Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Sci Adv ; 10(10): eadj8803, 2024 Mar 08.
Article En | MEDLINE | ID: mdl-38457494

Philadelphia chromosome-positive B cell acute lymphoblastic leukemia (B-ALL), characterized by the BCR::ABL1 fusion gene, remains a poor prognosis cancer needing new therapeutic approaches. Transcriptomic profiling identified up-regulation of oncogenic transcription factors ERG and c-MYC in BCR::ABL1 B-ALL with ERG and c-MYC required for BCR::ABL1 B-ALL in murine and human models. Profiling of ERG- and c-MYC-dependent gene expression and analysis of ChIP-seq data established ERG and c-MYC coordinate a regulatory network in BCR::ABL1 B-ALL that controls expression of genes involved in several biological processes. Prominent was control of ribosome biogenesis, including expression of RNA polymerase I (POL I) subunits, the importance of which was validated by inhibition of BCR::ABL1 cells by POL I inhibitors, including CX-5461, that prevents promoter recruitment and transcription initiation by POL I. Our results reveal an essential ERG- and c-MYC-dependent transcriptional network involved in regulation of metabolic and ribosome biogenesis pathways in BCR::ABL1 B-ALL, from which previously unidentified vulnerabilities and therapeutic targets may emerge.


Fusion Proteins, bcr-abl , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Transcriptional Regulator ERG , Animals , Humans , Mice , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Fusion Proteins, bcr-abl/therapeutic use , Gene Regulatory Networks , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Transcription Factors/genetics , Transcriptional Regulator ERG/genetics
2.
Leukemia ; 38(6): 1342-1352, 2024 Jun.
Article En | MEDLINE | ID: mdl-38491305

Thrombopoietin (Tpo), which binds to its specific receptor, the Mpl protein, is the major cytokine regulator of megakaryopoiesis and circulating platelet number. Tpo binding to Mpl triggers activation of Janus kinase 2 (Jak2) and phosphorylation of the receptor, as well as activation of several intracellular signalling cascades that mediate cellular responses. Three tyrosine (Y) residues in the C-terminal region of the Mpl intracellular domain have been implicated as sites of phosphorylation required for regulation of major Tpo-stimulated signalling pathways: Mpl-Y565, Mpl-Y599 and Mpl-Y604. Here, we have introduced mutations in the mouse germline and report a consistent physiological requirement for Mpl-Y599, mutation of which resulted in thrombocytopenia, deficient megakaryopoiesis, low hematopoietic stem cell (HSC) number and function, and attenuated responses to myelosuppression. We further show that in models of myeloproliferative neoplasms (MPN), where Mpl is required for pathogenesis, thrombocytosis was dependent on intact Mpl-Y599. In contrast, Mpl-Y565 was required for negative regulation of Tpo responses; mutation of this residue resulted in excess megakaryopoiesis at steady-state and in response to myelosuppression, and exacerbated thrombocytosis associated with MPN.


Hematopoiesis , Myeloproliferative Disorders , Receptors, Thrombopoietin , Thrombopoietin , Tyrosine , Animals , Receptors, Thrombopoietin/metabolism , Receptors, Thrombopoietin/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Mice , Thrombopoietin/metabolism , Tyrosine/metabolism , Tyrosine/genetics , Phosphorylation , Mice, Inbred C57BL , Hematopoietic Stem Cells/metabolism , Signal Transduction , Mutation , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Thrombopoiesis/genetics
3.
Nat Commun ; 15(1): 1135, 2024 Feb 07.
Article En | MEDLINE | ID: mdl-38326297

Thrombopoietin (Tpo) is the primary regulator of megakaryocyte and platelet numbers and is required for haematopoetic stem cell maintenance. Tpo functions by binding its receptor (TpoR, a homodimeric Class I cytokine receptor) and initiating cell proliferation or differentiation. Here we characterise the murine Tpo:TpoR signalling complex biochemically and structurally, using cryo-electron microscopy. Tpo uses opposing surfaces to recruit two copies of receptor, forming a 1:2 complex. Although it binds to the same, membrane-distal site on both receptor chains, it does so with significantly different affinities and its highly glycosylated C-terminal domain is not required. In one receptor chain, a large insertion, unique to TpoR, forms a partially structured loop that contacts cytokine. Tpo binding induces the juxtaposition of the two receptor chains adjacent to the cell membrane. The therapeutic agent romiplostim also targets the cytokine-binding site and the characterisation presented here supports the future development of improved TpoR agonists.


Receptors, Thrombopoietin , Thrombopoietin , Animals , Mice , Cryoelectron Microscopy , Receptors, Cytokine/metabolism , Receptors, Thrombopoietin/metabolism , Signal Transduction
5.
Nat Commun ; 13(1): 4739, 2022 08 12.
Article En | MEDLINE | ID: mdl-35961968

CRISPR technologies have advanced cancer modelling in mice, but CRISPR activation (CRISPRa) methods have not been exploited in this context. We establish a CRISPRa mouse (dCas9a-SAMKI) for inducing gene expression in vivo and in vitro. Using dCas9a-SAMKI primary lymphocytes, we induce B cell restricted genes in T cells and vice versa, demonstrating the power of this system. There are limited models of aggressive double hit lymphoma. Therefore, we transactivate pro-survival BCL-2 in Eµ-MycT/+;dCas9a-SAMKI/+ haematopoietic stem and progenitor cells. Mice transplanted with these cells rapidly develop lymphomas expressing high BCL-2 and MYC. Unlike standard Eµ-Myc lymphomas, BCL-2 expressing lymphomas are highly sensitive to the BCL-2 inhibitor venetoclax. We perform genome-wide activation screens in these lymphoma cells and find a dominant role for the BCL-2 protein A1 in venetoclax resistance. Here we show the potential of our CRISPRa model for mimicking disease and providing insights into resistance mechanisms towards targeted therapies.


Clustered Regularly Interspaced Short Palindromic Repeats , Lymphoma , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/pathology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sulfonamides
6.
Nat Commun ; 11(1): 3013, 2020 06 15.
Article En | MEDLINE | ID: mdl-32541654

B lymphoid development is initiated by the differentiation of hematopoietic stem cells into lineage committed progenitors, ultimately generating mature B cells. This highly regulated process generates clonal immunological diversity via recombination of immunoglobulin V, D and J gene segments. While several transcription factors that control B cell development and V(D)J recombination have been defined, how these processes are initiated and coordinated into a precise regulatory network remains poorly understood. Here, we show that the transcription factor ETS Related Gene (Erg) is essential for early B lymphoid differentiation. Erg initiates a transcriptional network involving the B cell lineage defining genes, Ebf1 and Pax5, which directly promotes expression of key genes involved in V(D)J recombination and formation of the B cell receptor. Complementation of Erg deficiency with a productively rearranged immunoglobulin gene rescued B lineage development, demonstrating that Erg is an essential and stage-specific regulator of the gene regulatory network controlling B lymphopoiesis.


B-Lymphocytes/metabolism , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Lymphopoiesis/genetics , Oncogene Proteins/genetics , Transcription, Genetic , Transcriptional Regulator ERG/genetics , Animals , B-Lymphocytes/cytology , Cell Lineage/genetics , Cells, Cultured , Gene Regulatory Networks/genetics , Hematopoietic Stem Cells/cytology , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/metabolism , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Regulator ERG/metabolism , V(D)J Recombination/genetics
7.
Growth Factors ; 36(3-4): 89-103, 2018 08.
Article En | MEDLINE | ID: mdl-30318940

Platelets are anuclear blood cells required for haemostasis and are implicated in other processes including inflammation and metastasis. Platelets are produced by megakaryocytes, specialized cells that are themselves generated by a process of controlled differentiation and maturation of bone-marrow stem and progenitor cells. This process of megakaryopoiesis involves the coordinated interplay of transcription factor-controlled cellular programming with extra-cellular cues produced locally in supporting niches or as circulating factors. This review focuses on these external cues, the cytokines and chemokines, that drive production of megakaryocytes and support the terminal process of platelet release. Emphasis is given to thrombopoietin (Tpo), the major cytokine regulator of steady-state megakaryopoiesis, and its specific cell surface receptor, the Mpl protein, including normal and pathological roles as well as clinical application. The potential for alternative or supplementary regulatory mechanisms for platelet production, particularly in times of acute need, or in states of infection or inflammation are also discussed.


Cytokines/metabolism , Hematopoiesis , Megakaryocyte Progenitor Cells/cytology , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cytokines/genetics , Humans , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism
8.
J Exp Med ; 214(3): 737-752, 2017 03 06.
Article En | MEDLINE | ID: mdl-28213513

Acute myeloid leukemia (AML) is induced by the cooperative action of deregulated genes that perturb self-renewal, proliferation, and differentiation. Internal tandem duplications (ITDs) in the FLT3 receptor tyrosine kinase are common mutations in AML, confer poor prognosis, and stimulate myeloproliferation. AML patient samples with FLT3-ITD express high levels of RUNX1, a transcription factor with known tumor-suppressor function. In this study, to understand this paradox, we investigated the impact of RUNX1 and FLT3-ITD coexpression. FLT3-ITD directly impacts on RUNX1 activity, whereby up-regulated and phosphorylated RUNX1 cooperates with FLT3-ITD to induce AML. Inactivating RUNX1 in tumors releases the differentiation block and down-regulates genes controlling ribosome biogenesis. We identified Hhex as a direct target of RUNX1 and FLT3-ITD stimulation and confirmed high HHEX expression in FLT3-ITD AMLs. HHEX could replace RUNX1 in cooperating with FLT3-ITD to induce AML. These results establish and elucidate the unanticipated oncogenic function of RUNX1 in AML. We predict that blocking RUNX1 activity will greatly enhance current therapeutic approaches using FLT3 inhibitors.


Core Binding Factor Alpha 2 Subunit/physiology , Leukemia, Myeloid, Acute/etiology , fms-Like Tyrosine Kinase 3/physiology , Animals , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Mice , Protein Processing, Post-Translational , Transcription Factors/genetics
9.
Blood ; 127(26): 3369-81, 2016 06 30.
Article En | MEDLINE | ID: mdl-27076172

Disrupting mutations of the RUNX1 gene are found in 10% of patients with myelodysplasia (MDS) and 30% of patients with acute myeloid leukemia (AML). Previous studies have revealed an increase in hematopoietic stem cells (HSCs) and multipotent progenitor (MPP) cells in conditional Runx1-knockout (KO) mice, but the molecular mechanism is unresolved. We investigated the myeloid progenitor (MP) compartment in KO mice, arguing that disruptions at the HSC/MPP level may be amplified in downstream cells. We demonstrate that the MP compartment is increased by more than fivefold in Runx1 KO mice, with a prominent skewing toward megakaryocyte (Meg) progenitors. Runx1-deficient granulocyte-macrophage progenitors are characterized by increased cloning capacity, impaired development into mature cells, and HSC and Meg transcription signatures. An HSC/MPP subpopulation expressing Meg markers was also increased in Runx1-deficient mice. Rescue experiments coupled with transcriptome analysis and Runx1 DNA-binding assays demonstrated that granulocytic/monocytic (G/M) commitment is marked by Runx1 suppression of genes encoding adherence and motility proteins (Tek, Jam3, Plxnc1, Pcdh7, and Selp) that support HSC-Meg interactions with the BM niche. In vitro assays confirmed that enforced Tek expression in HSCs/MPPs increases Meg output. Interestingly, besides this key repressor function of Runx1 to control lineage decisions and cell numbers in progenitors, our study also revealed a critical activating function in erythroblast differentiation, in addition to its known importance in Meg and G/M maturation. Thus both repressor and activator functions of Runx1 at multiple hematopoietic stages and lineages likely contribute to the tumor suppressor activity in MDS and AML.


Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Megakaryocytes/pathology , Mice , Mice, Knockout , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Tumor Suppressor Proteins/genetics
10.
Blood ; 125(23): 3570-9, 2015 Jun 04.
Article En | MEDLINE | ID: mdl-25911237

The activity of antagonizing transcription factors represents a mechanistic paradigm of bidirectional lineage-fate control during hematopoiesis. At the megakaryocytic/erythroid bifurcation, the cross-antagonism of krueppel-like factor 1 (KLF1) and friend leukemia integration 1 (FLI1) has such a decisive role. However, how this antagonism is resolved during lineage specification is poorly understood. We found that runt-related transcription factor 1 (RUNX1) inhibits erythroid differentiation of murine megakaryocytic/erythroid progenitors and primary human CD34(+) progenitor cells. We show that RUNX1 represses the erythroid gene expression program during megakaryocytic differentiation by epigenetic repression of the erythroid master regulator KLF1. RUNX1 binding to the KLF1 locus is increased during megakaryocytic differentiation and counterbalances the activating role of T-cell acute lymphocytic leukemia 1 (TAL1). We found that corepressor recruitment by RUNX1 contributes to a block of the KLF1-dependent erythroid gene expression program. Our data indicate that the repressive function of RUNX1 influences the balance between erythroid and megakaryocytic differentiation by shifting the balance between KLF1 and FLI1 in the direction of FLI1. Taken together, we show that RUNX1 is a key player within a network of transcription factors that represses the erythroid gene expression program.


Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation/physiology , Megakaryocytes/metabolism , Thrombopoiesis/physiology , Antigens, CD34/genetics , Antigens, CD34/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Humans , K562 Cells , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1
11.
Blood ; 123(14): 2229-37, 2014 Apr 03.
Article En | MEDLINE | ID: mdl-24523240

Severe congenital neutropenia (CN) is a preleukemic bone marrow failure syndrome with a 20% risk of evolving into leukemia or myelodysplastic syndrome (MDS). Patterns of acquisition of leukemia-associated mutations were investigated using next-generation deep-sequencing in 31 CN patients who developed leukemia or MDS. Twenty (64.5%) of the 31 patients had mutations in RUNX1. A majority of patients with RUNX1 mutations (80.5%) also had acquired CSF3R mutations. In contrast to their high frequency in CN patients who developed leukemia or MDS, RUNX1 mutations were found in only 9 of 307 (2.9%) patients with de novo pediatric acute myeloid leukemia. A sequential analysis at stages prior to overt leukemia revealed RUNX1 mutations to be late events in leukemic transformation. Single-cell analyses in 2 patients showed that RUNX1 and CSF3R mutations were present in the same malignant clone. Functional studies demonstrated elevated granulocyte colony-stimulating factor (G-CSF)-induced proliferation with diminished myeloid differentiation of hematopoietic CD34(+) cells coexpressing mutated forms of RUNX1 and CSF3R. The high frequency of cooperating RUNX1 and CSF3R mutations in CN patients suggests a novel molecular pathway of leukemogenesis: mutations in the hematopoietic cytokine receptor (G-CSFR) in combination with the second mutations in the downstream hematopoietic transcription fator (RUNX1). The detection of both RUNX1 and CSF3R mutations could be used as a marker for identifying CN patients with a high risk of progressing to leukemia or MDS.


Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid/genetics , Mutation , Neutropenia/congenital , Receptors, Colony-Stimulating Factor/genetics , Adolescent , Adult , Child , Child, Preschool , Congenital Bone Marrow Failure Syndromes , Cytogenetic Analysis , Female , Humans , Male , Neutropenia/genetics , Neutropenia/pathology , Signal Transduction/genetics , Young Adult
12.
Blood ; 122(3): 413-23, 2013 Jul 18.
Article En | MEDLINE | ID: mdl-23704093

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.


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
13.
Blood ; 119(18): 4152-61, 2012 May 03.
Article En | MEDLINE | ID: mdl-22411870

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.


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
...