Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 5: 3904, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24887457

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma and can be separated into two subtypes based upon molecular features with similarities to germinal centre B-cells (GCB-like) or activated B-cells (ABC-like). Here we identify gain of 3q27.2 as being significantly associated with adverse outcome in DLBCL and linked with the ABC-like subtype. This lesion includes the BCL6 oncogene, but does not alter BCL6 transcript levels or target-gene repression. Separately, we identify expression of BCL6 in a subset of human haematopoietic stem/progenitor cells (HSPCs). We therefore hypothesize that BCL6 may act by 'hit-and-run' oncogenesis. We model this hit-and-run mechanism by transiently expressing Bcl6 within murine HSPCs, and find that it causes mature B-cell lymphomas that lack Bcl6 expression and target-gene repression, are transcriptionally similar to post-GCB cells, and show epigenetic changes that are conserved from HSPCs to mature B-cells. Together, these results suggest that BCL6 may function in a 'hit-and-run' role in lymphomagenesis.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Animals , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , DNA Copy Number Variations , DNA Methylation , DNA-Binding Proteins/metabolism , Doxorubicin/therapeutic use , Epigenesis, Genetic , Female , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Male , Mice , Mice, Transgenic , Phenotype , Prednisone/therapeutic use , Prognosis , Proto-Oncogene Proteins c-bcl-6 , Rituximab , Vincristine/therapeutic use
2.
Cell Cycle ; 13(11): 1717-26, 2014.
Article in English | MEDLINE | ID: mdl-24675889

ABSTRACT

In hematopoietic malignancies, oncogenic alterations interfere with cellular differentiation and lead to tumoral development. Identification of the proteins regulating differentiation is essential to understand how they are altered in malignancies. Chronic myelogenous leukemia (CML) is a biphasic disease initiated by an alteration taking place in hematopoietic stem cells. CML progresses to a blast crisis (BC) due to a secondary differentiation block in any of the hematopoietic lineages. However, the molecular mechanisms of CML evolution to T-cell BC remain unclear. Here, we have profiled the changes in DNA methylation patterns in human samples from BC-CML, in order to identify genes whose expression is epigenetically silenced during progression to T-cell lineage-specific BC. We have found that the CpG-island of the ENGRAILED-2 (EN2) gene becomes methylated in this progression. Afterwards, we demonstrate that En2 is expressed during T-cell development in mice and humans. Finally, we further show that genetic deletion of En2 in a CML transgenic mouse model induces a T-cell lineage BC that recapitulates human disease. These results identify En2 as a new regulator of T-cell differentiation whose disruption induces a malignant T-cell fate in CML progression, and validate the strategy used to identify new developmental regulators of hematopoiesis.


Subject(s)
Blast Crisis/metabolism , Cell Differentiation/immunology , Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Nerve Tissue Proteins/metabolism , T-Lymphocytes/immunology , Animals , CpG Islands/genetics , DNA Methylation/genetics , DNA Primers/genetics , Disease Progression , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Transgenic , Microarray Analysis , Nerve Tissue Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
EMBO J ; 31(18): 3704-17, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22903061

ABSTRACT

Understanding the cellular origin of cancer can help to improve disease prevention and therapeutics. Human plasma cell neoplasias are thought to develop from either differentiated B cells or plasma cells. However, when the expression of Maf oncogenes (associated to human plasma cell neoplasias) is targeted to mouse B cells, the resulting animals fail to reproduce the human disease. Here, to explore early cellular changes that might take place in the development of plasma cell neoplasias, we engineered transgenic mice to express MafB in haematopoietic stem/progenitor cells (HS/PCs). Unexpectedly, we show that plasma cell neoplasias arise in the MafB-transgenic mice. Beyond their clinical resemblance to human disease, these neoplasias highly express genes that are known to be upregulated in human multiple myeloma. Moreover, gene expression profiling revealed that MafB-expressing HS/PCs were more similar to B cells and tumour plasma cells than to any other subset, including wild-type HS/PCs. Consistent with this, genome-scale DNA methylation profiling revealed that MafB imposes an epigenetic program in HS/PCs, and that this program is preserved in mature B cells of MafB-transgenic mice, demonstrating a novel molecular mechanism involved in tumour initiation. Our findings suggest that, mechanistically, the haematopoietic progenitor population can be the target for transformation in MafB-associated plasma cell neoplasias.


Subject(s)
Gene Expression Regulation, Neoplastic , MafB Transcription Factor/metabolism , Multiple Myeloma/metabolism , Animals , Antigens, CD34/biosynthesis , Antigens, Ly/metabolism , B-Lymphocytes/metabolism , DNA Methylation , DNA, Complementary/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Library , Hematopoietic Stem Cells/cytology , Humans , In Situ Hybridization, Fluorescence , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Translocation, Genetic
4.
Proc Natl Acad Sci U S A ; 109(26): 10534-9, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22689981

ABSTRACT

Chromosomal translocations involving the MALT1 gene are hallmarks of mucosa-associated lymphoid tissue (MALT) lymphoma. To date, targeting these translocations to mouse B cells has failed to reproduce human disease. Here, we induced MALT1 expression in mouse Sca1(+)Lin(-) hematopoietic stem/progenitor cells, which showed NF-κB activation and early lymphoid priming, being selectively skewed toward B-cell differentiation. These cells accumulated in extranodal tissues and gave rise to clonal tumors recapitulating the principal clinical, biological, and molecular genetic features of MALT lymphoma. Deletion of p53 gene accelerated tumor onset and induced transformation of MALT lymphoma to activated B-cell diffuse large-cell lymphoma (ABC-DLBCL). Treatment of MALT1-induced lymphomas with a specific inhibitor of MALT1 proteolytic activity decreased cell viability, indicating that endogenous Malt1 signaling was required for tumor cell survival. Our study shows that human-like lymphomas can be modeled in mice by targeting MALT1 expression to hematopoietic stem/progenitor cells, demonstrating the oncogenic role of MALT1 in lymphomagenesis. Furthermore, this work establishes a molecular link between MALT lymphoma and ABC-DLBCL, and provides mouse models to test MALT1 inhibitors. Finally, our results suggest that hematopoietic stem/progenitor cells may be involved in the pathogenesis of human mature B-cell lymphomas.


Subject(s)
Caspases/genetics , Hematopoietic Stem Cells/metabolism , Lymphoma/pathology , Neoplasm Proteins/genetics , Oncogenes , Animals , Humans , Mice , Mice, Transgenic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/metabolism , Transcription, Genetic
5.
Aging (Albany NY) ; 2(12): 908-13, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21164221

ABSTRACT

The incidence, malignancy and treatment resistance of many types of human B-cell leukaemias (B-ALL) are directly related to patient age. A major obstacle to elucidate the contribution of age to the development and evolution of leukaemias is the lack of appropriate mouse models where precise control of the timing of oncogene expression is possible. Here we present proof-of-principle experiments showing how a conditional transgenic mouse model of BCR-ABLp190-driven B-ALL offers the opportunity to test the hypothesis that the age of the leukemic cells-of-origin of B-ALL influences B-ALL malignancy. B-ALLs generated from 12- and 20-month-old progenitors gave rise to a more invasive B-ALL than the one developed from 4-month old precursors. This was evidenced by survival analysis revealing the increased malignancy of B-ALLs generated from 20 or 12-month-old transformed progenitors compared with the 4-month equivalents (median survival of 88 days versus 50.5 and 33 days, respectively). Our study shows that the age of target cells at the time of transformation affects B-ALL malignancy.


Subject(s)
Cellular Senescence , Leukemia, B-Cell/pathology , Leukemia, Experimental/pathology , Neoplastic Stem Cells/pathology , Age Factors , Animals , Bone Marrow Transplantation , Cell Line , Cell Survival , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Genes, abl , Humans , Leukemia, B-Cell/genetics , Leukemia, B-Cell/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Mice , Mice, Transgenic , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...