FOXO1-p300-Txn Circuit Regulates Oxidative Stress Responses in Diffuse Large B-Cell Lymphomas Characterized By Enhanced Oxidative Phosphorylation

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 466-466 ◽  
Author(s):  
Tomasz Sewastianik ◽  
Maciej Szydlowski ◽  
Emilia Bialopiotrowicz ◽  
Ewa Jablonska ◽  
Przemyslaw Kiliszek ◽  
...  

Abstract Diffuse large B-cell lymphoma (DLBCL) is a clinically and molecularly heterogeneous disease. The comparison of DLBCLs transcriptional profiles using multiple clustering algorithms led to the identification of distinct DLBCL subtypes reflecting tumor-intrinsic features. The OxPhos-DLBCL subtype is characterized by enhanced mitochondrial oxidative phosphorylation, a major source of potentially toxic reactive oxygen species (ROS). Therefore, we investigated the role of potential mechanisms attenuating ROS toxicity in this DLBCL subtype. We found significantly increased thioredoxin (TXN) mRNA abundance in DLBCLs classified as OxPhos subtype compared to other subtypes. The overall survival (OS) of patients with high TXN mRNA expression was significantly shorter than of those with low TXN mRNA expression, regardless of treatment regimen (R-CHOP and CHOP). TXN overexpression in OxPhos-DLBCLs was also confirmed in a cell line panel at protein level using immunoblotting. To explain transcriptional mechanisms responsible for differential TXN expression in different DLBCLs, we analyzed the TXN promoter and identified two BCL6 binding sites within 1kb upstream of TXN transcription start site. Unlike in other molecular subtypes, BCL6 does not exhibit a repressor activity in OxPhos-DLBCLs (PNAS, 2007; 104: 3207-12). Using luciferase reporter assays and shRNA-mediated gene expression knock-down, we demonstrated that relative differences in TXN abundance between DLBCL subtypes are at least in part caused by the lack of BCL6 transcription repressor activity. We next tested the consequences of TXN depletion in DLBCLs. We found that OxPhos cells with silenced TXN expression were uniformly more sensitive to apoptosis induced by ROS production than control cells. TXN inhibition sensitized all tested cell lines to doxorubicin, the fundamental drug used in DLBCL chemotherapy acting in part as a ROS inducer. In addition to its role in maintaining redox homeostasis, TXN also regulates transcriptional responses to ROS. For example, TXN reduces disulfide bonds between FOXO4 and acetyltransferase p300, which results in reduced FOXO4 acetylation and impaired proapoptotic signaling. For this reason, we assessed whether p300 and TXN are involved in acetylation of FOXO1, a major FOXO member expressed in DLBCLs. In cells with blocked TXN activity, FOXO1 acetylation was significantly higher compared to cells overexpressing wild-type TXN. TXN decreased p300-mediated FOXO1 acetylation, reduced its proapoptotic activity and expression of FOXO1-dependent genes (TRAIL, p27, BIM). We found that FOXO1 and p300 interact in redox and TXN-dependent manner and identified a conserved FOXO1's Cys612 to be responsible for FOXO1-p300 binding. We mutated FOXO1 C612 to Ala and found that when cotransfected with p300, C612A FOXO1 exhibited dramatically suppressed ROS-induced acetylation. Blockade of FOXO1 acetylation resulted in markedly lower expression of FOXO1 target genes, higher cellular proliferation and lower apoptosis. Furthermore, TXN inhibited FOXO1 nuclear translocation in response to oxidative stress in OxPhos-DLBCLs. Finally, silencing FOXO1 in OxPhos cells with knocked-down TXN expression markedly inhibited DLBCL cell line apoptosis in response to oxidative stress, suggesting that FOXO1 is an essential TXN-regulated sensor and effector of ROS toxicity in OxPhos-DLBCL cells. Taken together, these results demonstrate that TXN is overexpressed in a subset of DLBCLs, and high TXN mRNA abundance is related to shorter OS of DLBCL patients. TXN knock-down enhances oxidative stress toxicity in OxPhos cell lines at least in part by facilitating FOXO1 nuclear retention and increasing acetylation of this transcription factor, thus augmenting its proapoptotic activity. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2963-2963
Author(s):  
Goldi A Kozloski ◽  
Xiaoyu Jiang ◽  
Shruti Bhatt ◽  
Rita Shaknovich ◽  
Ari M Melnick ◽  
...  

Abstract Introduction: Diffuse large B-cell lymphoma (DLBCL) is subdivided into the germinal center B-like (GCB) and activated B cell-like (ABC) subtypes by gene expression profiling, and these subtypes exhibit different clinical outcomes and signaling pathway deregulations. Compared to the GCB, the ABC-DLBCL subtype displays a more aggressive clinical course and shorter patient survival. Constitutive nuclear factor kappa-B (NF-kB) activity is often associated with the ABC-DLBCL subtype, however recent studies suggest that NF-kB signaling activation is also observed to a lower extent in the GCB-DLBCL subtype (Lina Odqvist et al. 2014). miRNAs have diagnostic and prognostic value in disease classification, and growing evidence implicates miRNAs in tumorigenesis, tumor maintenance, and dissemination through their ability to modulate the expression of critical genes and signaling networks. We previously demonstrated that miRNA-181a expression correlates with longer survival in patients treated with R-CHOP, independent of established clinical and molecular predictors. However, the molecular and cellular mechanisms underlying the association between miRNA-181a expression and improved prognosis in DLBCL patients are currently unknown. Herein we analyzed the role of miRNA-181a in DLBCL pathogenesis. Results:Quantitative RT-PCR analyses demonstrate higher endogenous miRNA-181a levels in centroblasts than in plasmablasts. Concordantly, endogenous miRNA-181a levels were significantly higher in GCB DLBCL cell lines and primary tumors compared with ABC DLBCL. These expression differences could not be attributed to distinct DNA methylation signatures in the miRNA-181a promoters (Chromosomes 1, 9) or regulatory elements as analyzed by Mass Array Sequenom Epityping. In search for putative miRNA-181a targets we identified 5 genes (CARD11, NFKB1A (IKBα), NFKB1 (p105/p50), RELA (p65), and REL (CREL)) within the NF-kB signaling pathway. Analyses of these targets show a decrease in the levels of these proteins and mRNAs in ABC and GCB DLBCL cell lines ectopically expressing miRNA-181a compared with scramble control plasmid. Luciferase reporter analyses encoding the respective wild type or mutated 3′UTR sequences demonstrate direct and specific targeting of these transcripts with the exception of RELA. Analysis of the net effect of miRNA-181a on NF-kB signaling using NF-kB luciferase reporter demonstrate significant decrease in NF-kB signaling. Concordantly, anti-miRNA-181a transfection led to increased NF-kB luciferase reporter activity. Moreover, western blot analyses of cytoplasmic and nuclear fractions showed a decrease in the levels of the transcription factors CREL and p50 in both cellular compartments, a decrease in the binding to DNA at NF-kB binding motifs, and a consequent decrease in NF-kB target gene transcription in the miRNA-181a expressing cells compared with scramble control. Together these studies point to miRNA-181a-mediated repression of NF-kB signaling in DLBCLs. Ectopic miRNA-181a expression led to a decrease in cell proliferation and an increase in cell death in both DLBCL subtypes, but this effect was more pronounced in the ABC DLBCL cell lines. The miRNA-181a-mediated increase in cell apoptosis could not be rescued by BCL2 co-transfection, an anti-apoptotic protein that was previously established as a direct miRNA-181a target. Analyses of miRNA-181a effects in NOD/SCID mice demonstrated that in vivo miRNA-181a induction in GCB and ABC human DLBCL xenografts led to decreased tumor growth and significantly longer animal survival. Notably, survival was prolonged in both GCB and ABC DLBCL bearing animals. Figure 1 Figure 1. Conclusions: miRNA-181a directly suppress the NF-kB signaling pathway and lead to increased tumor cell death in both DLBCL subtypes suggesting that NF-kB deregulation is present in both tumor subtypes. However, the lower miRNA-181a expression level in the ABC DLBCL subtype may contribute to the higher NF-kB signaling activity that is observed in this subtype. Furthermore, our study provides a plausible explanation for the association between high miRNA-181a expression and longer survival of DLBCL patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1870-1870
Author(s):  
Brandon Willis ◽  
India Neveras ◽  
Hannah Dry ◽  
Wendan Xu ◽  
Yang Li ◽  
...  

Abstract Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell malignancy among adults and despite approximately 65% of patients with DLBCL being cured with RCHOP therapy, nonresponsive and relapsed patients have inadequate treatment options, highlighting the importance for innovative treatment regimens. Blockade of B-cell receptor (BCR) downstream signaling components with various targeted agents is emerging as a clinically tractable treatment strategy across multiple B-cell malignancies. Protein Kinase B (AKT) signaling downstream of the BCR complex has been shown to be a central node in germinal center B-cell (GCB) DLBCL and the potent, selective inhibitor of AKT1, AKT2, AKT3, capivasertib, currently being evaluated in multiple clinical trials by targeting AKT-driven solid cancers, has been shown to induce apoptosis in a subset of GCB-DLBCL cell lines and cause tumor stasis in xenograft mouse models (Erdman et al., 2017). Since the monotherapy capivasertib responses in GCB DLBCL models are partial and lack durability, we hypothesized a combination approach could deliver even greater therapeutic benefit. To identify optimal partners, we conducted a capivasertib centric in vitro combination screen with specific with BH3 family members across a panel of 15 DLBCL cell lines, which revealed a synergistically active combination with the BCL2 inhibitor, venetoclax which is currently being evaluated in DLBCL. The activity was specifically enhanced in cell lines of the GCB subtype, with 4 PTEN del and 2 PTEN wt cell line models showing combination benefit. To determine the ability of this combination to drive stronger and durable responses, we assessed capivasertib and ventoclax activity in xenograft mouse models using two GCB-DLBCL cell line lines, SUDHL4 (PTEN wt) and WSU-DLCL2 (PTEN del). Oral administration of either monotherapy capivasertib (130 mg/kg BID, 4-day on/3-day off) or venetoclax (100 mg/kg QD) provided partial tumor growth inhibition (capivasertib TGI = 74% in SUDHL4 and 29% in WSU-DLCL2, and venetoclax TGI = 46% in SUDHL4 and 0% in WSU-DLCL2), whereas the combination of capivasertib and venetoclax both on a 4-day on/3-day off schedule produced complete tumor regression (100% regression) in both xenograft GCB cell line models during the dosing period. Notably, in both xenograft models all mice (5/5 per model) remained tumor free for at least 30 days following dosing cessation demonstrating high durability of response for the combination. Additionally, this combination is currently being evaluated in clinically relevant GCB and non-GCB PDX mouse models. Taken together, our results provide preclinical evidence for the rational combination of AKT and BCL-2 blockade with capivasertib and venetoclax respectively in patients with relapsed/refractory GCB-DLBCL. Disclosures Willis: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Neveras: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Dry: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Mongeon: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Rosen: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Mettetal: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Barry: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options.


2016 ◽  
Vol 113 (5) ◽  
pp. E577-E586 ◽  
Author(s):  
Joseph D. Dekker ◽  
Daechan Park ◽  
Arthur L. Shaffer ◽  
Holger Kohlhammer ◽  
Wei Deng ◽  
...  

High expression of the forkhead box P1 (FOXP1) transcription factor distinguishes the aggressive activated B cell (ABC) diffuse large B-cell lymphoma (DLBCL) subtype from the better prognosis germinal center B-cell (GCB)-DLBCL subtype and is highly correlated with poor outcomes. A genetic or functional role for FOXP1 in lymphomagenesis, however, remains unknown. Here, we report that sustained FOXP1 expression is vital for ABC-DLBCL cell-line survival. Genome-wide analyses revealed direct and indirect FOXP1 transcriptional enforcement of ABC-DLBCL hallmarks, including the classical NF-κB and MYD88 (myeloid differentiation primary response gene 88) pathways. FOXP1 promoted gene expression underlying transition of the GCB cell to the plasmablast—the transient B-cell stage targeted in ABC-DLBCL transformation—by antagonizing pathways distinctive of GCB-DLBCL, including that of the GCB “master regulator,” BCL6 (B-cell lymphoma 6). Cell-line derived FOXP1 target genes that were highly correlated with FOXP1 expression in primary DLBCL accurately segregated the corresponding clinical subtypes of a large cohort of primary DLBCL isolates and identified conserved pathways associated with ABC-DLBCL pathology.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 244-244
Author(s):  
Izidore S. Lossos ◽  
XiaoQing Lu ◽  
Feiying Ding ◽  
Manuel Rosado ◽  
Ash A. Alizadeh ◽  
...  

Abstract Gene expression profiling studies sub-classified diffuse large B-cell lymphomas (DLBCL) into two clinically distinct types: germinal center B cell (GCB)-like and activated B-cell (ABC)-like tumors, characterized by long and short survival, respectively. At least two markers of the GCB-phenotype - BCL6 and HGAL - are IL-4 target genes whose high expression independently predicts better overall survival. Gene expression analysis of DLBCL also demonstrated higher levels of mRNA expression of components of the IL-4 signaling pathway (IL-4Rα, IRS, p110 subunit of PI-3 kinase, and PKC delta) in the GCB-like DLBCL (Alizadeh et al Nature. 2000;403:503). Identification of IL-4 inducible genes in normal B-lymphocytes revealed additional IL-4 target genes that are expressed at higher levels in GCB-like DLBCL compared to ABC-like DLBCL. Together, these observations support the distinct activity of the IL-4 signaling pathway in DLBCL subtypes. Accordingly, IL-4 stimulation of GCB-like (SUDHL6, SUDHL4 and OCILY19) and ABC-like (OCILY10 and OCILY3) DLBCL cell lines increased expression of its known target genes only in GCB-like, but not in ABC-like DLBCL. Further, IL-4 stimulation led to AKT phosphorylation in the ABC-like but not in the GCB-like cells. Conversely, IL-4 induced STAT6 phosphorylation (pSTAT6) in all the tested GCB-like and in the OCILY10 cell lines but not in the OCILY3 ABC-like cell-line. IL-4 induced progressive accumulation of large quantities of pSTAT6 in both the cytoplasm and in the nucleus in the GCB-like DLBCL. In contrast, in IL-4 treated ABC-like OCILY10 cells, pSTAT6 did not accumulate in either cytoplasm or nucleus, and much smaller amounts of pSTAT6 were detected in the nuclear extracts from stimulated cells. The latter observation was at least partially attributed to different extent of pSTAT6 nuclear deposphorylation and proteasomal degradation in the GCB-like and ABC-like DLBCL, as determined by exposure of these cell lines to STAT6 nuclear export inhibitor (leptomycin B) and phosphatase and proteasome inhibitors. Nuclear protein tyrosine phosphatase assays revealed significantly higher phosphatase activity in the ABC-like compared to the GCB-like DLBCL cell-lines. Evaluation of mRNA expression of 51 known tyrosine phosphatases in the GCB-like and ABC-like DLBCL tumors based on array data revealed that mRNA expression of 13 protein tyrosine phosphatases was significantly higher (p<0.001) in the ABC-like compared to GCB-like DLBCL. In vitro pSTAT6 dephosphorylation studies suggested that PTPN2, expressed in ABC-like but not in the GCB-like cell-lines, is a putative STAT6 phosphatase. Taken together with the independent prognostic significance of IL-4 target genes (e.g., BCL-6 and HGAL) and distinct effects of IL-4 on proliferation and immuno-chemosensitivity of GCB-like and ABC-like cells (Abstract by Nechushtan et al), the observed differences in the intracellular IL-4 signaling might contribute to different clinical outcome of patients with DLBCL.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2497-2497 ◽  
Author(s):  
Kui Nie ◽  
Taotao Zhang ◽  
Jiong Yan ◽  
Leonardo Boiocchi ◽  
Shuhua Cheng ◽  
...  

Abstract A novel IGH-associated reciprocal translocation, t(4;14)(q24;q32), was identified, along with trisomy 9, in 20 of 20 metaphases by conventional karyotyping in a case of malignant gastric post-transplant lymphoproliferative disorder (PTLD). Cloning of the translocation site by inverse PCR identified BANK1 (B-cell scaffold protein with ankyrin repeats 1), a B-cell-specific adaptor protein with putative functions in B-cell receptor and CD40 signaling, as a novel IGH translocation partner. The breakpoints were located at the Sα region of IGH and intron 1 of BANK1. The translocation juxtaposed the two genes in opposite orientations, and surprisingly, resulted in transcriptional inactivation of BANK1 as a result of dissociation of the major BANK1 promoter. While BANK1 isoforms were expressed in all tonsillar B-cells, with lower levels (∼ 5 fold) in the germinal centers (GC) compared to naïve and memory B-cells, transcription from the major promoter in the tumor was absent and transcription from the minor promoter was reduced 50% relative to GC B-cells, suggesting that the non-translocated BANK1 allele was also inactivated. The total BANK1 expression was very low (∼10% of normal GC B cells) and crytic promoter activation was not identified. Several genes (PPP3CA, MIR1255A, FLJ20021 and SLC39A8), located 180 to 440 kb away from BANK1, were analyzed for mRNA expression; there is no significant activation in any of these genes, further supporting that BANK1is indeed the target gene affected by the translocation. Interphase FISH using break-apart BANK1 probes confirmed breakpoint in the index case but did not identify translocations in additional 15 PTLDs and 68 diffuse large B-cell lymphomas (DLBCL), implying that BANK1 translocation may be a rare event. To determine if BANK1 inactivation may occur in B-cell lymphomas by other mechanisms, 23 B-cell lymphoma cell lines, including 8 Burkitt lymphoma (BL), 9 diffuse large B cell lymphoma (DLBCL), 3 primary effusion lymphoma (PEL), and 3 classical Hodgkin lymphoma (cHL) were bisulfite sequenced to assess the methylation status of 37 CpG dinucleotides in a 436 base-pair region at the 5’ end of BANK1, which extends across exon 1 into the 5’ portion of intron 1. High level of methylation (>60% methylation on average among all CpGs) was seen in all 3 cHL and 2 of 3 PEL cell lines. Regional methylation was seen in 3 of 8 BL lines and 1 of 3 PEL lines. No hypermemethylation was identified in the DLBCL lines or in normal tonsils. Hypermethylation was associated with almost complete silencing of BANK1 transcription. In the DLBCL lines and BL lines without BANK1 hypermethylation, BANK1mRNA expressions were variable, ranging from <5% to 130% of GCB cells. To confirm that BANK1 hypermethylation is present in primary lymphoma cases, methylation status of 17 of the 37 CpGs were assessed in 23 cHL cases using en bloc formalin-fixed, paraffin-embedded materials and also laser-capture micro-issected Hodgkin/Reed-Sternberg (HRS) cells. There was evidence of BANK1 hypermethylation in the tumor cells in 9 of 23 cHL. Tumor cell specificity of BANK1 hypermethylation was further confirmed in 4 cHL cases using micro-dissected HRS cells. HRS cells were negative for BANK1 in 28 of 29 cHL cases examined by immunohistochemistry, suggesting that other mechanisms other than DNA methylation may be responsible for silencing BANK1expression. To investigate whether BANK1 has biological effects on B-cells related to lymphoma development, exogenous BANK1 was re-introduced to BC3, a PEL cell line showing marked BANK1 hypermethylation with absence of BANK1 expression. We established a stable doxycycline-inducible BC3 cell line expressing BANK1. Inhibition of cell growth was observed 2 to 3 days after doxycyline induction, and the number of viable cells with transfected BANK1 was only 25% compared to BC3 cells carry vehicle alone at day 6. An analysis of 5-bromo-2’ deoxyuridine (BrdU) incorporation after 48 hours of doxycline induction revealed that the fraction of cells in S-phase was reduced by 50% in the BANK1 transfectants, suggesting that BANK1has a negative effect on cell proliferation in these B cells. In summary, we have identified a novel IGH translocation partner and provide an example of an unusual consequence (gene inactivation) of IGH-associated translocation. We provide for the first time evidence of a potential role of BANK1 down-regulation in the development of B-cell lymphomas. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 643-643
Author(s):  
Julie Marie Matthews ◽  
Li Tan ◽  
Shruti Bhatt ◽  
Matthew Patricelli ◽  
Tyzoon Nomanbhoy ◽  
...  

Abstract Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma (NHL). The pathogenesis of DLBCL represents a multi-step process that involves the accumulation of multiple genetic and molecular lesions. Marked advances in the understanding of DLBCL pathobiology have been made by the application of gene expression arrays, comparative genomic hybridization arrays and “next” generation sequencing. This led to the identification of previously unrecognized DLBCL subtypes (germinal center-like (GCB) and activated B cell-like (ABC)) as well as type specific-deregulation of particular signaling pathways. These approaches focused on genetic aberrations and mRNA expression profiles, whereas the crucial events transforming normal cells are executed by proteins. Kinases play an important role in neoplastic transformation. Herein, we have undertaken the task of profiling kinase activity in DLBCL to further delineate potential mechanisms of DLBCL pathogenesis and develop novel therapeutic agents. A comprehensive analysis of global kinase activity/protein expression was performed using KiNativ technology. Kinomic analysis of 8 DLBCL cell lines, as compared to non-cancerous primary B-cells, led to the discovery of 13 members of the MAPK cascade which were activated and/or overexpressed in DLBCL. Only three of the detected MAPK members were inactive or had reduced expression compared to their non-cancerous counterparts. To determine whether these findings could be extended to de novo primary human DLBCL tumors, we performed immunohistochemistry (IHC) of the proximally activated kinase, MAP4K2 or “Germinal Center Kinase” (GCK) and the phosphorylated forms of its downstream targets: MAP3K1, MAP2K4, MAP2K7, and C-jun N-terminal Kinase 1 (JNK1). Analyzed kinases were expressed and activated in more than 80% of primary DLBCL tumors, confirming the KiNativ cell line data. The kinase array data was further corroborated with classical immunoprecipitation-based JNK and p38 assays. Hierarchical clustering analysis of 36 DLBCL specimens stained for GCB and ABC markers demonstrated that GCK expression/activation is not DLBCL subtype specific. Notably, in a cohort of 151 primary DLBCL cases, we found that patients whose tumors did not express GCK had an estimated progression free survival (PFS) of 85% at 10 years of follow up, whereas those tumors expressing GCK were associated with significantly reduced PFS of 53% (p=0.04). While there was a similar trend in overall survival, it did not reach statistical significance, which may be due to the relatively small number of DLBCL cases not expressing GCK and the potential rescue of these patients with second line treatments. RNA interference studies in DLBCL cell lines confirmed the importance of GCK for the survival of these tumors, resulting in reduced viability and G0/G1 arrest. We next developed a small molecule inhibitor, HG6-64-1. KiNativ, Ambit and Invitrogen profiling of HG6-64-1 targets revealed that it potently inhibited GCK. In vitro treatment with the novel GCK inhibitor, HG6-64-1, led to cell cycle arrest and the induction of apoptosis in DLBCL cell lines and primary DLBCL tumors. G452, a DLBCL cell line minimally expressing GCK, was not affected by HG6-64-1. In vivo treatment with HG6-64-1, via intratumoral and intraperitoneal injections, significantly decreased the tumor growth rate resulting in a significantly extended lifespan of DLBCL xenograft mouse models. Overall our results identified a previously unrecognized activation of the GCK pathway which contributes to the proliferation and survival of DLBCLs and can be used as a therapeutic target using novel GCK inhibitors. Disclosures: Patricelli: ActivX Biosciences: Employment. Nomanbhoy:ActivX Biosciences: Employment.


2021 ◽  
Vol 22 (23) ◽  
pp. 12673
Author(s):  
Weibo Kong ◽  
Sina Sender ◽  
Leila Taher ◽  
Simon Villa-Perez ◽  
Yixuan Ma ◽  
...  

Bruton’s tyrosine kinase (BTK) and phosphoinositide 3-kinase (PI3K) in the B-cell receptor (BCR) signaling pathway are considered potential therapeutic targets for the treatment of B-cell lymphomas, among which, diffuse large B-cell lymphoma (DLBCL) is the most common type. Herein, we comparatively evaluated the single and combined application of the BTK inhibitor ibrutinib and the selective PI3Kγ inhibitor AS-605240 in the canine DLBCL cell line CLBL-1. For further comparison, key findings were additionally analyzed in canine B-cell leukemia GL-1 and human DLBCL cell line SU-DHL-4. While ibrutinib alone induced significant anti-proliferative effects on all cell lines in a dose-dependent manner, AS-605240 only induced anti-proliferative effects at high concentrations. Interestingly, ibrutinib and AS-605240 acted synergistically, reducing cell proliferation and increasing apoptosis/necrosis in all cell lines and inducing morphological changes in CLBL-1. Moreover, the combined application of ibrutinib and AS-605240 reduced relative phosphorylation and, in some instances, the levels of the BTK, AKT, GSK3β, and ERK proteins. Comparative variant analysis of RNA-seq data among canine B- and T-lymphoid cell lines and primary B-cell lymphoma samples revealed potentially high-impact somatic variants in the genes that encode PI3K, which may explain why AS-605240 does not singly inhibit the proliferation of cell lines. The combination of ibrutinib and AS-605240 represents a promising approach that warrants further in vivo evaluation in dogs, potentially bearing significant value for the treatment of human DLBCL.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2717-2717
Author(s):  
Lan V Pham ◽  
Alex Rollo ◽  
Archito T. Tamayo ◽  
John Lee ◽  
Zhuang Zuo ◽  
...  

Abstract Abstract 2717 The biological significance of the ubiquitin-proteasome system in the control of cellular processes has been well-recognized; however, the pathophysiological importance of the immunoproteasome, the inducible form of the proteasome, has not been well-appreciated in cancer cells, particularly in this common diffuse large B-cell lymphoma (DLBCL), a clinically challenging, aggressive B-cell non-Hodgkin's lymphoma (NHL-B). The primary function of the immunoproteasome was originally believed to be only in immune cells to improve MHC-I antigen presentation efficiency in adaptive immune responses. It has now becomes evident that the immunoproteasome possesses broader biological functions, and is associated with various types of cancer. Using the Oncomine database to analyze the mRNA expression levels of immunoproteasome subunits in DLBCL, we found that the subunits MECL-1 and LMP-2 are highly expressed in comparison to normal B lymphocytes. We then examined the clinical significance of MECL-1 and LMP-2 mRNA expression in primary DLBCL, and found that increased MECL-1 mRNA expression is significantly associated with decreased cumulative overall survival rate (P=0.019). We then analyzed the protein expressions of MECL-1 and LMP-2 in various (20) DLBCL cell lines, and discovered that most of the DLBCL cell lines highly expressed both MECL-1 and LMP-2 but there is a subset of cell lines that did not express MECL-1 and LMP-2. Further analysis indicated that MECL-1 and LMP-2 subunits of the immunoproteasome are not associated with constitutive NF-kB activation in DLBCL since MECL-1 and LMP-2-negative DLBCL cell lines also express constitutive NF-kB activation. RNA interference-mediated knock-down of MECL-1 or LMP-2 leads to cell growth inhibition in DLBCL cell lines in vitro. These results strongly suggest that the immunoproteasome has important biological function in controlling growth and survival mechanisms in DLBCL and thus selective targeting of the immunoproteasome may offer therapeutic opportunity for this deadly disease. Bortezomib (BZ) is the first in the class of proteasome inhibitor (PSI) and represents a major advance in NHL, particularly mantle cell lymphoma. However, with the emergence of a new class of PSIs, such as Carfilzomib (CFZ), we are presented with opportunities to improve patient care in relapsed/refractory NHL-B. To elucidate the role of proteasome inhibitors in DLBCL, we analyzed the effect of BZ and CFZ in our representative DLBCL cell lines. BZ and CFZ treatments in DLBCL cell lines (20) have shown strong responses, with IC50s in the low nM ranges (2–50 nM). We have shown that DLBCL cell lines lacking both MECL-1 and LMP-2 are more resistant to CFZ than DLBCL cell lines that have both MECL-1 and LMP-2. To investigate a potential CFZ resistance mechanism(s) in these cell lines, we measured the 20S proteasome activity and compared this activity to the CFZ sensitive DLBCL cell lines. The results indicated that DLBCL cells that are more sensitive to CFZ show higher immunoproteasomal activity. The immunoproteasome activity in the resistant DLBCL cell lines is comparable to the proteasome activity found in normal B cells. These results suggest that the immunoproteasome is deregulated in DLBCL and represents a potential target for therapy in personalized medicine. Our studies emphasize understanding the mechanisms responsible for abnormal proteasomal function in DLBCL, that are critical for establishing an etiologic link to chemo-resistance and the development of new specific therapies for DLBCL targeting defective proteolysis through the immunoproteasome. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2003 ◽  
Vol 101 (7) ◽  
pp. 2756-2761 ◽  
Author(s):  
Christiane Copie-Bergman ◽  
Marie-Laure Boulland ◽  
Catherine Dehoulle ◽  
Peter Möller ◽  
Jean-Pierre Farcet ◽  
...  

The molecular markers that distinguish primary mediastinal large B-cell lymphoma (PMBL) from nonmediastinal diffuse large B-cell lymphomas (NM-DLBLs) remain to be identified. Using cDNA representational difference analysis to compare PMBL and NM-DLBL transcripts, we isolated a cDNA fragment homologous to the mouse B-cell interleukin 4 (IL-4)–inducible gene FIG1(interleukin 4–induced gene 1) transcript. The human FIG1mRNA encodes a 567 amino acid protein that comprises a signal peptide and a large flavin-binding amino oxidase domain, and shares significant homology with secreted apoptosis-inducing L-amino acid oxidases. Northern blot studies showed that FIG1 mRNA expression is mainly restricted to lymphoid tissues. It is expressed at low levels in thymus, spleen, tonsils, and reactive lymph nodes, and is highly up-regulated in IL-4+CD40–activated tonsillar B cells. Interestingly, in human B-cell lines, FIG1 mRNA expression appeared restricted to the PMBL-derived MedB-1 and Karpas 1106 cell lines. Using real-time reverse transcriptase–polymerase chain reaction (RT-PCR), we demonstrated that all but one PMBL (16/17) displayed high FIG1 mRNA levels, whereas most NM-DLBLs (12/18) and all low-grade B-cell lymphomas tested (8/8) exhibited low FIG1 mRNA levels. The difference between PMBLs and NM-DLBLs was statistically significant (Fisher test;P = .0003). Southern blot studies did not show rearrangement of the FIG1 gene. FIG1 gene expression might be due to a constitutive activation of a cytokine signaling pathway in PMBL.


Blood ◽  
1990 ◽  
Vol 76 (11) ◽  
pp. 2311-2320 ◽  
Author(s):  
FM Lemoine ◽  
S Dedhar ◽  
GM Lima ◽  
CJ Eaves

Abstract Marrow stromal elements produce as yet uncharacterized soluble growth factors that can stimulate the proliferation of murine pre-B cells, although close contact between these two cell types appears to ensure a better pre-B cell response. We have now shown that freshly isolated normal pre-B cells (ie, the B220+, surface mu- fraction of adult mouse bone marrow) adhere to fibronectin (FN) via an RGD cell-attachment site, as shown in a serum-free adherence assay, and they lose this functional ability on differentiation in vivo into B cells (ie, the B220+, surface mu+ fraction). Similarly, cells from an immortalized but stromal cell-dependent and nontumorigenic murine pre-B cell line originally derived from a Whitlock-Witte culture were also found to adhere to fibronectin (FN) via an RGD cell-attachment site. Moreover, in the presence of anti-FN receptor antibodies, the ability of this immortalized pre-B cell line to proliferate when co-cultured with a supportive stromal cell line (M2–10B4 cells) was markedly reduced (down to 30% of control). This suggests that pre-B cell attachment to FN on stromal cells may be an important component of the mechanism by which stromal cells stimulate normal pre-B cell proliferation and one that is no longer operative to control their more differentiated progeny. Two differently transformed pre-B cell lines, both of which are autocrine, stromal-independent, tumorigenic in vivo, and partially or completely differentiation-arrested at a very early stage of pre-B cell development, did not bind to FN. In addition, anti-FN receptor antibodies were much less effective in diminishing the ability of these tumorigenic pre-B cells to respond to M2–10B4 cell stimulation, which could still be demonstrated when the tumorigenic pre-B cells were co- cultured with M2–10B4 cells at a sufficiently low cell density. Analysis of cell surface molecules immunoprecipitated from both the nontumorigenic and tumorigenic pre-B cell lines by an anti-FN receptor antibody showed an increase in very late antigen (VLA) alpha chain(s) in both tumorigenic pre-B cell lines and a decrease in the beta 1 chain in one. Interestingly, all of the pre-B cell lines expressed similar amounts of messenger RNA for the beta 1 chain of the FN receptor. These results suggest that alteration of FN receptor expression on pre-B cells may represent a mechanism contributing to the outgrowth of leukemic pre-B cells with an autocrine phenotype and capable of stromal cell-independent, autonomous growth.


Sign in / Sign up

Export Citation Format

Share Document