Expression of Germinal Center B Cell Markers in Bortezomib-Resistant Multiple Myeloma Cells

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 129-129 ◽  
Author(s):  
Holly Stessman ◽  
Linda B. Baughn ◽  
Aatif Mansoor ◽  
Brian Van Ness

Abstract Abstract 129 Multiple myeloma (MM), the second most common hematopoietic malignancy, is an incurable plasma cell neoplasm representing approximately 2% of all cancer deaths. The proteasome inhibitor, bortezomib (Bz), has been widely used to treat MM, and emerging next-generation proteasome inhibitors are being evaluated. Despite advances in therapies, patients treated with Bz eventually relapse due to the development of drug resistance. Thus, identifying signatures that distinguish Bz sensitive from resistant MM cells will more accurately predict Bz sensitivity and will aid in the development of novel approaches to reverse resistance. MM populations are characterized by phenotypic heterogeneity suggesting the possibility that certain MM subpopulations are associated with Bz resistance. In order to characterize these populations in greater detail, we have utilized isogenic pairs of Bz sensitive and resistant MM lines derived from the genetically engineered iMycCa/Bcl-xl mouse model of plasma cell malignancy. The iMycCa/Bcl-xl mouse is a highly robust model that very closely resembles human MM based on gene expression profiling (GEP), chromosomal abnormalities and progression of disease and response to therapy (Boylan, et al. Cancer Res (2007) 67: 4069). We have employed this mouse model because of its striking similarity to human MM, and MM cells isolated from these mice can be readily infected by lentivirus as well as adoptively transferred into syngeneic, immunocompetent recipients establishing a powerful system to directly test biologically relevant hypotheses relating to Bz resistance. Isogenic pairs of Bz sensitive and resistant lines were extensively characterized using GEP, quantitative PCR and flow cytometry. We observed increased expression of germinal B cell markers Pax5, AID and Bcl-6 and reduced expression of the plasma cell marker CD93 in Bz resistant lines compared to Bz sensitive pairs suggesting that Bz resistance is associated with germinal center-like B cell characteristics. Remarkably, we detect within the same cell co-expression of both CD138/Pax5 and CD138/AID demonstrating the tremendous plasticity of these cells and their intermediate B cell differentiation phenotype. Recently, both AID and Bcl-6 have been identified as crucial mediators of resistance to tyrosine kinase inhibitors in leukemia cells (Duy, et al. Nature (2011) 473: 384; Klemm, et al. Cancer Cell (2009) 16: 232) raising an intriguing possibility that these proteins thought to be restricted to germinal center B cells could play a role in Bz resistance in MM. These observations have led us to more closely characterize the apparent shift in developmental stage resulting from Bz-driven expression changes or selection. Time course analysis of gene and protein expression following low-dose Bz treatment in the absence of cell death indicates that Bz increases the expression and promotes the stabilization of Pax5, AID and Bcl-6. This demonstrates a drug-induced change in markers associated with B cell differentiation relating to the ultimate emergence of the resistance phenotype. We are now determining whether similar B cell differentiation phenotypes emerge in a recently published human drug trial where GEP was completed prior to and 48 hours after Bz treatment in patients (Shaughnessy, et al. Blood (2011), ahead of print). Thus, we will present phenotypic signatures correlating to Bz resistance in cell lines derived from our mouse model of MM and suggest that targeting B cell differentiation may represent a rational therapeutic approach to overcoming Bz resistance. Disclosures: Stessman: Millennium: The Takeda Oncology Company: Research Funding. Mansoor:Millennium: The Takeda Oncology Company: Research Funding. Van Ness:Millennium: The Takeda Oncology Company: Research Funding.

2016 ◽  
Vol 213 (6) ◽  
pp. 1095-1111 ◽  
Author(s):  
Renee Gloury ◽  
Dimitra Zotos ◽  
Malou Zuidscherwoude ◽  
Frederick Masson ◽  
Yang Liao ◽  
...  

The generation of high-affinity antibodies requires germinal center (GC) development and differentiation of long-lived plasma cells in a multilayered process that is tightly controlled by the activity of multiple transcription factors. Here, we reveal a new layer of complexity by demonstrating that dynamic changes in Id3 and E-protein activity govern both GC and plasma cell differentiation. We show that down-regulation of Id3 in B cells is essential for releasing E2A and E2-2, which in a redundant manner are required for antigen-induced B cell differentiation. We demonstrate that this pathway controls the expression of multiple key factors, including Blimp1, Xbp1, and CXCR4, and is therefore critical for establishing the transcriptional network that controls GC B cell and plasma cell differentiation.


2006 ◽  
Vol 203 (4) ◽  
pp. 1081-1091 ◽  
Author(s):  
Didrik Paus ◽  
Tri Giang Phan ◽  
Tyani D. Chan ◽  
Sandra Gardam ◽  
Antony Basten ◽  
...  

B cells responding to T-dependent antigen either differentiate rapidly into extrafollicular plasma cells or enter germinal centers and undergo somatic hypermutation and affinity maturation. However, the physiological cues that direct B cell differentiation down one pathway versus the other are unknown. Here we show that the strength of the initial interaction between B cell receptor (BCR) and antigen is a primary determinant of this decision. B cells expressing a defined BCR specificity for hen egg lysozyme (HEL) were challenged with sheep red blood cell conjugates of a series of recombinant mutant HEL proteins engineered to bind this BCR over a 10,000-fold affinity range. Decreasing either initial BCR affinity or antigen density was found to selectively remove the extrafollicular plasma cell response but leave the germinal center response intact. Moreover, analysis of competing B cells revealed that high affinity specificities are more prevalent in the extrafollicular plasma cell versus the germinal center B cell response. Thus, the effectiveness of early T-dependent antibody responses is optimized by preferentially steering B cells reactive against either high affinity or abundant epitopes toward extrafollicular plasma cell differentiation. Conversely, responding clones with weaker antigen reactivity are primarily directed to germinal centers where they undergo affinity maturation.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 914-914
Author(s):  
Salvador Alonso ◽  
William Matsui ◽  
Richard J Jones ◽  
Gabriel Ghiaur

Abstract Bortezomib (BTZ), a proteasome inhibitor that induces accumulation of misfolded proteins and apoptosis in highly secretory cells, has improved survival in multiple myeloma (MM) but fails to achieve cure. It has been postulated that a population of MM cells, phenotypically similar to B-cells, are intrinsically resistant to BTZ, and contribute to minimal residual disease (MRD) and relapse (Matsui et al., 2008). Bone marrow (BM) stroma expresses CYP26 enzymes and creates a retinoic acid (RA)-low environment that prevents differentiation of normal and malignant cells (Meng et al., 2015). Since RA promotes plasma cell differentiation, Ig secretion (Ertesvag et al., 2007) and thus, ER-stress (Xu et al., 2007), we tested if the BM niche induces BTZ resistance by promoting a B-cell program in MM. Consistent with this, MM cells (H929 and MM1S) cultured under low RA conditions (co-cultured with BM stroma or treated with AGN194310, a pan-RA receptor inhibitor) up regulated B-cell markers (BCL6), and down regulated genes associated with plasma cell differentiation and BTZ sensitivity (XBP1s, Blimp, CHOP) (Fig 1A). In addition, inhibition of stromal CYP26 via R115866 reversed these gene expression changes in stroma co-culture conditions (Fig 1A). To verify whether a low-RA environment promotes BTZ resistance, we incubated MM cells with BM mesenchymal cells for 5 days. Then, MM cells were separated from stroma, and treated with BTZ. Incubation with BM stroma induced BTZ resistance of MM cells, which was completely overcome by CYP26 inhibition or by the CYP26 resistant retinoid IRX5183 (Fig 1B). Moreover, MM cells pretreated with AGN for 5 days were also resistant to BTZ (Fig 1B). Interestingly, this BTZ resistant phenotype was preserved for up to 48h upon removal of MM cells from stromal co-culture (Fig 1C). Recent studies suggest that malignant cells remodel their microenvironments to build a more protective niche (Schepers et al., 2013). Consistent with this, we found that stromal CYP26A1 was highly upregulated after co-culture with MM cells. Supernatant from MM cells had similar effects, implicating a soluble factor on this interaction. Since secretion of SHH was implicated in chemotherapy resistance in MM (Liu et al., 2014), we tested if MM-induced CYP26 up-regulation was dependent on SHH signaling. As hypothesized, treatment with the Smo antagonist cyclopamine, or the use of SMO knockout (KO) stromal cells, overcame CYP26A1 up regulation by MM cells (Fig 1D) To test if cell extrinsic SHH signaling in stromal niche contributes to BTZ resistance by modulating RA pathway in MM cells, we modeled MM-BM stroma interactions in a xenograft setting. Each mouse carried two subcutaneous tumors consisting of Luciferase expressing MM1S cells and SmoFl/Fl BM stroma cells, transduced with either a control vector (WT stroma) or Cre-recombinase (Smo KO stroma) (Fig E). Mice were treated with IRX5183, BTZ, or the combination. Consistent with our in vitro data, tumors with Smo KO stroma showed a significant response to BTZ, while those with WT stroma were refractory, as determined by exponential increase in bioluminescence. However, the combination of IRX+BTZ resulted in a dramatic and equivalent response regardless of the phenotype of the stromal compartment (Fig 2A, 2B). Moreover, tumors with Smo KO stroma, or treated with IRX, showed decrease expression of B-cell markers, and up-regulation of genes associated with plasma cell differentiation and BTZ sensitivity. In conclusion, we show that the BM niche promotes a B-cell program in MM, characterized by a low ER-stress and BTZ resistance. This niche-induced program is maintained even after displacement from the BM, which may have important implications for the use of mobilization-sensitization strategies. Finally, we propose the existence of a bi-directional crosstalk between MM cells and their respective niches. In our model, MM cells via secretion of SHH modify BM stroma to create RA-low environments, induce BTZ resistance and contribute to MRD. Figure 1. Figure 1. Figure 2. Figure 2. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 2266
Author(s):  
Marta Cuenca ◽  
Victor Peperzak

B-cell malignancies arise from different stages of B-cell differentiation and constitute a heterogeneous group of cancers including B-cell lymphomas, B-cell leukemias, and plasma cell dyscrasias [...]


Blood ◽  
2011 ◽  
Vol 117 (22) ◽  
pp. 5907-5917 ◽  
Author(s):  
Katerina Vrzalikova ◽  
Martina Vockerodt ◽  
Sarah Leonard ◽  
Andrew Bell ◽  
Wenbin Wei ◽  
...  

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


Sign in / Sign up

Export Citation Format

Share Document