The Irf4 Gene, a Susceptibility Locus for Chronic Lymphocytic Leukemia (CLL), Controls Establishment of Follicular and Marginal Zone B Cell Compartments in Mice

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 285-285
Author(s):  
Giorgia Simonetti ◽  
Amanda Carette ◽  
Haowei Wang ◽  
Mark Shlomchik ◽  
Ulf Klein

Abstract Abstract 285 Chronic lymphocytic leukemia (CLL) originates from the malignant transformation of mature B cells. Recently, single nucleotide polymorphisms (SNPs) in the 3'UTR region of the Irf4 gene have been associated with an increased risk of developing CLL in independent patient cohorts. IRF4 is a member of the interferon regulatory factor (IRF) family of transcription factors, and in the B lineage is essential for plasma cell differentiation in T-dependent immune responses. Irf4 has been demonstrated to act as an oncogene in multiple myeloma. Conversely, evidence suggests that the corresponding SNPs in the Irf4 gene lead to a reduction of IRF4 mRNA expression, and it was reported that tumor cells in half of CLL cases show reduced IRF4 protein expression compared to normal B cells. Together, these observations suggest that aberrant downregulation of IRF4 expression in mature B cells may contribute to CLL development. However, the normal function of IRF4 in mature B cells is incompletely understood. In order to investigate how IRF4 deficiency affects the biology of mature B cells, we investigated the consequences of deleting Irf4 specifically in B cells in vivo using a conditional Irf4 knockout mouse line. We and others had previously observed that Irf4−/− mice develop an expansion of B cells with a marginal zone (MZ) phenotype (IgMhiIgDloCD23–CD21+). By demarcating the MZ with the MOMA-1 marker, we here show that B cells in Irf4−/− mice localized preferentially in the MZ area, causing MZ hyperplasia. In contrast, the area where follicular B cells normally home contained few B cells. B cell autonomy of the observed phenotype was ascertained by crossing a “floxed” Irf4 allele with CD19-Cre mice to achieve B cell conditional deletion. We then crossed the floxed Irf4 allele with a transgenic mouse that allows inducible deletion of Irf4 specifically in B cells. Whereas flow-cytometric analysis revealed an unchanged ratio between cells with a follicular (IgMloIgDhiCD23+CD21int) vs. a MZ B cell surface phenotype upon Irf4 deletion, immunohistochemical (IHC) stainings of spleen sections for a marker protein (eGFP) that signals gene deletion demonstrated that the Irf4-deleted cells localized preferentially in the MZ, leading to MZ hyperplasia. Together, these results suggest that deletion of Irf4 in B cells alters chemokine responsiveness and migratory capacity. In agreement, global gene expression profile analysis of B cells purified from Irf4−/− and Irf4+/+ mice identified a set of differentially expressed genes with known functions in cell migration and homing. Notably, PLXND1 and the chemokine receptor CXCR7 showed 4.5 and 6.5-fold upregulation, respectively, while the G protein coupled receptor RGS13 and the adhesion molecule ALCAM (CD166) showed 3.5 and 5-fold downregulation in Irf4−/− vs. Irf4+/+ B cells. Unexpectedly, we observed in the profiling analysis that expression of the known NOTCH target genes Deltex1 and Hes5 was significantly upregulated (3 and 5-fold) in Irf4−/− vs. Irf4+/+ B cells. Despite unchanged NOTCH1 and NOTCH2 mRNA levels, Western blot and immunofluorescence analysis showed that Notch2, a gene known to be indispensible for MZ B cell development, was strongly upregulated in Irf4−/− B cells, suggesting that IRF4 is indirectly involved in NOTCH2 repression at a post-transcriptional level. Together with the altered migratory properties of Irf4−/− vs. Irf4+/+ B cells, these findings indicate that a balanced expression of IRF4 and NOTCH2 in B cells is required for establishing the follicular and MZ B cell compartments in mice, and suggest that IRF4 maintains the cellular identity of follicular B cells. The results may have implications for understanding CLL pathogenesis, as both NOTCH1 and NOTCH2 transmembrane receptors were reported to be expressed and activated in CLL B cells, and since NOTCH1 was recently found to be aberrantly activated in a fraction of CLL cases due to genetic mutations. Alterations in the balance of the transcriptional network established by NOTCH and IRF4 either through mutations, polymorphisms, or microenvironmental factors may disrupt normal B cell physiology and thereby contribute to tumorigenesis by an as yet unknown mechanism. Recently, a small fraction of CLL patients were identified that have a recurrent heterozygous somatic mutation in exon 2 of Irf4, providing additional rationale for determining how alterations in IRF4 function may promote CLL pathogenesis. Disclosures: No relevant conflicts of interest to declare.

Author(s):  
Sarah Wilmore ◽  
Karly-Rai Rogers-Broadway ◽  
Joe Taylor ◽  
Elizabeth Lemm ◽  
Rachel Fell ◽  
...  

AbstractSignaling via the B-cell receptor (BCR) is a key driver and therapeutic target in chronic lymphocytic leukemia (CLL). BCR stimulation of CLL cells induces expression of eIF4A, an initiation factor important for translation of multiple oncoproteins, and reduces expression of PDCD4, a natural inhibitor of eIF4A, suggesting that eIF4A may be a critical nexus controlling protein expression downstream of the BCR in these cells. We, therefore, investigated the effect of eIF4A inhibitors (eIF4Ai) on BCR-induced responses. We demonstrated that eIF4Ai (silvestrol and rocaglamide A) reduced anti-IgM-induced global mRNA translation in CLL cells and also inhibited accumulation of MYC and MCL1, key drivers of proliferation and survival, respectively, without effects on upstream signaling responses (ERK1/2 and AKT phosphorylation). Analysis of normal naïve and non-switched memory B cells, likely counterparts of the two main subsets of CLL, demonstrated that basal RNA translation was higher in memory B cells, but was similarly increased and susceptible to eIF4Ai-mediated inhibition in both. We probed the fate of MYC mRNA in eIF4Ai-treated CLL cells and found that eIF4Ai caused a profound accumulation of MYC mRNA in anti-IgM treated cells. This was mediated by MYC mRNA stabilization and was not observed for MCL1 mRNA. Following drug wash-out, MYC mRNA levels declined but without substantial MYC protein accumulation, indicating that stabilized MYC mRNA remained blocked from translation. In conclusion, BCR-induced regulation of eIF4A may be a critical signal-dependent nexus for therapeutic attack in CLL and other B-cell malignancies, especially those dependent on MYC and/or MCL1.


Blood ◽  
2012 ◽  
Vol 119 (9) ◽  
pp. 2110-2113 ◽  
Author(s):  
Marek Mraz ◽  
Dasa Dolezalova ◽  
Karla Plevova ◽  
Katerina Stano Kozubik ◽  
Veronika Mayerova ◽  
...  

AbstractMicroRNAs (miRNAs) play a key role in chronic lymphocytic leukemia as well as in normal B cells. Notably, miRNA gene encoding miR-650 and its homologs overlap with several variable (V) subgenes coding for lambda immunoglobulin (IgLλ). Recent studies describe the role of miR-650 in solid tumors, but its role in chronic lymphocytic leukemia (CLL) has not yet been studied. Our experiments demonstrate that miR-650 expression is regulated by coupled expression with its host gene for IgLλ. This coupling provides a unique yet unobserved mechanism for microRNA gene regulation. We determine that higher expression of miR-650 is associated with a favorable CLL prognosis and influences the proliferation capacity of B cells. We also establish that in B cells, miR-650 targets proteins important in cell proliferation and survival: cyclin dependent kinase 1 (CDK1), inhibitor of growth 4 (ING4), and early B-cell factor 3 (EBF3). This study underscores the importance of miR-650 in CLL biology and normal B-cell physiology.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Markus Lechner ◽  
Thomas Engleitner ◽  
Tea Babushku ◽  
Marc Schmidt-Supprian ◽  
Roland Rad ◽  
...  

AbstractFollicular B (FoB) and marginal zone B (MZB) cells are functionally and spatially distinct mature B cell populations in the spleen, originating from a Notch2-dependent fate decision after splenic influx of immature transitional B cells. In the B cell follicle, a Notch2-signal is provided by DLL-1-expressing fibroblasts. However, it is unclear whether FoB cells, which are in close contact with these DLL-1 expressing fibroblasts, can also differentiate to MZB cells if they receive a Notch2-signal. Here, we show induced Notch2IC-expression in FoB cells re-programs mature FoB cells into bona fide MZB cells as is evident from the surface phenotype, localization, immunological function and transcriptome of these cells. Furthermore, the lineage conversion from FoB to MZB cells occurs in immunocompetent wildtype mice. These findings demonstrate plasticity between mature FoB and MZB cells that can be driven by a singular signaling event, the activation of Notch2.


2005 ◽  
Vol 46 (9) ◽  
pp. 1369-1374 ◽  
Author(s):  
Lucile Baseggio ◽  
Sophie Gazzo ◽  
Evelyne Callet-Bauchu ◽  
Alexandra Traverse-Glehen ◽  
Catherine Thieblemont ◽  
...  

2017 ◽  
Vol 39 (2) ◽  
pp. 141-144
Author(s):  
S V Andreieva ◽  
K V Korets ◽  
O E Ruzhinska ◽  
I M Skorokhod ◽  
O G Alkhimova

Aim: The genetic mechanisms of resistance to chemotherapy in B-cell chronic lymphocytic leukemia/small lymphocytic lymphoma (B-CLL/SLL) are not clear. We aimed to determine the peculiarities of abnormal karyotype formation in bone marrow (BM) cells and peripheral blood (PB) blast transformed B-cells in relapse of B-CLL/SLL. Materials and Methods: Cytogenetic GTG banding technique and molecular cytogenetic in interphase cells (i-FISH) studies of BM cells and PB blast transformed B-lymphocytes were performed in 14 patients (10 males and 4 females) with B-CLL/SLL. Results: The results of karyotyping BM and PB cells revealed the heterogeneity of cytogenetic abnormalities in combined single nosological group of B-CLL/SLL. In PB B-cells, chromosome abnormalities related to a poor prognosis group were registered 2.5 times more often than in BM cells. Additional near tetraploid clones that occurred in 57.1% cases were the peculiar feature of BM cell karyotypes. Chromosomal rearrangements characteristic of the group of adverse cytogenetic prognosis were revealed in all cases from which in 2 cases by karyotyping BM cells, in 6 cases in PB B-cells and in 8 cases by the i-FISH method in BM cells, i.e. their detection frequency was 3 times higher in PB B-cells and 4 times higher when analyzing by i-FISH in BM cells. Conclusions: Mismatch in abnormal karyotypes in BM and PB B-cells by the presence of quantitative and structural chromosomal rearrangements may be indicative of simultaneous and independent processes of abnormal clone formation in the lymph nodes and BM hematopoietic cells. Accumulation the information about previously unidentified chromosomal rearrangements in relapse of the disease may help to understand the ways of resistance formation to chemotherapy.


Blood ◽  
1988 ◽  
Vol 71 (4) ◽  
pp. 1012-1020 ◽  
Author(s):  
JS Moore ◽  
MB Prystowsky ◽  
RG Hoover ◽  
EC Besa ◽  
PC Nowell

The consistent occurrence of T cell abnormalities in patients with B cell chronic lymphocytic leukemia (B-CLL) suggest that the non- neoplastic host T cells may be involved in the pathogenesis of this B cell neoplasm. Because potential defects of immunoglobulin regulation are evident in B-CLL patients, we investigated one aspect of this by studying the T cell-mediated immunoglobulin isotype-specific immunoregulatory circuit in B-CLL. The existence of class-specific immunoglobulin regulatory mechanisms mediated by Fc receptor-bearing T cells (FcR + T) through soluble immunoglobulin binding factors (IgBFs) has been well established in many experimental systems. IgBFs can both suppress and enhance B cell activity in an isotype-specific manner. We investigated the apparently abnormal IgA regulation in a B-CLL patient (CLL249) whose B cells secrete primarily IgA in vitro. Enumeration of FcR + T cells showed a disproportionate increase in IgA FcR + T cells in the peripheral blood of this patient. Our studies showed that the neoplastic B cells were not intrinsically unresponsive to the suppressing component of IgABF produced from normal T cells, but rather the IgABF produced by the CLL249 host T cells was defective. CLL249 IgABF was unable to suppress IgA secretion by host or normal B cells and enhanced the in vitro proliferation of the host B cells. Size fractionation of both normal and CLL249 IgABF by gel-filtration high- performance liquid chromatography (HPLC) demonstrated differences in the ultraviolet-absorbing components of IgABF obtained from normal T cells v that from our patient with defective IgA regulation. Such T cell dysfunction may not be restricted to IgA regulation, since we have found similar expansion of isotype-specific FcR + T cells associated with expansion of the corresponding B cell clone in other patients with B-CLL. These data suggest that this T cell-mediated regulatory circuit could be significantly involved in the pathogenesis of B-CLL.


Blood ◽  
2005 ◽  
Vol 106 (7) ◽  
pp. 2391-2398 ◽  
Author(s):  
Elena Vigorito ◽  
Laure Gambardella ◽  
Francesco Colucci ◽  
Simon McAdam ◽  
Martin Turner

AbstractMice lacking all 3 Vav proteins fail to produce significant numbers of recirculating follicular or marginal zone B cells. Those B cells that do mature have shortened lifespans. The constitutive nuclear factor-kappaB (NF-κB) activity of resting naive B cells required Vav function and expression of cellular reticuloendotheliosis (c-Rel). Rel-A was reduced in Vav-deficient B cells. Furthermore, expression of the NF-κB-regulated antiapoptotic genes A1 and Bcl-2 was reduced in mature Vav-deficient B cells. Overexpression of Bcl-2 restored the number of mature follicular B cells in the spleens of Vav-deficient mice. When activated by B-cell receptor (BCR) cross-linking, Vav-deficient B cells failed to activate NF-κB. Vav proteins thus regulate an NF-κB-dependent survival signal in naive B cells and are required for NF-κB function after BCR cross-linking.


Blood ◽  
1997 ◽  
Vol 89 (8) ◽  
pp. 2833-2841 ◽  
Author(s):  
Lyda M. Osorio ◽  
Angelina De Santiago ◽  
Miguel Aguilar-SantelisesHå ◽  
kan Mellstedt ◽  
Mikael Jondal

Abstract CD6 and CD5 belong to a scavenger-receptor cysteine-rich (SRCR) super family of membrane glycoproteins that are expressed on chronic lymphocytic leukemia B (B-CLL) cells, normal T cells, and a small subset of normal B cells. CD6 configures in the membrane in relation to the cellular activation level and can act as a coreceptor for T-cell activation. We have examined a group of progressive and nonprogressive B-CLL cells. Most B-CLL cells were positive for CD6 and the expression of CD6 was increased after activation with Staphylococcus aureus Cowan I plus interleukin-2 or 12-O-tetradecanoylphorbol 13-acetate, although anti-CD6 antibodies did not increase proliferative responses to these stimuli. However, anti-CD6 stimulation was found to protect against anti-IgM–induced apoptosis in B-CLL. baxα upregulation and bcl-2 downregulation were found in anti-IgM– and glucocorticoid (GCC)-induced apoptotic cells, respectively. Furthermore, CD6 cross-linking downregulated baxα mRNA levels in anti-IgM–treated cells, resulting in an increased bcl-2/baxα ratio. CD6 activation also prevented bcl-2 mRNA downregulation and apoptosis induced by GCC in one of six GCC-sensitive patients. These data suggest that an interaction between CD6 and its ligand might contribute to B-CLL survival through the modulation of the Bcl-2/Bax ratio.


Blood ◽  
2002 ◽  
Vol 100 (8) ◽  
pp. 2973-2979 ◽  
Author(s):  
Anne J. Novak ◽  
Richard J. Bram ◽  
Neil E. Kay ◽  
Diane F. Jelinek

B-cell chronic lymphocytic leukemia (B-CLL) is defined by the accumulation of CD5+ B cells in the periphery and bone marrow. This disease is not characterized by highly proliferative cells but rather by the presence of leukemic cells with significant resistance to apoptosis and, therefore, prolonged survival. B-lymphocyte stimulator (BLyS) is a newly identified tumor necrosis factor (TNF) family member shown to be critical for maintenance of normal B-cell development and homeostasis and it shares significant homology with another TNF superfamily member, APRIL. The striking effects of BLyS on normal B-cell maintenance and survival raises the possibility that it may be involved in pathogenesis and maintenance of hematologic malignancies, including B-CLL. In this study, we investigated the status of APRIL and BLyS expression, as well as their receptors, in this disease. All B-CLL patient cells studied expressed one or more of 3 known receptors for BLyS; however, the pattern of expression was variable. In addition, we demonstrate for the first time that B-CLL cells from a subset of patients aberrantly express BLyS and APRIL mRNA, whereas these molecules were not detectable in normal B cells. Furthermore, we provide in vitro evidence that BLyS protects B-CLL cells from apoptosis and enhances cell survival. Because these molecules are key regulators of B-cell homeostasis and tumor progression, leukemic cell autocrine expression of BLyS and APRIL may be playing an important role in the pathogenesis of this disease.


Sign in / Sign up

Export Citation Format

Share Document