The BCL6 Oncoprotein Forms Distinct Transcriptional Repression Complexes on Cohorts of Target Genes Involved in Specific Cellular Functions.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2615-2615
Author(s):  
Jose M. Polo ◽  
Stella Maris Ranuncolo ◽  
Samir Parek ◽  
Jose Arcaya ◽  
Ari M. Melnick

Abstract The BCL6 (B-Cell-Lymphoma-6) oncoprotein is required for formation of germinal centers by normal B-cells and is frequently constitutively activated in B-cell lymphomas. BCL6 is a transcriptional repressor of the BTB/POZ - zinc finger family of proteins. Transcriptional repressors are generally believed to function by binding to specific DNA sequences in target promoters and then recruiting a cohort of corepressor proteins that physically alter the chromatin structure of the locus leading to silencing. BCL6 can recruit several different corepressors. Accordingly, N-CoR, SMRT and BCoR bind to the BTB domain of BCL6, the NuRD complex binds to the second repression domain and ETO binds to the C-terminal zinc finger domain. These proteins recruit additional corepressors such as HDACS, thus forming multi-protein complexes. BCL6 is involved in both licensing germinal center B-cells for survival as well as blocking differentiation to memory or plasma cells. However, we found that blockade of the BCL6 BTB domain with a specific inhibitor causes only apoptosis but not differentiation of B-cells. In order to identify BCL6 target genes and the mechanism through which they are silenced, we performed extensive ChIP on chip analysis of BCL6, its corepressors, and their chromatin signatures using tiled oligonucleotide arrays containing 1.5 KB of 24,000 promoters. These were performed in the presence or absence of BCL6 shRNA and other BCL6 inhibitor molecules, in tandem with expression arrays as a functional readout. We also performed in depth ChIP on chip experiments using custom arrays, where sets of entire BCL6 target loci were tiled through with overlapping oligos. From these studies we i) identified a large cohort of direct BCL6 target genes involved in apoptosis, cell damage, protein degradation and differentiation that provide critical insight into the mechanism of action of BCL6 in normal and malignant B-cells; ii) discovered that cohorts of BCL6 direct target genes involved in different pathways are regulated by specific corepressors, which are mutually exclusive in their binding to BCL6 target loci, iii) that BCL6 can repress genes through a variety of different mechanisms with unique chromatin signatures. Thus, previously unrecognized mechanisms exist in transcriptional repression, that determine formation of specific corepressor complexes. Using this information, we have been able to re-activate discrete cohorts of BCL6 target genes controlled by specific corepressors, resulting in specific biological effects such as apoptosis or differentiation of lymphoma cells. These results provide fundamental insights into the transcriptional and biological mechanism of action of BCL6 in B-cells.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5-5 ◽  
Author(s):  
Jose M. Polo ◽  
Tania Dell’Oso ◽  
Stella Maris Ranuncolo ◽  
Leandro Cerchietti ◽  
Gustavo F. Da Silva ◽  
...  

Abstract The Bcl-6 transcriptional repressor is required for the establishment of germinal centers during B-cell maturation. Regulatory elements of the BCL6 gene are frequently mutated in diffuse large B-cell lymphomas (DLBCL), leading to inappropriately timed expression of Bcl-6. However, it is unknown whether Bcl-6 is involved in lymphomagenesis. Within Bcl-6, the N-terminal BTB domain is involved in mediating transcriptional repression. We investigated the mechanism of action of the Bcl-6 BTB domain using X-ray crystallography and functional assays. We found that the SMRT, N-CoR and BCoR corepressors bind directly to Bcl-6 through a 16 residue “BBD motif”, which fits into a highly specific “lateral groove” surface feature of the Bcl-6 BTB domain. To determine the contribution of BTB lateral groove recruitment of corepressors to transcriptional and biological functions of Bcl-6, we engineered small Bcl-6 BTB peptide inhibitors (BPI). BPI penetrate cells and localize to the nuclei where they specifically bound to Bcl-6 and blocked its interaction and co-localization with co-repressors. This resulted in dose-dependent blockade of repression by Bcl-6 in reporter assays and reactivation of endogenous Bcl-6 target genes in Bcl-6 expressing B-cells measured by real-time PCR. Loss of repression was caused by disruption of the endogenous Bcl-6 transcriptional repression complex. We found that BPI specifically excluded corepressors from the promoters of endogenous Bcl-6 target genes, resulting in a switch from repressed to activated histone code settings with consequent gene reactivation. From the biological standpoint, BPI injection into mice reproduced the B-cell phenotype of Bcl-6 null animals, as germinal center formation in response to T-cell dependent antigens was abrogated. To address the question of whether Bcl-6 is required to maintain the malignant phenotype of DLBCL, a panel of Bcl-6 positive and negative DLBCL cells were exposed to BPI. BPI induced growth suppression, cell cycle arrest and apoptosis in Bcl-6 positive but not negative DLBCL cells. Expression array analysis showed immediate upregulation of checkpoint genes involved in proliferation and apoptosis, allowing us to identify novel direct Bcl-6 target genes. In contrast, BPI did not upregulate differentiation related genes and did not induce differentiation in lymphoma cells, indicating that the Bcl-6 BTB domain lateral groove controls proliferation and survival pathways but not differentiation. Finally, BPI completely suppressed growth of Bcl-6 positive human DLBCL xenotransplants without any toxicity to other organs. In summary, specific disruption of the Bcl-6 BTB domain repression mechanism allowed us to reveal the contribution of specific corepressor recruitment to the activities of Bcl-6 in B-cells and to demonstrate that Bcl-6 is in fact an oncogene, required to maintain the malignant phenotype of Bcl-6 positive DLBCL. Our pre-clinical studies indicate that Bcl-6 is a bona fide therapeutic target and that BPI is a specific, potent and non-toxic anti-lymphoma therapeutic agent.


Blood ◽  
2010 ◽  
Vol 116 (24) ◽  
pp. 5247-5255 ◽  
Author(s):  
Irina Velichutina ◽  
Rita Shaknovich ◽  
Huimin Geng ◽  
Nathalie A. Johnson ◽  
Randy D. Gascoyne ◽  
...  

Abstract EZH2 is the catalytic subunit of the PRC2 Polycomb complex and mediates transcriptional repression through its histone methyltransferase activity. EZH2 is up-regulated in normal germinal center (GC) B cells and is implicated in lymphomagenesis. To explore the transcriptional programs controlled by EZH2, we performed chromatin immunoprecipitation (ChIP-on-chip) in GC cells and found that it binds approximately 1800 promoters, often associated with DNA sequences similar to Droso-phila Polycomb response elements. While EZH2 targets overlapped extensively between GC B cells and embryonic stem cells, we also observed a large GC-specific EZH2 regulatory program. These genes are preferentially histone 3 lysine 27–trimethylated and repressed in GC B cells and include several key cell cycle–related tumor suppressor genes. Accordingly, siRNA-mediated down-regulation of EZH2 in diffuse large B-cell lymphoma (DLBCL) cells resulted in acute cell cycle arrest at the G1/S transition and up-regulation of its tumor suppressor target genes. At the DNA level, EZH2-bound promoters are hypomethylated in GC B cells, but many of them are aberrantly hypermethylated in DLBCL, suggesting disruption of normal epigenetic processes in these cells. EZH2 is thus involved in regulating a specific epigenetic program in normal GCs, including silencing of antiproliferative genes, which may contribute to the malignant transformation of GC B cells into DLBCLs.


2005 ◽  
Vol 203 (1) ◽  
pp. 63-72 ◽  
Author(s):  
Chang Hoon Lee ◽  
Mark Melchers ◽  
Hongsheng Wang ◽  
Ted A. Torrey ◽  
Rebecca Slota ◽  
...  

Interferon (IFN) consensus sequence-binding protein/IFN regulatory factor 8 (IRF8) is a transcription factor that regulates the differentiation and function of macrophages, granulocytes, and dendritic cells through activation or repression of target genes. Although IRF8 is also expressed in lymphocytes, its roles in B cell and T cell maturation or function are ill defined, and few transcriptional targets are known. Gene expression profiling of human tonsillar B cells and mouse B cell lymphomas showed that IRF8 transcripts were expressed at highest levels in centroblasts, either from secondary lymphoid tissue or transformed cells. In addition, staining for IRF8 was most intense in tonsillar germinal center (GC) dark-zone centroblasts. To discover B cell genes regulated by IRF8, we transfected purified primary tonsillar B cells with enhanced green fluorescent protein–tagged IRF8, generated small interfering RNA knockdowns of IRF8 expression in a mouse B cell lymphoma cell line, and examined the effects of a null mutation of IRF8 on B cells. Each approach identified activation-induced cytidine deaminase (AICDA) and BCL6 as targets of transcriptional activation. Chromatin immunoprecipitation studies demonstrated in vivo occupancy of 5′ sequences of both genes by IRF8 protein. These results suggest previously unappreciated roles for IRF8 in the transcriptional regulation of B cell GC reactions that include direct regulation of AICDA and BCL6.


Blood ◽  
2007 ◽  
Vol 110 (6) ◽  
pp. 2121-2127 ◽  
Author(s):  
Hai-Jun Zhou ◽  
Lan V. Pham ◽  
Archito T. Tamayo ◽  
Yen-Chiu Lin-Lee ◽  
Lingchen Fu ◽  
...  

Abstract CD40 is an integral plasma membrane–associated member of the TNF receptor family that has recently been shown to also reside in the nucleus of both normal B cells and large B-cell lymphoma (LBCL) cells. However, the physiological function of CD40 in the B-cell nucleus has not been examined. In this study, we demonstrate that nuclear CD40 interacts with the NF-κB protein c-Rel, but not p65, in LBCL cells. Nuclear CD40 forms complexes with c-Rel on the promoters of NF-κB target genes, CD154, BLyS/BAFF, and Bfl-1/A1, in various LBCL cell lines. Wild-type CD40, but not NLS-mutated CD40, further enhances c-Rel–mediated Blys promoter activation as well as proliferation in LBCL cells. Studies in normal B cells and LBCL patient cells further support a nuclear transcriptional function for CD40 and c-Rel. Cooperation between nuclear CD40 and c-Rel appears to be important in regulating cell growth and survival genes involved in lymphoma cell proliferation and survival mechanisms. Modulating the nuclear function of CD40 and c-Rel could reveal new mechanisms in LBCL pathophysiology and provide potential new targets for lymphoma therapy.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 783-783
Author(s):  
Chuanxin Huang ◽  
Ann Haberman ◽  
Ari M. Melnick

Abstract The transcriptional repressor Bcl6 is a master regulator of the germinal center (GC) reaction through directing naïve B cells and CD4+ T cells to differentiate into GC B cells and follicular T helper (TFH) cells respectively. Bcl6 mediates its action largely by recruitment of co-repressors through its N-terminal BTB domain and its middle second repression domain (RD2). The BTB domain repression function is critical for GC B cell survival and proliferation, but not important for TFH cell differentiation. However, the in vivobiological function of RD2 remains unknown. To explore the specific role of RD2 transcriptional repression in the GC reaction, we generated a knockin mouse model in which the endogenous Bcl6 locus encodes a mutant form of the protein that specifically disrupts RD2 mediated transcriptional repression. RD2 mutant mice were developmentally indistinguishable from wild-type mice and displayed normal B cell development prior to the GC phase. However, these mice failed to accumulate GCs after immunization with sheep blood cells and exhibited remarkably impaired production of high-affinity antibodies 21 days after T-cell dependent antigen immunization, indicative of severe deficiency of the GC reaction. Mixed bone marrow transplantation experiments showed that RD2 loss of function led to complete loss of GC B cells and partial impairment of TFH cell differentiation in cell-intrinsic manner. Intravital imaging analysis indicated that RD2-deficent antigen-engaged B cells migrate normally to the inter-follicular zone of lymph nodes and interacted normally with cognate T helper cells. To further understand the nature of the functional defect of RD2 mutant B-cells, hen egg lysosome (HEL)-specific RD2-deficient GFP B cells and wild type RFP B cells (with the ratio 1:1) were transferred together with non-fluorescent ovalbumin (OVA)-specific T cells into SMARTA hosts, which were then immunized at the footpad with HEL-OVA two days later. On day 5 after immunization, draining popliteal lymph nodes were harvested and subjected for immunofluorescence histology analysis. At this time point, wild-type RFP B cells have started to cluster into tiny GC, whereas RD2-deficient GFP B cells did not form GCs. Moreover, wild-type B cells in the follicular interior were predominantly Bcl6hi, a characteristic of pre-GC B cells, suggesting that they could serve as a source of GC B cells. By contrast, RD2-deficient GFP B cells were primarily extra-follicular, and infrequently observed in the follicle interior. Most importantly, these cells were typically Bcl6lo, demonstrating that RD2 repression function is essential for pre-GC B cell differentiation. BCL6 knockout mice display a lethal inflammatory phenotype due to aberrant T-cell and macrophage activation. In striking contrast, RD2-deficient mice experienced normal healthy lives with no inflammation, and had nearly normal inflammation cytokine production in B cells and macrophages as well as differentiation of Th1,Th2 and Th17 subtypes. Hence the RD2 repression domain is specifically involved in humoral immunity but has minimal participation in the anti-inflammatory functions of BCL6. Instead we observed that the BCL6 zing finger domain plays the key role in anti-inflammatory functions in macrophages, and through ChIP-competition assays show that this is mediated by directly competing with STATs for binding to chemokine genes. These results highlight an essential role of RD2-mediated transcriptional repression in pre-GC B cell development specifically at the early B-cell activation phase. This is different than mice with BCL6 BTB mutations where early activation is normal and the defect occurs later on in the proliferative phase of GCs. The data suggest a surprising development and cellular context-specific biochemical functions of Bcl6 governing each distinct phase of the humoral immune response and inflammation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2003 ◽  
Vol 101 (5) ◽  
pp. 1934-1940 ◽  
Author(s):  
Søren Warming ◽  
Pentao Liu ◽  
Takeshi Suzuki ◽  
Keiko Akagi ◽  
Susan Lindtner ◽  
...  

Retroviral insertional mutagenesis in inbred mouse strains provides a powerful method for cancer gene discovery. Here, we show that a common retroviral integration site (RIS) in AKXD B-cell lymphomas, termed Evi3, encodes a novel zinc finger protein with 30 Krüppel-like zinc finger repeats. Most integrations atEvi3 are located upstream of the first translated exon and result in 3′ long-terminal repeat (LTR)–driven overexpression ofEvi3. Evi3 is highly related to the early B-cell factor–associated zinc finger gene (Ebfaz), and all 30 zinc fingers found in EVI3 are conserved in EBFAZ. EBFAZ binds to and negatively regulates early B-cell factor (EBF) (also known as olfactory-1, OLF1), a basic helix-loop-helix (bHLH) transcription factor required for B-lineage commitment and the development of the olfactory epithelium. EBFAZ also binds to SMA- and MAD-related protein–1 (SMAD1) and SMAD4 in response to bone morphogenetic protein–2 (BMP2) signaling, which in turn activates the homeobox regulator of Xenopus mesoderm and neural development Xvent-2. Surprisingly, while Ebfazand Evi3 are coexpressed in many tissues, and both proteins are nuclear, we could not detect Ebfaz expression in B cells by reverse transcriptase–polymerase chain reaction (RT-PCR), whereas Evi3 expression could be detected at all stages of B-cell development. Our results suggest that EVI3, like EBFAZ, is a multifunctional protein that participates in many signaling pathways via its multiple zinc fingers. Furthermore, our results suggest that EVI3, not EBFAZ, is the member of this protein family that interacts with and regulates EBF in B cells.


1991 ◽  
Vol 11 (11) ◽  
pp. 5551-5561
Author(s):  
P Rothman ◽  
S C Li ◽  
B Gorham ◽  
L Glimcher ◽  
F Alt ◽  
...  

Treatment of splenic B lymphocytes and certain B-lineage cell lines with the mitogen lipopolysaccharide (LPS) and the lymphokine interleukin-4 (IL-4) induces expression of germ line immunoglobulin C epsilon transcripts and class switching to the C epsilon gene. We show that LPS-plus-IL-4 induction of germ line epsilon transcripts (termed I epsilon transcripts) occurs at the transcriptional level in an Abelson murine leukemia virus-transformed pre-B-cell line. A 1.1-kb region of DNA surrounding the I epsilon promoter endows inducible transcription to a heterologous reporter gene stably transfected into these cells; such inducible expression depends on combined treatment with LPS and IL-4. Analyses of constructs transiently introduced into a B-cell lymphoma line demonstrated that LPS-plus-IL-4-inducible expression can be conferred by a 179-bp segment of DNA spanning the I epsilon transcriptional initiation site. Mutational analyses demonstrated that this expression depended on DNA sequences within a conserved region directly upstream from the I epsilon transcriptional initiation region. One nuclear protein that is constitutively expressed in normal B cells binds to the downstream end of the conserved sequence; its binding specificity correlates with the functional effect of several mutations. Two additional proteins, which are induced by IL-4 treatment of splenic B cells, bind to the transcription initiation sites of I epsilon. These proteins are indistinguishable in binding assays from proteins previously shown to bind an enhancer region of the class II major histocompatibility complex gene A alpha.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 212-212 ◽  
Author(s):  
Ryan T. Phan ◽  
Huifeng Niu ◽  
Masumichi Saito ◽  
Katia Basso ◽  
Giorgio Cattoretti ◽  
...  

Abstract The proto-oncogene BCL6 encodes a BTB/POZ-zinc finger transcriptional repressor that is necessary for germinal center (GC) formation and is implicated in the pathogenesis of B-cell lymphoma. In ~50% diffuse large cell lymphoma and 10% follicular lymphoma, BCL6 gene expression is deregulated by chromosomal translocations or mutations that affect its 5′ regulatory region. The precise function of BCL6 in GC development and lymphomagenesis is unclear since very few BCL6 direct target genes have been identified. We report that BCL6 suppresses p53-dependent and p53-indepenent growth arrest and apoptosis responses in GC B cells. BCL6 directly suppresses the transcription of the p53 gene, as demonstrated by (1) chromatin immunoprecipitation (ChIP) assays showing that BCL6 binds the p53 promoter region in vivo; and (2) transient transfection/reporter assays identifying within the p53 promoter region two BCL6-binding sites that mediate BCL6-mediated suppression of p53 transcription. Accordingly, suppression of BCL6 expression via specific siRNA leads to increased expression of p53 both under basal condition and in response to DNA damage. Consistent with a physiological role for BCL6-mediated p53 suppression, immunohistochemical analysis shows that p53 expression is absent in GC B cells where BCL6 is highly expressed. In addition, our data reveal that BCL6 inhibits the p53-independent activation of the p21/WAF1 cell cycle arrest gene by binding to Miz-1, a transcription factor involved in p21 activation. Consistent with a role of BCL6 in inhibiting p53-related cell cycle arrest and apoptotic responses, constitutive expression of BCL6 suppresses p53 expression and p53-target genes (P21 and PUMA) and protects B cell lines from apoptosis induced by DNA damage. These results indicate that one function of BCL6 is to allow GC B cells (centroblasts) to constitutively proliferate and to sustain the physiologic DNA breaks required for immunoglobulin switch recombination and somatic hypermutation without inducing p53-related responses. These findings also imply that B cell lymphoma with deregulated BCL6 expression are functionally p53-negative and impaired in apoptotic responses.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3129-3129
Author(s):  
Ryan T Phan ◽  
DanVy Nguyen ◽  
Phillipp Nham ◽  
Riccardo Dalla-Favera

Abstract Abstract 3129 The proto-oncogene BCL6 encodes for a BTB/POZ-zinc finger transcriptional repressor which plays an essential role in germinal center (GC) formation and is implicated in the pathogenesis of B-cell lymphoma. BCL6 is shown to be a critical regulator modulating many important functions in B cells, including activation, differentiation, cell cycle arrest and apoptosis. Tight regulation of BCL6 is, therefore, critical for the control of GC reaction as well as for the phenotype of GC-derived lymphomas. In response to B cell receptor activation or DNA damage induction, BCL6 protein is phosphorylated and subsequently degraded through distinct mechanisms that involve ubiquitination process. Here we report that BCL6 interacts with Cullin-3 (Cul3), major component of a multimeric E3 ubiquitin ligase complex. Consistent with previous reports indicating that BTB/POZ domain proteins interact with Cul3, the BCL6-POZ domain is sufficient for BCL6 interaction with Cul3. Importantly, BCL6 and Cul3 interaction is also readily detected in native B cells. Interestingly, this interaction does not affect BCL6 transcriptional repression activity nor does it affect basal level of BCL6 stability. The pathophysiological consequence of this interaction is investigated, including a possibility that BCL6 functions as a specific substrate adaptor for the Cullin-3 ubiquitin complex. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2279-2279
Author(s):  
Hakim Bouamar ◽  
Long Wang ◽  
Manoela Ortega ◽  
An-Ping Lin ◽  
Daifeng Jiang ◽  
...  

Abstract The germinal center (GC) reaction includes two critical events: somatic hypermutation (SHM) and class switch DNA recombination (CSR). These processes are tightly controlled, thus preventing excessive DNA injury which could lead to loss of normal B lymphocytes as well as the survival of cells with oncogenic DNA lesions. MicroRNA-155 (miR-155) plays an important role in immune cell biology; miR-155 knock-out (KO) mice have a defective mature B cell development characterized by a decreased number of GC B cells, whereas the Eμ-miR-155 transgenic mouse model develops and oligoclonal proliferation which evolves to B cell lymphoma. These observations suggest that miR-155 may regulate B cell sensitivity and response to DNA damage, which could mechanistically explain the phenotypes observed in the gain and loss-of-function animal models. To examine this concept, we purified mature B cells from multiple pairs of miR-155 KO and WT littermates, and stimulated them with LPS and IL4 thus recapitulating the GC reaction in vitro. Next, we used immunohistochemistry to quantify γH2AX at the DNA double-strand breaks (DSBs) foci, western blot to evaluate the phospho-p53 (Ser15) levels, and real-time RT-PCR to quantify the expression of p53 target genes (p21, GADD45a, cdc25c, PCNA). In these assays, we found a significantly higher γH2AX staining in B cells null for miR-155 than in WT controls (number of foci per nucleus: 4.1±0.9 vs. 1.4±0.2, p<0.01), and a markedly elevated p53 activity, defined by its phospho-level and target genes expression. To confirm these observations in a more physiologic setting, we immunized miR-155 WT and KO mice with NP-CGG, collected spleens and purified mature B-cells. Quantification of subpopulation by FACS confirmed that miR-155 mice developed fewer GC B cells, and the examination of DSBs foci and p53 target genes expression supported our in vitro observation of a heightened sensitivity to DNA damage and p53 engagement in miR-155 null cells. We concluded that miR-155 deficiency in B cells associates with excessive DNA damage and p53 activity. To further isolate define the role of miR-155 in these events, we used a retrovirus system to rescue miR-155 expression in B cells derived from the KO mouse, and defined the pattern of DNA damage response. In these assays, cells transduced with a MSCV-miR-155 construct formed fewer DSBs foci than their control counterparts transduced with an empty vector (9.0±2.1 vs. 4.9±1.8, p<0.05) and showed significantly lower p53 activity, defined by target gene expression. To test if miR-155 controls the DNA damage induced by broad genotoxic stresses, we exposed miR-155 KO or WT B cells, thymocytes and bone marrow cells to 5Gy of ionizing radiation (IR) or etoposide (4µM). In these assays, albeit a robust induction of γH2AX foci formation and p53 activation were detected, there was no significant difference between WT and miR-155 KO mice, in any of the cell types analyzed. We concluded that the heightened sensitivity to genotoxic stress in miR-155 KO mice is specific to B cells undergoing the GC reaction. This observation suggests a potential role in this process for activation-induced cytidine deaminase (AID), a key regulator of the DNA damage inducing SHM and CSR, which is also a miR-155 target gene. We confirmed that AID expression is higher in miR-155 KO than in WT B cells, and to test its contribution to the excessive DSB and p53 activity found miR-155 null cells we used an RNAi approach. We found that the inhibition of AID levels in miR-155 KO B cells significantly reduced γH2AX foci formation and expression of p53 target genes. Together, our data highlight a hitherto unappreciated interaction between miR-155 and the p53 pathway, involving DNA lesions that are germane to the GC reaction and the control of AID expression/activity. The excessive engagement of p53 is this setting may explain, at least in part, the loss of normal GC B cells found in the miR-155 KO mice. Conversely, it is possible that cells expressing abnormally high miR-155 levels show a subpar activation of the DNA damage response thus leading to the accumulation of oncogenic mutations. This paradigm would provide a mechanistic explanation for the initial poly/oligoclonal proliferation reported in the Eμ-miR-155 mice, which eventually evolves into a B-cell lymphoma Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document