delayed, Runx1∆/∆ mice survived postnatally and subsequent growth and maturation of the lung proceeded normally. Increased respiratory distress, inflammation, and proinflammatory cytokines were observed in the Runx1-deleted mice after pulmonary LPS exposure. RUNX1 deletion was associated with the activation of NF-κB in respiratory epithelial cells. RUNX1 was required for the suppression of NF-κB signaling pathway via inhibition of IkB kinase ß in in vitro studies. RUNX1 plays a critical role in the lung inflammation after LPS-induced injury.


Subject(s)
Acute Lung Injury/metabolism , Core Binding Factor Alpha 2 Subunit/physiology , NF-kappa B/metabolism , Signal Transduction , Acute Lung Injury/chemically induced , Alveolar Epithelial Cells/metabolism , Animals , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Endotoxins/toxicity , Gene Expression Regulation, Developmental , I-kappa B Kinase/metabolism , Inflammation , Lung/embryology , Lung/growth & development , Lung/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Specific Pathogen-Free Organisms
6.
Development ; 141(17): 3319-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25139854

ABSTRACT

Haematopoiesis in adult animals is maintained by haematopoietic stem cells (HSCs), which self-renew and can give rise to all blood cell lineages. The AGM region is an important intra-embryonic site of HSC development and a wealth of evidence indicates that HSCs emerge from the endothelium of the embryonic dorsal aorta and extra-embryonic large arteries. This, however, is a stepwise process that occurs through sequential upregulation of CD41 and CD45 followed by emergence of fully functional definitive HSCs. Although largely dispensable at later stages, the Runx1 transcription factor is crucially important during developmental maturation of HSCs; however, exact points of crucial involvement of Runx1 in this multi-step developmental maturation process remain unclear. Here, we have investigated requirements for Runx1 using a conditional reversible knockout strategy. We report that Runx1 deficiency does not preclude formation of VE-cad+CD45-CD41+ cells, which are phenotypically equivalent to precursors of definitive HSCs (pre-HSC Type I) but blocks transition to the subsequent CD45+ stage (pre-HSC Type II). These data emphasise that developmental progression of HSCs during a very short period of time is regulated by precise stage-specific molecular mechanisms.


Subject(s)
Cell Lineage , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Platelet Membrane Glycoprotein IIb/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/deficiency , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Nature ; 464(7285): 116-20, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20154729

ABSTRACT

Haematopoietic stem cells (HSCs), responsible for blood production in the adult mouse, are first detected in the dorsal aorta starting at embryonic day 10.5 (E10.5). Immunohistological analysis of fixed embryo sections has revealed the presence of haematopoietic cell clusters attached to the aortic endothelium where HSCs might localize. The origin of HSCs has long been controversial and several candidates of the direct HSC precursors have been proposed (for review see ref. 7), including a specialized endothelial cell population with a haemogenic potential. Such cells have been described both in vitro in the embryonic stem cell (ESC) culture system and retrospectively in vivo by endothelial lineage tracing and conditional deletion experiments. Whether the transition from haemogenic endothelium to HSC actually occurs in the mouse embryonic aorta is still unclear and requires direct and real-time in vivo observation. To address this issue we used time-lapse confocal imaging and a new dissection procedure to visualize the deeply located aorta. Here we show the dynamic de novo emergence of phenotypically defined HSCs (Sca1(+), c-kit(+), CD41(+)) directly from ventral aortic haemogenic endothelial cells.


Subject(s)
Aorta/cytology , Cell Differentiation , Cell Lineage , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Animals , Aorta/embryology , Aorta/surgery , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Dissection , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Endothelium, Vascular/embryology , Female , Male , Mice , Microscopy, Confocal , Phenotype , Pregnancy
8.
Nature ; 464(7285): 112-5, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20154732

ABSTRACT

The ontogeny of haematopoietic stem cells (HSCs) during embryonic development is still highly debated, especially their possible lineage relationship to vascular endothelial cells. The first anatomical site from which cells with long-term HSC potential have been isolated is the aorta-gonad-mesonephros (AGM), more specifically the vicinity of the dorsal aortic floor. But although some authors have presented evidence that HSCs may arise directly from the aortic floor into the dorsal aortic lumen, others support the notion that HSCs first emerge within the underlying mesenchyme. Here we show by non-invasive, high-resolution imaging of live zebrafish embryos, that HSCs emerge directly from the aortic floor, through a stereotyped process that does not involve cell division but a strong bending then egress of single endothelial cells from the aortic ventral wall into the sub-aortic space, and their concomitant transformation into haematopoietic cells. The process is polarized not only in the dorso-ventral but also in the rostro-caudal versus medio-lateral direction, and depends on Runx1 expression: in Runx1-deficient embryos, the exit events are initially similar, but much rarer, and abort into violent death of the exiting cell. These results demonstrate that the aortic floor is haemogenic and that HSCs emerge from it into the sub-aortic space, not by asymmetric cell division but through a new type of cell behaviour, which we call an endothelial haematopoietic transition.


Subject(s)
Aorta/cytology , Cell Differentiation , Cell Lineage , Cell Movement , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Zebrafish/blood , Animals , Animals, Genetically Modified , Aorta/embryology , Cell Death , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/embryology , Zebrafish/embryology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Blood ; 122(3): 413-23, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23704093

ABSTRACT

The t(12;21) chromosomal translocation, targeting the gene encoding the RUNX1 transcription factor, is observed in 25% of pediatric acute lymphoblastic leukemia (ALL) and is an initiating event in the disease. To elucidate the mechanism by which RUNX1 disruption initiates leukemogenesis, we investigated its normal role in murine B-cell development. This study revealed 2 critical functions of Runx1: (1) to promote survival and development of progenitors specified to the B-cell lineage, a function that can be substituted by ectopic Bcl2 expression, and (2) to enable the developmental transition through the pre-B stage triggered by the pre-B-cell antigen receptor (pre-BCR). Gene expression analysis and genomewide Runx1 occupancy studies support the hypothesis that Runx1 reinforces the transcription factor network governing early B-cell survival and development and specifically regulates genes encoding members of the Lyn kinase subfamily (key integrators of interleukin-7 and pre-BCR signaling) and the stage-specific transcription factors SpiB and Aiolos (critical downstream effectors of pre-BCR signaling). Interrogation of expression databases of 257 ALL samples demonstrated the specific down-regulation of the SPIB and IKZF3 genes (the latter encoding AIOLOS) in t(12;21) ALL, providing novel insight into the mechanism by which the translocation blocks B-cell development and promotes leukemia.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Core Binding Factor Alpha 2 Subunit/metabolism , Animals , Apoptosis/genetics , Binding Sites , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 21/genetics , Core Binding Factor Alpha 2 Subunit/deficiency , Enhancer Elements, Genetic/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Gene Targeting , Genome/genetics , Humans , Ikaros Transcription Factor , Mice , Mice, Inbred C57BL , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Translocation, Genetic
10.
Blood ; 120(2): 314-22, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22668850

ABSTRACT

Recent studies have established that during embryonic development, hematopoietic progenitors and stem cells are generated from hemogenic endothelium precursors through a process termed endothelial to hematopoietic transition (EHT). The transcription factor RUNX1 is essential for this process, but its main downstream effectors remain largely unknown. Here, we report the identification of Gfi1 and Gfi1b as direct targets of RUNX1 and critical regulators of EHT. GFI1 and GFI1B are able to trigger, in the absence of RUNX1, the down-regulation of endothelial markers and the formation of round cells, a morphologic change characteristic of EHT. Conversely, blood progenitors in Gfi1- and Gfi1b-deficient embryos maintain the expression of endothelial genes. Moreover, those cells are not released from the yolk sac and disseminated into embryonic tissues. Taken together, our findings demonstrate a critical and specific role of the GFI1 transcription factors in the first steps of the process leading to the generation of hematopoietic progenitors from hemogenic endothelium.


Subject(s)
DNA-Binding Proteins/physiology , Hemangioblasts/cytology , Hemangioblasts/physiology , Hematopoiesis/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Line , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/physiology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Knockout , Models, Biological , Neovascularization, Physiologic , Pregnancy , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Yolk Sac/cytology , Yolk Sac/embryology , Yolk Sac/physiology
11.
Blood ; 119(18): 4152-61, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22411870

ABSTRACT

The transcription factor runt-related transcription factor 1 (Runx1) is essential for the establishment of definitive hematopoiesis during embryonic development. In adult blood homeostasis, Runx1 plays a pivotal role in the maturation of lymphocytes and megakaryocytes. Furthermore, Runx1 is required for the regulation of hematopoietic stem and progenitor cells. However, how Runx1 orchestrates self-renewal and lineage choices in combination with other factors is not well understood. In the present study, we describe a genome-scale RNA interference screen to detect genes that cooperate with Runx1 in regulating hematopoietic stem and progenitor cells. We identify the polycomb group protein Pcgf1 as an epigenetic regulator involved in hematopoietic cell differentiation and show that simultaneous depletion of Runx1 and Pcgf1 allows sustained self-renewal while blocking differentiation of lineage marker-negative cells in vitro. We found an up-regulation of HoxA cluster genes on Pcgf1 knock-down that possibly accounts for the increase in self-renewal. Moreover, our data suggest that cells lacking both Runx1 and Pcgf1 are blocked at an early progenitor stage, indicating that a concerted action of the transcription factor Runx1, together with the epigenetic repressor Pcgf1, is necessary for terminal differentiation. The results of the present study uncover a link between transcriptional and epigenetic regulation that is required for hematopoietic differentiation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , DNA-Binding Proteins/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Transplantation , Cell Division , Cells, Cultured/cytology , Chromatin Immunoprecipitation , Colony-Forming Units Assay , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polycomb Repressive Complex 1 , RNA, Small Interfering/pharmacology , Radiation Chimera , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/physiology , Specific Pathogen-Free Organisms , Transduction, Genetic
12.
Brain Behav Immun ; 35: 96-106, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24041578

ABSTRACT

Colitis is a group of inflammatory and auto-immune disorders that affect the tissue lining of the gastrointestinal (GI) system. Studies of chemically-induced animal models of colitis have indicated that nociceptive afferents or neuropeptides have differing effects on GI inflammation. However, the molecular mechanisms involved in visceral pain and the role of visceral sensory afferents involved in the modulation of colitis remains unclear. A previous study demonstrated that Runx1, a Runt domain transcription factor, is restricted to nociceptors. In these neurons, Runx1 regulates the expression of numerous ion channels and receptors, controlling the lamina-specific innervation patterns of nociceptive afferents in the spinal cord. Moreover, mice that lack Runx1 exhibit specific defects in thermal and neuropathic pain. To examine the function of Runx1 in visceral nociception, we employed double-transgenic mice (WntCre: Runx1(F/F)), in which the expression of Runx1 was specifically disrupted in the sensory neurons. To determine the role of Runx1 in visceral pain sensation, the WntCre: Runx1(F/F) mice and their control littermates (Runx1(F/F)) were treated using dextran sodium sulfate (DSS) to induce colitis. The results indicated that disrupted Runx1 in the sensory afferents resulted in: (1) impairment of the visceral pain sensation in murine DSS-induced colitis; (2) exacerbating the phenotypes in murine DSS-induced colitis; (3) a differential effect on the production of pro- and anti-inflammatory cytokines in the colon tissues isolated from mice treated using DSS and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis; and (4) alteration of the distribution of lymphocytes and mast cells in mucosa. These results show that the function of Runx1 in sensory afferents is vital for modulating visceral pain and the neuro-immune axis.


Subject(s)
Colitis/physiopathology , Core Binding Factor Alpha 2 Subunit/physiology , Neurons, Afferent/physiology , Nociception/physiology , Visceral Pain/physiopathology , Animals , Colitis/chemically induced , Colitis/complications , Core Binding Factor Alpha 2 Subunit/deficiency , Dextran Sulfate/pharmacology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Sensory Receptor Cells/physiology , Visceral Pain/etiology
13.
J Immunol ; 188(11): 5408-20, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22551552

ABSTRACT

The Runx1 transcription factor is abundantly expressed in naive T cells but rapidly downregulated in activated T cells, suggesting that it plays an important role in a naive stage. In the current study, Runx1(-/-)Bcl2(tg) mice harboring Runx1-deleted CD4(+) T cells developed a fatal autoimmune lung disease. CD4(+) T cells from these mice were spontaneously activated, preferentially homed to the lung, and expressed various cytokines, including IL-17 and IL-21. Among these, the deregulation of IL-21 transcription was likely to be associated with Runx binding sites located in an IL-21 intron. IL-17 produced in Runx1-deleted cells mobilized innate immune responses, such as those promoted by neutrophils and monocytes, whereas IL-21 triggered humoral responses, such as plasma cells. Thus, at an initial stage, peribronchovascular regions in the lung were infiltrated by CD4(+) lymphocytes, whereas at a terminal stage, interstitial regions were massively occupied by immune cells, and alveolar spaces were filled with granular exudates that resembled pulmonary alveolar proteinosis in humans. Mice suffered from respiratory failure, as well as systemic inflammatory responses. Our data indicate that Runx1 plays an essential role in repressing the transcription of cytokine genes in naive CD4(+) T cells and, thereby, maintains cell quiescence.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 2 Subunit/deficiency , Lung Diseases/immunology , Lymphocyte Activation/immunology , Animals , Autoimmune Diseases/mortality , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/mortality , Jurkat Cells , Lung Diseases/mortality , Lung Diseases/pathology , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology
14.
Int J Hematol ; 120(2): 186-193, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38702444

ABSTRACT

Acute myeloid leukemia (AML) with t(8;21)(q22;q22.1);RUNX1-ETO is one of the most common subtypes of AML. Although t(8;21) AML has been classified as favorable-risk, only about half of patients are cured with current therapies. Several genetic abnormalities, including TP53 mutations and deletions, negatively impact survival in t(8;21) AML. In this study, we established Cas9+ mouse models of t(8;21) AML with intact or deficient Tpr53 (a mouse homolog of TP53) using a retrovirus-mediated gene transfer and transplantation system. Trp53 deficiency accelerates the in vivo development of AML driven by RUNX1-ETO9a, a short isoform of RUNX1-ETO with strong leukemogenic potential. Trp53 deficiency also confers resistance to genetic depletion of RUNX1 and a TP53-activating drug in t(8;21) AML. However, Trp53-deficient t(8;21) AML cells were still sensitive to several drugs such as dexamethasone. Cas9+ RUNX1-ETO9a cells with/without Trp53 deficiency can produce AML in vivo, can be cultured in vitro for several weeks, and allow efficient gene depletion using the CRISPR/Cas9 system, providing useful tools to advance our understanding of t(8;21) AML.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit , Disease Models, Animal , Leukemia, Myeloid, Acute , Translocation, Genetic , Tumor Suppressor Protein p53 , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/etiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/deficiency , Animals , Mice , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 21/genetics , Humans , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/deficiency , Oncogene Proteins, Fusion/genetics
15.
J Neurosci ; 32(33): 11285-98, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895712

ABSTRACT

Microglia are the immune cells of the nervous system, where they act as resident macrophages during inflammatory events underlying many neuropathological conditions. Microglia derive from primitive myeloid precursors that colonize the nervous system during embryonic development. In the postnatal brain, microglia are initially mitotic, rounded in shape (amoeboid), and phagocytically active. As brain development proceeds, they gradually undergo a transition to a surveillant nonphagocytic state characterized by a highly branched (ramified) morphology. This ramification process is almost recapitulated in reverse during the process of microglia activation in the adult brain, when surveillant microglia undergo a ramified-to-amoeboid morphological transformation and become phagocytic in response to injury or disease. Little is known about the mechanisms controlling amoeboid microglial cell proliferation, activation, and ramification during brain development, despite the critical role of these processes in the establishment of the adult microglia pool and their relevance to microglia activation in the adult brain. Here we show that the mouse transcription factor Runx1, a key regulator of myeloid cell proliferation and differentiation, is expressed in forebrain amoeboid microglia during the first two postnatal weeks. Runx1 expression is then downregulated in ramified microglia. Runx1 inhibits mouse amoeboid microglia proliferation and promotes progression to the ramified state. We show further that Runx1 expression is upregulated in microglia following nerve injury in the adult mouse nervous system. These findings provide insight into the regulation of postnatal microglia activation and maturation to the ramified state and have implications for microglia biology in the developing and injured brain.


Subject(s)
Cell Proliferation , Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation, Developmental/physiology , Microglia/metabolism , Prosencephalon/cytology , Prosencephalon/growth & development , Animals , Animals, Newborn , Antigens, Differentiation/metabolism , Bromodeoxyuridine/metabolism , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cell Line, Transformed , Cells, Cultured , Chromatin Immunoprecipitation , Core Binding Factor Alpha 2 Subunit/deficiency , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/metabolism , Humans , Intermediate Filament Proteins/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Nitric Oxide Synthase Type II/metabolism , Phosphatidylethanolamines , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Spinal Cord/cytology
16.
Blood ; 118(9): 2541-50, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21757616

ABSTRACT

Dysfunction of AML1/Runx1, a transcription factor, plays a crucial role in the development of many types of leukemia. Additional events are often required for AML1 dysfunction to induce full-blown leukemia; however, a mechanistic basis of their cooperation is still elusive. Here, we investigated the effect of AML1 deficiency on the development of MLL-ENL leukemia in mice. Aml1 excised bone marrow cells lead to MLL-ENL leukemia with shorter duration than Aml1 intact cells in vivo. Although the number of MLL-ENL leukemia-initiating cells is not affected by loss of AML1, the proliferation of leukemic cells is enhanced in Aml1-excised MLL-ENL leukemic mice. We found that the enhanced proliferation is the result of repression of p19(ARF) that is directly regulated by AML1 in MLL-ENL leukemic cells. We also found that down-regulation of p19(ARF) induces the accelerated onset of MLL-ENL leukemia, suggesting that p19(ARF) is a major target of AML1 in MLL-ENL leukemia. These results provide a new insight into a role for AML1 in the progression of leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Gene Expression Regulation, Leukemic/genetics , Leukemia, Biphenotypic, Acute/genetics , Neoplasm Proteins/physiology , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Bone Marrow Transplantation , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Radiation Chimera , Recombinant Fusion Proteins/physiology , Transcription, Genetic
17.
J Immunol ; 186(3): 1450-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21178013

ABSTRACT

Hematopoietic lymphoid tissue inducer (LTi) cells are essential for the development of secondary lymphoid tissues including lymph nodes and Peyer's patches. Two transcription factors, the helix-loop-helix inhibitor Id2 and the retinoic acid-related orphan receptor γt (Rorγt), have been shown to be crucial for LTi cell development. However, it remains unclear how the specification of multipotent hematopoietic progenitor cells toward the LTi lineage is programmed. In this study, we report impaired lymphoid tissue organogenesis in mice in which the function of Runx1/Cbfß transcription factor complexes was attenuated by the loss of either the distal promoter-derived Runx1 or Cbfß2 variant protein. We found that LTi progenitors in fetal liver, defined previously as a lineage marker-negative α4ß7 integrin (α4ß7)(+) IL-7R α-chain (IL-7Rα)(+) population, can be subdivided into Rorγt-expressing IL-7Rα(high) cells and nonexpressing IL-7Rα(mid) cells. Whereas Id2 and Rorγt are required to direct α4ß7(+)IL-7Rα(mid) cells to become α4ß7(+)IL-7Rα(high) cells, Runx1/Cbfß2 complexes are necessary for the emergence of α4ß7(+)IL-7Rα(mid) cells. In addition, the loss of Cbfß2, but not P1-Runx1, resulted in an inefficient upregulation of Rorγt in residual α4ß7(+)IL-7Rα(+) LTi cells at anlagen. Our results thus revealed that Runx1/Cbfß2 complexes regulate the differentiation of LTi cells at two stages: an early specification of hematopoietic progenitors toward the LTi lineage and a subsequent activation of Rorγt expression at anlagen.


Subject(s)
Cell Differentiation/immunology , Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/physiology , Lymphoid Tissue/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/genetics , Genetic Variation/immunology , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/physiology , Liver/embryology , Liver/immunology , Liver/pathology , Lymphoid Tissue/cytology , Lymphoid Tissue/embryology , Mice , Mice, Mutant Strains , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology
18.
Nature ; 446(7139): 1056-61, 2007 Apr 26.
Article in English | MEDLINE | ID: mdl-17377529

ABSTRACT

The first haematopoietic stem cells (HSCs) appear in the aorta-gonad-mesonephros (AGM) region, major vitelline and umbilical vessels, and placenta; however, whether they arise locally or from immigrant yolk sac precursor cells remains unclear. This issue is best addressed by measuring cell-lineage relationships rather than cell potentials. To undertake long-term in vivo tracing of yolk sac cells, we designed a non-invasive pulse-labelling system based on Cre/loxP recombination. Here we show that in Runx1(+/-) (runt-related transcription factor 1) heterozygous mice, yolk sac cells expressing Runx1 at embryonic day 7.5 develop into fetal lymphoid progenitors and adult HSCs. During mid-gestation the labelled (embryonic day 7.5) yolk sac cells colonize the umbilical cord, the AGM region and subsequently the embryonic liver. This raises the possibility that some HSCs associated with major embryonic vasculature are derived from yolk sac precursors. We observed virtually no contribution of the labelled cells towards the yolk sac vasculature, indicating early segregation of endothelial and haematopoietic lineages.


Subject(s)
Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Yolk Sac/cytology , Animals , Aorta , Core Binding Factor Alpha 2 Subunit/analysis , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Female , Fetus/cytology , Fetus/immunology , Gene Expression Regulation, Developmental , Genes, Reporter/genetics , Hematopoietic Stem Cells/metabolism , Heterozygote , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Time Factors , Umbilical Arteries , Umbilical Veins , Yolk Sac/metabolism
19.
Blood ; 115(15): 3042-50, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20139099

ABSTRACT

The transcription factor Runx1 is a pivotal regulator of definitive hematopoiesis in mouse ontogeny. Vertebrate Runx1 is transcribed from 2 promoters, the distal P1 and proximal P2, which provide a paradigm of the complex transcriptional and translational control of Runx1 function. However, very little is known about the biologic relevance of alternative Runx1 promoter usage in definitive hematopoietic cell emergence. Here we report that both promoters are active at the very onset of definitive hematopoiesis, with a skewing toward the P2. Moreover, functional and morphologic analysis of a novel P1-null and an attenuated P2 mouse model revealed that although both promoters play important nonredundant roles in the emergence of definitive hematopoietic cells, the proximal P2 was most critically required for this. The nature of the observed phenotypes is indicative of a differential contribution of the P1 and P2 promoters to the control of overall Runx1 levels, where and when this is most critically required. In addition, the dynamic expression of P1-Runx1 and P2-Runx1 points at a requirement for Runx1 early in development, when the P2 is still the prevalent promoter in the emerging hemogenic endothelium and/or first committed hematopoietic cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/genetics , Promoter Regions, Genetic/genetics , Aging/genetics , Alleles , Animals , Aorta/metabolism , Aorta/pathology , Bone Marrow Cells/metabolism , Cadherins/metabolism , Cell Aggregation , Cell Count , Colony-Forming Units Assay , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo Loss/genetics , Embryo Loss/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Expression Regulation, Developmental , Genes, Reporter , Genetic Loci/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Leukocyte Common Antigens/metabolism , Mice , Models, Genetic , Mutation/genetics
20.
Blood ; 115(15): 3128-35, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20181616

ABSTRACT

Haploinsufficiency of RUNX1 (also known as CBFA2/AML1) is associated with familial thrombocytopenia, platelet dysfunction, and predisposition to acute leukemia. We have reported on a patient with thrombocytopenia and impaired agonist-induced aggregation, secretion, and protein phosphorylation associated with a RUNX1 mutation. Expression profiling of platelets revealed approximately 5-fold decreased expression of 12-lipoxygenase (12-LO, gene ALOX12), which catalyzes 12-hydroxyeicosatetraenoic acid production from arachidonic acid. We hypothesized that ALOX12 is a direct transcriptional target gene of RUNX1. In present studies, agonist-induced platelet 12-HETE production was decreased in the patient. Four RUNX1 consensus sites were identified in the 2-kb promoter region of ALOX12 (at -1498, -1491, -708, -526 from ATG). In luciferase reporter studies in human erythroleukemia cells, mutation of each site decreased activity; overexpression of RUNX1 up-regulated promoter activity, which was abolished by mutation of RUNX1 sites. Gel shift studies, including with recombinant protein, revealed RUNX1 binding to each site. Chromatin immunoprecipitation revealed in vivo RUNX1 binding in the region of interest. siRNA knockdown of RUNX1 decreased RUNX1 and 12-LO proteins. ALOX12 is a direct transcriptional target of RUNX1. Our studies provide further proof of principle that platelet expression profiling can elucidate novel alterations in platelets with inherited dysfunction.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Blood Platelets/enzymology , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/metabolism , Haploidy , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/biosynthesis , Arachidonate 12-Lipoxygenase/metabolism , Arachidonic Acid/pharmacology , Base Sequence , Binding Sites , Blood Platelets/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , Consensus Sequence , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Genes, Reporter , Humans , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Luciferases/metabolism , Molecular Sequence Data , Platelet Activation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL