Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Blood ; 140(23): 2463-2476, 2022 12 08.
Article in English | MEDLINE | ID: mdl-35960849

ABSTRACT

Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of hematological cancers arising from the malignant transformation of mature T cells. In a cohort of 28 PTCL cases, we identified recurrent overexpression of MYCN, a member of the MYC family of oncogenic transcription factors. Approximately half of all PTCL cases was characterized by a MYC expression signature. Inducible expression of MYCN in lymphoid cells in a mouse model caused T-cell lymphoma that recapitulated human PTCL with an MYC expression signature. Integration of mouse and human expression data identified EZH2 as a key downstream target of MYCN. Remarkably, EZH2 was found to be an essential cofactor for the transcriptional activation of the MYCN-driven gene expression program, which was independent of methyltransferase activity but dependent on phosphorylation by CDK1. MYCN-driven T-cell lymphoma was sensitive to EZH2 degradation or CDK1 inhibition, which displayed synergy with US Food and Drug Administration-approved histone deacetylase (HDAC) inhibitors.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Lymphoma, T-Cell, Peripheral , N-Myc Proto-Oncogene Protein , Humans , Enhancer of Zeste Homolog 2 Protein/genetics , Lymphoma, T-Cell, Peripheral/genetics , N-Myc Proto-Oncogene Protein/genetics
2.
Haematologica ; 107(10): 2304-2317, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35354248

ABSTRACT

TAL1 is ectopically expressed in about 30% of T-cell acute lymphoblastic leukemia (T-ALL) due to chromosomal rearrangements leading to the STIL-TAL1 fusion genes or due to non-coding mutations leading to a de novo enhancer driving TAL1 expression. Analysis of sequence data from T-ALL cases demonstrates a significant association between TAL1 expression and activating mutations of the PI3K-AKT pathway. We investigated the oncogenic function of TAL1 and the possible cooperation with PI3K-AKT pathway activation using isogenic pro-T-cell cultures ex vivo and in vivo leukemia models. We found that TAL1 on its own suppressed T-cell growth, in part by affecting apoptosis genes, while the combination with AKT pathway activation reduced apoptosis and was strongly driving cell proliferation ex vivo and leukemia development in vivo. As a consequence, we found that TAL1+AKTE17K transformed cells are more sensitive to PI3K-AKT pathway inhibition compared to AKTE17K transformed cells, related to the negative effect of TAL1 in the absence of activated PI3K-AKT signaling. We also found that both TAL1 and PI3K-AKT signaling increased the DNA-repair signature in T cells resulting in synergy between PARP and PI3K-AKT pathway inhibition. In conclusion, we have developed a novel mouse model for TAL1+AKTE17K driven T-ALL development and have identified a vulnerability of these leukemia cells to PI3K-AKT and PARP inhibitors.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Animals , DNA , Mice , Phosphatidylinositol 3-Kinases/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , T-Lymphocytes/metabolism
4.
Nat Commun ; 12(1): 3705, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140493

ABSTRACT

Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of non-Hodgkin lymphomas with poor prognosis. Up to 30% of PTCL lack distinctive features and are classified as PTCL, not otherwise specified (PTCL-NOS). To further improve our understanding of the genetic landscape and biology of PTCL-NOS, we perform RNA-sequencing of 18 cases and validate results in an independent cohort of 37 PTCL cases. We identify FYN-TRAF3IP2, KHDRBS1-LCK and SIN3A-FOXO1 as new in-frame fusion transcripts, with FYN-TRAF3IP2 as a recurrent fusion detected in 8 of 55 cases. Using ex vivo and in vivo experiments, we demonstrate that FYN-TRAF3IP2 and KHDRBS1-LCK activate signaling pathways downstream of the T cell receptor (TCR) complex and confer therapeutic vulnerability to clinically available drugs.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphoma, T-Cell, Peripheral/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-fyn/genetics , RNA-Binding Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cohort Studies , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-fyn/metabolism , RNA-Binding Proteins/metabolism , RNA-Seq , Signal Transduction/genetics , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/metabolism
5.
Clin Cancer Res ; 26(21): 5747-5758, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32826328

ABSTRACT

PURPOSE: KPT-8602 (Eltanexor) is a second-generation exportin-1 (XPO1) inhibitor with potent activity against acute lymphoblastic leukemia (ALL) in preclinical models and with minimal effects on normal cells. In this study, we evaluated whether KPT-8602 would synergize with dexamethasone, vincristine, or doxorubicin, three drugs currently used for the treatment of ALL. EXPERIMENTAL DESIGN: First, we searched for the most synergistic combination of KPT-8602 with dexamethasone, vincristine, or doxorubicin in vitro in both B-ALL and T-ALL cell lines using proliferation and apoptosis as a readout. Next, we validated this synergistic effect by treatment of clinically relevant B- and T-ALL patient-derived xenograft models in vivo. Finally, we performed RNA-sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) to determine the mechanism of synergy. RESULTS: KPT-8602 showed strong synergism with dexamethasone on human B-ALL and T-ALL cell lines as well as in vivo in three patient-derived ALL xenografts. Compared with single-drug treatment, the drug combination caused increased apoptosis and led to histone depletion. Mechanistically, integration of ChIP-seq and RNA-seq data revealed that addition of KPT-8602 to dexamethasone enhanced the activity of the glucocorticoid receptor (NR3C1) and led to increased inhibition of E2F-mediated transcription. We observed strong inhibition of E2F target genes related to cell cycle, DNA replication, and transcriptional regulation. CONCLUSIONS: Our preclinical study demonstrates that KPT-8602 enhances the effects of dexamethasone to inhibit B-ALL and T-ALL cells via NR3C1- and E2F-mediated transcriptional complexes, allowing to achieve increased dexamethasone effects for patients.


Subject(s)
Dexamethasone/pharmacology , Doxorubicin/pharmacology , Karyopherins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Karyopherins/antagonists & inhibitors , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Vincristine/pharmacology , Xenograft Model Antitumor Assays , Exportin 1 Protein
6.
Blood ; 134(16): 1323-1336, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31492675

ABSTRACT

The polycomb repressive complex 2, with core components EZH2, SUZ12, and EED, is responsible for writing histone 3 lysine 27 trimethylation histone marks associated with gene repression. Analysis of sequence data from 419 T-cell acute lymphoblastic leukemia (T-ALL) cases demonstrated a significant association between SUZ12 and JAK3 mutations. Here we show that CRISPR/Cas9-mediated inactivation of Suz12 cooperates with mutant JAK3 to drive T-cell transformation and T-ALL development. Gene expression profiling integrated with ChIP-seq and ATAC-seq data established that inactivation of Suz12 led to increased PI3K/mammalian target of rapamycin (mTOR), vascular endothelial growth factor (VEGF), and WNT signaling. Moreover, a drug screen revealed that JAK3/Suz12 mutant leukemia cells were more sensitive to histone deacetylase (HDAC)6 inhibition than JAK3 mutant leukemia cells. Among the broad genome and gene expression changes observed on Suz12 inactivation, our integrated analysis identified the PI3K/mTOR, VEGF/VEGF receptor, and HDAC6/HSP90 pathways as specific vulnerabilities in T-ALL cells with combined JAK3 and SUZ12 mutations.


Subject(s)
Cell Transformation, Neoplastic/genetics , Polycomb Repressive Complex 2/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction/physiology , Animals , Humans , Janus Kinase 3/genetics , Mice , Mutation , Neoplasm Proteins , Transcription Factors
7.
Cancer Cell ; 34(2): 271-285.e7, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30107177

ABSTRACT

The NUP214-ABL1 fusion is a constitutively activated tyrosine kinase that is significantly associated with overexpression of the TLX1 and TLX3 transcription factors in T cell acute lymphoblastic leukemia (T-ALL). Here we show that NUP214-ABL1 cooperates with TLX1 in driving T-ALL development using a transgenic mouse model and human T-ALL cells. Using integrated ChIP-sequencing, ATAC-sequencing, and RNA-sequencing data, we demonstrate that TLX1 and STAT5, the downstream effector of NUP214-ABL1, co-bind poised enhancer regions, and cooperatively activate the expression of key proto-oncogenes such as MYC and BCL2. Inhibition of STAT5, downregulation of TLX1 or MYC, or interference with enhancer function through BET-inhibitor treatment leads to reduction of target gene expression and induction of leukemia cell death.


Subject(s)
Enhancer Elements, Genetic , Homeodomain Proteins/physiology , Nuclear Pore Complex Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins/physiology , STAT5 Transcription Factor/physiology , Animals , Gene Fusion , Homeodomain Proteins/genetics , Humans , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/physiology , STAT5 Transcription Factor/genetics
9.
Haematologica ; 102(9): 1605-1616, 2017 09.
Article in English | MEDLINE | ID: mdl-28659337

ABSTRACT

Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma is characterized by 2p23/ALK aberrations, including the classic t(2;5)(p23;q35)/NPM1-ALK rearrangement present in ~80% of cases and several variant t(2p23/ALK) occurring in the remaining cases. The ALK fusion partners play a key role in the constitutive activation of the chimeric protein and its subcellular localization. Using various molecular technologies, we have characterized ALK fusions in eight recently diagnosed anaplastic large cell lymphoma cases with cytoplasmic-only ALK expression. The identified partner genes included EEF1G (one case), RNF213/ALO17 (one case), ATIC (four cases) and TPM3 (two cases). Notably, all cases showed copy number gain of the rearranged ALK gene, which is never observed in NPM1-ALK-positive lymphomas. We hypothesized that this could be due to lower expression levels and/or lower oncogenic potential of the variant ALK fusions. Indeed, all partner genes, except EEF1G, showed lower expression in normal and malignant T cells, in comparison with NPM1 In addition, we investigated the transformation potential of endogenous Npm1-Alk and Atic-Alk fusions generated by clustered regularly interspaced short palindromic repeats/Cas9 genome editing in Ba/F3 cells. We found that Npm1-Alk has a stronger transformation potential than Atic-Alk, and we observed a subclonal gain of Atic-Alk after a longer culture period, which was not observed for Npm1-Alk Taken together, our data illustrate that lymphomas driven by the variant ATIC-ALK fusion (and likely by RNF213-ALK and TPM3-ALK), but not the classic NPM1-ALK, require an increased dosage of the ALK hybrid gene to compensate for the relatively low and insufficient expression and signaling properties of the chimeric gene.


Subject(s)
Adenosine Triphosphatases/genetics , Gene Rearrangement , Hydroxymethyl and Formyl Transferases/genetics , Lymphoma, Large-Cell, Anaplastic/genetics , Multienzyme Complexes/genetics , Nucleotide Deaminases/genetics , Oncogene Proteins, Fusion/genetics , Receptor Protein-Tyrosine Kinases/genetics , Translocation, Genetic , Tropomyosin/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Aged , Anaplastic Lymphoma Kinase , Child, Preschool , Female , Humans , Lymphoma, Large-Cell, Anaplastic/pathology , Male , Middle Aged , Nucleophosmin
10.
Blood ; 124(20): 3092-100, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25193870

ABSTRACT

JAK3 is a tyrosine kinase that associates with the common γ chain of cytokine receptors and is recurrently mutated in T-cell acute lymphoblastic leukemia (T-ALL). We tested the transforming properties of JAK3 pseudokinase and kinase domain mutants using in vitro and in vivo assays. Most, but not all, JAK3 mutants transformed cytokine-dependent Ba/F3 or MOHITO cell lines to cytokine-independent proliferation. JAK3 pseudokinase mutants were dependent on Jak1 kinase activity for cellular transformation, whereas the JAK3 kinase domain mutant could transform cells in a Jak1 kinase-independent manner. Reconstitution of the IL7 receptor signaling complex in 293T cells showed that JAK3 mutants required receptor binding to mediate downstream STAT5 phosphorylation. Mice transplanted with bone marrow progenitor cells expressing JAK3 mutants developed a long-latency transplantable T-ALL-like disease, characterized by an accumulation of immature CD8(+) T cells. In vivo treatment of leukemic mice with the JAK3 selective inhibitor tofacitinib reduced the white blood cell count and caused leukemic cell apoptosis. Our data show that JAK3 mutations are drivers of T-ALL and require the cytokine receptor complex for transformation. These results warrant further investigation of JAK1/JAK3 inhibitors for the treatment of T-ALL.


Subject(s)
Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Janus Kinase 1/metabolism , Janus Kinase 3/genetics , Leukemia, T-Cell/genetics , Mice , Acute Disease , Animals , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enzyme Activation/drug effects , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Male , Mice/genetics , Mice/metabolism , Mice, Inbred BALB C , Mutation , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
11.
Haematologica ; 99(1): 85-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23872305

ABSTRACT

The NUP214-ABL1 fusion protein is a constitutively active protein tyrosine kinase that is found in 6% of patients with T-cell acute lymphoblastic leukemia and that promotes proliferation and survival of T-lymphoblasts. Although NUP214-ABL1 is sensitive to ABL1 kinase inhibitors, development of resistance to these compounds is a major clinical problem, underlining the need for additional drug targets in the sparsely studied NUP214-ABL1 signaling network. In this work, we identify and validate the SRC family kinase LCK as a protein whose activity is absolutely required for the proliferation and survival of T-cell acute lymphoblastic leukemia cells that depend on NUP214-ABL1 activity. These findings underscore the potential of SRC kinase inhibitors and of the dual ABL1/SRC kinase inhibitors dasatinib and bosutinib for the treatment of NUP214-ABL1-positive T-cell acute lymphoblastic leukemia. In addition, we used mass spectrometry to identify protein interaction partners of NUP214-ABL1. Our results strongly support that the signaling network of NUP214-ABL1 is distinct from that previously reported for BCR-ABL1. Moreover, we found that three NUP214-ABL1-interacting proteins, MAD2L1, NUP155, and SMC4, are strictly required for the proliferation and survival of NUP214-ABL1-positive T-cell acute lymphoblastic leukemia cells. In conclusion, this work identifies LCK, MAD2L1, NUP155 and SMC4 as four new potential drug targets in NUP214-ABL1-positive T-cell acute lymphoblastic leukemia.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Humans , Phosphorylation , Protein Binding , Protein Interaction Mapping , RNA Interference , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
12.
PLoS Genet ; 9(12): e1003997, 2013.
Article in English | MEDLINE | ID: mdl-24367274

ABSTRACT

RNA-seq is a promising technology to re-sequence protein coding genes for the identification of single nucleotide variants (SNV), while simultaneously obtaining information on structural variations and gene expression perturbations. We asked whether RNA-seq is suitable for the detection of driver mutations in T-cell acute lymphoblastic leukemia (T-ALL). These leukemias are caused by a combination of gene fusions, over-expression of transcription factors and cooperative point mutations in oncogenes and tumor suppressor genes. We analyzed 31 T-ALL patient samples and 18 T-ALL cell lines by high-coverage paired-end RNA-seq. First, we optimized the detection of SNVs in RNA-seq data by comparing the results with exome re-sequencing data. We identified known driver genes with recurrent protein altering variations, as well as several new candidates including H3F3A, PTK2B, and STAT5B. Next, we determined accurate gene expression levels from the RNA-seq data through normalizations and batch effect removal, and used these to classify patients into T-ALL subtypes. Finally, we detected gene fusions, of which several can explain the over-expression of key driver genes such as TLX1, PLAG1, LMO1, or NKX2-1; and others result in novel fusion transcripts encoding activated kinases (SSBP2-FER and TPM3-JAK2) or involving MLLT10. In conclusion, we present novel analysis pipelines for variant calling, variant filtering, and expression normalization on RNA-seq data, and successfully applied these for the detection of translocations, point mutations, INDELs, exon-skipping events, and expression perturbations in T-ALL.


Subject(s)
Base Sequence/genetics , Gene Expression Regulation, Leukemic , High-Throughput Nucleotide Sequencing/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcriptome/genetics , Adolescent , Adult , Aged , Cell Line, Tumor , Child , Child, Preschool , Exome/genetics , Female , Gene Fusion , Humans , INDEL Mutation/genetics , Infant , Male , Middle Aged , Polymorphism, Single Nucleotide , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
13.
Nat Genet ; 45(2): 186-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263491

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


Subject(s)
Exome/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Ribosomal Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , Drosophila melanogaster , High-Throughput Nucleotide Sequencing , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Polyribosomes/genetics , RNA Interference , Ribosomal Protein L10 , Saccharomyces cerevisiae , Sequence Alignment
14.
Blood ; 117(26): 7090-8, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21551237

ABSTRACT

We have recently reported inactivation of the tyrosine phosphatase PTPN2 (also known as TC-PTP) through deletion of the entire gene locus in ∼ 6% of T-cell acute lymphoblastic leukemia (T-ALL) cases. T-ALL is an aggressive disease of the thymocytes characterized by the stepwise accumulation of chromosomal abnormalities and gene mutations. In the present study, we confirmed the strong association of the PTPN2 deletion with TLX1 and NUP214-ABL1 expression. In addition, we found cooperation between PTPN2 deletion and activating JAK1 gene mutations. Activating mutations in JAK1 kinase occur in ∼ 10% of human T-ALL cases, and aberrant kinase activity has been shown to confer proliferation and survival advantages. Our results reveal that some JAK1 mutation-positive T-ALLs harbor deletions of the tyrosine phosphatase PTPN2, a known negative regulator of the JAK/STAT pathway. We provide evidence that down-regulation of Ptpn2 sensitizes lymphoid cells to JAK1-mediated transformation and reduces their sensitivity to JAK inhibition.


Subject(s)
Gene Expression Regulation, Leukemic , Janus Kinase 1/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , T-Lymphocytes/metabolism , Adult , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic , Child , Comparative Genomic Hybridization , Female , Gene Deletion , Gene Silencing , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/chemistry , Janus Kinase 1/genetics , Male , Middle Aged , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering , Young Adult
15.
Haematologica ; 96(5): 779-83, 2011 May.
Article in English | MEDLINE | ID: mdl-21193420

ABSTRACT

The mouse pro-B cell line Ba/F3 has gained major interest as a model system to investigate oncogenic tyrosine kinases and to determine the efficacy of kinase inhibitors. While Ba/F3 cells are suitable to study oncogenic kinases derived from various cell types, the signaling networks in Ba/F3 cells are B-cell specific. We have established a mouse CD4+CD8+ double positive T-cell line (named MOHITO, for MOuse Hematopoietic Interleukin-dependent cell line of T-cell Origin) that has many features of human T-cell acute lymphoblastic leukemia (Notch1 and Jak1 mutation, TCR rearrangement) and is dependent on interleukin-7. The MOHITO cell line can be transformed to cytokine independent proliferation by BCR-ABL1 or mutant JAK1. This mouse T-cell line is a novel model system to investigate protein signaling and inhibition in a T-cell specific context and is a valuable tool to study and verify oncogenic capacity of mutations in the kinome and phosphatome in T-cell malignancies.


Subject(s)
Carcinogens/metabolism , Interleukin-7/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , HEK293 Cells , Humans , In Situ Hybridization, Fluorescence , Interleukin-7/pharmacology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Karyotyping , Mice , Mice, Inbred BALB C , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Nat Genet ; 42(6): 530-5, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20473312

ABSTRACT

PTPN2 (protein tyrosine phosphatase non-receptor type 2, also known as TC-PTP) is a cytosolic tyrosine phosphatase that functions as a negative regulator of a variety of tyrosine kinases and other signaling proteins. In agreement with its role in the regulation of the immune system, PTPN2 was identified as a susceptibility locus for autoimmune diseases. In this work, we describe the identification of focal deletions of PTPN2 in human T-cell acute lymphoblastic leukemia (T-ALL). Deletion of PTPN2 was specifically found in T-ALLs with aberrant expression of the TLX1 transcription factor oncogene, including four cases also expressing the NUP214-ABL1 tyrosine kinase. Knockdown of PTPN2 increased the proliferation and cytokine sensitivity of T-ALL cells. In addition, PTPN2 was identified as a negative regulator of NUP214-ABL1 kinase activity. Our study provides genetic and functional evidence for a tumor suppressor role of PTPN2 and suggests that expression of PTPN2 may modulate response to treatment.


Subject(s)
Gene Deletion , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Animals , Benzamides , Cell Line , Cell Line, Tumor , Chemokine CCL1/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Imatinib Mesylate , Interleukin-2/metabolism , Interleukin-7/metabolism , Leukemia, Experimental/genetics , Mice , Piperazines/therapeutic use , Proto-Oncogene Proteins/genetics , Pyrimidines/therapeutic use
17.
Haematologica ; 93(4): 533-42, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18322257

ABSTRACT

BACKGROUND: Activating NOTCH1 mutations are common in T-cell acute lymphoblastic leukemia. Inhibition of NOTCH1 signaling with gamma-secretase inhibitors causes cell cycle block, but only after treatment for several days. We further documented the effects of gamma-secretase inhibitor treatment on T-cell acute lymphoblastic leukemia cell lines and tested whether combining gamma-secretase inhibitors with other anti-cancer drugs offers a therapeutic advantage. DESIGN AND METHODS: The effect of gamma-secretase inhibitor treatment and combinations of gamma-secretase inhibitors with chemotherapy or glucocorticoids was assessed on T-cell acute lymphoblastic leukemia cell lines. We sequenced NOTCH1 in T-cell acute lymphoblastic leukemia cases with ABL1 fusions and tested combinations of gamma-secretase inhibitors and the ABL1 inhibitor imatinib in a T-cell acute lymphoblastic leukemia cell line. RESULTS: gamma-secretase inhibitor treatment for 5-7 days reversibly inhibited cell proliferation, caused cell cycle block in sensitive T-cell acute lymphoblastic leukemia cell lines, and caused differentiation of some T-cell acute lymphoblastic leukemia cell lines. Treatment for 14 days or longer was required to induce significant apoptosis. The cytotoxic effects of the chemotherapeutic agent vincristine were not significantly enhanced by addition of gamma-secretase inhibitors to T-cell acute lymphoblastic leukemia cell lines, but gamma-secretase inhibitor treatment sensitized cells to the effect of dexamethasone. NOTCH1 mutations were identified in all T-cell acute lymphoblastic leukemia patients with ABL1 fusions and in a T-cell acute lymphoblastic leukemia cell line expressing NUP214-ABL1. In this cell line, the anti-proliferative effect of imatinib was increased by pre-treatment with gamma-secretase inhibitors. CONCLUSIONS: Short-term treatment of T-cell acute lymphoblastic leukemia cell lines with gamma-secretase inhibitors had limited effects on cell proliferation and survival. Combinations of gamma-secretase inhibitors with other drugs may be required to obtain efficient therapeutic effects in T-cell acute lymphoblastic leukemia, and not all combinations may be useful.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Benzodiazepinones/pharmacology , Carbamates/pharmacology , Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Receptor, Notch1/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Benzamides , Carbamates/administration & dosage , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , DNA, Neoplasm/genetics , Daunorubicin/administration & dosage , Daunorubicin/pharmacology , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Dipeptides/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Inhibitors/administration & dosage , Humans , Imatinib Mesylate , In Vitro Techniques , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Mutation , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Piperazines/administration & dosage , Piperazines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Receptor, Notch1/genetics , Sequence Analysis, DNA , Vincristine/administration & dosage , Vincristine/pharmacology
18.
Haematologica ; 93(1): 49-56, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18166785

ABSTRACT

BACKGROUND: Translocations involving region 5q31-32 (PDGFRB) have been reported in a variety of myeloproliferative diseases and are often associated with significant peripheral eosinophilia. We report an unusual case of a patient presenting with peripheral basophilia and systemic mastocytosis in whom cytogenetic analysis revealed a t(4;5)(q21.1;q31.3). DESIGN AND METHODS: We used molecular analyses to determine the role of PDGFRB in this case. The patient was treated with imatinib. RESULTS: Fluorescence in situ hybridization (FISH) documented a breakpoint in PDGFRB. In agreement with this, the patient responded very well to imatinib with resolution of clinical symptoms, basophilia, and mast cell disease. Molecular analyses revealed that PDGFRB, encoding an imatinib-sensitive tyrosine kinase, was fused to PRKG2. The fusion gene incorporates the first two exons of PRKG2 fused to the truncated exon 12 of PDGFRB, resulting in the disruption of its juxtamembrane domain. Functional studies confirmed that the activity and transforming properties of PRKG2-PDGFRbeta were dependent on the disruption of the auto-inhibitory juxtamembrane domain. CONCLUSIONS: Our results identify a second case of the PRKG2-PDGFRB fusion and confirm the unusual PDGFRB breakpoint associated with this fusion. This work also illustrates the use of imatinib for the treatment of specific cases of systemic mastocytosis.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/genetics , Oncogene Proteins, Fusion/genetics , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/chemistry , Animals , Benzamides , Bone Marrow/pathology , Cytogenetics , Humans , Imatinib Mesylate , Immunophenotyping , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Male , Mastocytosis, Systemic/complications , Mice , Middle Aged
19.
Nat Genet ; 39(5): 593-5, 2007 May.
Article in English | MEDLINE | ID: mdl-17435759

ABSTRACT

We identified a duplication of the MYB oncogene in 8.4% of individuals with T cell acute lymphoblastic leukemia (T-ALL) and in five T-ALL cell lines. The duplication is associated with a threefold increase in MYB expression, and knockdown of MYB expression initiates T cell differentiation. Our results identify duplication of MYB as an oncogenic event and suggest that MYB could be a therapeutic target in human T-ALL.


Subject(s)
Cell Differentiation/genetics , Gene Duplication , Genes, myb/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , T-Lymphocytes/pathology , Cell Differentiation/immunology , Cell Line, Tumor , Chromosomes, Artificial/genetics , Flow Cytometry , Gene Dosage , Gene Expression Regulation, Neoplastic/genetics , Genetic Testing , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mutation/genetics , Nucleic Acid Hybridization/genetics , RNA, Small Interfering/genetics , Statistics, Nonparametric
20.
Haematologica ; 92(1): 27-34, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17229632

ABSTRACT

BACKGROUND AND OBJECTIVES: Activated tyrosine kinases are implicated in the pathogenesis of chronic and acute leukemia, and represent attractive targets for therapy. Sorafenib (BAY43-9006, Nexavar) is a small molecule B-RAF inhibitor that is used for the treatment of renal cell carcinoma, and has been shown to have activity against receptor tyrosine kinases from the platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR) families. We investigated the efficacy of sorafenib at inhibiting mutants of the receptor tyrosine kinases PDGFRbeta, KIT, and FLT3, which are implicated in the pathogenesis of myeloid malignancies. DESIGN AND METHODS: We tested the effect of sorafenib on the proliferation of hematopoietic cells transformed by ETV6-PDGFRbeta, FLT3 with an internal tandem duplication or D835Y point mutation, and the KIT(D816V) mutant. The direct effect of sorafenib on the activity of these kinases and their downstream signaling was tested using phospho-specific antibodies. RESULTS: We show that sorafenib is a potent inhibitor of ETV6-PDGFRbeta and FLT3 mutants, including some of the mutants that confer resistance to PKC412 and other FLT3 inhibitors. Sorafenib induced a cell cycle block and apoptosis in the acute myeloid leukemia cell lines MV4-11 and MOLM-13, both expressing FLT3 with an internal tandem duplication, whereas no effect was observed on four other acute myeloid leukemia cell lines. The imatinib-resistant KIT(D816V) mutant, associated with systemic mastocytosis, was found to be resistant to sorafenib. INTERPRETATION AND CONCLUSIONS: These results warrant further clinical studies of sorafenib for the treatment of myeloid malignancies expressing activated forms of PDGFRbeta and FLT3.


Subject(s)
Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Drug Resistance, Neoplasm , Pyridines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , fms-Like Tyrosine Kinase 3/biosynthesis , fms-Like Tyrosine Kinase 3/genetics , Apoptosis , Cell Cycle , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Humans , Mutation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Sorafenib
SELECTION OF CITATIONS
SEARCH DETAIL
...