scholarly journals Patterns of microRNA expression characterize stages of human B-cell differentiation

Blood ◽  
2009 ◽  
Vol 113 (19) ◽  
pp. 4586-4594 ◽  
Author(s):  
Jenny Zhang ◽  
Dereje D. Jima ◽  
Cassandra Jacobs ◽  
Randy Fischer ◽  
Eva Gottwein ◽  
...  

Abstract Mature B-cell differentiation provides an important mechanism for the acquisition of adaptive immunity. Malignancies derived from mature B cells constitute the majority of leukemias and lymphomas. These malignancies often maintain the characteristics of the normal B cells that they are derived from, a feature that is frequently used in their diagnosis. The role of microRNAs in mature B cells is largely unknown. Through concomitant microRNA and mRNA profiling, we demonstrate a potential regulatory role for microRNAs at every stage of the mature B-cell differentiation process. In addition, we have experimentally identified a direct role for the microRNA regulation of key transcription factors in B-cell differentiation: LMO2 and PRDM1 (Blimp1). We also profiled the microRNA of B-cell tumors derived from diffuse large B-cell lymphoma, Burkitt lymphoma, and chronic lymphocytic leukemia. We found that, in contrast to many other malignancies, common B-cell malignancies do not down-regulate microRNA expression. Although these tumors could be distinguished from each other with use of microRNA expression, each tumor type maintained the expression of the lineage-specific microRNAs. Expression of these lineage-specific microRNAs could correctly predict the lineage of B-cell malignancies in more than 95% of the cases. Thus, our data demonstrate that microRNAs may be important in maintaining the mature B-cell phenotype in normal and malignant B cells.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 446-446 ◽  
Author(s):  
Jonathan Mandelbaum ◽  
Govind Bhagat ◽  
Tongwei Mo ◽  
Alexander Tarakhovsky ◽  
Laura Pasqualucci ◽  
...  

Abstract Abstract 446 The PRDM1/ BLIMP1 gene encodes a zinc finger transcriptional repressor that is expressed in a subset of germinal center (GC) B cells and in all plasma cells, and is required for terminal B cell differentiation. The BLIMP1 locus is biallelically inactivated by structural alterations in approximately one third of activated B cell-like diffuse large B cell lymphoma (ABC-DLBCL) (Pasqualucci et al, J Exp Med 2006). Moreover, the expression of the Blimp1 protein is absent in up to 80% of ABC-DLBCL due to alternative genetic and epigenetic mechanisms. These findings suggest that BLIMP1 may function as a tumor suppressor gene whose loss may contribute to the pathogenesis of this lymphoma type by blocking terminal B cell differentiation. To investigate the role of BLIMP1 inactivation in lymphomagenesis in vivo, we tested whether conditional deletion of the Blimp1 gene in mouse B cells can promote the growth of lymphomas recapitulating the features of ABC-DLBCL. Toward this end, a mouse model carrying a loxP-flanked exon 5 of the Blimp1 gene that can be deleted by Cre-mediated recombination (Ohinata et al, Nature 2005) was crossed with a CD19-Cre deletor strain, expressing the Cre recombinase in all B cells. The resulting mice were monitored for tumor development and survival. Consistent with previous observations in a similar model (Shapiro-Shelef et al, Immunity 2003), Blimp1 conditional knockout (Blimp1CD19KO) mice showed a severe impairment in the generation of CD138+ plasma cells and had decreased serum immunoglobulin levels of all isotypes, together with a two-fold increase in the number of PNAhiCD95+ GC B cells. Over time, significantly reduced survival was observed in the Blimp1CD19KO cohort, with only 27% of the animals being alive at 15 months of age (LogRank p value<0.0001). Macroscopic and flow cytometric analysis of the lymphoid compartments revealed the presence of splenomegaly in 32/38 (84%) Blimp1CD19KO, as compared to 1/25 (4%) age-matched wildtype (WT) littermates, and a significant increase in IgM+IgD-CD21+CD23lo splenic B cells, indicative of marginal zone B cell expansion. In addition, 79% (n=30/38) of Blimp1CD19KO mice showed markedly hyperplastic bronchus-associated lymphoid tissue (BALT). Notably, between 10 and 16 months of age 34% (13/38) of these animals developed clonal lymphoproliferative disorders with a mature B cell phenotype (B220+Pax5+) and histologic features of DLBCL (n=6) or less aggressive lymphoid proliferations (LPD: n=6; marginal zone lymphoma: n=1), in contrast with 1/27 heterozygous and 0/25 WT animals. Sequencing analysis of the rearranged immunoglobulin variable region genes in lymphoma biopsies revealed the presence of somatic mutations in 6/8 samples investigated, demonstrating their origin from a GC-experienced B cell. Moreover, immunohistochemical staining for Bcl6 and Irf4 documented a late-GC “activated” B cell phenotype (Bcl6-Irf4+) in all tumors tested (n=4), consistent with the expansion of cells that had been committed to plasma cell differentiation. These data demonstrate that Blimp1 is a bona-fide tumor suppressor gene whose B-cell specific inactivation in vivo promotes the development of lymphomas sharing features of the human ABC-DLBCL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1995 ◽  
Vol 85 (7) ◽  
pp. 1836-1842 ◽  
Author(s):  
MY Mapara ◽  
K Bommert ◽  
RC Bargou ◽  
C Leng ◽  
C Beck ◽  
...  

Recently G alpha 16, a new guanosine triphosphate (GTP) binding protein alpha subunit has been described to be specifically expressed in human hematopoietic cells. Expression of G alpha 16 was observed in human cell lines of myelomonocytic and T-lymphocytic origin, but not in human B-cell lines Raji and IM9. We studied the expression of G alpha 16 in human B cells corresponding to different stages of B-cell differentiation by means of reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting. The human Burkitt's lymphoma cell lines Raji, Ramos, BJAB, the lymphoblastoid cell line SKW6.4, and the plasmocytoma cell line U266 were devoid of G alpha 16. In contrast, G alpha 16 was detected in the human progenitor B cell lines Reh and Nalm-6. Using the mu+, k-cell line BLIN-1 (pre-B cell phenotype) and its derived subclone 1E8 (surface mu+, k+; B-cell phenotype) G alpha 16 expression was found to disappear on transition from pre-B to B-cell differentiation stage. The analysis of a broad panel of human neoplastic B lymphocytes ranging from progenitor B-acute lymphatic leukemia (pre-pre-B-ALL), common acute leukemias (cALL), pre-B-ALL, mature B-ALL to low grade B-cell lymphoma (chronic lymphocytic leukemia of B-cell type, leukemic centrocytic non-Hodgkins lymphoma [NHL], hairy cell leukemia) showed that G alpha 16 expression is limited to progenitor and pre-B-ALL cells. Therefore, we conclude that within B-cell differentiation, G alpha 16 is expressed solely during early B cell ontogeny and downregulated during differentiation. Thus, G alpha 16 might be an important regulator involved in signaling processes in progenitor B cells.


1993 ◽  
Vol 13 (7) ◽  
pp. 3929-3936
Author(s):  
T D Randall ◽  
F E Lund ◽  
J W Brewer ◽  
C Aldridge ◽  
R Wall ◽  
...  

Interleukin-5 (IL-5) and IL-6 have both been reported to act as B-cell differentiation factors by stimulating activated B cells to secrete antibody. However, it has not been possible to directly compare the effects of these two lymphokines because of the lack of a suitable B-cell line capable of responding to both. We have identified a clonal, inducible B-cell lymphoma, CH12, that has this property. Both IL-5 and IL-6 can independently stimulate increases in steady-state levels of immunoglobulin and J-chain mRNA and proteins, and they both induce the differentiation of CH12 into high-rate antibody-secreting cells. Nevertheless, there are significant differences in the activities of these two lymphokines. First, while IL-6 acts only as a differentiation factor, IL-5 also augments the proliferation of CH12 cells. Second, the differentiation stimulated by IL-5 but not by IL-6 is partially inhibited by IL-4. Inhibition of IL-5-induced differentiation was not at the level of IL-5 receptor expression, since IL-4 did not inhibit IL-5-induced proliferation. Third, IL-5 but not IL-6 stimulated increased mouse mammary tumor proviral gene expression in CH12 cells. These results demonstrate that while both IL-5 and IL-6 may act as differentiation factors for B cells, they induce differentiation by using at least partially distinct molecular pathways. Our results also establish that B cells characteristic of a single stage of development can independently respond to IL-4, IL-5, and IL-6.


1993 ◽  
Vol 13 (7) ◽  
pp. 3929-3936 ◽  
Author(s):  
T D Randall ◽  
F E Lund ◽  
J W Brewer ◽  
C Aldridge ◽  
R Wall ◽  
...  

Interleukin-5 (IL-5) and IL-6 have both been reported to act as B-cell differentiation factors by stimulating activated B cells to secrete antibody. However, it has not been possible to directly compare the effects of these two lymphokines because of the lack of a suitable B-cell line capable of responding to both. We have identified a clonal, inducible B-cell lymphoma, CH12, that has this property. Both IL-5 and IL-6 can independently stimulate increases in steady-state levels of immunoglobulin and J-chain mRNA and proteins, and they both induce the differentiation of CH12 into high-rate antibody-secreting cells. Nevertheless, there are significant differences in the activities of these two lymphokines. First, while IL-6 acts only as a differentiation factor, IL-5 also augments the proliferation of CH12 cells. Second, the differentiation stimulated by IL-5 but not by IL-6 is partially inhibited by IL-4. Inhibition of IL-5-induced differentiation was not at the level of IL-5 receptor expression, since IL-4 did not inhibit IL-5-induced proliferation. Third, IL-5 but not IL-6 stimulated increased mouse mammary tumor proviral gene expression in CH12 cells. These results demonstrate that while both IL-5 and IL-6 may act as differentiation factors for B cells, they induce differentiation by using at least partially distinct molecular pathways. Our results also establish that B cells characteristic of a single stage of development can independently respond to IL-4, IL-5, and IL-6.


2020 ◽  
Vol 21 (6) ◽  
pp. 2206 ◽  
Author(s):  
Andrea Härzschel ◽  
Antonella Zucchetto ◽  
Valter Gattei ◽  
Tanja Nicole Hartmann

Lineage commitment and differentiation of hematopoietic cells takes place in well-defined microenvironmental surroundings. Communication with other cell types is a vital prerequisite for the normal functions of the immune system, while disturbances in this communication support the development and progression of neoplastic disease. Integrins such as the integrin very late antigen-4 (VLA-4; CD49d/CD29) control the localization of healthy as well as malignant B cells within the tissue, and thus determine the patterns of organ infiltration. Malignant B cells retain some key characteristics of their normal counterparts, with B cell receptor (BCR) signaling and integrin-mediated adhesion being essential mediators of tumor cell homing, survival and proliferation. It is thus not surprising that targeting the BCR pathway using small molecule inhibitors has proved highly effective in the treatment of B cell malignancies. Attenuation of BCR-dependent lymphoma–microenvironment interactions was, in this regard, described as a main mechanism critically contributing to the efficacy of these agents. Here, we review the contribution of VLA-4 to normal B cell differentiation on the one hand, and to the pathophysiology of B cell malignancies on the other hand. We describe its impact as a prognostic marker, its interplay with BCR signaling and its predictive role for novel BCR-targeting therapies, in chronic lymphocytic leukemia and beyond.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 705-705
Author(s):  
Jenny Zhang ◽  
Dereje D. Jima ◽  
Cassandra L. Jacobs ◽  
Eva Gottwein ◽  
Grace Huang ◽  
...  

Abstract Background: Mature B cell differentiation provides an important mechanism for the acquisition of adaptive immunity. Malignancies derived from mature B cells are common and constitute the majority of leukemias and lymphomas. MicroRNAs are known to play a role in oncogenesis, lineage-selection, and immune cell function, including early B cell differentiation. However, the full extent and function of microRNA expression during mature B cell differentiation and in B cell malignancies are not known. Methods: From normal young patients undergoing tonsillectomies, we sorted the mature B cell subsets (naive, germinal center, memory and plasma) using FACS, based on their expression of CD19, CD38, IgD and CD27. These sorted B cells were profiled for microRNA expression using a highly sensitive multiplexed real-time PCR assay, as well as for gene expression at the whole genome level using Affymetrix U133plus microarrays. miRNA targets can be predicted based on seed sequence matching of their 2–8 nt to the 3′UTR of gene transcripts. For each B cell stage, we experimentally validated microRNA regulation of predicted target genes of interest, LMO2, MYBL1 and PRDM1, by microRNA over-expression experiments and luciferase assays. Results: We found that microRNAs have a characteristic expression pattern that defines each mature B cell stage. Examination of both microRNA and mRNA expression showed that in each B cell population, the target genes predicted based on seed matching were expressed at lower levels, results that were highly significant (P<1E-10). We found that differential microRNA expression is important at every B cell stage transition, and differentially expressed microRNAs frequently target differentially expressed transcription factors. In the naive to germinal center B cell and germinal center B cell to memory cell transitions, we found that miR-223 had an inverse relationship with its predicted target genes LMO2 and MYBL1. To test this relationship predicted based on seed pairing, in Germinal Center-derived BJAB cells, we over-expressed miR-223 by introducing its precursor, and saw a subsequent knockdown of LMO2 and MYBL1 at both the mRNA and protein level. We confirmed seed sequence specificity by comparing miR-223 knockdown of luciferase reporter activity on the LMO2 3′UTR compared to its seed sequence mutant. We further found that miR-9 and miR-30 family members directly regulate PRDM1 (blimp1), a master regulator of the GC to PC transition. In U266 cells (PC-derived), introduction of miR-9 and miR-30 family precursor resulted in decreased PRDM1 protein expression, although transcript levels were not changed, consistent with previous evidence that miRNA can regulate at the post-transcriptional steps. We further profiled over 50 tumors derived from various B cell malignancies (small lymphocytic lymphoma, Burkitt lymphoma, and the molecular subsets of diffuse large B cell lymphoma) and found that these malignancies maintain the expression patterns of their respective lineage; microRNA expression profiles of normal B cells could correctly classify the lineage of these tumors in over 80% of the cases. In contrast to other malignancies, common lymphomas do not down-regulate microRNAs, but rather maintain the microRNA-expression patterns of their normal B-cell counterparts. Conclusion: Through concomitant microRNA and mRNA-profiling, we demonstrate a regulatory role for microRNAs at every stage in mature B-cell differentiation. Further, we have experimentally identified a direct role for the microRNA-regulation of key transcription factors in B-cell differentiation: LMO2, MYBL1 and PRDM1 (Blimp1). Thus, our data demonstrate that microRNAs may be important in maintaining the mature B-cell phenotype in normal and malignant B-cells.


Blood ◽  
2008 ◽  
Vol 111 (1) ◽  
pp. 275-284 ◽  
Author(s):  
Qing Liu ◽  
Xiaobin Zhao ◽  
Frank Frissora ◽  
Yihui Ma ◽  
Ramasamy Santhanam ◽  
...  

FTY720 is an immunosuppressant developed to prevent organ transplant rejection. Recent studies indicate an additional role for FTY720 in inducing cell apoptosis. We demonstrate here that FTY720 mediates toxic effects in cell lines representing different B-cell malignancies and primary B cells from patients with chronic lymphocytic leukemia (CLL). In contrast to previous reports in T-cell lines, FTY720-induced toxicity in the Raji cell line and primary CLL B cells is independent of activation of caspases or poly(ADP-ribose) polymerase processing. Further, pancaspase inhibitor Z-VAD-fmk failed to rescue these cells from apoptosis mediated by FTY720. FTY720 induced down-regulation of Mcl-1 but not Bcl-2 in CLL B cells. Overexpression of Bcl-2 failed to protect transformed B cells from FTY720-induced apoptosis, suggesting a Bcl-2–independent mechanism. Interestingly, FTY720 induced protein phosphatase 2a (PP2a) activation and downstream dephosphorylation of ERK1/2, whereas okadaic acid at concentrations that inhibited the FTY720-induced PP2a activation also resulted in inhibition of FTY720-mediated apoptosis and restoration of baseline ERK1/2 phosphorylation in primary CLL cells, indicating a role for PP2a activation in FTY720-induced cytotoxicity. Further, FTY720 treatment resulted in significant prolonged survival in a xenograft severe combined immunodeficiency (SCID) mouse model of disseminated B-cell lymphoma/leukemia. These results provide the first evidence for the potential use of FTY720 as a therapeutic agent in a variety of B-cell malignancies, including CLL.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 805-805 ◽  
Author(s):  
Raquel Malumbres ◽  
Robert Tibshirani ◽  
Elena Cubedo ◽  
Kristopher A Sarosiek ◽  
Xiaoyu Jiang ◽  
...  

Abstract B-cell development and differentiation are complex processes controlled by distinct programs of transcriptional control. A large set of transcriptional factors together or in succession control this process and their deregulation may result in block of differentiation or malignant transformation. MicroRNAs are small RNAs that orchestrate cellular functions by modulating the level of their targeted proteins by either translational arrest or transcript degradation, and play a key role in cell differentiation, apoptosis, proliferation and cancer development. An increasing number of transcription factors are being found targeted by microRNAs. Emerging evidence suggests that differentiation stage-specific expression of microRNAs occurs in the hematopoietic system and during T cell differentiation. Only limited information exists on microRNA expression in normal B cell differentiation and its malignant counterparts. Herein we analyzed microRNA expression profiles in distinct peripheral B cell differentiation stages-naïve, germinal center (GC) centroblasts and memory cells as well as tonsilar T cells. Furthermore, microRNA profiling was performed in germinal center-like (GCB-like) and activated B-cell-like (ABC-like) DLBCL cell lines originating from distinct B-cell differentiation stages. RNA, extracted with mirVana kit (AMBION) from B cell subsets and T cells enriched from normal tonsils was hybridized on LC Sciences (Houston, TX) microarrays harboring 470 human microRNAs probes (Sanger miRBase Release 9.1). Expression of selected microRNAs was confirmed by ABI RT-PCR methodology. Unsupervised clustering of microRNAs with values present in at least 50% of the samples (122 probes) resulted in perfect differentiation-stage clustering of samples. Application of Statistical Analysis of Microarrays (SAM) and Prediction Analysis of Microarrays (PAM) methods (FDR= 10%) identified a 47 microRNA cell of origin classifier for B-cells differentiation stage; 27 of these microRNAs were upregulated and 20 downregulated in centroblasts compared to memory B-cells. MicroRNAs belonging to paralog microRNA clusters (e.g. miR17-92-1, miR363-106a and miR25-106b) demonstrated similar patterns of expression in specific differentiation stages. To identify specific microRNA targets, miRanda, TargetScan and PicTar programs were used. To experimentally confirm the targets, we assessed the effects of specific microRNAs on the expression levels of targeted proteins and on the luciferase reporter under the control of the wild type and mutated 3′ UTR regions of putative target genes. Using this experimental approach we identified lymphocyte-stage-specific microRNAs which expression inversely correlated and might regulate the expression of LMO2, BLIMP1 and IRF4 proteins distinctively expressed at different differentiation stages of B lymphocytes. For example, miR223, which expression is low in GC cells but is high in naïve and memory B cells, downregulates the expression of LMO2. We next analyzed microRNA expression in DLBCL cell lines. Clustering analysis, using the 47 microRNA cell of origin classifier perfectly classified GCB-like and ABC-like cell lines. Interestingly, the expression of microRNAs in both GCB-like and ABC-like DLBCL cell lines was more similar to normal centroblasts than to memory B cells, suggesting that both may originate from distinct subpopulations of GC lymphocytes. The similarity of microRNA expression in cell lines to centroblasts was striking, with only 16 microRNAs (1 upregulated and 15 downregulated in cell lines) showing noticeable differences in levels of expression compared to normal cells. These microRNAs might be involved in the process of lymphoma transformation. SAM analysis aimed to differentiate GCB-like and ABC-like cell lines identified 11 microRNAs, only 3 of which were present in the cell of origin classifier. This observation suggests that there is also a difference in expression of microRNAs not directly related to the distinct cell of origin between the DLBCL subtypes. In summary, our results demonstrate that the microRNA profile changes during the GC reaction as well as during malignant transformation. Specific microRNAs can regulate key transcription factors controlling the processes of lymphocyte differentiation and transformation.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2403-2403
Author(s):  
Cassandra L. Jacobs ◽  
Dereje D Jima ◽  
Jenny Zhang ◽  
Cherie Dunphy ◽  
Kristy L. Richards ◽  
...  

Abstract Abstract 2403 Poster Board II-380 Background MicroRNAs are 18-22 nucleotide-long RNA molecules that regulate expression of genes. We and others have previously demonstrated a role for microRNAs in the pathogenesis of B cell malignancies. Computational predictions suggest that the human genome encodes several thousand microRNAs. Thus far, about 700 microRNAs have been discovered in humans, including over 200 new microRNAs in the past year alone. The ongoing discovery of microRNAs makes it difficult to comprehensively study their role in a disease group. The advent of high throughput sequencing allows the simultaneous identification of millions of transcripts, thereby providing a sensitivity that is several orders of magnitude higher than conventional methods. We hypothesized that high throughput sequencing would be an effective tool to comprehensively identify microRNAs in normal and malignant B cells. While there is an overlap between diffuse large B-cell lymphoma (DLBCL) and Burkitt lymphoma (BL) in morphology, immunophenotype and cytogenetics, distinguishing between BL and DLBCL is critical because there are important differences in their clinical management. We investigated whether microRNA expression could be used to reliably distinguish BL from DLBCL. Methods and Results We carefully chose 31 human samples to represent the spectrum of normal and malignant B cells including FACS-sorted naive, germinal center, memory, plasma cells, EBV transformed and activated B cells. Samples derived from B cell malignancies included B-lymphoblastic lymphoma, chronic lymphocytic leukemia (immunoglobulin gene mutated and unmutated), mantle cell lymphoma, marginal zone lymphomas, HIV-related lymphoma, BL, DLBCL (activated and germinal center type), primary mediastinal B cell lymphoma, Hodgkin lymphoma, and multiple myeloma. We applied massively parallel, high-throughput sequencing of the 18-22 nt RNAs from these cases and generated a total of 255,624,785 sequences (∼5 billion bases). Using a computational approach that we have previously validated with normal B cells, we identified the expression of 429 known microRNAs in normal and malignant B cells, a number that is over three times higher than previously recognized in any tissue type. We also identified the expression of 302 novel microRNAs in normal and malignant B cells. The vast majority of these microRNAs were highly conserved in multiple species. As a proof of principle, we generated a custom microarray that included all the known human, and viral microRNAs, as well as 302 novel microRNAs identified by sequencing, and applied it to the clinically important distinction of BL from DLBCL. Biopsy samples were collected from 104 patients (BL, N=25, DLBCL, N=79) treated at 9 institutions that comprise an international consortium. All cases were reviewed for pathology diagnosis and profiled for microRNA expression. We constructed a Bayesian predictor to distinguish BL from DLBCL based on the microRNA expression. The predictor performance was tested using leave-one-out cross-validation. We also applied gene expression profiling to the cases of DLBCL to identify the molecular subsets of DLBCL: activated B cell like and germinal center B cell like DLBCL. The microRNA profiles of these cases were equally efficacious in distinguishing the DLBCL subsets. The predictor constructed based on microRNA expression was over 90% accurate in distinguishing BL from DLBCL, using pathology diagnosis as the gold standard. Further, microRNA-based predictor was also over 90% accurate in the distinction of the molecular subsets of DLBCL, compared to the gold standard of gene expression-profiling. As additional validation, we performed in situ hybridization of selected microRNAs to directly visualize their expression using methods that are easily accessible in conventional pathology laboratories. We found excellent concordance between the expression results derived from microarrays and in situ hybridization suggesting a ready path to clinical translation. Conclusion Our study represents the first comprehensive delineation of microRNA expression in B cell malignancies using high throughput sequencing. Our data suggest that microRNAs are a promising marker for the distinction of aggressive lymphomas. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1852-1852
Author(s):  
Eva Gentner ◽  
Andrea Nicola Mazzarello ◽  
Martin Becker ◽  
Antonella Nicolò ◽  
Valerio Renna ◽  
...  

Abstract B cell malignancies including chronic lymphocytic leukemia (CLL) and diffuse large B cell lymphoma (DLBCL) are age-associated diseases driven by clonal B cell proliferation. Signaling through B cell antigen receptor (BCRs) is dysregulated in these diseases. In addition to BCRs, chemokine receptors, such as CXCR4 and CXCR5, are used to predict clinical course. The chemokine receptor CXCR4 is expressed at different developmental stages of B cells, serving different homeostatic functions including migration. We previously reported that the cross-talk of CXCR4 and the BCR isotype IgD is supporting survival and activation of mature B cells in mice. In this process, the B cell marker and co-activator CD19 plays a pivotal role. Nevertheless, interaction of BCR with CXCR4 has not been analyzed in detail in B cell malignancies. In this study, we further elucidated the CXCR4 signaling in mouse as well as human B cell subsets including immature and mature B cells. Consistent with murine B cells, CXCR4 signaling in human B cells from healthy donors remains tightly linked to surface IgD-BCR expression, although CXCR4 is highly expressed in human IgG positive memory B cell compartment. Furthermore, proximity of CXCR4 and IgD in human mature B cells is reminiscent of that of mouse B cells. In contrast, IgD:CXCR4 proximity is skewed towards IgM:CXCR4 in CLL cells. In in vitro assays, unmutated (U)-CLL cells migrate better compared to mutated (M)-CLL. Nevertheless, our analyses reveal a frequent association of IgM:CXCR4 in M-CLL. Taking together our murine and human data, we propose that IgD:CXCR4 association is crucial for CXCR4 signaling in both CLL and healthy B cells. Apart from CXCR4, mutations within the immunoreceptor tyrosine-based activation motif (ITAM) residues of CD79a and CD79b are frequently associated with B cell malignancies including DLBCL. Knowing the potential role of BCR and its isotypes in CXCR4 induced signaling, we further analyzed the role of CD79a and CD79b. Here, we took advantage of transgenic mice, whose CD79a and CD79b cytoplasmic tails carrying ITAM motifs can be inducibly deleted. Our analysis reveals the indispensable role of the CD79b cytoplasmic tail, whose loss of function causes complete impairment of CXCR4 induced signaling in murine B cells. In contrast, loss of CD79a cytoplasmic tail partially blocks CXCR4 induced signaling, which could be rescued by CD19 co-stimulation. Extending our murine results, we established an in vitro read-out system to test the role of ITAM mutants derived from DLBCLs, as well as DLBCL-derived isotypes for analyzing their impact on CXCR4 signaling. Taking our findings together, IgD:CXCR4 association is crucial for CXCR4 signaling in CLL and healthy B cells. An increased association of IgM:CXCR4 in M-CLL compared to U-CLL suggests the necessity of IgD:CXCR4 for functional CXCR4 signaling. Furthermore, CD79b is crucial for CXCR4 induced signaling in mature B cells and loss of CD79b function abrogates CXCR4 signaling in mature B cells. Disclosures Chiorazzi: AR Pharma: Equity Ownership; Janssen, Inc: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document