Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 287
Filter
1.
J Thromb Haemost ; 14(12): 2524-2535, 2016 12.
Article in English | MEDLINE | ID: mdl-27685868

ABSTRACT

Essentials The role of the cytoskeleton during megakaryocyte differentiation was examined. Human megakaryocytes are derived from in vitro cultured CD34+ cells. Cell division control protein 42 (CDC42) positively regulates proplatelet formation (PPF). Neural Wiskott-Aldrich syndrome protein, the main effector of CDC42 with Src positively regulates PPF. SUMMARY: Background Cytoskeletal rearrangements are essential for platelet release. The RHO small GTPase family, as regulators of the actin cytoskeleton, play an important role in proplatelet formation (PPF). In the neuronal system, CDC42 is involved in axon formation, a process that combines elongation and branching as for PPF. Objective To analyze the role of CDC42 and its effectors of the Wiskott-Aldrich syndrome protein (WASP) family in PPF. Methods Human megakaryocytes (MKs) were obtained from CD34+ cells. Inhibition of CDC42 in MKs was performed with the chemical inhibitor CASIN or with an active or a dominant-negative form of CDC42. The knock-down of N-WASP was obtained with a small hairpin RNA strategy Results Herein, we show that CDC42 activity increased during MK differentiation. The use of the chemical inhibitor CASIN or of an active or a dominant-negative form of CDC42 demonstrated that CDC42 positively regulated PPF in vitro. We determined that N-WASP, but not WASP, regulated PPF. We found that N-WASP knockdown led to a marked decrease in PPF, owing to a defect in the demarcation membrane system (DMS). This was associated with RHOA activation, and a concomitant augmentation in the phosphorylation of mysosin light chain 2. Phosphorylation of N-WASP, creating a primed form of N-WASP, increased during MK differentiation. Phosphorylation inhibition by two Src family kinase inhibitors decreased PPF. Conclusions We conclude that N-WASP positively regulates DMS development and PPF, and that the Src family kinases in association with CDC42 regulate PPF through N-WASP.


Subject(s)
Antigens, CD34/metabolism , Blood Platelets/cytology , Wiskott-Aldrich Syndrome Protein/metabolism , cdc42 GTP-Binding Protein/metabolism , Axons/metabolism , Cell Differentiation , Cytoskeleton/metabolism , Genes, Dominant , Humans , Lentivirus/genetics , Megakaryocytes/cytology , Neurons/metabolism , Phosphorylation , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , src-Family Kinases/metabolism
4.
J Thromb Haemost ; 13(5): 851-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25736522

ABSTRACT

BACKGROUND: At the end of maturation, megakaryocytes (MKs) form long cytoplasmic extensions called proplatelets (PPT). Enormous changes in cytoskeletal structures cause PPT to extend further, to re-localize organelles such as mitochondria and to fragment, leading to platelet release. Two non-muscle myosin IIs (NMIIs) are expressed in MKs; however, only NMII-A (MYH9), but not NMII-B (MYH10), is expressed in mature MKs and is implicated in PPT formation. OBJECTIVES: To provide in vivo evidence on the specific role of NMII-A and IIB in MK PPT formation. METHODS: We studied two transgenic mouse models in which non-muscle myosin heavy chain (NMHC) II-A was genetically replaced either by II-B or by a chimeric NMHCII that combined the head domain of II-A with the rod and tail domains of II-B. RESULTS AND CONCLUSIONS: This work demonstrates that the kinetic properties of NM-IIA, depending on the N-terminal domain, render NMII-A the better NMII candidate to control PPT formation. Furthermore, the carboxyl-terminal domain determines myosin II localization in the constriction region of PPT and is responsible for the specific role of NMII in platelet release.


Subject(s)
Blood Platelets/metabolism , Myosin Type II/metabolism , Animals , Mice , Mice, Inbred C57BL
6.
Oncogene ; 34(10): 1323-32, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-24681953

ABSTRACT

STAT (Signal Transducer and Activator of Transcription) transcription factors are constitutively activated in most hematopoietic cancers. We previously identified a target gene, LPP/miR-28 (LIM domain containing preferred translocation partner in lipoma), induced by constitutive activation of STAT5, but not by transient cytokine-activated STAT5. miR-28 exerts negative effects on thrombopoietin receptor signaling and platelet formation. Here, we demonstrate that, in transformed hematopoietic cells, STAT5 and p53 must be synergistically bound to chromatin for induction of LPP/miR-28 transcription. Genome-wide association studies show that both STAT5 and p53 are co-localized on the chromatin at 463 genomic positions in proximal promoters. Chromatin binding of p53 is dependent on persistent STAT5 activation at these proximal promoters. The transcriptional activity of selected promoters bound by STAT5 and p53 was significantly changed upon STAT5 or p53 inhibition. Abnormal expression of several STAT5-p53 target genes (LEP, ATP5J, GTF2A2, VEGFC, NPY1R and NPY5R) is frequently detected in platelets of myeloproliferative neoplasm (MPN) patients, but not in platelets from healthy controls. In conclusion, persistently active STAT5 can recruit normal p53, like in the case of MPN cells, but also p53 mutants, such as p53 M133K in human erythroleukemia cells, leading to pathologic gene expression that differs from canonical STAT5 or p53 transcriptional programs.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Binding Sites , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Humans , Promoter Regions, Genetic , Protein Binding , Protein Transport
7.
Leukemia ; 28(2): 338-48, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23689514

ABSTRACT

Extranodal, nasal-type natural killer (NK)/T-cell lymphoma (NKCL) is an aggressive malignancy with poor prognosis in which, usually, signal transducer and activator of transcription 3 (STAT3) is constitutively activated and oncogenic. Here, we demonstrate that STAT3 activation mostly results from constitutive Janus kinase (JAK)3 phosphorylation on tyrosine 980, as observed in three of the four tested NKCL cell lines and in 20 of the 23 NKCL tumor samples under study. In one of the cell lines and in 4 of 19 (21%) NKCL primary tumor samples, constitutive JAK3 activation was related to an acquired mutation (A573V or V722I) in the JAK3 pseudokinase domain. We then show that constitutive activation of the JAK3/STAT3 pathway has a major role in NKCL cell growth and survival and in the invasive phenotype. Indeed, NKCL cell growth was slowed down in vitro by targeting JAK3 with chemical inhibitors or small-interfering RNAs. In a human NKCL xenograft mouse model, tumor growth was significantly delayed by the JAK3 inhibitor CP-690550. Altogether, the constitutive activation of JAK3, which can result from JAK3-activating mutations, is a frequent feature of NKCL that deserves to be tested as a therapeutic target.


Subject(s)
Janus Kinase 3/genetics , Lymphoma, Extranodal NK-T-Cell/genetics , Lymphoma, Extranodal NK-T-Cell/pathology , Mutation , Adult , Aged , Aged, 80 and over , Animals , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Lymphoma, Extranodal NK-T-Cell/drug therapy , Lymphoma, Extranodal NK-T-Cell/metabolism , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Phosphorylation , Piperidines/administration & dosage , Piperidines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
9.
Leukemia ; 28(3): 485-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24220273

ABSTRACT

Ten-Eleven Translocation-2 (TET2) inactivation through loss-of-function mutation, deletion and IDH1/2 (Isocitrate Dehydrogenase 1 and 2) gene mutation is a common event in myeloid and lymphoid malignancies. TET2 gene mutations similar to those observed in myeloid and lymphoid malignancies also accumulate with age in otherwise healthy subjects with clonal hematopoiesis. TET2 is one of the three proteins of the TET (Ten-Eleven Translocation) family, which are evolutionarily conserved dioxygenases that catalyze the conversion of 5-methyl-cytosine (5-mC) to 5-hydroxymethyl-cytosine (5-hmC) and promote DNA demethylation. TET dioxygenases require 2-oxoglutarate, oxygen and Fe(II) for their activity, which is enhanced in the presence of ascorbic acid. TET2 is the most expressed TET gene in the hematopoietic tissue, especially in hematopoietic stem cells. In addition to their hydroxylase activity, TET proteins recruit the O-linked ß-D-N-acetylglucosamine (O-GlcNAc) transferase (OGT) enzyme to chromatin, which promotes post-transcriptional modifications of histones and facilitates gene expression. The TET2 level is regulated by interaction with IDAX, originating from TET2 gene fission during evolution, and by the microRNA miR-22. TET2 has pleiotropic roles during hematopoiesis, including stem-cell self-renewal, lineage commitment and terminal differentiation of monocytes. Analysis of Tet2 knockout mice, which are viable and fertile, demonstrated that Tet2 functions as a tumor suppressor whose haploinsufficiency initiates myeloid and lymphoid transformations. This review summarizes the recently identified TET2 physiological and pathological functions and discusses how this knowledge influences our therapeutic approaches in hematological malignancies and possibly other tumor types.


Subject(s)
DNA-Binding Proteins/genetics , Hematologic Diseases/genetics , Hematopoiesis/genetics , Proto-Oncogene Proteins/genetics , Aging/genetics , Ascorbic Acid/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Genes, Tumor Suppressor , Humans , MicroRNAs/physiology , Mutation , Proto-Oncogene Proteins/metabolism
10.
Eur J Cancer ; 50(1): 159-69, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24135685

ABSTRACT

In this study, we investigated the role of NF-κB (canonical and alternative pathways) in the survival or proliferation of mantle cell lymphoma (MCL) cell lines. P50/p65 complexes were detectable by EMSA assays in 4/5 cell lines. Stable expression of a dominant-negative form of IkBa had no effect on proliferation nor on apoptosis in EBV-negative cell lines. Three out of 4 of the cell lines tested exhibited Phospho-p65 (Ser(536)). The alternative NF-κB pathway was not activated in 4/5 cell lines tested. Patient samples were also studied by Western blot, EMSA and Immunohistochemistry (IHC). No p50/p65 complexes were detected in cells freshly collected from 7 patients, but 1/7 cells exhibited Phospho-p65 (Ser(536)). We investigated immunohistochemically, the expression of NF-κB in 86 patients enrolled in two multicentre prospective trials. Patients with MCL exhibiting negative or positive cytoplasmic expression of NF-κB had a median overall survival of 35.7months compared to 22.4months for patients with nuclear NF-κB expression (p=0.0193). All these data suggest that NF-κB does not play a key role in proliferation and apoptotic processes in MCL cell lines. In patient samples, the presence of p65 in the nucleus reflecting NF-κB activation is rare but associated with a poor outcome.


Subject(s)
Lymphoma, Mantle-Cell/metabolism , NF-kappa B/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/isolation & purification , Humans , Immunohistochemistry , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/virology , Middle Aged
11.
J Thromb Haemost ; 11(12): 2163-75, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24165359

ABSTRACT

BACKGROUND: Mutations in the MYH9 gene cause autosomal dominant MYH9-related diseases (MYH9-RD) that associate macrothrombocytopenia with various other clinical conditions. The mechanisms giving rise to giant platelets remain poorly understood. OBJECTIVES/PATIENTS: To study the proplatelet formation (PPF) derived from megakaryocytes (MKs) generated in vitro from 11 patients with MYH9-RD with different mutations, compared with controls. METHODS: Proplatelet formation from cultured patients' MKs was evaluated with or without blebbistatin or the ROCK inhibitor Y27632. Myosin IIA and actin distribution were studied in spreading MKs on different surfaces by immunoconfocal analysis. Kinetic studies of contractility were performed on spreading MKs and the impact of blebbistatin on the maturation of the patients' MKs was evaluated by electron microscopy. RESULTS AND CONCLUSIONS: We show that in vitro MKs of 11 patients formed significantly fewer proplatelets than controls. MKs from MYH9-RD displayed an abnormal spreading on polylysine, fibronectin and collagen, with a disorganized actin network and a marked increase in stress fiber formation. Traction force microscopy studies demonstrated an elevated level of contractile forces in adherent mutated MKs. The myosin II inhibitor blebbistatin and the ROCK inhibitor Y27632 both rescued the proplatelet formation defect and normalized the ultrastructural characteristics of MYH9-RD MKs. Altogether, our results show that in MYH9-RD, mutations modify the overall MYH9 function and provoke a proplatelet defect through an excess of actomyosin contractility in spreading MKs. These results may promote new therapeutic strategies aimed at reducing this actomyosin contractility.


Subject(s)
Actomyosin/metabolism , Blood Platelets/cytology , Molecular Motor Proteins/physiology , Myosin Heavy Chains/physiology , Nonmuscle Myosin Type IIA/antagonists & inhibitors , Thrombocytopenia/pathology , Blood Platelets/metabolism , Cells, Cultured , Heterocyclic Compounds, 4 or More Rings/metabolism , Humans , Molecular Motor Proteins/genetics , Mutation , Myosin Heavy Chains/genetics , Thrombocytopenia/metabolism
12.
Cell Death Dis ; 4: e738, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23887629

ABSTRACT

Abexinostat is a pan histone deacetylase inhibitor (HDACi) that demonstrates efficacy in malignancy treatment. Like other HDACi, this drug induces a profound thrombocytopenia whose mechanism is only partially understood. We have analyzed its effect at doses reached in patient plasma on in vitro megakaryopoiesis derived from human CD34(+) cells. When added at day 0 in culture, abexinostat inhibited CFU-MK growth, megakaryocyte (MK) proliferation and differentiation. These effects required only a short incubation period. Decreased proliferation was due to induction of apoptosis and was not related to a defect in TPO/MPL/JAK2/STAT signaling. When added later (day 8), the compound induced a dose-dependent decrease (up to 10-fold) in proplatelet (PPT) formation. Gene profiling from MK revealed a silencing in the expression of DNA repair genes with a marked RAD51 decrease at protein level. DNA double-strand breaks were increased as attested by elevated γH2AX phosphorylation level. Moreover, ATM was phosphorylated leading to p53 stabilization and increased BAX and p21 expression. The use of a p53 shRNA rescued apoptosis, and only partially the defect in PPT formation. These results suggest that HDACi induces a thrombocytopenia by a p53-dependent mechanism along MK differentiation and a p53-dependent and -independent mechanism for PPT formation.


Subject(s)
Benzofurans/adverse effects , Histone Deacetylase Inhibitors/adverse effects , Hydroxamic Acids/adverse effects , Thrombocytopenia/chemically induced , Thrombocytopenia/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Benzofurans/administration & dosage , Cell Growth Processes/physiology , DNA Repair , Histone Deacetylase Inhibitors/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Phosphorylation , Signal Transduction , Thrombocytopenia/genetics , Tumor Suppressor Protein p53/genetics
13.
J Thromb Haemost ; 11(9): 1730-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23782903

ABSTRACT

BACKGROUND: The molecular bases of the cellular changes that occur during human megakaryocyte (MK) ontogeny remain unknown, and may be important for understanding the significance of MK differentiation from human embryonic stem cells (hESCs) METHODS: We optimized the differentiation of MKs from hESCs, and compared these with MKs obtained from primary human hematopoietic tissues at different stages of development. RESULTS: Transcriptome analyses revealed a close relationship between hESC-derived and fetal liver-derived MKs, and between neonate-derived and adult-derived MKs. Major changes in the expression profiles of cell cycle and transcription factors (TFs), including MYC and LIN28b, and MK-specific regulators indicated that MK maturation progresses during ontogeny towards an increase in MK ploidy and a platelet-forming function. Important genes, including CXCR4, were regulated by an on-off mechanism during development. DISCUSSION: Our analysis of the pattern of TF network and signaling pathways was consistent with a growing specialization of MKs towards hemostasis during ontogeny, and support the idea that MKs derived from hESCs reflect primitive hematopoiesis.


Subject(s)
Hematopoiesis , Megakaryocytes/cytology , Flow Cytometry , Gene Expression Profiling , Humans , Megakaryocytes/metabolism , Real-Time Polymerase Chain Reaction
15.
Leukemia ; 27(11): 2187-95, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23558526

ABSTRACT

Although other mutations may predate the acquisition of the JAK2(V617F) mutation, the latter is sufficient to drive the disease phenotype observed in BCR-ABL-negative myeloproliferative neoplasms (MPNs). One of the consequences of JAK2(V617F) is genetic instability that could explain JAK2(V617F)-mediated MPN progression and heterogeneity. Here, we show that JAK2(V617F) induces the accumulation of reactive oxygen species (ROS) in the hematopoietic stem cell compartment of a knock-in (KI) mouse model and in patients with JAK2(V617F) MPNs. JAK2(V617F)-dependent ROS elevation was partly mediated by an AKT-induced decrease in catalase expression and was accompanied by an increased number of 8-oxo-guanines and DNA double-strand breaks (DSBs). Moreover, there was evidence for a mitotic recombination event in mice resulting in loss of heterozygosity of Jak2(V617F). Mice engrafted with 30% of Jak2(V617F) KI bone marrow (BM) cells developed a polycythemia vera-like disorder. Treatment with the anti-oxidant N-acetylcysteine (NAC) substantially restored blood parameters and reduced damages to DNA. Furthermore, NAC induced a marked decrease in splenomegaly with reduction in the frequency of the Jak2(V617F)-positive hematopoietic progenitors in BM and spleen. Altogether, overproduction of ROS is a mediator of JAK2(V617F)-induced DNA damages that promote disease progression. Targeting ROS accumulation might prevent the development of JAK2(V617F) MPNs.


Subject(s)
Janus Kinase 2/physiology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Point Mutation/genetics , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Blotting, Western , Bone Marrow Transplantation , Case-Control Studies , DNA Damage/drug effects , Disease Progression , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Oncogene ; 32(21): 2601-13, 2013 May 23.
Article in English | MEDLINE | ID: mdl-22869151

ABSTRACT

The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is central to signaling by cytokine receptors, a superfamily of more than 30 transmembrane proteins that recognize specific cytokines, and is critical in blood formation and immune response. Many of those receptors transmit anti-apoptotic, proliferative and differentiation signals, and their expression and functions are critical for the formation of blood lineages. Several cancers, including blood malignancies, have been associated with constitutive activation of members of the STAT family, which normally require JAK-mediated tyrosine phosphorylation for transcriptional activation. More recently, human myeloproliferative neoplasms were discovered to be associated with a unique acquired somatic mutation in JAK2 (JAK2 V617F), rare exon 12 JAK2 mutations, or thrombopoietin receptor mutations that constitutively activate wild-type JAK2. Prompted by these observations, many studies have explored the possibility that JAKs, cytokine receptors, or other components of the JAK/STAT pathway are mutated or upregulated in several hematological malignancies. This has been observed in certain pediatric acute lymphoblastic leukemias and adult T-cell lymphoblastic leukemias, and overexpression of JAK2 seems to be important in Hodgkin lymphoma. Here we discuss the nature and respective contribution of mutations dysregulating the JAK/STAT pathway in hematological malignancies and present examples in which such mutations drive the disease, contribute to the phenotype, or provide a survival and proliferative advantage. JAK inhibitors are making their way into the therapeutic arsenal (for example, in myelofibrosis), and we discuss the possibility that other hematological diseases might benefit from treatment with these inhibitors in combination with other agents.


Subject(s)
Hematologic Neoplasms/metabolism , Hodgkin Disease/metabolism , Janus Kinase 2/biosynthesis , Mutation, Missense , Neoplasm Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Amino Acid Substitution , Animals , Exons/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hodgkin Disease/drug therapy , Hodgkin Disease/genetics , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Neoplasm Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/therapeutic use , STAT Transcription Factors/genetics
17.
Cell Death Dis ; 3: e396, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-23034331

ABSTRACT

The chemokine receptor CXCR4 favors the interaction of acute myeloid leukemia (AML) cells with their niche but the extent to which it participates in pathogenesis is unclear. Here, we show that CXCR4 expression at the surface of leukemic cells allowed distinguishing CXCR4 (high) from CXCR4(neg/low) AML patients. When high levels of CXCR4 are expressed at the surface of AML cells, blocking the receptor function with small molecule inhibitors could promote leukemic cell death and reduce NOD/Shi-scid/IL-2Rγ(null) (NOG) leukemia-initiating cells (LICs). Conversely, these drugs had no efficacy when AML cells do not express CXCR4 or when they do not respond to chemokine CXC motif ligand 12 (CXCL12). Functional analysis showed a greater mobilization of leukemic cells and LICs in response to drugs, suggesting that they target the interaction between leukemic cells and their supportive bone marrow microenvironment. In addition, increased apoptosis of leukemic cells in vitro and in vivo was observed. CXCR4 expression level on AML blast cells and their migratory response to CXCL12 are therefore predictive of the response to the inhibitors and could be used as biomarkers to select patients that could potentially benefit from the drugs.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Receptors, CXCR4/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Apoptosis/drug effects , Benzylamines , Chemokine CXCL12/metabolism , Child, Preschool , Cyclams , Disease Models, Animal , Female , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Middle Aged , Receptors, CXCR4/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
19.
Cell Death Dis ; 3: e356, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22833095

ABSTRACT

Diamond-Blackfan anemia (DBA) is caused by aberrant ribosomal biogenesis due to ribosomal protein (RP) gene mutations. To develop mechanistic understanding of DBA pathogenesis, we studied CD34⁺ cells from peripheral blood of DBA patients carrying RPL11 and RPS19 ribosomal gene mutations and determined their ability to undergo erythroid differentiation in vitro. RPS19 mutations induced a decrease in proliferation of progenitor cells, but the terminal erythroid differentiation was normal with little or no apoptosis. This phenotype was related to a G0/G1 cell cycle arrest associated with activation of the p53 pathway. In marked contrast, RPL11 mutations led to a dramatic decrease in progenitor cell proliferation and a delayed erythroid differentiation with a marked increase in apoptosis and G0/G1 cell cycle arrest with activation of p53. Infection of cord blood CD34⁺ cells with specific short hairpin (sh) RNAs against RPS19 or RPL11 recapitulated the two distinct phenotypes in concordance with findings from primary cells. In both cases, the phenotype has been reverted by shRNA p53 knockdown. These results show that p53 pathway activation has an important role in pathogenesis of DBA and can be independent of the RPL11 pathway. These findings shed new insights into the pathogenesis of DBA.


Subject(s)
Anemia, Diamond-Blackfan/metabolism , Erythroid Cells/metabolism , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Antigens, CD34/metabolism , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Child, Preschool , Erythroid Cells/cytology , Female , G1 Phase Cell Cycle Checkpoints , Humans , Infant , Infant, Newborn , Male , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Oncogene ; 31(10): 1323-33, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-21785463

ABSTRACT

JAK2(V617F) is a gain of function mutation that promotes cytokine-independent growth of myeloid cells and accounts for a majority of myeloproliferative neoplasms (MPN). Mutations in p53 are rarely found in these diseases before acute leukemia transformation, but this does not rule out a role for p53 deregulation in disease progression. Using Ba/F3-EPOR cells and ex vivo cultured CD34(+) cells from MPN patients, we demonstrate that expression of JAK2(V617F) affected the p53 response to DNA damage. We show that E3 ubiquitin ligase MDM2 accumulated in these cells, due to an increased translation of MDM2 mRNA. Accumulation of the La autoantigen, which interacts with MDM2 mRNA and promotes its translation, was responsible for the increase in MDM2 protein level and the subsequent degradation of p53 after DNA damage. Downregulation of La protein or cell treatment with nutlin-3, a MDM2 antagonist, restored the p53 response to DNA damage and the cytokine-dependence of Ba/F3-EPOR-JAK2(V617F) cells. Altogether, these data indicate that the JAK2(V617F) mutation affects p53 response to DNA damage through the upregulation of La antigen and accumulation of MDM2. They also suggest that p53 functional inactivation accounts for the cytokine hypersensitivity of JAK2(V617F) MPN and might have a role in disease progression.


Subject(s)
Autoantigens/physiology , Hematologic Neoplasms/etiology , Janus Kinase 2/physiology , Myeloproliferative Disorders/etiology , Proto-Oncogene Proteins c-mdm2/physiology , Ribonucleoproteins/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Autoantigens/analysis , Cell Line , Cytokines/physiology , DNA Damage , Humans , Janus Kinase 2/genetics , Mice , Mutation , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/analysis , Ribonucleoproteins/analysis , SS-B Antigen
SELECTION OF CITATIONS
SEARCH DETAIL