Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Leukemia ; 35(3): 679-690, 2021 03.
Article in English | MEDLINE | ID: mdl-32606318

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes and is largely driven by the NOTCH/MYC pathway. Yet, additional oncogenic drivers are required for transformation. Here, we identify protein tyrosine phosphatase type 4 A3 (PRL3) as a collaborating oncogenic driver in T-ALL. PRL3 is expressed in a large fraction of primary human T-ALLs and is commonly co-amplified with MYC. PRL3 also synergized with MYC to initiate early-onset ALL in transgenic zebrafish and was required for human T-ALL growth and maintenance. Mass-spectrometry phosphoproteomic analysis and mechanistic studies uncovered that PRL3 suppresses downstream T-cell phosphorylation signaling pathways, including those modulated by VAV1, and subsequently suppresses apoptosis in leukemia cells. Taken together, our studies have identified new roles for PRL3 as a collaborating oncogenic driver in human T-ALL and suggest that therapeutic targeting of the PRL3 phosphatase will likely be a useful treatment strategy for T-ALL.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Tyrosine Phosphatases/metabolism , T-Lymphocytes/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , Protein Tyrosine Phosphatases/genetics , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zebrafish
3.
Leukemia ; 29(12): 2317-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26108691

ABSTRACT

The TLX1 transcription factor is critically involved in the multi-step pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL) and often cooperates with NOTCH1 activation during malignant T-cell transformation. However, the exact molecular mechanism by which these T-cell specific oncogenes cooperate during transformation remains to be established. Here, we used chromatin immunoprecipitation followed by sequencing to establish the genome-wide binding pattern of TLX1 in human T-ALL. This integrative genomics approach showed that ectopic TLX1 expression drives repression of T cell-specific enhancers and mediates an unexpected transcriptional antagonism with NOTCH1 at critical target genes, including IL7R and NOTCH3. These phenomena coordinately trigger a TLX1-driven pre-leukemic phenotype in human thymic precursor cells, reminiscent of the thymus regression observed in murine TLX1 tumor models, and create a strong genetic pressure for acquiring activating NOTCH1 mutations as a prerequisite for full leukemic transformation. In conclusion, our results uncover a functional antagonism between cooperative oncogenes during the earliest phases of tumor development and provide novel insights in the multi-step pathogenesis of TLX1-driven human leukemia.


Subject(s)
Homeodomain Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Homeodomain Proteins/physiology , Humans , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Proto-Oncogene Proteins/physiology , Receptor, Notch1/genetics , Receptor, Notch1/physiology
4.
Leukemia ; 29(4): 798-806, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25231743

ABSTRACT

The MYB oncogene is a leucine zipper transcription factor essential for normal and malignant hematopoiesis. In T-cell acute lymphoblastic leukemia (T-ALL), elevated MYB levels can arise directly through T-cell receptor-mediated MYB translocations, genomic MYB duplications or enhanced TAL1 complex binding at the MYB locus or indirectly through the TAL1/miR-223/FBXW7 regulatory axis. In this study, we used an unbiased MYB 3'untranslated region-microRNA (miRNA) library screen and identified 33 putative MYB-targeting miRNAs. Subsequently, transcriptome data from two independent T-ALL cohorts and different subsets of normal T-cells were used to select miRNAs with relevance in the context of normal and malignant T-cell transformation. Hereby, miR-193b-3p was identified as a novel bona fide tumor-suppressor miRNA that targets MYB during malignant T-cell transformation thereby offering an entry point for efficient MYB targeting-oriented therapies for human T-ALL.


Subject(s)
Gene Expression Regulation, Leukemic , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myb/genetics , T-Lymphocytes/metabolism , 3' Untranslated Regions , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Profiling , Genomic Library , Humans , Mice , MicroRNAs/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1 , T-Lymphocytes/pathology , Transcriptome , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Leukemia ; 28(4): 830-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24091848

ABSTRACT

Peripheral blood T cells transduced with a tumor-specific T-cell receptor (TCR) face problems of auto-reactivity and lack of efficacy caused by cross-pairing of exogenous and endogenous TCR chains, as well as short term in vivo survival due to activation and growth factor-induced differentiation. We here studied an alternative strategy for the efficient generation of naive CD8(+) T cells with a single TCR. TCR-transduced human postnatal thymus-derived and adult mobilized blood-derived hematopoietic progenitor cells (HPCs) were differentiated to CD4(+)CD8(+) double-positive T cells using OP9-Delta-like 1 (OP9-DL1) cultures. Addition of the agonist peptide induced double positive cells to cross-present the peptide, leading, in the absence of co-stimulation, to cell cycle arrest and differentiation into mature CD8(+) T cells. Comprehensive phenotypic, molecular and functional analysis revealed the generation of naive and resting CD8(+) T cells through a process similar to thymic positive selection. These mature T cells show a near complete inhibition of endogenous TCRA and TCRB rearrangements and express high levels of the introduced multimer-reactive TCR. Upon activation, specific cytokine production and efficient killing of tumor cells were induced. Using this strategy, large numbers of high-avidity tumor-specific naive T cells can be generated from readily available HPCs without TCR chain cross-pairing.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/physiology , Adult , Cell Differentiation , Cell Line, Tumor , Child , Child, Preschool , Gene Rearrangement, T-Lymphocyte , Humans , Immunotherapy, Adoptive , Infant , Infant, Newborn , Receptors, Antigen, T-Cell/agonists
6.
Leukemia ; 26(1): 127-38, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22051534

ABSTRACT

In wild-type mice, T-cell receptor (TCR) γδ(+) cells differentiate along a CD4 CD8 double-negative (DN) pathway whereas TCRαß(+) cells differentiate along the double-positive (DP) pathway. In the human postnatal thymus (PNT), DN, DP and single-positive (SP) TCRγδ(+) populations are present. Here, the precursor-progeny relationship of the various PNT TCRγδ(+) populations was studied and the role of the DP TCRγδ(+) population during T-cell differentiation was elucidated. We demonstrate that human TCRγδ(+) cells differentiate along two pathways downstream from an immature CD1(+) DN TCRγδ(+) precursor: a Notch-independent DN pathway generating mature DN and CD8αα SP TCRγδ(+) cells, and a Notch-dependent, highly proliferative DP pathway generating immature CD4 SP and subsequently DP TCRγδ(+) populations. DP TCRγδ(+) cells are actively rearranging the TCRα locus, and differentiate to TCR(-) DP cells, to CD8αß SP TCRγδ(+) cells and to TCRαß(+) cells. Finally, we show that the γδ subset of T-cell acute lymphoblastic leukemias (T-ALL) consists mainly of CD4 SP or DP phenotypes carrying significantly more activating Notch mutations than DN T-ALL. The latter suggests that activating Notch mutations in TCRγδ(+) thymocytes induce proliferation and differentiation along the DP pathway in vivo.


Subject(s)
CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Proliferation , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Notch/physiology , Thymocytes/immunology , Base Sequence , Cell Differentiation , Coculture Techniques , DNA Primers , Humans , Real-Time Polymerase Chain Reaction , Thymocytes/cytology
7.
Verh K Acad Geneeskd Belg ; 71(5): 301-14, 2009.
Article in English | MEDLINE | ID: mdl-20232786

ABSTRACT

Notch signalling is a critical mediator of T cell development. However, while this signalling pathway is essential, it is not sufficient to induce T cell fate into multipotent hematopoietic precursor cells. Although Notch signalling events are crucial at all of the initial stages of T-lineage differentiation, T cell development also depends on other regulatory factors that are required at precise levels in order to preserve the well-balanced network that drives this process. Miss-expression of one of the factors profoundly perturbs this balance and results in alternative lineage cell fate. In this process, Notch plays an essential role as a gate-keeper of T-lineage fidelity by either enforcing the T cell fate or by inducing cell death if alternative lineages are induced in its presence. Notch also plays a critical role in the further lineage choices that occur within the T cell lineage. Importantly however, this seems to be different between mouse and human. While in the mouse it is clear that TCR-alphabeta T cells are much more dependent on Notch signalling compared to TCR-gammadelta T cells, the requirement for Notch signalling during human T cell development seems to be the opposite for both T cell lineages. Thus, while it is clear that Notch signalling plays a critical role during the early stages of T cell development, further work is essential to delineate the precise molecular network that controls T cell differentiation and this might be different between mouse and human.


Subject(s)
Cell Differentiation/physiology , Receptors, Notch/physiology , Signal Transduction , T-Lymphocytes/physiology , Animals , Humans , Mice , Receptors, Notch/immunology , Species Specificity , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
8.
Semin Immunopathol ; 30(4): 411-23, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18925396

ABSTRACT

Human T lymphocytes can be generated from CD34 progenitor cells from different sources. This can be obtained in an in vivo model wherein human thymic tissue and fetal liver is transplanted in an immunodeficient mouse. However, human T cells are also generated in immunodeficient mice without co-transplantation of human thymus or in in vitro hybrid human-mouse fetal thymus organ culture. This shows that xenogeneic mouse thymus tissue supports human T cell differentiation. Finally, human T cells are generated on co-culture with murine stromal cells that express the Delta-like1 ligand for the Notch receptor. How these different environments influence the human T cell repertoire is reviewed and discussed.


Subject(s)
Cell Differentiation/immunology , Hematopoietic Stem Cells/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Antigens, CD/immunology , Hematopoietic Stem Cells/immunology , Humans , Mice , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology
9.
Leukemia ; 17(6): 1157-63, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12764384

ABSTRACT

Class I homeobox (HOX) genes comprise a large family of transcription factors that have been implicated in normal and malignant hematopoiesis. However, data on their expression or function during T-cell development is limited. Using degenerated RT-PCR and Affymetrix microarray analysis, we analyzed the expression pattern of this gene family in human multipotent stem cells from fetal liver (FL) and adult bone marrow (ABM), and in T-cell progenitors from child thymus. We show that FL and ABM stem cells are similar in terms of HOX gene expression, but significant differences were observed between these two cell types and child thymocytes. As the most immature thymocytes are derived from immigrated FL and ABM stem cells, this indicates a drastic change in HOX gene expression upon entry into the thymus. Further analysis of HOX-A7, HOX-A9, HOX-A10, and HOX-A11 expression with specific RT-PCR in all thymocyte differentiation stages showed a sequential loss of 3' region HOX-A cluster genes during intrathymic T-cell development and an unexpected expression of HOX-A11, previously not recognized to play a role in hematopoiesis. Also HOX-B3 and HOX-C4 were expressed throughout thymocyte development. Overall, these data provide novel evidence for an important role of certain HOX genes in human T-cell development.


Subject(s)
Gene Expression , Genes, Homeobox/physiology , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Stem Cells/metabolism , T-Lymphocytes/metabolism , Adult , Cell Differentiation/genetics , Cell Lineage/genetics , Child , DNA Primers/chemistry , Fetus , Gene Expression Profiling , Humans , Liver/cytology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/cytology
10.
J Immunol ; 167(8): 4468-75, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591773

ABSTRACT

Following bone marrow transplantation, patients often suffer from immune incompetence by reduced or late T cell development. Moreover, adult bone marrow stem cells have a lower capacity to generate T cells compared with fetal liver- and umbilical cord blood-derived progenitors. Therefore, enhancing thymic-dependent T cell generation might hold great therapeutic potential. GATA-3 is a transcription factor that is essential in T cell development. In this study we examined the therapeutic potential of GATA-3 to enhance T cell generation by overexpressing GATA-3 in T cell progenitors followed by fetal thymic organ culture (FTOC). We observed that early during FTOC, there was an enhanced differentiation toward the double positive stage of T cell development. From day 10 of FTOC, however, overexpression of GATA-3 induced a severe reduction in thymic cellularity, which probably correlates with the absence of a functional TCR-beta chain. We further show that the frequency of apoptosis was increased in GATA-3-transduced thymocytes. Despite the absence of a functional TCR-beta chain, GATA-3 transduced progenitors were able to differentiate into CD8beta(+) double positive thymocytes. This study shows that a strictly regulated expression of GATA-3 is essential for normal T cell development and this puts severe restrictions on the potential therapeutic use of continuously overexpressed GATA-3.


Subject(s)
DNA-Binding Proteins/biosynthesis , Hematopoietic Stem Cells/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology , Trans-Activators/biosynthesis , Antigens, Differentiation, T-Lymphocyte , Apoptosis , CD3 Complex , CD4 Antigens , CD8 Antigens , Cell Differentiation , Cloning, Molecular , DNA-Binding Proteins/genetics , GATA3 Transcription Factor , Gene Expression Regulation , Gene Transfer Techniques , Genes, T-Cell Receptor beta , Hematopoietic Stem Cells/immunology , Humans , Organ Culture Techniques , T-Lymphocytes/immunology , Thymus Gland/immunology , Trans-Activators/genetics
11.
J Immunol ; 165(2): 645-53, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10878336

ABSTRACT

Langerhans cells (LCs) are immature dendritic cells (DCs) present in the skin epithelium. Upon Ag exposure, they migrate to the draining lymph nodes where they mature into potent stimulators of naive T cells. The aim of this study was to investigate the influence of T cells on LC migration and maturation. Therefore, the in vivo migration and maturation of LCs after sensitization with the hapten FITC was compared between C57BL/6 or BALB/c mice used as positive controls, and recombination activating gene (RAG) 1 knockout (-/-) mice or SCID mice used as T cell-deficient mice. Phenotypically, there was no difference between migrated LCs from RAG1-/- or SCID mice vs normal C57BL/6 or BALB/c mice: both populations of FITC+ cells had a dendritic morphology and a mature phenotype as they expressed high levels of MHC class II molecules and costimulatory molecules CD80, CD86, and CD54. Sorted migrated LCs of RAG1-/- or SCID mice were efficient stimulators of allogeneic T cells and Ag-specific CD4+ T cells. The same results were found if migrated LCs were fixed instead of irradiated, excluding the possibility that LCs derived from RAG1-/- or SCID mice would mature in the presence of T cells during the stimulation tests. Importantly, fixed migrated LCs of RAG1-/- mice were also efficient stimulators of cytotoxic CD8+ T cells. These data suggest that T cells are not required for full maturation of LCs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Langerhans Cells/cytology , Langerhans Cells/immunology , Lymphocyte Activation , Lymphopenia/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Fluorescein-5-isothiocyanate/administration & dosage , Haptens/administration & dosage , Haptens/immunology , Homeodomain Proteins/genetics , Immunization , Immunophenotyping , Lymphocyte Activation/genetics , Lymphopenia/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Organ Culture Techniques , Skin/cytology , Skin/immunology , T-Lymphocytes/pathology , Transposases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...