Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cancer Cell ; 41(10): 1817-1828.e9, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37683639

ABSTRACT

The dysregulated expression of immune checkpoint molecules enables cancer cells to evade immune destruction. While blockade of inhibitory immune checkpoints like PD-L1 forms the basis of current cancer immunotherapies, a deficiency in costimulatory signals can render these therapies futile. CD58, a costimulatory ligand, plays a crucial role in antitumor immune responses, but the mechanisms controlling its expression remain unclear. Using two systematic approaches, we reveal that CMTM6 positively regulates CD58 expression. Notably, CMTM6 interacts with both CD58 and PD-L1, maintaining the expression of these two immune checkpoint ligands with opposing functions. Functionally, the presence of CMTM6 and CD58 on tumor cells significantly affects T cell-tumor interactions and response to PD-L1-PD-1 blockade. Collectively, these findings provide fundamental insights into CD58 regulation, uncover a shared regulator of stimulatory and inhibitory immune checkpoints, and highlight the importance of tumor-intrinsic CMTM6 and CD58 expression in antitumor immune responses.


Subject(s)
B7-H1 Antigen , MARVEL Domain-Containing Proteins , Myelin Proteins , Neoplasms , T-Lymphocytes , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Immunity , Immunotherapy , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/immunology , Myelin Proteins/metabolism , MARVEL Domain-Containing Proteins/metabolism
2.
Cancers (Basel) ; 14(17)2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36077655

ABSTRACT

Most people infected by EBV acquire specific immunity, which then controls latent infection throughout their life. Immune surveillance of EBV-infected cells by cytotoxic CD4+ T cells has been recognized; however, the molecular mechanism of generating cytotoxic effector T cells of the CD4+ subset remains poorly understood. Here we compared phenotypic features and the transcriptome of EBV-specific effector-memory CD4+ T cells and CD8+ T cells in mice and found that both T cell types show cytotoxicity and, to our surprise, widely similar gene expression patterns relating to cytotoxicity. Similar to cytotoxic CD8+ T cells, EBV-specific cytotoxic CD4+ T cells from human peripheral blood expressed T-bet, Granzyme B, and Perforin and upregulated the degranulation marker, CD107a, immediately after restimulation. Furthermore, T-bet expression in cytotoxic CD4+ T cells was highly correlated with Granzyme B and Perforin expression at the protein level. Thus, differentiation of EBV-specific cytotoxic CD4+ T cells is possibly controlled by mechanisms shared by cytotoxic CD8+ T cells. T-bet-mediated transcriptional regulation may explain the similarity of cytotoxic effector differentiation between CD4+ T cells and CD8+ T cells, implicating that this differentiation pathway may be directed by environmental input rather than T cell subset.

3.
Blood Cancer Discov ; 2(1): 70-91, 2021 01.
Article in English | MEDLINE | ID: mdl-33447829

ABSTRACT

Based on gene expression profiles, diffuse large B cell lymphoma (DLBCL) is sub-divided into germinal center B cell-like (GCB) and activated B cell-like (ABC) DLBCL. Two of the most common genomic aberrations in ABC-DLBCL are mutations in MYD88, as well as BCL2 copy number gains. Here, we employ immune phenotyping, RNA-Seq and whole exome sequencing to characterize a Myd88 and Bcl2-driven mouse model of ABC-DLBCL. We show that this model resembles features of human ABC-DLBCL. We further demonstrate an actionable dependence of our murine ABC-DLBCL model on BCL2. This BCL2 dependence was also detectable in human ABC-DLBCL cell lines. Moreover, human ABC-DLBCLs displayed increased PD-L1 expression, compared to GCB-DLBCL. In vivo experiments in our ABC-DLBCL model showed that combined venetoclax and RMP1-14 significantly increased the overall survival of lymphoma bearing animals, indicating that this combination may be a viable option for selected human ABC-DLBCL cases harboring MYD88 and BCL2 aberrations.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Myeloid Differentiation Factor 88 , Animals , Genes, bcl-2 , Germinal Center/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Myeloid Differentiation Factor 88/genetics , Proto-Oncogene Proteins c-bcl-2/genetics
4.
JCI Insight ; 4(15)2019 08 08.
Article in English | MEDLINE | ID: mdl-31391340

ABSTRACT

Aberrant activity of the glycoprotein 130 130/JAK/STAT3 (gp130/JAK/STAT3) signaling axis is a recurrent event in inflammation and cancer. In particular, it is associated with a wide range of hematological malignancies, including multiple myeloma and leukemia. Novel targeted therapies have only been successful for some subtypes of these malignancies, underlining the need for developing robust mouse models to better dissect the role of this pathway in specific tumorigenic processes. Here, we investigated the role of selective gp130/JAK/STAT3 activation by generating a conditional mouse model. This model targeted constitutively active, cell-autonomous gp130 activity to B cells, as well as to the entire hematopoietic system. We found that regardless of the timing of activation in B cells, constitutively active gp130 signaling resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders with full penetrance, only with different latencies, where infiltrating CD138+ cells were a dominant feature in every tumor. Furthermore, constitutively active gp130 signaling in all adult hematopoietic cells also led to the development specifically of largely mature, aggressive B cell cancers, again with a high penetrance of CD138+ tumors. Importantly, gp130 activity abrogated the differentiation block induced by a B cell-targeted Myc transgene and resulted in a complete penetrance of the gp130-associated, CD138+, mature B cell lymphoma phenotype. Thus, gp130 signaling selectively provides a strong growth and differentiation advantage for mature B cells and directs lymphomagenesis specifically toward terminally differentiated B cell cancers.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cytokine Receptor gp130/metabolism , Lymphoma/immunology , Plasmacytoma/immunology , Signal Transduction/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Differentiation/genetics , Cytokine Receptor gp130/genetics , Disease Models, Animal , Female , Humans , Janus Kinases/metabolism , Lymphocyte Activation/genetics , Lymphoma/genetics , Lymphoma/pathology , Male , Mice , Plasmacytoma/genetics , Plasmacytoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism
5.
Science ; 363(6428): 748-753, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30765568

ABSTRACT

B2 cells engage in classical antibody responses, whereas B1 cells are considered carriers of innate immunity, biased toward recognizing epitopes present on the surfaces of common pathogens and self antigens. To explore the role of B cell antigen receptor (BCR) specificity in driving B1 cell differentiation, we developed a transgenic system allowing us to change BCR specificity in B cells in an inducible and programmed manner. Mature B2 cells differentiated into bona fide B1 cells upon acquisition of a B1 cell-typical self-reactive BCR through a phase of proliferative expansion. Thus, B2 cells have B1 cell differentiation potential in addition to their classical capacity to differentiate into memory and plasma cells, and B1 differentiation can be instructed by BCR-mediated self-reactivity, in the absence of B1-lineage precommitment.


Subject(s)
B-Lymphocyte Subsets/cytology , Cell Differentiation/immunology , Cell Plasticity/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Cell Differentiation/genetics , Cell Lineage , Cell Plasticity/genetics , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Mice , Mice, Transgenic , Receptors, Antigen, B-Cell/genetics , Transcriptome
6.
Blood ; 132(25): 2670-2683, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30333121

ABSTRACT

Forkhead box class O1 (FOXO1) acts as a tumor suppressor in solid tumors. The oncogenic phosphoinositide-3-kinase (PI3K) pathway suppresses FOXO1 transcriptional activity by enforcing its nuclear exclusion upon AKT-mediated phosphorylation. We show here abundant nuclear expression of FOXO1 in Burkitt lymphoma (BL), a germinal center (GC) B-cell-derived lymphoma whose pathogenesis is linked to PI3K activation. Recurrent FOXO1 mutations, which prevent AKT targeting and lock the transcription factor in the nucleus, are used by BL to circumvent mutual exclusivity between PI3K and FOXO1 activation. Using genome editing in human and mouse lymphomas in which MYC and PI3K cooperate synergistically in tumor development, we demonstrate proproliferative and antiapoptotic activity of FOXO1 in BL and identify its nuclear localization as an oncogenic event in GC B-cell-derived lymphomagenesis.


Subject(s)
B-Lymphocytes , Burkitt Lymphoma , Cell Nucleus , Cell Transformation, Neoplastic , Forkhead Box Protein O1 , Germinal Center , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Editing , Germinal Center/metabolism , Germinal Center/pathology , Humans , Mice , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
8.
Methods Mol Biol ; 1623: 233-242, 2017.
Article in English | MEDLINE | ID: mdl-28589360

ABSTRACT

Germinal centers (GC) are the predominant origin of human B cell lymphomagenesis. Transgenic mice in which gene expression is altered specifically in GC B cells have broadened our knowledge about the mechanisms of malignant transformation. However, extensive resources are needed due to the genetic complexity of these mouse models. Thus, bone marrow (BM)-derived chimerism is an attractive approach to study GC B cell derived lymphomagenesis, as it allows for an efficient allocation of resources and reduces the number of animals used.


Subject(s)
Germinal Center/pathology , Lymphoma/pathology , Animals , Bone Marrow Cells , Bone Marrow Transplantation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Rearrangement, B-Lymphocyte , Germinal Center/immunology , Germinal Center/metabolism , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/metabolism , Mice , V(D)J Recombination
9.
Immunity ; 43(6): 1075-86, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26620760

ABSTRACT

Phosphatidylinositol 3' OH kinase (PI3K) signaling and FOXO transcription factors play opposing roles at several B cell developmental stages. We show here abundant nuclear FOXO1 expression in the proliferative compartment of the germinal center (GC), its dark zone (DZ), and PI3K activity, downregulating FOXO1, in the light zone (LZ), where cells are selected for further differentiation. In the LZ, however, FOXO1 was expressed in a fraction of cells destined for DZ reentry. Upon FOXO1 ablation or induction of PI3K activity, GCs lost their DZ, owing at least partly to downregulation of the chemokine receptor CXCR4. Although this prevented proper cyclic selection of cells in GCs, somatic hypermutation and proliferation were maintained. Class switch recombination was partly lost due to a failure of switch region targeting by activation-induced deaminase (AID).


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Germinal Center/immunology , Phosphatidylinositol 3-Kinases/immunology , Animals , B-Lymphocytes/cytology , Cell Separation , Chromatography, Liquid , Cytidine Deaminase/immunology , Flow Cytometry , Fluorescent Antibody Technique , Forkhead Box Protein O1 , Gene Expression Regulation/immunology , Germinal Center/cytology , Immunoglobulin Class Switching/immunology , Lymphocyte Activation/immunology , Mice , Mice, Mutant Strains , Polymerase Chain Reaction , Somatic Hypermutation, Immunoglobulin/immunology , Tandem Mass Spectrometry
10.
Nat Biotechnol ; 33(5): 543-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25803306

ABSTRACT

The insertion of precise genetic modifications by genome editing tools such as CRISPR-Cas9 is limited by the relatively low efficiency of homology-directed repair (HDR) compared with the higher efficiency of the nonhomologous end-joining (NHEJ) pathway. To enhance HDR, enabling the insertion of precise genetic modifications, we suppressed the NHEJ key molecules KU70, KU80 or DNA ligase IV by gene silencing, the ligase IV inhibitor SCR7 or the coexpression of adenovirus 4 E1B55K and E4orf6 proteins in a 'traffic light' and other reporter systems. Suppression of KU70 and DNA ligase IV promotes the efficiency of HDR 4-5-fold. When co-expressed with the Cas9 system, E1B55K and E4orf6 improved the efficiency of HDR up to eightfold and essentially abolished NHEJ activity in both human and mouse cell lines. Our findings provide useful tools to improve the frequency of precise gene modifications in mammalian cells.


Subject(s)
CRISPR-Cas Systems/genetics , DNA End-Joining Repair/genetics , Genetic Engineering/methods , Adenoviridae/genetics , Adenovirus E4 Proteins/biosynthesis , Adenovirus E4 Proteins/genetics , Animals , Cell Line , DNA Breaks, Double-Stranded , DNA Ligase ATP , DNA Ligases/genetics , Gene Expression Regulation , Genome, Human , Homologous Recombination/genetics , Humans , Mice , Viral Proteins/biosynthesis , Viral Proteins/genetics
12.
Cancer Cell ; 22(2): 167-79, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22897848

ABSTRACT

In Burkitt lymphoma (BL), a germinal center B-cell-derived tumor, the pro-apoptotic properties of c-MYC must be counterbalanced. Predicting that survival signals would be delivered by phosphoinositide-3-kinase (PI3K), a major survival determinant in mature B cells, we indeed found that combining constitutive c-MYC expression and PI3K activity in germinal center B cells of the mouse led to BL-like tumors, which fully phenocopy human BL with regard to histology, surface and other markers, and gene expression profile. The tumors also accumulate tertiary mutational events, some of which are recurrent in the human disease. These results and our finding of recurrent PI3K pathway activation in human BL indicate that deregulated c-MYC and PI3K activity cooperate in BL pathogenesis.


Subject(s)
Burkitt Lymphoma/enzymology , Burkitt Lymphoma/pathology , Cell Transformation, Neoplastic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Base Sequence , Burkitt Lymphoma/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation , Gene Expression Regulation, Neoplastic , Germinal Center/enzymology , Germinal Center/pathology , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics
13.
Haematologica ; 96(12): 1783-91, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21880628

ABSTRACT

BACKGROUND: MicroRNAs are regulators of gene expression, which act mainly by decreasing mRNA levels of their multiple targets. Deregulated microRNA expression has been shown for acute myeloid leukemia, a disease also characterized by altered gene expression associated with distinct genomic aberrations such as nucleophosmin (NPM1) mutations. To shed further light on the role of deregulated microRNA and gene expression in cytogenetically normal acute myeloid leukemia with NPM1 mutation we performed an integrative analysis of microRNA and mRNA expression data sets. DESIGN AND METHODS: Both microRNA and gene expression profiles were investigated in samples from a cohort of adult cytogenetically normal acute myeloid leukemia patients (n=43; median age 46 years, range 23-60 years) with known NPM1 mutation status (n=23 mutated, n=20 wild-type) and the data were integratively analyzed. Putative microRNA-mRNA interactions were validated by quantitative reverse transcriptase polymerase chain reaction, western blotting and luciferase reporter assays. For selected microRNAs, sensitivity of microRNA-overexpressing cells to cytarabine treatment was tested by FACS viability and cell proliferation assays. RESULTS: Our integrative approach of analyzing both microRNA- and gene expression profiles in parallel resulted in a refined list of putative target genes affected by NPM1 mutation-associated microRNA deregulation. Of 177 putative microRNA - target mRNA interactions we identified and validated 77 novel candidates with known or potential involvement in leukemogenesis, such as IRF2-miR-20a, KIT-miR-20a and MN1-miR-15a. Furthermore, our data showed that deregulated expression of tumor suppressor microRNAs, such as miR-29a and miR-30c, might contribute to sensitivity to cytarabine, which is observed in NPM1 mutated acute myeloid leukemia. CONCLUSIONS: Overall, our observations highlight that integrative data analysis approaches can improve insights into leukemia biology, and lead to the identification of novel microRNA - target gene interactions of potential relevance for acute myeloid leukemia treatment.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/biosynthesis , Mutation , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Neoplasm/biosynthesis , Adult , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cohort Studies , Cytarabine/pharmacology , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Nucleophosmin , RNA, Neoplasm/genetics
14.
Blood ; 114(12): 2448-58, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19628709

ABSTRACT

Deregulated c-MYC is found in a variety of cancers where it promotes proliferation as well as apoptosis. In many hematologic malignancies, enhanced NF-kappaB exerts prosurvival functions. Here we investigated the role of NF-kappaB in mouse and human c-MYC-transformed lymphomas. The NF-kappaB pathway is extinguished in murine lymphoma cells, and extrinsic stimuli typically inducing NF-kappaB activity fail to activate this pathway. Genetic activation of the NF-kappaB pathway induces apoptosis in these cells, whereas inhibition of NF-kappaB by an IkappaBalpha superrepressor provides a selective advantage in vivo. Furthermore, in human Burkitt lymphoma cells we find that NF-kappaB activation induces apoptosis. NF-kappaB up-regulates Fas and predisposes to Fas-induced cell death, which is caspase-8 mediated and can be prevented by CFLAR overexpression. We conclude that c-MYC overexpression sensitizes cells to NF-kappaB-induced apoptosis, and persistent inactivity of NF-kappaB signaling is a prerequisite for MYC-mediated tumorigenesis. We could also show that low immunogenicity and Fas insensitivity of MYC-driven lymphoma cells are reversed by activation of NF-kappaB. Our observations provide a molecular explanation for the described absence of the NF-kappaB signaling in Burkitt lymphoma and question the applicability of NF-kappaB inhibitors as candidates for treatment of this cancer.


Subject(s)
Apoptosis , Burkitt Lymphoma/pathology , I-kappa B Kinase/physiology , Lymphoma, B-Cell/pathology , NF-kappa B/physiology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/physiology , Animals , Blotting, Western , Burkitt Lymphoma/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, B-Cell/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection
16.
Cell Cycle ; 8(4): 556-9, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19197161

ABSTRACT

The MYC oncogene is often mutated or amplified in human tumors resulting in increased activity of the transcription factor. Recent evidence suggested that MYC not only regulates expression of protein-coding genes directly, but also controls expression of miRNAs thereby using a second mode to impact on gene expression programs. The importance of miRNAs in MYC-driven tumorigenesis has been enlightened by studying cell line and murine lymphoma models with conditional expression of MYC. The application of microarray technology revealed both MYC-induced and MYC-repressed miRNAs. A miRNA consistently repressed by MYC in multiple tumors was miR-26a indicating that this miRNA might have strong tumor suppressor function for MYC-induced lymphomas. Indeed, ectopic miR-26a expression in MYC-dependent cells resulted in attenuated proliferation and impaired cell cycle progression. When the effector pathway for miR-26a was elucidated, the Polycomb complex protein EZH2, a global regulator of gene expression, was identified as a direct target. The suppression of miR-26a mediated attenuation of EZH2 expression by MYC was shown to play a critical role in lymphomagenesis. Thus, MYC-induced oncogenesis does not only depend on direct targeting of protein-coding genes, but also on modulating them via deregulation of their targeting miRNAs, thereby significantly impacting lymphomagenesis.


Subject(s)
Genes, myc , Lymphoma , Gene Expression Regulation, Neoplastic , Humans , Lymphoma/metabolism , Lymphoma/pathology , Lymphoma/physiopathology , MicroRNAs/genetics , MicroRNAs/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology
17.
Blood ; 113(19): 4690-701, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19228926

ABSTRACT

Loss of neurofibromin or interferon consensus sequence binding protein (Icsbp) leads to a myeloproliferative disorder. Transcription of NF1 is directly controlled by ICSBP. It has been postulated that loss of NF1 expression resulting from loss of transcriptional activation by ICSBP contributes to human hematologic malignancies. To investigate the functional cooperation of these 2 proteins, we have established Icsbp-deficient mice with Nf1 haploinsufficiency. We here demonstrate that loss of Icsbp and Nf1 haploinsufficiency synergize to induce a forced myeloproliferation in Icsbp-deficient mice because of an expansion of a mature myeloid progenitor cell. Furthermore, Nf1 haploinsufficiency and loss of Icsbp contribute synergistically to progression of the myeloproliferative disorder toward transplantable leukemias. Leukemias are characterized by distinct phenotypes, which correlate with progressive genetic abnormalities. Loss of Nf1 heterozygosity is not mandatory for disease progression, but its occurrence with other genetic abnormalities indicates progressive genetic alterations in a defined subset of leukemias. These data show that loss of the 2 tumor suppressor genes Nf1 and Icsbp synergize in the induction of leukemias.


Subject(s)
Gene Expression Regulation, Leukemic , Interferon Regulatory Factors/physiology , Leukemia/etiology , Myeloproliferative Disorders/etiology , Neurofibromin 1/physiology , Animals , Bone Marrow Cells , Colony-Forming Units Assay , Drug Synergism , Female , Flow Cytometry , Hematopoietic Stem Cells , Leukemia/metabolism , Leukemia/pathology , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Blood ; 112(10): 4202-12, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18713946

ABSTRACT

The MYC oncogene, which is commonly mutated/amplified in tumors, represents an important regulator of cell growth because of its ability to induce both proliferation and apoptosis. Recent evidence links MYC to altered miRNA expression, thereby suggesting that MYC-regulated miRNAs might contribute to tumorigenesis. To further analyze the impact of MYC-regulated miRNAs, we investigated a murine lymphoma model harboring the MYC transgene in a Tet-off system to control its expression. Microarray-based miRNA expression profiling revealed both known and novel MYC targets. Among the miRNAs repressed by MYC, we identified the potential tumor suppressor miR-26a, which possessed the ability to attenuate proliferation in MYC-dependent cells. Interestingly, miR-26a was also found to be deregulated in primary human Burkitt lymphoma samples, thereby probably being of clinical relevance. Although today only few miRNA targets have been identified in human disease, we could show that ectopic expression of miR-26a influenced cell cycle progression by targeting the bona fide oncogene EZH2, a Polycomb protein and global regulator of gene expression yet unknown to be regulated by miRNAs. Thus, in addition to directly targeting protein-coding genes, MYC modulates genes important to oncogenesis via deregulation of miRNAs, thereby vitally contributing to MYC-induced lymphomagenesis.


Subject(s)
Burkitt Lymphoma/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Leukemic , MicroRNAs/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Neoplasm/metabolism , Transcription Factors/biosynthesis , Animals , Burkitt Lymphoma/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Leukemic/genetics , Histone-Lysine N-Methyltransferase , Humans , Mice , MicroRNAs/genetics , Polycomb Repressive Complex 2 , Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Neoplasm/genetics , Transcription Factors/genetics
19.
Br J Haematol ; 142(4): 538-50, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18564361

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive lymphoid tumour characterized by the translocation t(11;14)(q13;q32) and a poor clinical outcome (median survival: 3-4 years). Recent studies revealed that increased proliferation of the tumour cells and certain chromosomal aberrations, such as deletions of 17p13 and 9p21 represent major adverse biological markers in this disease, although the molecular targets of chromosomal imbalances in MCL have not been identified for the large majority of loci affected. To correlate histomorphological and proliferation features of MCL with genetic findings, we investigated 223 MCL by fluorescence in situ hybridization (FISH) (n = 157) and/or classical cytogenetic banding analysis (n = 129). FISH analysis turned out to be distinctly more sensitive in the delineation of aberrations. Complex karyotypic alterations were associated with higher proliferation indices and inferior prognosis. A comprehensive analysis of biological features including genetic alterations in MCL by hierarchical clustering resulted in the delineation of four tumour subgroups differing with respect to their genetic constitution and suggesting different transformation or progression pathways. Moreover, in one of the groups identified, a more indolent clinical behaviour was associated with few secondary aberrations and fewer known high-risk chromosomal aberrations, which points to the importance of the quality of karyotypic evolution in MCL tumours.


Subject(s)
Interphase/genetics , Lymphoma, Mantle-Cell/genetics , Adult , Aged , Aged, 80 and over , Antigens, CD/blood , Biomarkers, Tumor/blood , Cytogenetics , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Survival Analysis , Translocation, Genetic , Tumor Suppressor Protein p53/metabolism
20.
Haematologica ; 93(5): 680-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18367489

ABSTRACT

BACKGROUND: The genetic hallmark of mantle cell lymphoma is a t(11;14)(q13;q32). However, additional genomic alterations are likely involved in the pathogenesis of this lymphoma. DESIGN AND METHODS: To determine the incidence and clinical relevance of these aberrations, we analyzed 103 well-characterized samples of mantle cell lymphoma by fluorescence in situ hybridization for the most common recurrent additional genomic findings. RESULTS: Screening 16 different regions we detected additional genomic aberrations in 92% of the cases of mantle cell lymphoma. Common gains included 3q26, 8q24, 15q23, 7p15, and common losses 13q14, 11q22-q23, 9p21, 1p22, 17p13, 6q27, and 8p22. Deletions 8p22, 9p21, 13q14, and gain of 7p15 were associated with evidence of clonal heterogeneity. While there was no correlation of additional genomic aberrations and VH-mutation status, gain of 15q23 and deletion 6q27 were associated with lower disease stage (p=0.01 and p=0.04, respectively). Patients with deletion 13q14 had shorter overall survival times (p=0.01), and there was a strong trend towards inferior outcome in patients with deletion 9p21 (p=0.07). In multivariable analysis, loss of 13q14 and an International Prognosis Index score >/= 3 turned out to be significantly associated with inferior clinical outcome (p=0.002 and p<0.001, respectively). CONCLUSIONS: The comprehensive analysis of additional genomic aberrations in mantle cell lymphoma provided further evidence for the prognostic relevance of loss of 13q14, which warrants evaluation within prospective trials. Furthermore, our analysis gave novel insights into the pathogenesis of mantle cell lymphoma with regard to the detection of clonal heterogeneity, possibly indicating clonal evolution in this type of lymphoma.


Subject(s)
Chromosome Aberrations , In Situ Hybridization, Fluorescence/methods , Interphase , Lymphoma, Mantle-Cell/epidemiology , Lymphoma, Mantle-Cell/genetics , Adult , Aged , Aged, 80 and over , Female , Genomics , Humans , Incidence , Lymphoma, Mantle-Cell/diagnosis , Male , Middle Aged , Prognosis , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...