C-Rel in the Regulation of BAFF-R Expression in Malignant Human B Cells.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2400-2400
Author(s):  
Stephen A. Mihalcik ◽  
Diane F. Jelinek

Abstract Abstract 2400 Poster Board II-377 Tumor necrosis factor superfamily member BAFF-R has been shown to transduce a powerful survival signal in B lineage cells upon ligation by BAFF, its soluble high affinity ligand. The availability of BAFF in lymphoid organs and its detectable levels in the blood suggest that it is the control of receptor expression by the B cell rather than ligand production by supportive cells that limits the activity of this survival signal. The presence of BAFF-R on the surface of B cells from the transitional stages of development through maturity mirrors the expression of BAFF-R on the malignancies thought to arise from these various developmental stages and may enable B lineage malignancies to receive an anti-apoptotic signal from BAFF that reinforces and supports the expansion of the malignant cells. This additional survival signal may be crucial in indolent malignancies like chronic lymphocytic leukemia (CLL), a cancer characterized more by the prolonged survival of the cancerous cells than by their abundant proliferation. While there are ongoing efforts to thwart this survival pathway with antibody-based therapeutics directed toward BAFF and BAFF-R, understanding the regulation of BAFF-R expression may open a more proximal, potent, and novel therapeutic avenue. Rel/NF-kB family member c-Rel is an appealing candidate BAFF-R regulator, since its nuclear localization and expression within B lineage cells roughly correlate with BAFF-R expression, predominating at nuclear kB sites as a dimer with p50 in mature B cells and declining in plasma cells. Recently published evidence has shown that B cell receptor (BCR)-induced c-Rel expression can modulate BAFF-R in murine transitional B cells, providing a critical increase in BAFF-R expression and thus, BAFF-responsiveness, during B cell development (Castro, I. J Immunol. 2009; 182(12): 7729). A role for the BCR is especially provocative in the context of CLL, which is commonly characterized by lower levels of surface BAFF-R despite a generally activated phenotype and, in the unmutated subtype, typically increased expression of genes downstream of the BCR. In related studies, we have analyzed BAFF-R promoter activity in BAFF-R expressing B cells vs. myeloma cell lines, which generally lack expression of this receptor, and have indeed demonstrated differential promoter activity of the region directly upstream of the BAFF-R gene. The goal of our current study was to focus on the role of the transcription factor, c-Rel, in regulating BAFF-R expression. While in silico tools identifying transcription factor binding sites in the three kilobases upstream of the BAFF-R gene found numerous possible c-Rel binding sites, only three c-Rel sites remained under the most stringent search conditions. An initial chromatin immunoprecipitation (ChIP) experiment with an anti-c-Rel antibody suggested that the binding site within the first five hundred bases upstream of the transcriptional start site may be bound by c-Rel: ChIP with RAMOS B cells showed a 6-fold increase in the precipitated DNA of the region over the isotype-matched antibody control. Electrophoretic mobility shift assays (EMSAs) using peripheral blood (PB) B cell nuclear extracts both from normal donors and CLL patients as well as malignant cell lines, demonstrate the different levels of binding to the putative c-Rel sites identified in silico and support the hypothesis that c-Rel directly regulates BAFF-R expression in these cells. These assays, which demonstrated the affinity of c-Rel for the putative promoter sites upstream of the BAFF-R gene, led to the transfection of new luciferase reporter constructs into malignant B and plasma cell lines, which demonstrate the effects both of amplifying and of mutating the c-Rel binding sites in our reporter assays. We supplemented these assays with experiments measuring the production of c-Rel in these cells both by quantitative RT-PCR, which demonstrated a twenty-fold decrease in plasma cell line ALMC-1 and a two-fold decrease in CLL cell c-Rel mRNA as compared to normal PB B cells, and by Western blot. Finally, we used a c-Rel expression vector to directly increase BAFF-R expression in the malignant cell lines. By identifying c-Rel expression and activation as a possible mechanism of direct control of BAFF-R expression in human B cells, we reveal a critical new rationale for the use of NF-kB inhibitors as chemotherapeutic adjuvants in B cell cancers. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4765-4765
Author(s):  
Stephen A. Mihalcik ◽  
Renee C. Tschumper ◽  
Diane F. Jelinek

Abstract Throughout differentiation, mature B cells express distinct combinations of the BAFF and APRIL receptors, BAFF-R, TACI, and BCMA. The patterns of B lineage cell receptor expression reflect their stage of differentiation and impart the ability to respond to ligands, in some cases delivering a powerful anti-apoptotic signal. B cell malignancies arise from each stage of differentiation, typically exhibit the patterns of receptor expression that reflect their cell of origin, and have been shown to exploit the generally anti-apoptotic effects of BAFF and/or APRIL. For example, there is evidence for a role for BAFF in mature B cell cancers, including B cell chronic lymphocytic leukemia (B-CLL). As the majority of circulating CLL B cells are quiescent cells, prolonged survival is a significant hallmark, a trait that signals through BAFF-R could initiate or reinforce. Therapeutic strategies designed to interrupt this pro-survival pathway have thus far primarily focused on blocking ligand binding. Therapeutic modalities impacting receptor expression may be similarly effective. However, despite the apparent precise activation stage-dependent orchestration of B cell BAFF-R, TACI, and BCMA expression, the genetic mechanisms regulating expression of the three receptors remain undefined, and the question of whether each receptor governs expression of the other two remains unanswered. In agreement with previous studies in our own lab and others, analyses of the receptor profiles of CLL B cells continue to show BAFF-R surface expression, albeit at lower levels than seen on normal peripheral B cells, and the curious variable presence of BCMA and TACI. Similarly, multiple myeloma (MM) plasma cells (PCs), like normal PCs, uniformly lack BAFF-R expression, express BCMA, and variably express TACI. Our current study explores the mechanisms of receptor regulation in B cells, with an emphasis on BAFF-R, the receptor that is most consistently expressed on the CLL B cell population and that has the most clearly defined survival function. We began by analyzing the BAFF-R gene’s genomic context. We identified a possible regulatory element 2 kb upstream of the first exon with significant homology across seven mammalian species that overlapped a cluster of B cell lineage transcription factor binding sites, and, thus, we called the 2.5 kb directly upstream of the gene the putative BAFF-R promoter. We cloned the region and created promoter-reporter vectors in which the full-length promoter and 0.5 kb 5’ truncations thereof drive firefly luciferase production. While studies of primary B cells continue, studies with malignant B cell lines suggest that we have successfully cloned a powerful positive regulatory region. Specifically, B cell lines that express surface BAFF-R show positive inductions of firefly to control renilla luciferase activity in all of the promoter constructs over the empty construct with the greatest promoter activity in the longest constructs: 6-, 18-, and 3-fold inductions with the 2.5 kb promoter in RAJI, Loukes, and MEC-1 cells, respectively. To further test the promoter specificity, we transfected malignant PC lines, ALMC-1, ALMC-2, and KAS-6/1, which are negative by RT-PCR and surface staining for BAFF-R. These lines showed little promoter activity over baseline, with fold inductions between 0.5 and 2.5 for all of the promoter constructs. These results suggest that the MM lines no longer express the transcription factors required to drive BAFF-R expression and underscore our conclusion that we have identified the BAFF-R promoter. At the same time, investigations into epigenetic modification may reveal a crucial level of control. The transcriptional start site of the BAFF-R gene falls within a region of high CG content, and may be a possible CpG island. Upon treatment with the methyltransferase inhibitor, 5-azacytidine, primary blood B cells and MM cells showed no change in receptor expression. However, in CLL B cells, treatment of cultured cells caused a slight (9%) decrease in BAFF-R expression and prevented TACI upregulation in cells stimulated with CpG (a 76% increase fell to 36%). This evidence suggests that methylation indirectly suppresses expression of BAFF-R and TACI. It is essential to understand the regulation of survival receptors critical to normal B lineage cell survival, which may also be crucial for their malignant counterparts, in order to target those mechanisms as therapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4133-4133
Author(s):  
Elena Cubedo ◽  
Michelle Maurin ◽  
Ken Wright ◽  
Izidore S Lossos

Abstract Abstract 4133 Human germinal center–associated lymphoma (HGAL) and LIM domain only-2 (LMO2) are proteins highly expressed in germinal center (GC) B lymphocytes. HGAL and LMO2 are also expressed in GC-derived lymphomas and distinguish biologically distinct subgroups of diffuse large B-cell lymphomas (DLBCL) associated with improved survival. HGAL is involved in motility regulation of both normal and malignant B cells, while the function of LMO2 in GC B cells and lymphomas is unknown. However, LMO2, functioning as a transcription factor, is frequently activated in childhood T-cell acute lymphoblastic leukemias by chromosomal rearrangement or retroviral integration inducing thymocyte self renewal (McCormack, 2010) leading to oncogenic transformation. LMO2 is also necessary for embryonic erythropoiesis and adult hematopoiesis. PRDM1/BLIMP1 encodes a zinc finger transcriptional repressor that is expressed in a subset of GC B cells and in all plasma cells. It is a master regulator of terminal B cell differentiation to plasma cells. BLIMP1 may also function as a tumor suppressor in the pathogenesis of DLBCL, where it is frequently inactivated by mutations and deletions. Blimp1 promotes plasma cell differentiation and exerts its tumor suppressive effects by inhibiting the expression of genes important for GC B cell functions like c-myc, CIITA, PAX5, Bcl6, Spi-B, and ID-3 (Shaffer, 2002). Here, we demonstrate that Blimp1 also inhibits LMO2 and HGAL expression by direct binding to their promotors. Transient over-expression of BLIMP1 in B lymphoma cell lines results in a decrease in RNA and protein levels of HGAL and LMO2, as assessed by quantitative real time PCR and immunoblotting. To confirm a direct effect of BLIMP1 on expression of the HGAL and LMO2 genes, we have cloned the corresponding 2046 bp and 2549 bp DNA promoter sequences containing putative binding sites of BLIMP1 into luciferase reporter constructs. Over-expression of BLIMP1 in HeLa, VAL and CA-46 cell lines induces a statistically significant decrease in the promoter activity of both HGAL and LMO2. Site-specific mutagenesis of the BLIMP binding sites partially reverses the inhibitory effect of the BLIMP1 protein on the promoter activity, thus confirming specificity of the observed BLIMP1 inhibitory effects. To further demonstrate existence of the BLIMP1 inhibitory effect in a physiological context, we demonstrated in vivo binding of the endogenous BLIMP1 protein to HGAL and LMO2 promoters in plasma cells lines by chromatin immunoprecipitation assays. These findings demonstrate that Blimp1 is a physiological transcriptional repressor of the expression of LMO2 and HGAL genes. This inhibitory effect may contribute to the disappearance of HGAL and LMO2 expression upon differentiation of GC B cell to plasma cells and may also lead to absence of HGAL and LMO2 expression in post-GC lymphoid tumors. Additional studies are necessary to demonstrate whether inhibition of LMO2 and HGAL expression is necessary for oncogene suppressor functions of the BLIMP1 in DLBCL pathogenesis. Disclosures: No relevant conflicts of interest to declare.


1990 ◽  
Vol 10 (7) ◽  
pp. 3562-3568
Author(s):  
M Principato ◽  
J L Cleveland ◽  
U R Rapp ◽  
K L Holmes ◽  
J H Pierce ◽  
...  

Murine bone marrow cells infected with replication-defective retroviruses containing v-raf alone or v-myc alone yielded transformed pre-B cell lines, while a retroviral construct containing both v-raf and v-myc oncogenes produced clonally related populations of mature B cells and mature macrophages. The genealogy of these transformants demonstrates that mature myeloid cells were derived from cells with apparent B-lineage commitment and functional immunoglobulin rearrangements. This system should facilitate studies of developmental relationships in hematopoietic differentiation and analysis of lineage determination.


2016 ◽  
Vol 2016 ◽  
pp. 1-9 ◽  
Author(s):  
Kristien Van Belle ◽  
Jean Herman ◽  
Louis Boon ◽  
Mark Waer ◽  
Ben Sprangers ◽  
...  

B cell specific immunomodulatory drugs still remain an unmet medical need. Utilisation of validated simplified in vitro models would allow readily obtaining new insights in the complexity of B cell regulation. For this purpose we investigated which human B lymphocyte stimulation assays may be ideally suited to investigate new B lymphocyte immunosuppressants. Primary polyclonal human B cells underwent in vitro stimulation and their proliferation, production of immunoglobulins (Igs) and of cytokines, and expression of cell surface molecules were analysed using various stimuli. ODN2006, a toll-like receptor 9 (TLR9) agonist, was the most potent general B cell stimulus. Subsequently, we investigated on which human B cell lines ODN2006 evoked the broadest immunostimulatory effects. The Namalwa cell line proved to be the most responsive upon TLR9 stimulation and hence may serve as a relevant, homogeneous, and stable B cell model in an in vitro phenotypic assay for the discovery of new targets and inhibitors of the B cell activation processes. As for the read-out for such screening assay, it is proposed that the expression of activation and costimulatory surface markers reliably reflects B lymphocyte activation.


1992 ◽  
Vol 22 (10) ◽  
pp. 2547-2553 ◽  
Author(s):  
Anna Biró ◽  
Gabriella Sármay ◽  
Zoltán Rozsnyay ◽  
Eva Klein ◽  
János Gergely

Lipids ◽  
1996 ◽  
Vol 31 (10) ◽  
pp. 1051-1058 ◽  
Author(s):  
Cheikh M. Nguer ◽  
Dominique Treton ◽  
Marek Rola-Pleszczynski ◽  
Zohair Mishal ◽  
Yolène Thomas ◽  
...  

2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Elham Ahmadi ◽  
Mehrdad Ravanshad ◽  
Jun Xie ◽  
Rajesh Panigrahi ◽  
Sandeep S. Jubbal ◽  
...  

Abstract Background B-cell proliferative disorders, such as post-transplant lymphoproliferative disease (PTLD), are increased among persons afflicted by T-cell compromise. Most are Epstein–Barr virus (EBV) + and can first present with a focal lesion. Direct introduction of oncolytic viruses into localized tumors provides theoretical advantages over chemotherapy, immunotherapy and radiation therapy by reducing systemic toxicity. Despite extensive study as a vehicle for gene therapy, adeno-associated viruses (AAV) have rarely been applied to human cancer research due to technical and theoretical obstacles. Moreover, human B-cells have historically been described as resistant to AAV infection. Nonetheless, advances using different recombinant (r)AAV serotypes with unique tropisms to deliver cytotoxic therapy suggested a localized anti-tumor approach was feasible. Methods As a prelude to the development of a therapeutic vehicle, the ability of fifteen distinct EGFP-bearing rAAV serotypes to transduce human B-cells, including primary, immortalized, and B-cell tumor lines ± EBV was assessed by confocal microscopy, flow cytometry and subsequently cell viability assay. Results Rank order analysis revealed augmented transduction by rAAV6.2 and closely related virions. EBV infection of EBV-negative B-cell tumor lines and EBV immortalization of primary B-cells increased susceptibility to rAAV6.2 transduction. As a proof of concept, transduction by rAAV6.2 encoding herpes simplex virus type 1 (HSV1)-thymidine kinase (TK) eliminated TK-negative rhabdomyosarcoma cells and diminished viability of transduced B-cell lines upon incubation with ganciclovir. Conclusions rAAV serotypes differentially transduce human B-cell lines reversing the dogma that human B-cells are refractory to AAV infection. EBV + B-cells display increased susceptibility to rAAV6.2 infection, uncovering a new method for improved nucleic acid transfer into transfection-resistant B-cell lines. The introduction of a functional suicide gene into the rAAV6.2 genome identifies a candidate vector for the development of rAAV-based oncolytic therapy targeting focal EBV-bearing B-lymphoproliferative disorders.


2006 ◽  
Vol 104 (6) ◽  
pp. 1191-1201 ◽  
Author(s):  
Leslie C. McKinney ◽  
Thomas Butler ◽  
Shawn P. Mullen ◽  
Michael G. Klein

Background Mutations in the ryanodine type 1 receptor (RyR1) are causative for malignant hyperthermia. Studies in human B lymphocytes have shown that measurement of RyR1-mediated intracellular Ca(2+) (Ca(2+)(i)) release can differentiate between normal and malignant hyperthermia-susceptible individuals. The authors have further developed the B-cell assay by pharmacologically characterizing RyR1-mediated Ca release in two normal human B-cell lines and demonstrating increased sensitivity of lymphocytes to the RyR1 agonist 4-chloro-m-cresol (4-CmC) in the porcine model of MH. Methods Ca(2+)(i) was measured fluorometrically using fura-2 in populations of cells in suspension or with fluo-4 in single cells using confocal microscopy. The Dakiki and PP normal human B cell lines were used, as well as lymphocytes obtained from normal and malignant hyperthermia-susceptible pigs. 4-CmC was used to elicit RyR1-mediated Ca release; all experiments were performed in the absence of external Ca(2+). Results EC(50) values for 4-CmC were 0.98 and 1.04 mm for Dakiki and PP cells, respectively, demonstrating reproducibility. The 4-CmC-induced increase in Ca(2+)(i) was eliminated by thapsigargin and was unaffected by xestospongin C. The Ca(2+)(i) increase was separable from mitochondrial stores and was inhibited by azumolene. Caffeine did not induce Ca(2+)(i) release, but ryanodine depleted intracellular stores by 50%. Lymphocytes from pigs carrying the Arg614Cys mutation in RyR1 showed increased sensitivity to 4-CmC (EC(50) = 0.47 vs. 0.81 mm for cells derived from normal animals). Conclusions RyR1-mediated Ca(2+) signals can be pharmacologically distinguished from other intracellular sources in human B cells, and alterations of RyR1 function can be successfully detected using Ca(2+) release from intracellular stores as an end point.


Blood ◽  
2003 ◽  
Vol 101 (4) ◽  
pp. 1505-1512 ◽  
Author(s):  
Ines Schwering ◽  
Andreas Bräuninger ◽  
Ulf Klein ◽  
Berit Jungnickel ◽  
Marianne Tinguely ◽  
...  

Hodgkin and Reed-Sternberg (HRS) cells represent the malignant cells in classical Hodgkin lymphoma (HL). Because their immunophenotype cannot be attributed to any normal cell of the hematopoietic lineage, the origin of HRS cells has been controversially discussed, but molecular studies established their derivation from germinal center B cells. In this study, gene expression profiles generated by serial analysis of gene expression (SAGE) and DNA chip microarrays from HL cell lines were compared with those of normal B-cell subsets, focusing here on the expression of B-lineage markers. This analysis revealed decreased mRNA levels for nearly all established B-lineage–specific genes. For 9 of these genes, lack of protein expression was histochemically confirmed. Down-regulation of genes affected multiple components of signaling pathways active in B cells, including B-cell receptor (BCR) signaling. Because several genes down-regulated in HRS cells are positively regulated by the transcriptional activator Pax-5, which is expressed in most HRS cells, we studied HL cell lines for mutations in the Pax-5gene. However, no mutations were found. We propose that the lost B-lineage identity in HRS cells may explain their survival without BCR expression and reflect a fundamental defect in maintaining the B-cell differentiation state in HRS cells, which is likely caused by a novel, yet unknown, pathogenic mechanism.


1990 ◽  
Vol 10 (7) ◽  
pp. 3562-3568 ◽  
Author(s):  
M Principato ◽  
J L Cleveland ◽  
U R Rapp ◽  
K L Holmes ◽  
J H Pierce ◽  
...  

Murine bone marrow cells infected with replication-defective retroviruses containing v-raf alone or v-myc alone yielded transformed pre-B cell lines, while a retroviral construct containing both v-raf and v-myc oncogenes produced clonally related populations of mature B cells and mature macrophages. The genealogy of these transformants demonstrates that mature myeloid cells were derived from cells with apparent B-lineage commitment and functional immunoglobulin rearrangements. This system should facilitate studies of developmental relationships in hematopoietic differentiation and analysis of lineage determination.


Sign in / Sign up

Export Citation Format

Share Document