Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(36): 9701-9706, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28831000

ABSTRACT

The genes encoding the histone acetyl-transferases (HATs) CREB binding protein (CREBBP) and EP300 are recurrently mutated in the activated B cell-like and germinal center (GC) B cell-like subtypes of diffuse large B cell lymphoma (DLBCL). Here, we introduced a patient mutation into a human DLBCL cell line using CRISPR and deleted Crebbp and Ep300 in the GC B cell compartment of mice. CREBBP-mutant DLBCL clones exhibited reduced histone H3 acetylation, expressed significantly less MHCII, and grew faster than wild-type clones in s.c. and orthotopic xenograft models. Mice lacking Crebbp in GC B cells exhibited hyperproliferation of their GC compartment upon immunization, had reduced MHCII surface expression on GC cells, and developed accelerated MYC-driven lymphomas. Ep300 inactivation reproduced some, but not all, consequences of Crebbp inactivation. MHCII deficiency phenocopied the effects of CREBBP loss in spontaneous and serial transplantation models of MYC-driven lymphomagenesis, supporting the idea that the mutational inactivation of CREBBP promotes immune evasion. Indeed, the depletion of CD4+ T cells greatly facilitated the engraftment of lymphoma cells in serial transplantation models. In summary, we provide evidence that both HATs are bona fide tumor suppressors that control MHCII expression and promote tumor immune control; mutational inactivation of CREBBP, but not of EP300, has additional cell-intrinsic engraftment and growth-promoting effects.


Subject(s)
CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CREB-Binding Protein/deficiency , CREB-Binding Protein/immunology , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/deficiency , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/immunology , Gene Deletion , Genes, MHC Class II , Germinal Center/immunology , Germinal Center/pathology , HLA Antigens/genetics , Heterografts , Histone Code/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
2.
Blood ; 127(11): 1438-48, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26729899

ABSTRACT

Aberrant expression of the oncogenic transcription factor forkhead box protein 1 (FOXP1) is a common feature of diffuse large B-cell lymphoma (DLBCL). We have combined chromatin immunoprecipitation and gene expression profiling after FOXP1 depletion with functional screening to identify targets of FOXP1 contributing to tumor cell survival. We find that the sphingosine-1-phosphate receptor 2 (S1PR2) is repressed by FOXP1 in activated B-cell (ABC) and germinal center B-cell (GCB) DLBCL cell lines with aberrantly high FOXP1 levels; S1PR2 expression is further inversely correlated with FOXP1 expression in 3 patient cohorts. Ectopic expression of wild-type S1PR2, but not a point mutant incapable of activating downstream signaling pathways, induces apoptosis in DLBCL cells and restricts tumor growth in subcutaneous and orthotopic models of the disease. The proapoptotic effects of S1PR2 are phenocopied by ectopic expression of the small G protein Gα13 but are independent of AKT signaling. We further show that low S1PR2 expression is a strong negative prognosticator of patient survival, alone and especially in combination with high FOXP1 expression. The S1PR2 locus has previously been demonstrated to be recurrently mutated in GCB DLBCL; the transcriptional silencing of S1PR2 by FOXP1 represents an alternative mechanism leading to inactivation of this important hematopoietic tumor suppressor.


Subject(s)
Forkhead Transcription Factors/physiology , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Proteins/physiology , Receptors, Lysosphingolipid/physiology , Repressor Proteins/physiology , Signal Transduction/physiology , Animals , Apoptosis/physiology , Cell Line, Tumor , Chromatin Immunoprecipitation , Forkhead Transcription Factors/genetics , GTP-Binding Protein alpha Subunits, G12-G13/biosynthesis , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Germinal Center/pathology , Heterografts , Humans , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/classification , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/mortality , Mice , Neoplasm Transplantation , Prognosis , Proto-Oncogene Proteins c-akt/analysis , RNA Interference , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/biosynthesis , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics , Repressor Proteins/genetics , Sphingosine-1-Phosphate Receptors
3.
J Exp Med ; 212(5): 775-92, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25847947

ABSTRACT

The epigenetic dysregulation of tumor suppressor genes is an important driver of human carcinogenesis. We have combined genome-wide DNA methylation analyses and gene expression profiling after pharmacological DNA demethylation with functional screening to identify novel tumor suppressors in diffuse large B cell lymphoma (DLBCL). We find that a CpG island in the promoter of the dual-specificity phosphatase DUSP4 is aberrantly methylated in nodal and extranodal DLBCL, irrespective of ABC or GCB subtype, resulting in loss of DUSP4 expression in 75% of >200 examined cases. The DUSP4 genomic locus is further deleted in up to 13% of aggressive B cell lymphomas, and the lack of DUSP4 is a negative prognostic factor in three independent cohorts of DLBCL patients. Ectopic expression of wild-type DUSP4, but not of a phosphatase-deficient mutant, dephosphorylates c-JUN N-terminal kinase (JNK) and induces apoptosis in DLBCL cells. Pharmacological or dominant-negative JNK inhibition restricts DLBCL survival in vitro and in vivo and synergizes strongly with the Bruton's tyrosine kinase inhibitor ibrutinib. Our results indicate that DLBCL cells depend on JNK signaling for survival. This finding provides a mechanistic basis for the clinical development of JNK inhibitors in DLBCL, ideally in synthetic lethal combinations with inhibitors of chronic active B cell receptor signaling.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Dual-Specificity Phosphatases/deficiency , Lymphoma, Large B-Cell, Diffuse/metabolism , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinase Phosphatases/deficiency , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Signal Transduction , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Survival , CpG Islands , DNA, Neoplasm/genetics , Female , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , MAP Kinase Kinase 4/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics
4.
J Exp Med ; 210(5): 1003-19, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23589569

ABSTRACT

The molecular etiology of myeloproliferative neoplasms (MPNs) remains incompletely understood, despite recent advances incurred through the discovery of several different mutations in MPN patients. We have recently described overexpression of the transcription factor NF-E2 in MPN patients and shown that elevated NF-E2 levels in vivo cause an MPN phenotype and predispose to leukemic transformation in transgenic mice. We report the presence of acquired insertion and deletion mutations in the NF-E2 gene in MPN patients. These result in truncated NF-E2 proteins that enhance wild-type (WT) NF-E2 function and cause erythrocytosis and thrombocytosis in a murine model. NF-E2 mutant cells acquire a proliferative advantage, witnessed by clonal dominance over WT NF-E2 cells in MPN patients. Our data underscore the role of increased NF-E2 activity in the pathophysiology of MPNs.


Subject(s)
Bone Marrow Neoplasms/genetics , Mutation/genetics , Myeloproliferative Disorders/genetics , NF-E2 Transcription Factor, p45 Subunit/genetics , Animals , Bone Marrow Neoplasms/pathology , Bone Marrow Transplantation , Cell Lineage/genetics , Cell Proliferation , Clone Cells , DNA/metabolism , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Janus Kinase 2/metabolism , Mice , Mutant Proteins/metabolism , Myeloproliferative Disorders/pathology , NF-E2 Transcription Factor, p45 Subunit/metabolism , Protein Binding/genetics , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation/genetics
5.
Curr Pharm Des ; 19(7): 1201-10, 2013.
Article in English | MEDLINE | ID: mdl-23092340

ABSTRACT

microRNAs (miRNAs) comprise a recently discovered class of non-coding RNAs with regulatory functions in post-transcriptional gene expression control. Many miRNAs are located in genomic regions that are frequently deleted in cancer, or are subject to epigenetic and transcriptional deregulation in cancer cells. The miRNA transcriptome of cancer cells is very different from that of their normal cell counterparts. miRNAs can exhibit oncogenic or tumor suppressive or even both properties depending on the specific targets and cellular context. It is becoming increasingly clear that miRNAs not only serve as useful tumor biomarkers with implications for diagnosis, prognosis and the prediction of treatment responses, but may also be used for targeted cancer treatment and even as therapeutics. In this review, we provide an overview of recent advances in our understanding of the tumor suppressor miRNAs and oncomiRs involved in the pathogenesis of leukemias and lymphomas, and their target transcripts in cancer signaling networks. In particular, we focus on the role of miRNAs in chronic lymphocytic and acute lymphoblastic leukemia and in B-cell lymphomas. In the second part, we review the various alternative strategies of targeting miRNAs in cancer therapy. Methods of oncomiR antagonization by antagomiRs or locked nucleid acids are contrasted with strategies that harness the tumor suppressive properties of certain miRNAs for cancer treatment. Preclinical progress, also with regard to delivery strategies, possible side effects and other pharmacological aspects, is presented along with results from the first human trials assessing the safety and efficacy of miRNA-targeting therapeutics.


Subject(s)
Hematologic Neoplasms/genetics , Hematologic Neoplasms/therapy , MicroRNAs/physiology , Genes, Tumor Suppressor , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...