Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Oncol ; 14(3): 571-589, 2020 03.
Article in English | MEDLINE | ID: mdl-31825135

ABSTRACT

Macrophages (Mφ) are abundantly present in the tumor microenvironment and may predict outcome in solid tumors and defined lymphoma subtypes. Mφ heterogeneity, the mechanisms of their recruitment, and their differentiation into lymphoma-promoting, alternatively activated M2-like phenotypes are still not fully understood. Therefore, further functional studies are required to understand biological mechanisms associated with human tumor-associated Mφ (TAM). Here, we show that the global mRNA expression and protein abundance of human Mφ differentiated in Hodgkin lymphoma (HL)-conditioned medium (CM) differ from those of Mφ educated by conditioned media from diffuse large B-cell lymphoma (DLBCL) cells or, classically, by macrophage colony-stimulating factor (M-CSF). Conditioned media from HL cells support TAM differentiation through upregulation of surface antigens such as CD40, CD163, CD206, and PD-L1. In particular, RNA and cell surface protein expression of mannose receptor 1 (MRC1)/CD206 significantly exceed the levels induced by classical M-CSF stimulation in M2-like Mφ; this is regulated by interleukin 13 to a large extent. Functionally, high CD206 enhances mannose-dependent endocytosis and uptake of type I collagen. Together with high matrix metalloprotease9 secretion, HL-TAMs appear to be active modulators of the tumor matrix. Preclinical in ovo models show that co-cultures of HL cells with monocytes or Mφ support dissemination of lymphoma cells via lymphatic vessels, while tumor size and vessel destruction are decreased in comparison with lymphoma-only tumors. Immunohistology of human HL tissues reveals a fraction of cases feature large numbers of CD206-positive cells, with high MRC1 expression being characteristic of HL-stage IV. In summary, the lymphoma-TAM interaction contributes to matrix-remodeling and lymphoma cell dissemination.


Subject(s)
Culture Media, Conditioned/pharmacology , Hodgkin Disease/metabolism , Lymphoma, B-Cell/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Tumor Microenvironment , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , B7-H1 Antigen/metabolism , CD40 Antigens/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Collagen Type I/metabolism , Culture Media, Conditioned/metabolism , Fluorescent Antibody Technique , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Interleukin-13/metabolism , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Macrophages/drug effects , Membrane Glycoproteins/immunology , Monocytes/metabolism , Neoplasm Metastasis/immunology , Proteome/genetics , Proteome/metabolism , RNA-Seq , Receptors, Cell Surface/metabolism , Receptors, Immunologic/immunology , Up-Regulation , Xenograft Model Antitumor Assays
2.
J Pathol ; 248(2): 142-154, 2019 06.
Article in English | MEDLINE | ID: mdl-30666658

ABSTRACT

The Epstein-Barr virus (EBV) is found almost exclusively in the activated B-cell (ABC) subtype of diffuse large B-cell lymphoma (DLBCL), yet its contribution to this tumour remains poorly understood. We have focused on the EBV-encoded latent membrane protein-1 (LMP1), a constitutively activated CD40 homologue expressed in almost all EBV-positive DLBCLs and which can disrupt germinal centre (GC) formation and drive lymphomagenesis in mice. Comparison of the transcriptional changes that follow LMP1 expression with those that follow transient CD40 signalling in human GC B cells enabled us to define pathogenic targets of LMP1 aberrantly expressed in ABC-DLBCL. These included the down-regulation of S1PR2, a sphingosine-1-phosphate (S1P) receptor that is transcriptionally down-regulated in ABC-DLBCL, and when genetically ablated leads to DLBCL in mice. Consistent with this, we found that LMP1-expressing primary ABC-DLBCLs were significantly more likely to lack S1PR2 expression than were LMP1-negative tumours. Furthermore, we showed that the down-regulation of S1PR2 by LMP1 drives a signalling loop leading to constitutive activation of the phosphatidylinositol-3-kinase (PI3-K) pathway. Finally, core LMP1-PI3-K targets were enriched for lymphoma-related transcription factors and genes associated with shorter overall survival in patients with ABC-DLBCL. Our data identify a novel function for LMP1 in aggressive DLBCL. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Viral Matrix Proteins/metabolism , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cell Line, Tumor , Cell Transformation, Viral , Databases, Genetic , Epstein-Barr Virus Infections/mortality , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Host-Pathogen Interactions , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/virology , Phosphatidylinositol 3-Kinase/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors/genetics , Viral Matrix Proteins/genetics
3.
Cancers (Basel) ; 10(9)2018 Aug 24.
Article in English | MEDLINE | ID: mdl-30149502

ABSTRACT

The Epstein-Barr virus (EBV) is present in the tumour cells of a subset of patients with classic Hodgkin lymphoma (cHL), yet the contribution of the virus to the pathogenesis of these tumours remains only poorly understood. The EBV genome in virus-associated cHL expresses a limited subset of genes, restricted to the non-coding Epstein-Barr virus-encoded RNAs (EBERs) and viral miRNA, as well as only three virus proteins; the Epstein-Barr virus nuclear antigen-1 (EBNA1), and the two latent membrane proteins, known as LMP1 and LMP2, the latter of which has two isoforms, LMP2A and LMP2B. LMP1 and LMP2A are of particular interest because they are co-expressed in tumour cells and can activate cellular signalling pathways, driving aberrant cellular transcription in infected B cells to promote lymphomagenesis. This article seeks to bring together the results of recent studies of the latent membrane proteins in different B cell systems, including experiments in animal models as well as a re-analysis of our own transcriptional data. In doing so, we summarise the potentially co-operative and antagonistic effects of the LMPs that are relevant to B cell lymphomagenesis.

4.
Oncotarget ; 7(30): 47061-47081, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27166259

ABSTRACT

To discover new regulatory pathways in B lymphoma cells, we performed a combined analysis of experimental, clinical and global gene expression data. We identified a specific cluster of genes that was coherently expressed in primary lymphoma samples and suppressed by activation of the B cell receptor (BCR) through αIgM treatment of lymphoma cells in vitro. This gene cluster, which we called BCR.1, includes numerous cell cycle regulators. A reduced expression of BCR.1 genes after BCR activation was observed in different cell lines and also in CD10+ germinal center B cells. We found that BCR activation led to a delayed entry to and progression of mitosis and defects in metaphase. Cytogenetic changes were detected upon long-term αIgM treatment. Furthermore, an inverse correlation of BCR.1 genes with c-Myc co-regulated genes in distinct groups of lymphoma patients was observed. Finally, we showed that the BCR.1 index discriminates activated B cell-like and germinal centre B cell-like diffuse large B cell lymphoma supporting the functional relevance of this new regulatory circuit and the power of guided clustering for biomarker discovery.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Lymphoma, Large B-Cell, Diffuse/genetics , Receptors, Antigen, B-Cell/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cohort Studies , Gene Expression Profiling , Germinal Center/metabolism , Germinal Center/pathology , Humans , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-6 , Proto-Oncogene Proteins c-myc , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/genetics , Tumor Microenvironment
5.
J Pathol ; 235(2): 312-22, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25294567

ABSTRACT

Since the discovery in 1964 of the Epstein-Barr virus (EBV) in African Burkitt lymphoma, this virus has been associated with a remarkably diverse range of cancer types. Because EBV persists in the B cells of the asymptomatic host, it can easily be envisaged how it contributes to the development of B-cell lymphomas. However, EBV is also found in other cancers, including T-cell/natural killer cell lymphomas and several epithelial malignancies. Explaining the aetiological role of EBV is challenging, partly because the virus probably contributes differently to each tumour and partly because the available disease models cannot adequately recapitulate the subtle variations in the virus-host balance that exist between the different EBV-associated cancers. A further challenge is to identify the co-factors involved; because most persistently infected individuals will never develop an EBV-associated cancer, the virus cannot be working alone. This article will review what is known about the contribution of EBV to lymphoma development.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Lymphoma/virology , Animals , Biopsy , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/immunology , Host-Pathogen Interactions , Humans , Lymphoma/immunology , Lymphoma/pathology , Pathology, Molecular/methods , Predictive Value of Tests , Prognosis , Risk Factors , Virology/methods , Virulence
6.
Chin J Cancer ; 33(12): 591-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25418190

ABSTRACT

Although Epstein-Barr virus (EBV) is present in the malignant Hodgkin/Reed-Sternberg (HRS) cells of a proportion of cases of classical Hodgkin lymphoma (cHL), how the virus contributes to the pathogenesis of this disease remains poorly defined. It is clear from the studies of other EBV-associated cancers that the virus is usually not sufficient for tumor development and that other oncogenic co-factors are required. This article reviews what is known about the contribution of EBV to the pathogenesis of cHL and focuses on emerging evidence implicating chronic inflammation as a potential oncogenic co-factor in this malignancy.


Subject(s)
Herpesvirus 4, Human , Hodgkin Disease/virology , Epstein-Barr Virus Infections , Humans , Reed-Sternberg Cells
7.
J Gen Virol ; 95(Pt 9): 1861-1869, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24893782

ABSTRACT

The relationship between Epstein-Barr virus (EBV) and the germinal centre (GC) of the asymptomatic host remains an enigma. The occasional appearance of EBV-positive germinal centres in some patients, particularly those with a history of immunosuppression, suggests that EBV numbers in the GC are subject to immune control. The relationship, if any, between lymphoid hyperplasia with EBV-positive germinal centres and subsequent or concurrent lymphomagenesis remains to be clarified. As far as the development of EBV-associated Hodgkin's lymphoma is concerned, the suppression of virus replication, mediated by LMP1 on the one hand, and the loss of B-cell receptor signalling on the other, appears to be an important pathogenic mechanism. A further important emerging concept is that alterations in the microenvironment of the EBV-infected B-cell may be important for lymphomagenesis.


Subject(s)
B-Lymphocytes/virology , Germinal Center/immunology , Germinal Center/virology , Herpesvirus 4, Human/immunology , Hodgkin Disease/virology , Adult , B-Lymphocytes/immunology , Cell Differentiation/immunology , Epstein-Barr Virus Infections/virology , Female , Hodgkin Disease/immunology , Humans , Male , Middle Aged , Pseudolymphoma/virology , Receptors, Antigen, B-Cell/immunology , Viral Matrix Proteins , Virus Replication/immunology , Young Adult
8.
Blood ; 122(26): 4237-45, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24136166

ABSTRACT

The malignant Hodgkin and Reed-Sternberg (HRS) cells of Hodgkin lymphoma are surrounded by a tumor microenvironment that is composed of a variety of cell types, as well as noncellular components such as collagen. Although HRS cells harbor oncogenic Epstein-Barr virus (EBV) in approximately 50% of cases, it is not known if the tumor microenvironment contributes to EBV-driven lymphomagenesis. We show that expression of the EBV-encoded latent membrane protein-1 (LMP1) in primary human germinal center B cells, the presumed progenitors of HRS cells, upregulates discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase activated by collagen. We also show that HRS cells intimately associated with collagen frequently overexpress DDR1 and that short-term exposure to collagen is sufficient to activate DDR1 in Hodgkin lymphoma-derived cell lines. The ectopic expression of DDR1 significantly increased the survival of collagen-treated DG75 Burkitt lymphoma cells, following etoposide treatment. Conversely, knockdown of DDR1 significantly decreased the survival of collagen-treated L428 Hodgkin lymphoma cells in the absence of specific apoptotic stimulus, suggesting that DDR1 also influences baseline survival. Our results identify a hitherto unknown function for collagen in protecting Hodgkin lymphoma cells from apoptosis and suggest an important contribution of the tumor microenvironment in promoting the oncogenic effects of EBV.


Subject(s)
B-Lymphocytes/cytology , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Hodgkin Disease/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Viral Matrix Proteins/genetics , B-Lymphocytes/physiology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Death/physiology , Cells, Cultured , Collagen/metabolism , Discoidin Domain Receptor 1 , Epstein-Barr Virus Infections/pathology , Gene Expression Regulation, Viral , Gene Knockdown Techniques , Germinal Center/cytology , Hodgkin Disease/pathology , Humans , Phosphorylation/physiology , Receptor Protein-Tyrosine Kinases/genetics , Reed-Sternberg Cells/cytology , Reed-Sternberg Cells/physiology , Tumor Microenvironment/physiology
9.
PLoS One ; 8(5): e64868, 2013.
Article in English | MEDLINE | ID: mdl-23724103

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic virus that is associated with the pathogenesis of several human lymphoid malignancies, including Hodgkin's lymphoma. Infection of normal resting B cells with EBV results in activation to lymphoblasts that are phenotypically similar to those generated by physiological stimulation with CD40L plus IL-4. One important difference is that infection leads to the establishment of permanently growing lymphoblastoid cell lines, whereas CD40L/IL-4 blasts have finite proliferation lifespans. To identify early events which might later determine why EBV infected blasts go on to establish transformed cell lines, we performed global transcriptome analyses on resting B cells and on EBV and CD40L/IL-4 blasts after 7 days culture. As anticipated there was considerable overlap in the transcriptomes of the two types of lymphoblasts when compared to the original resting B cells, reflecting common changes associated with lymphocyte activation and proliferation. Of interest to us was a subset of 255 genes that were differentially expressed between EBV and CD40L/IL-4 blasts. Genes which were more highly expressed in EBV blasts were substantially and significantly enriched for a set of interferon-stimulated genes which on further in silico analyses were found to be repressed by IL-4 in other cell contexts and to be up-regulated in micro-dissected malignant cells from Hodgkin's lymphoma biopsies when compared to their normal germinal center cell counterparts. We hypothesized that EBV and IL-4 were targeting and discordantly regulating a common set of genes. This was supported experimentally in our B cell model where IL-4 stimulation partially reversed transcriptional changes which follow EBV infection and it impaired the efficiency of EBV-induced B cell transformation. Taken together, these data suggest that the discordant regulation of interferon and IL-4 pathway genes by EBV that occurs early following infection of B cells has relevance to the development or maintenance of an EBV-associated malignancy.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Transformation, Viral/genetics , Gene Expression Regulation , Herpesvirus 4, Human/physiology , Interferon Regulatory Factors/genetics , Interleukin-4/metabolism , B-Lymphocytes/pathology , CD40 Ligand/immunology , CD40 Ligand/pharmacology , Cell Line, Tumor , Cell Transformation, Viral/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hodgkin Disease/genetics , Hodgkin Disease/metabolism , Humans , Interferons/metabolism , Interleukin-4/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Signal Transduction , Transcriptome , Viral Matrix Proteins/metabolism
10.
J Pathol ; 230(4): 399-409, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23592216

ABSTRACT

Hodgkin's lymphoma is unusual among B cell lymphomas, in so far as the malignant Hodgkin/Reed-Sternberg (HRS) cells lack a functional B cell receptor (BCR), as well as many of the required downstream signalling components. In Epstein-Barr virus (EBV)-positive cases of Hodgkin's lymphoma, HRS cells express the viral latent membrane proteins (LMP)-1 and -2A. LMP2A is thought to contribute to the pathogenesis of Hodgkin's lymphoma by providing a surrogate BCR-like survival signal. However, LMP2A has also been shown to induce the virus-replicative cycle in B cells, an event presumably incompatible with lymphomagenesis. In an attempt to resolve this apparent paradox, we compared the transcriptional changes observed in primary HRS cells with those induced by LMP2A and by BCR activation in primary human germinal centre (GC) B cells, the presumed progenitors of HRS cells. We found a subset of genes that were up-regulated by both LMP2A expression and BCR activation but which were down-regulated in primary HRS cells. These genes included EGR1, an immediate-early gene that is required for BCR-induced entry to the virus-replicative cycle. We present data supporting a model for the pathogenesis of EBV-positive Hodgkin's lymphoma in which LMP2A-expressing HRS cells lacking BCR signalling functions cannot induce EGR1 and are consequently protected from entry to the virus lytic cycle. The primary microarray data are available from GEO (http://www.ncbi.nlm.nih.gov/geo/) under series Accession No 46143.


Subject(s)
Early Growth Response Protein 1/metabolism , Herpesvirus 4, Human/metabolism , Hodgkin Disease/metabolism , Reed-Sternberg Cells/metabolism , Viral Matrix Proteins/metabolism , B-Lymphocytes/metabolism , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Cell Line, Tumor , Cytopathogenic Effect, Viral , Early Growth Response Protein 1/genetics , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Hodgkin Disease/virology , Humans , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, B-Cell/metabolism , Reed-Sternberg Cells/pathology , Reed-Sternberg Cells/virology , Trans-Activators/metabolism , Transfection , Up-Regulation , Viral Matrix Proteins/genetics
11.
Am J Pathol ; 182(4): 1092-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23375451

ABSTRACT

Burkitt's lymphoma (BL) is a highly malignant, aggressive non-Hodgkin's lymphoma derived from germinal center B cells. Recently, global gene expression profiling of patient samples led to a molecular definition of BL with lymphocyte enhancer-binding factor 1 (LEF1) as a signature gene. Herein, we report the expression of nucleic LEF1 in 15 of 18 patients with BL and the identification of LEF1 target genes. Germinal center B cells were devoid of detectable nuclear LEF1 expression, as were mantle cell lymphoma (0 of 5), marginal zone lymphoma (0 of 6), follicular lymphoma (0 of 12), and diffuse large B-cell lymphoma (1 of 31). Whole-genome gene expression profiling after transient knockdown of LEF1 in BL cell lines identified new LEF1 target genes; these LEF1 targets are enriched with genes associated with cancers. The expression of LEF1 and LEF1-regulated genes in primary BL suggests that LEF1 is not only aberrantly expressed but also transcriptionally active. This study supports a functionally important role for LEF1 and its target genes in BLs.


Subject(s)
Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Lymphoid Enhancer-Binding Factor 1/metabolism , Burkitt Lymphoma/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Oligonucleotide Array Sequence Analysis , Palatine Tonsil/metabolism , Palatine Tonsil/pathology
12.
Cell Commun Signal ; 10(1): 43, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23253402

ABSTRACT

BACKGROUND: Aggressive Non-Hodgkin lymphomas (NHL) are a group of lymphomas derived from germinal centre B cells which display a heterogeneous pattern of oncogenic pathway activation. We postulate that specific immune response associated signalling, affecting gene transcription networks, may be associated with the activation of different oncogenic pathways in aggressive Non-Hodgkin lymphomas (NHL). METHODOLOGY: The B cell receptor (BCR), CD40, B-cell activating factor (BAFF)-receptors and Interleukin (IL) 21 receptor and Toll like receptor 4 (TLR4) were stimulated in human transformed germinal centre B cells by treatment with anti IgM F(ab)2-fragments, CD40L, BAFF, IL21 and LPS respectively. The changes in gene expression following the activation of Jak/STAT, NF-кB, MAPK, Ca2+ and PI3K signalling triggered by these stimuli was assessed using microarray analysis. The expression of top 100 genes which had a change in gene expression following stimulation was investigated in gene expression profiles of patients with Aggressive non-Hodgkin Lymphoma (NHL). RESULTS: αIgM stimulation led to the largest number of changes in gene expression, affecting overall 6596 genes. While CD40L stimulation changed the expression of 1194 genes and IL21 stimulation affected 902 genes, only 283 and 129 genes were modulated by lipopolysaccharide or BAFF receptor stimulation, respectively. Interestingly, genes associated with a Burkitt-like phenotype, such as MYC, BCL6 or LEF1, were affected by αIgM. Unique and shared gene expression was delineated. NHL-patients were sorted according to their similarity in the expression of TOP100 affected genes to stimulated transformed germinal centre B cells The αIgM gene module discriminated individual DLBCL in a similar manner to CD40L or IL21 gene modules. DLBCLs with low module activation often carry chromosomal MYC aberrations. DLBCLs with high module activation show strong expression of genes involved in cell-cell communication, immune responses or negative feedback loops. Using chemical inhibitors for selected kinases we show that mitogen activated protein kinase- and phosphoinositide 3 kinase-signalling are dominantly involved in regulating genes included in the αIgM gene module. CONCLUSION: We provide an in vitro model system to investigate pathway activation in lymphomas. We defined the extent to which different immune response associated pathways are responsible for differences in gene expression which distinguish individual DLBCL cases. Our results support the view that tonic or constitutively active MAPK/ERK pathways are an important part of oncogenic signalling in NHL. The experimental model can now be applied to study the therapeutic potential of deregulated oncogenic pathways and to develop individual treatment strategies for lymphoma patients.

13.
Int J Cancer ; 131(4): E348-61, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-21913186

ABSTRACT

Gene expression profiling has recently enabled the reclassification of aggressive non-Hodgkin lymphomas (aNHL) into distinct subgroups. In Burkitt lymphoma (BL) aberrant c-Myc activity results from IG-MYC translocations. However, MYC aberrations are not limited to BLs and then have a negative prognostic impact. In this study, we investigated to which extent aberrant c-Myc activity plays a functional role in other aNHL and whether it is independent from MYC translocations. Based on a combined microarray analysis of human germinal center (GC) B cells transfected with c-Myc and 220 aNHLs cases, we developed a "c-Myc index." This index measures the extent to which lymphomas express c-Myc responsive genes. It comprises genes that are affected in a variety of tumors compared to normal tissue. This supports the view that aberrant c-Myc expression in GC B cells triggers a tumor-like expression pattern. As expected, the "c-Myc index" is very high in molecular Burkitt lymphoma (mBL), but more importantly also high within other aNHL. It constitutes a negative prognostic marker independent of established risk factors and of the presence of a MYC translocation. Our data provide new insights into the role of c-Myc activity in different lymphomas and raises the question of treatment changes for those patients under risk.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Fluorescent Antibody Technique , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Real-Time Polymerase Chain Reaction
14.
Pathogens ; 1(2): 83-101, 2012 Oct 08.
Article in English | MEDLINE | ID: mdl-25436766

ABSTRACT

B-lymphocyte-induced maturation protein 1 (BLIMP1) exists as two major isoforms, α and ß, which arise from alternate promoters. Inactivation of the full length BLIMP1α isoform is thought to contribute to B cell lymphomagenesis by blocking post-germinal centre (GC) B cell differentiation. In contrast, the shorter ß isoform is functionally impaired and over-expressed in several haematological malignancies, including diffuse large B cell lymphomas (DLBCL). We have studied the influence on BLIMP1ß expression of the Epstein-Barr virus (EBV), a human herpesvirus that is implicated in the pathogenesis of several GC-derived lymphomas, including a subset of DLBCL and Hodgkin's lymphoma (HL). We show that BLIMP1ß expression is increased following the EBV infection of normal human tonsillar GC B cells. We also show that this change in expression is accompanied by hypomethylation of the BLIMP1ß-specific promoter. Furthermore, we confirmed previous reports that the BLIMP1ß promoter is hypomethylated in DLBCL cell lines and show for the first time that BLIMP1ß is hypomethylated in the Hodgkin/Reed-Sternberg (HRS) cells of HL. Our results provide evidence in support of a role for BLIMP1ß in the pathogenesis of EBV-associated B cell lymphomas.

15.
J Virol ; 85(18): 9568-77, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21752916

ABSTRACT

Although Epstein-Barr virus (EBV) usually establishes an asymptomatic lifelong infection, it is also implicated in the development of germinal center (GC) B-cell-derived malignancies, including Hodgkin's lymphoma (HL). Following primary infection, EBV remains latent in the memory B-cell population, where host-driven methylation of viral DNA contributes to the repression of viral gene expression. However, it is still unclear how EBV harnesses the cell's methylation machinery in B cells, how this contributes to viral persistence, and what impact this has on the methylation of cellular genes. We show that EBV infection of GC B cells is followed by upregulation of the DNA methyltransferase DNMT3A and downregulation of DNMT3B and DNMT1. We show that the EBV latent membrane protein 1 (LMP1) oncogene downregulates DNMT1 and that DNMT3A binds to the viral promoter Wp. Genome-wide promoter arrays performed with these cells showed that EBV-associated methylation changes in cellular genes were not randomly distributed across the genome but clustered at chromosomal locations, consistent with an instructive pattern of methylation, and were in part determined by promoter CpG content. Both DNMT3B and DNMT1 were downregulated and DNMT3A was upregulated in HL cell lines, recapitulating the pattern of expression observed following EBV infection of GC B cells. We also found, by using gene expression profiling, that genes differentially expressed following EBV infection of GC B cells were significantly enriched for those reported to be differentially expressed in HL. These observations suggest that EBV-infected GC B cells are a useful model for studying virus-associated changes contributing to the pathogenesis of HL.


Subject(s)
B-Lymphocytes/immunology , Epigenesis, Genetic , Gene Expression Regulation , Germinal Center/immunology , Herpesvirus 4, Human/pathogenicity , Hodgkin Disease/virology , Transcription, Genetic , B-Lymphocytes/virology , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation , DNA Methyltransferase 3A , Gene Expression Profiling , Germinal Center/virology , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Viral Matrix Proteins/metabolism , DNA Methyltransferase 3B
16.
Blood ; 117(22): 5907-17, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21411757

ABSTRACT

An important pathogenic event in Epstein-Barr virus (EBV)-associated lymphomas is the suppression of virus replication, which would otherwise lead to cell death. Because virus replication in B cells is intimately linked to their differentiation toward plasma cells, we asked whether the physiologic signals that drive normal B-cell differentiation are absent in EBV-transformed cells. We focused on BLIMP1α, a transcription factor that is required for plasma cell differentiation and that is inactivated in diffuse large B-cell lymphomas. We show that BLIMP1α expression is down-regulated after EBV infection of primary germinal center B cells and that the EBV oncogene, latent membrane protein-1 (LMP-1), is alone capable of inducing this down-regulation in these cells. Furthermore, the down-regulation of BLIMP1α by LMP-1 was accompanied by a partial disruption of the BLIMP1α transcriptional program, including the aberrant induction of MYC, the repression of which is required for terminal differentiation. Finally, we show that the ectopic expression of BLIMP1α in EBV-transformed cells can induce the viral lytic cycle. Our results suggest that LMP-1 expression in progenitor germinal center B cells could contribute to the pathogenesis of EBV-associated lymphomas by down-regulating BLIMP1α, in turn preventing plasma cell differentiation and induction of the viral lytic cycle.


Subject(s)
B-Lymphocytes/virology , Cell Differentiation , Herpesvirus 4, Human/physiology , Lymphoma, B-Cell/etiology , Plasma Cells/pathology , Repressor Proteins/metabolism , Viral Matrix Proteins/metabolism , B-Lymphocytes/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Transformation, Viral , Cells, Cultured , Child , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Gene Expression Profiling , Germinal Center , Humans , Immunoenzyme Techniques , Lymphoma, B-Cell/pathology , Oligonucleotide Array Sequence Analysis , Palatine Tonsil/cytology , Palatine Tonsil/metabolism , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , RNA, Messenger/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Matrix Proteins/genetics , Virus Latency , Virus Replication
17.
Cancer Treat Rev ; 36(4): 348-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20193983

ABSTRACT

The Epstein-Barr virus (EBV) is a lymphotropic herpes virus with oncogenetic properties which can lead to the development of lymphomas such as Burkitt's lymphoma (BL), Hodgkin's lymphoma (HL), or post-transplant lymphoma. This review discusses our current understanding of lymphomagenesis in relation to EBV and the potential for targeted therapies.


Subject(s)
Herpesvirus 4, Human/isolation & purification , Lymphoma/etiology , Antiviral Agents/therapeutic use , Burkitt Lymphoma/etiology , Burkitt Lymphoma/virology , Child , Hodgkin Disease/etiology , Hodgkin Disease/virology , Humans , Immunotherapy, Adoptive , Lymphoma/virology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/virology , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/virology
18.
Int J Cancer ; 123(10): 2285-93, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18704935

ABSTRACT

Classical Hodgkin lymphoma (cHL) is characterized histologically by a minority of malignant Hodgkin Reed-Sternberg cells surrounded by abundant inflammatory cells, generally believed to be of major importance in the pathophysiology of the disease. Here, we present data that link inflammatory cell-derived arachidonic acid metabolites, the cysteinyl leukotrienes (CysLT), to the pathogenesis of cHL. Two HL cell lines, L1236 and KMH2, were shown to express functional CysLT(1) receptors, responding with a robust calcium signal upon leukotriene (LT) D(4) challenge. LTD(4) stimulated protein release of tumor necrosis factor-alpha, interleukin-6 and -8 by L1236 cells and interleukin-8 by KMH2 cells. Importantly, all these LTD(4)-induced effects were blocked by the CysLT(1) receptor-specific antagonist zafirlukast. Immunohistochemical studies of cHL biopsies and microarray analysis of microdissected cells revealed that the CysLT(1) receptor is expressed also by primary Hodgkin Reed-Sternberg cells. As these cells are surrounded by CysLT-producing eosinophils, macrophages and mast cells, our results suggest the CysLTs as mediators in the pathogenesis of cHL, contributing to the aberrant cytokine network of this lymphoma.


Subject(s)
Cysteine/chemistry , Hodgkin Disease/physiopathology , Leukotrienes/physiology , Adolescent , Adult , Aged , Calcium Signaling , Cell Line, Tumor , Child , Child, Preschool , Female , Hodgkin Disease/metabolism , Humans , Immunohistochemistry , Leukotriene D4/pharmacology , Leukotrienes/chemistry , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Receptors, Leukotriene/genetics , Receptors, Leukotriene/metabolism
19.
Blood ; 109(6): 2597-603, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17148591

ABSTRACT

Polycomb group (PcG) proteins are chromatin modifiers that are necessary for the maintenance and renewal of embryonic and adult stem cells. However, overexpression of the PcG protein, Bmi-1, causes lymphoma in transgenic mice. We show that Bmi-1 is up-regulated in Hodgkin lymphoma (HL) cells by the Epstein-Barr virus (EBV) oncogene latent membrane protein-1 (LMP1) and that this up-regulation is mediated by NF-kappaB signaling. We also show that Bmi-1 is up-regulated by NF-kappaB in EBV-negative HL cells. Down-regulation of LMP1 and Bmi-1 decreased the survival of HL cells, suggesting that Bmi-1 may mediate the prosurvival effects of LMP1-induced NF-kappaB signaling in HL cells. Transcriptional targets of Bmi-1 were identified after its knockdown in an HL cell line. We show here that Bmi-1 and LMP1 down-regulate the ataxia telangiectasia-mutated (ATM) tumor suppressor and conclude that Bmi-1 contributes to LMP1-induced oncogenesis in HL.


Subject(s)
Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/metabolism , Hodgkin Disease/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Viral Matrix Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Survival , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Expression Profiling , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Humans , NF-kappa B/metabolism , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation , Viral Matrix Proteins/genetics
20.
Science ; 307(5709): 593-6, 2005 Jan 28.
Article in English | MEDLINE | ID: mdl-15591165

ABSTRACT

CD4+ T cells classically recognize antigens that are endocytosed and processed in lysosomes for presentation on major histocompatibility complex (MHC) class II molecules. Here, endogenous Epstein-Barr virus nuclear antigen 1 (EBNA1) was found to gain access to this pathway by autophagy. On inhibition of lysosomal acidification, EBNA1, the dominant CD4+ T cell antigen of latent Epstein-Barr virus infection, slowly accumulated in cytosolic autophagosomes. In addition, inhibition of autophagy decreased recognition by EBNA1-specific CD4+ T cell clones. Thus, lysosomal processing after autophagy may contribute to MHC class II-restricted surveillance of long-lived endogenous antigens including nuclear proteins relevant to disease.


Subject(s)
Antigen Presentation , Autophagy , Epstein-Barr Virus Nuclear Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Phagosomes/metabolism , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Chloroquine/pharmacology , Epstein-Barr Virus Nuclear Antigens/immunology , Humans , Hydrogen-Ion Concentration , Lysosomes/immunology , Lysosomes/metabolism , Microsomes/metabolism , Phagosomes/immunology , Phagosomes/ultrastructure , Proteasome Endopeptidase Complex/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...