Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34930825

ABSTRACT

SF3B1 is the most frequently mutated RNA splicing factor in cancer, including in ∼25% of myelodysplastic syndromes (MDS) patients. SF3B1-mutated MDS, which is strongly associated with ringed sideroblast morphology, is characterized by ineffective erythropoiesis, leading to severe, often fatal anemia. However, functional evidence linking SF3B1 mutations to the anemia described in MDS patients harboring this genetic aberration is weak, and the underlying mechanism is completely unknown. Using isogenic SF3B1 WT and mutant cell lines, normal human CD34 cells, and MDS patient cells, we define a previously unrecognized role of the kinase MAP3K7, encoded by a known mutant SF3B1-targeted transcript, in controlling proper terminal erythroid differentiation, and show how MAP3K7 missplicing leads to the anemia characteristic of SF3B1-mutated MDS, although not to ringed sideroblast formation. We found that p38 MAPK is deactivated in SF3B1 mutant isogenic and patient cells and that MAP3K7 is an upstream positive effector of p38 MAPK. We demonstrate that disruption of this MAP3K7-p38 MAPK pathway leads to premature down-regulation of GATA1, a master regulator of erythroid differentiation, and that this is sufficient to trigger accelerated differentiation, erythroid hyperplasia, and ultimately apoptosis. Our findings thus define the mechanism leading to the severe anemia found in MDS patients harboring SF3B1 mutations.


Subject(s)
Anemia/metabolism , Erythropoiesis , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mutation , Myelodysplastic Syndromes/metabolism , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , Anemia/genetics , Anemia/pathology , Cell Differentiation/genetics , Erythroid Cells/metabolism , Erythroid Cells/pathology , Humans , K562 Cells , MAP Kinase Kinase Kinases/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Phosphoproteins/genetics , RNA Splicing Factors/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Mol Cell ; 76(1): 82-95.e7, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31474574

ABSTRACT

SF3B1, which encodes an essential spliceosomal protein, is frequently mutated in myelodysplastic syndromes (MDS) and many cancers. However, the defect of mutant SF3B1 is unknown. Here, we analyzed RNA sequencing data from MDS patients and confirmed that SF3B1 mutants use aberrant 3' splice sites. To elucidate the underlying mechanism, we purified complexes containing either wild-type or the hotspot K700E mutant SF3B1 and found that levels of a poorly studied spliceosomal protein, SUGP1, were reduced in mutant spliceosomes. Strikingly, SUGP1 knockdown completely recapitulated the splicing errors, whereas SUGP1 overexpression drove the protein, which our data suggest plays an important role in branchsite recognition, into the mutant spliceosome and partially rescued splicing. Other hotspot SF3B1 mutants showed similar altered splicing and diminished interaction with SUGP1. Our study demonstrates that SUGP1 loss is a common defect of spliceosomes with disease-causing SF3B1 mutations and, because this defect can be rescued, suggests possibilities for therapeutic intervention.


Subject(s)
Leukemia, Erythroblastic, Acute/metabolism , Mutation , Myelodysplastic Syndromes/metabolism , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , RNA Splicing , Spliceosomes/metabolism , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , HEK293 Cells , Humans , K562 Cells , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Phenotype , Phosphoproteins/genetics , Protein Binding , RNA Splicing Factors/genetics , Spliceosomes/genetics , Spliceosomes/pathology
3.
Nat Med ; 22(12): 1488-1495, 2016 12.
Article in English | MEDLINE | ID: mdl-27841873

ABSTRACT

Although the majority of patients with acute myeloid leukemia (AML) initially respond to chemotherapy, many of them subsequently relapse, and the mechanistic basis for AML persistence following chemotherapy has not been determined. Recurrent somatic mutations in DNA methyltransferase 3A (DNMT3A), most frequently at arginine 882 (DNMT3AR882), have been observed in AML and in individuals with clonal hematopoiesis in the absence of leukemic transformation. Patients with DNMT3AR882 AML have an inferior outcome when treated with standard-dose daunorubicin-based induction chemotherapy, suggesting that DNMT3AR882 cells persist and drive relapse. We found that Dnmt3a mutations induced hematopoietic stem cell expansion, cooperated with mutations in the FMS-like tyrosine kinase 3 gene (Flt3ITD) and the nucleophosmin gene (Npm1c) to induce AML in vivo, and promoted resistance to anthracycline chemotherapy. In patients with AML, the presence of DNMT3AR882 mutations predicts minimal residual disease, underscoring their role in AML chemoresistance. DNMT3AR882 cells showed impaired nucleosome eviction and chromatin remodeling in response to anthracycline treatment, which resulted from attenuated recruitment of histone chaperone SPT-16 following anthracycline exposure. This defect led to an inability to sense and repair DNA torsional stress, which resulted in increased mutagenesis. Our findings identify a crucial role for DNMT3AR882 mutations in driving AML chemoresistance and highlight the importance of chromatin remodeling in response to cytotoxic chemotherapy.


Subject(s)
Anthracyclines/therapeutic use , Chromatin Assembly and Disassembly/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival , DNA Methyltransferase 3A , Daunorubicin/therapeutic use , Hematopoietic Stem Cells , Humans , Immunoblotting , Immunoprecipitation , Leukemia, Myeloid, Acute/drug therapy , Mass Spectrometry , Mice , Mutation , Nuclear Proteins/genetics , Nucleophosmin , Nucleosomes/metabolism , fms-Like Tyrosine Kinase 3/genetics
4.
Proc Natl Acad Sci U S A ; 112(34): E4726-34, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261309

ABSTRACT

Serine/arginine-rich splicing factor 2 (SRSF2) is an RNA-binding protein that plays important roles in splicing of mRNA precursors. SRSF2 mutations are frequently found in patients with myelodysplastic syndromes and certain leukemias, but how these mutations affect SRSF2 function has only begun to be examined. We used clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 nuclease to introduce the P95H mutation to SRSF2 in K562 leukemia cells, generating an isogenic model so that splicing alterations can be attributed solely to mutant SRSF2. We found that SRSF2 (P95H) misregulates 548 splicing events (<1% of total). Of these events, 374 involved the inclusion of cassette exons, and the inclusion was either increased (206) or decreased (168). We detected a specific motif (UCCA/UG) enriched in the more-included exons and a distinct motif (UGGA/UG) in the more-excluded exons. RNA gel shift assays showed that a mutant SRSF2 derivative bound more tightly than its wild-type counterpart to RNA sites containing UCCAG but bound less tightly to UGGAG sites. Thus in most cases the pattern of exon inclusion or exclusion correlated with stronger or weaker RNA binding, respectively. We further show that the P95H mutation does not affect other functions of SRSF2, i.e., protein-protein interactions with key splicing factors. Our results thus demonstrate that the P95H mutation positively or negatively alters the binding affinity of SRSF2 for cognate RNA sites in target transcripts, leading to misregulation of exon inclusion. Our findings shed light on the mechanism of the disease-associated SRSF2 mutation in splicing regulation and also reveal a group of misspliced mRNA isoforms for potential therapeutic targeting.


Subject(s)
Mutation , Nuclear Proteins/genetics , RNA Splicing , RNA/metabolism , Ribonucleoproteins/genetics , Binding Sites , CRISPR-Associated Proteins/genetics , Exons , Humans , Nuclear Proteins/physiology , Ribonucleoproteins/physiology , Serine-Arginine Splicing Factors
5.
Eur J Cancer ; 50(11): 1982-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24835034

ABSTRACT

Heat shock protein (Hsp) 90 is a key component of the super-chaperone complex that maintains functionally active conformation of various client proteins. Many of these client proteins regulate important nodal points in multiple signalling pathways that promote cancer cell growth and survival. Inhibitors of Hsp90, therefore, have the potential of functioning as anti-cancer agents with pleiotropic effects. Identification of novel Hsp90 inhibitors with more favourable pharmacological properties is a priority in cancer therapy. To achieve this goal, we screened a compound library using a biochemical assay based on refolding of denatured firefly luciferase. The assay revealed high sensitivity, reliability and reproducibility with a Z-factor of 0.81 ± 0.17. Six Hsp90 inhibitory compounds identified by this screening with IC50 values between 1.0 and 6 µM were further characterised for anti-proliferative activity by Cell Titer-Blue Cell Viability Assay using multiple tumour cell lines. Of particular interest was ONO4140 with lowest GI50 values in three different cancer cell lines viz; DU-145, BT-474 and K562 cell lines. This study also revealed that short-term exposure of tumour cells with ONO4140 is sufficient to inhibit the catalytic activity of Hsp90, evaluated through disruption of Hsp90-p23 association by immunoprecipitation. This short term exposure appears to initiate events like degradation of Hsp90 client proteins such as ErbB2/Her-2 and Akt with concomitant inhibition of survival signalling leading to the apoptotic death of tumour cells as seen by western blotting and Caspase Glow-3,7 assay. The study also reveals that apoptosis following Hsp90 inhibition with ONO4140 occurs via Caspase9-Caspase3 intrinsic apoptotic pathway, a process that is likely triggered by inactivation of Akt. In conclusion, we have identified a novel class of synthetic compounds which show potent Hsp90 inhibitory action in preclinical studies. The discovery of this novel class of synthetic Hsp90 inhibitors with simple chemical backbone allows us to conduct further structural modifications to improve their potency and pharmacokinetic properties for use in cancer therapy.


Subject(s)
Chalcones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Sulfones/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Humans , K562 Cells , Luciferases, Firefly/chemistry , Luciferases, Firefly/metabolism , Rabbits , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 111(8): 3122-7, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516162

ABSTRACT

The B-myb (MYBL2) gene is a member of the MYB family of transcription factors and is involved in cell cycle regulation, DNA replication, and maintenance of genomic integrity. However, its function during adult development and hematopoiesis is unknown. We show here that conditional inactivation of B-myb in vivo results in depletion of the hematopoietic stem cell (HSC) pool, leading to profound reductions in mature lymphoid, erythroid, and myeloid cells. This defect is autonomous to the bone marrow and is first evident in stem cells, which accumulate in the S and G2/M phases. B-myb inactivation also causes defects in the myeloid progenitor compartment, consisting of depletion of common myeloid progenitors but relative sparing of granulocyte-macrophage progenitors. Microarray studies indicate that B-myb-null LSK(+) cells differentially express genes that direct myeloid lineage development and commitment, suggesting that B-myb is a key player in controlling cell fate. Collectively, these studies demonstrate that B-myb is essential for HSC and progenitor maintenance and survival during hematopoiesis.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Myeloid Progenitor Cells/physiology , Trans-Activators/metabolism , Animals , Bone Marrow Transplantation , Crosses, Genetic , DNA Primers/genetics , Flow Cytometry , Immunoblotting , Mice , Mice, Inbred C57BL , Microarray Analysis , Reverse Transcriptase Polymerase Chain Reaction
7.
Cell Cycle ; 11(18): 3504-12, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22918254

ABSTRACT

The differentiation of myeloid progenitors to mature, terminally differentiated cells is a highly regulated process. Here, we showed that conditional disruption of the c-myb proto-oncogene in adult mice resulted in dramatic reductions in CMP, GMP and MEP myeloid progenitors, leading to a reduction of neutrophils, basophils, monocytes and platelets in peripheral blood. In addition, c-myb plays a critical role at multiple stages of myeloid development, from multipotent CMP and bipotent GMP to unipotent CFU-G and CFU-M progenitor cells. c-myb controls the differentiation of these cells and is required for the proper commitment, maturation and normal differentiation of CMPs and GMPs. Specifically, c-myb regulates the precise commitment to the megakaryocytic and granulo-monocytic pathways and governs the granulocytic-monocytic lineage choice. c-myb is also required for the commitment along the granulocytic pathway for early myeloid progenitor cells and for the maturation of committed precursor cells along this pathway. On the other hand, disruption of the c-myb gene favors the commitment to the monocytic lineage, although monocytic development was abnormal with cells appearing more mature with atypical CD41 surface markers. These results demonstrate that c-myb plays a pivotal role in the regulation of multiple stages in adult myelogenesis.


Subject(s)
Aging/metabolism , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Animals , Blood Cell Count , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Differentiation/genetics , Cell Proliferation , Gene Expression Regulation , Integrases/metabolism , Megakaryocyte-Erythroid Progenitor Cells/cytology , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Neutrophils/cytology , Neutrophils/metabolism
8.
Cancer Cell ; 20(6): 728-40, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22137796

ABSTRACT

We report that diffuse large B cell lymphoma (DLBCL) commonly fails to express cell-surface molecules necessary for the recognition of tumor cells by immune-effector cells. In 29% of cases, mutations and deletions inactivate the ß2-Microglobulin gene, thus preventing the cell-surface expression of the HLA class-I (HLA-I) complex that is necessary for recognition by CD8(+) cytotoxic T cells. In 21% of cases, analogous lesions involve the CD58 gene, which encodes a molecule involved in T and natural killer cell-mediated responses. In addition to gene inactivation, alternative mechanisms lead to aberrant expression of HLA-I and CD58 in >60% of DLBCL. These two events are significantly associated in this disease, suggesting that they are coselected during lymphomagenesis for their combined role in escape from immune-surveillance.


Subject(s)
CD58 Antigens/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Mutation , beta 2-Microglobulin/genetics , CD58 Antigens/immunology , CD58 Antigens/metabolism , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , DNA Copy Number Variations , DNA Mutational Analysis , Genetic Association Studies , Genotype , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/physiology , Lymphoma, Large B-Cell, Diffuse/genetics , Polymorphism, Single Nucleotide , Protein Stability , Transcription, Genetic , beta 2-Microglobulin/metabolism
9.
Proc Natl Acad Sci U S A ; 106(51): 21689-94, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19955420

ABSTRACT

Hematopoietic stem cells (HSCs) have a unique capacity to undergo self-renewal and multi-lineage differentiation to provide a lifetime supply of mature blood cells. By using conditional knockout technology, we disrupted the c-myb proto-oncogene specifically in adult bone marrow (BM) to demonstrate that this transcription factor is a regulator of self-renewal and multi-lineage differentiation of adult HSCs. Targeted disruption of the c-myb gene resulted in a critical depletion of the HSC pool. In addition, BM hematopoiesis in adult mice was impaired, resulting in profound reductions of various hematopoietic lineages including neutrophilic, monocytic, B lymphoid, erythroid, and, unexpectedly, megakaryocytic cells. Serial BM transplantation into lethally irradiated recipient mice indicated an essential role for c-myb in the self-renewal process. Furthermore, in vitro functional assays demonstrated that deletion of the c-myb gene leads to a slightly reduced proliferative capacity and an aberrant and accelerated differentiation of HSCs. In addition to long-term HSCs, functional studies also show that c-myb plays a critical role in short-term HSCs and multi-potential progenitors. Collectively, our data indicate a critical role for c-myb in adult BM hematopoiesis and in self-renewal and multi-lineage differentiation of adult HSCs.


Subject(s)
Cell Differentiation , Cell Proliferation , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-myb/genetics , Animals , Gene Expression , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout
10.
Proc Natl Acad Sci U S A ; 101(41): 14853-8, 2004 Oct 12.
Article in English | MEDLINE | ID: mdl-15466706

ABSTRACT

Previous reports have suggested that the protooncogene c-myb participates in T cell development in the thymus and mature T cell proliferation. We have generated two T cell-specific c-myb knockout mouse models, myb/LckCre and myb/CD4Cre. We have demonstrated that c-myb is required for the development of thymocytes at the DN3 stage, for survival and proliferation of double-positive thymocytes, for differentiation of single-positive CD4 and CD8 T cells, and for the proliferative responses of mature T cells. In addition, our data show that c-myb is directly involved in the formation of double-positive CD4+CD8+CD25+, CD4+CD25+, and CD8+CD25+ T cells, developmental processes that may imply a role for c-myb in autoimmune dysfunction.


Subject(s)
Genes, myb/genetics , Proto-Oncogene Proteins c-myb/genetics , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Animals , Autoimmune Diseases/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Division , Cell Survival , Flow Cytometry , Gene Expression Regulation , Genotype , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myb/deficiency , Restriction Mapping , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...