Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Proc Natl Acad Sci U S A ; 121(25): e2312499121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857395

ABSTRACT

Ex vivo expansion of human CD34+ hematopoietic stem and progenitor cells remains a challenge due to rapid differentiation after detachment from the bone marrow niche. In this study, we assessed the capacity of an inducible fusion protein to enable sustained ex vivo proliferation of hematopoietic precursors and their capacity to differentiate into functional phagocytes. We fused the coding sequences of an FK506-Binding Protein 12 (FKBP12)-derived destabilization domain (DD) to the myeloid/lymphoid lineage leukemia/eleven nineteen leukemia (MLL-ENL) fusion gene to generate the fusion protein DD-MLL-ENL and retrovirally expressed the protein switch in human CD34+ progenitors. Using Shield1, a chemical inhibitor of DD fusion protein degradation, we established large-scale and long-term expansion of late monocytic precursors. Upon Shield1 removal, the cells lost self-renewal capacity and spontaneously differentiated, even after 2.5 y of continuous ex vivo expansion. In the absence of Shield1, stimulation with IFN-γ, LPS, and GM-CSF triggered terminal differentiation. Gene expression analysis of the obtained phagocytes revealed marked similarity with naïve monocytes. In functional assays, the novel phagocytes migrated toward CCL2, attached to VCAM-1 under shear stress, produced reactive oxygen species, and engulfed bacterial particles, cellular particles, and apoptotic cells. Finally, we demonstrated Fcγ receptor recognition and phagocytosis of opsonized lymphoma cells in an antibody-dependent manner. Overall, we have established an engineered protein that, as a single factor, is useful for large-scale ex vivo production of human phagocytes. Such adjustable proteins have the potential to be applied as molecular tools to produce functional immune cells for experimental cell-based approaches.


Subject(s)
Cell Differentiation , Phagocytes , Humans , Phagocytes/metabolism , Hematopoietic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Leukemia/genetics , Leukemia/pathology , Leukemia/metabolism , Protein Engineering/methods , Phagocytosis
2.
IUBMB Life ; 72(1): 151-158, 2020 01.
Article in English | MEDLINE | ID: mdl-31785092

ABSTRACT

The GATA family of transcription factors are zinc finger (ZF) DNA-binding proteins that regulate transcription during development and cell differentiation. GATA2 plays an essential role in the regulation of hematopoiesis. As a result, mutations in this gene or alterations in its expression level or function have been linked to a variety of human hematologic disorders. In this review, we summarize the findings and developments over the recent years regarding the clinical correlations and functional properties of distinct GATA2 mutations in hematopoietic malignancies, with particular focus on the mutational hotspots in the ZF domains.


Subject(s)
GATA2 Transcription Factor/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Zinc Fingers/genetics , Animals , Cell Differentiation , GATA2 Transcription Factor/metabolism , Humans
3.
Hemasphere ; 7(10): e958, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37841755

ABSTRACT

Activating colony-stimulating factor-3 receptor gene (CSF3R) mutations are recurrent in acute myeloid leukemia (AML) with t(8;21) translocation. However, the nature of oncogenic collaboration between alterations of CSF3R and the t(8;21) associated RUNX1-RUNX1T1 fusion remains unclear. In CD34+ hematopoietic stem and progenitor cells from healthy donors, double oncogene expression led to a clonal advantage, increased self-renewal potential, and blast-like morphology and distinct immunophenotype. Gene expression profiling revealed hedgehog signaling as a potential mechanism, with upregulation of GLI2 constituting a putative pharmacological target. Both primary hematopoietic cells and the t(8;21) positive AML cell line SKNO-1 showed increased sensitivity to the GLI inhibitor GANT61 when expressing CSF3R T618I. Our findings suggest that during leukemogenesis, the RUNX1-RUNXT1 fusion and CSF3R mutation act in a synergistic manner to alter hedgehog signaling, which can be exploited therapeutically.

4.
Exp Hematol ; 108: 26-35, 2022 04.
Article in English | MEDLINE | ID: mdl-35181392

ABSTRACT

GATA2 zinc-finger (ZF) mutations are associated with distinct entities of myeloid malignancies. The specific distribution of these mutations points toward different mechanisms of leukemogenesis depending on the ZF domain affected. In this study, we compared recurring somatic mutations in ZF1 and ZF2. All tested ZF mutants disrupted DNA binding in vitro. In transcription assays, co-expression of FOG1 counteracted GATA2-dependent transcriptional activation, while a variable response to FOG1-mediated repression was observed for individual GATA2 mutants. In primary murine bone marrow cells, GATA2 wild-type (WT) expression inhibited colony formation, while this effect was reduced for both mutants A318T (ZF1) and L359V (ZF2) with a shift toward granulopoiesis. In primary human CD34+ bone marrow cells and in the myeloid cell line K562, ectopic expression of GATA2 L359V, but not A318T or G320D, caused a block of erythroid differentiation accompanied by downregulation of GATA1, STAT5B, and PLCG1. Our findings may explain the role of GATA2 L359V during the progression of chronic myeloid leukemia and the collaboration of GATA2 ZF1 alterations with CEBPA double mutations in erythroleukemia.


Subject(s)
GATA2 Transcription Factor , Leukemia, Erythroblastic, Acute , Leukemia, Myeloid , Animals , Cell Differentiation/genetics , GATA2 Transcription Factor/genetics , Humans , K562 Cells , Leukemia, Erythroblastic, Acute/genetics , Mice , Mutation , Zinc Fingers
5.
Exp Hematol ; 87: 20-24.e1, 2020 07.
Article in English | MEDLINE | ID: mdl-32525064

ABSTRACT

ZBTB7A is a transcription factor that regulates all three branches of hematopoietic differentiation while repressing the expression of key glycolytic enzymes and glucose transporters. Here, we propose that ZBTB7A acts as a link between differentiation and metabolism, two interconnected cellular processes. In particular, ZBTB7A can activate or repress metabolic programs necessary for the differentiation of specific cell lineages while controlling key pathways such as Notch signaling. Finally, the dual role of ZBTB7A has implications for the treatment of myeloid malignancies, where the block of differentiation could potentially be overcome by metabolic therapies with low toxicity.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Hematologic Neoplasms/metabolism , Myeloproliferative Disorders/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasm Proteins/genetics , Transcription Factors/genetics
6.
Oncogene ; 39(15): 3195-3205, 2020 04.
Article in English | MEDLINE | ID: mdl-32115572

ABSTRACT

ZBTB7A is frequently mutated in acute myeloid leukemia (AML) with t(8;21) translocation. However, the oncogenic collaboration between mutated ZBTB7A and the RUNX1-RUNX1T1 fusion gene in AML t(8;21) remains unclear. Here, we investigate the role of ZBTB7A and its mutations in the context of normal and malignant hematopoiesis. We demonstrate that clinically relevant ZBTB7A mutations in AML t(8;21) lead to loss of function and result in perturbed myeloid differentiation with block of the granulocytic lineage in favor of monocytic commitment. In addition, loss of ZBTB7A increases glycolysis and hence sensitizes leukemic blasts to metabolic inhibition with 2-deoxy-D-glucose. We observed that ectopic expression of wild-type ZBTB7A prevents RUNX1-RUNX1T1-mediated clonal expansion of human CD34+ cells, whereas the outgrowth of progenitors is enabled by ZBTB7A mutation. Finally, ZBTB7A expression in t(8;21) cells lead to a cell cycle arrest that could be mimicked by inhibition of glycolysis. Our findings suggest that loss of ZBTB7A may facilitate the onset of AML t(8;21), and that RUNX1-RUNX1T1-rearranged leukemia might be treated with glycolytic inhibitors.


Subject(s)
Carcinogenesis/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/genetics , Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Transcription Factors/genetics , Animals , Bone Marrow/pathology , Carcinogenesis/drug effects , Cell Cycle Checkpoints/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/genetics , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/metabolism , Deoxyglucose/pharmacology , Deoxyglucose/therapeutic use , Gene Knockout Techniques , Glycolysis/drug effects , Glycolysis/genetics , Hematopoiesis/drug effects , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Loss of Function Mutation , Mice , Myeloid Progenitor Cells/pathology , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
7.
Oncogene ; 38(2): 261-272, 2019 01.
Article in English | MEDLINE | ID: mdl-30093631

ABSTRACT

Chromosomal translocations represent frequent events in leukemia. In t(8;21)+ acute myeloid leukemia, RUNX1 is fused to nearly the entire ETO protein, which contains four conserved nervy homology regions, NHR1-4. Furthermore RUNX1/ETO interacts with ETO-homologous proteins via NHR2, thereby multiplying NHR domain contacts. As shown recently, RUNX1/ETO retains oncogenic activity upon either deletion of the NHR3 + 4 N-CoR/SMRT interaction domain or substitution of the NHR2 tetramer domain. Thus, we aimed to clarify the specificities of the NHR domains. A C-terminally NHR3 + 4 truncated RUNX1/ETO containing a heterologous, structurally highly related non-NHR2 tetramer interface translocated into the nucleus and bound to RUNX1 consensus motifs. However, it failed to interact with ETO-homologues, repress RUNX1 targets, and transform progenitors. Surprisingly, transforming capacity was fully restored by C-terminal fusion with ETO's NHR4 zinc-finger or the repressor domain 3 of N-CoR, while other repression domains failed. With an inducible protein assembly system, we further demonstrated that NHR4 domain activity is critically required early in the establishment of progenitor cultures expressing the NHR2 exchanged truncated RUNX1/ETO. Together, we can show that NHR2 and NHR4 domains can be replaced by heterologous protein domains conferring tetramerization and repressor functions, thus showing that the NHR2 and NHR4 domain structures do not have irreplaceable functions concerning RUNX1/ETO activity for the establishment of human CD34+ cell expansion. We could resemble the function of RUNX1/ETO through modular recomposition with protein domains from RUNX1, ETO, BCR and N-CoR without any NHR2 and NHR4 sequences. As most transcriptional repressor proteins do not comprise tetramerization domains, our results provide a possible explanation as to the reason that RUNX1 is recurrently found translocated to ETO family members, which all contain tetramer together with transcriptional repressor moieties.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Antigens, CD34 , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Protein Domains , RUNX1 Translocation Partner 1 Protein/chemistry , RUNX1 Translocation Partner 1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL