Inhibitors Of Src Family and AKT Regulate The Activity Of CD20 Promoter

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1838-1838
Author(s):  
Beata Pyrzynska ◽  
Kamil Bojarczuk ◽  
Magdalena Winiarska ◽  
Jacek Bil ◽  
Nina Miazek ◽  
...  

Abstract Introduction The monoclonal antibodies against CD20 antigen (rituximab and ofatumumab) have been developed and used in a clinic as a therapeutic strategy in B-cell malignancies. These antibodies eliminate B cells by triggering indirect effector mechanisms of the immune system, namely complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), or immunophagocytosis. Unfortunately, in some patients the resistance to anti-CD20 therapy has been detected. Therefore the molecular mechanisms responsible for the therapy failure should be urgently elucidated. One of the major mechanisms responsible for the resistance to anti-CD20 therapy seems to be the reduced level of CD20 antigen on the surface of tumor B-cells. We have previously discovered that CD20 expression is strictly dependent on the activity of Src family tyrosine kinases and AKT kinase. We have noticed that treatment of B cells with Src family inhibitors or AKT inhibitors leads to a dose-dependent reduction of CD20 at both transcript and protein level. On the other hand the overexpression of constitutively active AKT (CA-AKT) leads to a significant increase in CD20 mRNA level. To uncover the transcriptional mechanisms governing the CD20 expression we employed the construct encoding the promoter region of CD20 cloned upstream of the firefly luciferase gene. The truncated and mutated versions of the CD20 promoter were used in the luciferase assays to elucidate the role of particular transcription factors binding sites in the regulation of CD20 expression. Objectives The aim of this study was to explore the molecular mechanisms governing the transcriptional regulation of CD20 expression in lymphoma cells as a potential explanation of the resistance to anti-CD20 therapy. Results In the initial experiments we observed a significant reduction of CD20 protein level in Raji cells treated with Src family tyrosine kinase inhibitor, dasatinib (Fig. 1A,B). This reduction correlated with the impaired binding of anti-CD20 monoclonal antibodies as estimated by FACS analysis. Quantitative PCR analysis revealed that the transcriptional regulation is the major mechanism responsible for the reduction of CD20 level upon dasatinib treatment (Fig. 1C). Consistently, the exogenously expressed CD20 under the control of CMV promoter was not sensitive to dasatinib treatment. To further elucidate the mechanism of transcriptional regulation of CD20 we performed the luciferase assays to estimate the activity of CD20 promoter and its truncated forms (Fig. 1D). Dasatinib or AKT kinase inhibitor (MK-2206) strongly decreased the activity of CD20 promoter (Fig. 1E,F) while the overexpression of CA-AKT partially blocked the inhibition caused by dasatinib (Fig. 1G). Using the truncated versions of the CD20 promoter we found that lack of the region (-313/-198) made the promoter insensitive to dasatinib treatment. Since this particular region is known to contain a putative Octamer transcription factor binding site (BAT-box, Thevenin et al., 1993), we introduced mutations in the BAT-box sequence. Although basal promoter activity was indeed decreased (Fig. 1H), dasatinib was equally effective in reducing the activity of both wild-type and mutated CD20 promoter. Collectively, our results indicate that Octamer transcription factor is an important regulator of basal CD20 expression, but it is not the major mediator of the effects caused by Src family inhibitors. Conclusions Our studies indicate that the Src family tyrosine kinases and AKT kinase are involved in the transcriptional regulation of CD20 antigen in lymphoma cells. The activity of CD20 promoter is significantly reduced upon treatment with Src family inhibitors, namely dasatinib. The particular region of CD20 promoter (-313/-198) was identified as the major region sensitive to dasatinib treatment. The transcriptional machinery responsible for the reduction of CD20 expression by dasatinib needs further investigation since the expected Octamer transcription factor does not mediate the effects caused by dasatinib. Disclosures: No relevant conflicts of interest to declare.

2013 ◽  
Vol 2013 ◽  
pp. 1-8
Author(s):  
Xiong-Fei Zhang ◽  
Yi Zhu ◽  
Wen-Biao Liang ◽  
Jing-Jing Zhang

Cyclooxygenase-2 (COX-2) expression is associated with many aspects of physiological and pathological conditions, including pancreaticβ-cell dysfunction. Prostaglandin E2 (PGE2) production, as a consequence of COX-2 gene induction, has been reported to impairβ-cell function. The molecular mechanisms involved in the regulation of COX-2 gene expression are not fully understood. We previously demonstrated that transcription factor Elk-1 significantly upregulated COX-2 gene promoter activity. In this report, we used pancreaticβ-cell line (INS-1) to explore the relationships between Elk-1 and COX-2. We first investigated the effects of Elk-1 on COX-2 transcriptional regulation and expression in INS-1 cells. We thus undertook to study the binding of Elk-1 to its putative binding sites in the COX-2 promoter. We also analysed glucose-stimulated insulin secretion (GSIS) in INS-1 cells that overexpressed Elk-1. Our results demonstrate that Elk-1 efficiently upregulates COX-2 expression at least partly through directly binding to the −82/−69 region of COX-2 promoter. Overexpression of Elk-1 inhibits GSIS in INS-1 cells. These findings will be helpful for better understanding the transcriptional regulation of COX-2 in pancreaticβ-cell. Moreover, Elk-1, the transcriptional regulator of COX-2 expression, will be a potential target for the prevention ofβ-cell dysfunction mediated by PGE2.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1661-1661
Author(s):  
Magdalena Winiarska ◽  
Jacek Bil ◽  
Kamil Bojarczuk ◽  
Dominika Nowis ◽  
Malgorzata Wanczyk ◽  
...  

Abstract Abstract 1661 Introduction: Anti-CD20 monoclonal antibodies (mAbs) have considerably improved the outcomes of patients with B-cell malignancies and reveal promising therapeutic activity in some autoimmune diseases. They eliminate B cells by triggering indirect effector mechanisms of the immune system, namely complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), or immunophagocytosis. Unfortunately, the resistance to anti-CD20 mAb-based first line therapies has been a considerable clinical problem. The mechanisms of this resistance are still poorly understood. In an elegant in vitro study by van Meerten et al. a direct positive correlation between rituximab antitumor activity and CD20 levels has been demonstrated. Although for many years CD20 has been described as a stable antigen, accumulating evidence indicates that CD20 can be modulated at several levels, both transcriptional and posttranscriptional. The processes that lead to CD20 downregulation could potentially impair antitumor activity of rituximab-based therapies and lead to rituximab resistance. Src family tyrosine kinases (SFTKs) including Lyn, Fyn and Lck have been already reported by Deans et al. to associate with CD20 localized to lipid rafts. They were shown to be activated during anti-CD20 mAb-mediated apoptosis upon clustering of rafts. However, to the best of our knowledge, the role of SFTKs in the regulation of CD20 expression has not been studied so far. Objectives: The aim of this study was to explore the molecular basis for Src family tyrosine kinases- dependent regulation of CD20 levels in lymphoma cells. Results: In the initial experiments performed using flow cytometry we observed a significantly reduced binding of anti-CD20 mAb to Raji cells incubated for 48h with various inhibitors of Src kinases (dasatinib, PP2, nilotynib, bosutinib, saracatinib) (Fig.1A-E). Dasatinib also impaired the binding of rituximab to Raji cells (Fig.1F). Decreased binding of anti-CD20 mAb upon dasatinib treatment was observed in three additional lymphoma cell lines and primary cells isolated from patients with chronic lymphocytic leukemia. All tested SFTKs inhibitors impaired rituximab-mediated CDC (R-CDC) over a dose range of rituximab concentrations (1–100 ug/ml) in all lymphoma cell lines. Interestingly, in Raji cells incubated for 48h with dasatinib we also observed a dose-dependent reduction of total CD20 protein levels, when assayed by Western blotting (Fig.2A). Moreover, a 48-h incubation with dasatinib significantly reduced the transcription of cd20 gene, as assessed with RT-PCR (Fig.2B). To further elucidate the mechanism of transcriptional regulation of CD20 we performed qRT-PCR studies. A strongly reduced transcription of cd20 gene was observed in Raji cells over a dose range of dasatinib (20–200 nM) after 24- (Fig.2C) and 48h- incubation (Fig.2D). Additionally, the CD20 promoter activity measured with reporter Firefly luciferase assay has been reduced as early as 1 hour after dasatinib treatment (Fig.2E). To elucidate in more detail binding of transcription factors to the promoter of cd20 gene, a chromatin immunoprecipitation assay was performed. Our early results indicate that dasatinib impairs binding of PU.1 transcription factor to its consensus site within cd20 promoter in Raji cells. Conclusions: Our studies indicate for the first time that SFTKs are involved in the transcriptional regulation of CD20 levels in lymphoma cells. Elucidation of the exact mechanism of this phenomena needs further studies. Results of these experiments will help to understand the biology and regulation of CD20 levels in lymphoma cells. The research was supported by Polish Ministry of Science and Higher Education [N N402 352938 (MW), IP2010/046570 (MW), IP2010/028670 (DN)]. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4493-4493 ◽  
Author(s):  
Keisuke Watanabe ◽  
Seitaro Terakura ◽  
Tatsunori Goto ◽  
Ryo Hanajiri ◽  
Nobuhiko Imahashi ◽  
...  

Background In chimeric antigen receptor transduced T-cell (CAR-T cell) therapy, setting of the target antigen is critical in terms of both the efficacy and the possible adverse effects. However, how low expressing antigen CAR-T cells can recognize and present cytotoxicity, and how low expressing antigen would be a candidate for the target or should be avoided for the concern of off-organ effect is still unclear. Thus we developed novel anti-CD20 CAR-T cells, and investigated the threshold antigen expression level required to activate CAR-T cells. Methods In this study, we generated a retrovirus vector that encodes a novel CAR consisting of anti-CD20 single chain variable fragments linked to a CD3-zeta chain, a CD28 costimulatory domain, and a truncated epidermal growth factor receptor (tEGFR) as a transduction and selection marker. CD3 positive T cells from a healthy donor were activated with anti-CD3/28 beads, transduced with the CAR on day 3, enriched by selection with anti-EGFR mAb, and expanded by stimulation with gamma-irradiated B-lymphoblastoid cells. CD20 expression level of the target cells was evaluated with CD20 mean fluorescence intensity (CD20-MFI) and was quantified as CD20 specific antibody biding capacity (CD20-ABC). Results To determine the threshold expression level of CD20 antigen to induce cytotoxicity, we performed 51Cr release assay by anti-CD20 CAR-T cells against 30 clones of CD20 transduced CCRF-CEM (CD20-CEM) expressing variable levels of CD20 (CD20-MFI: 126-6,924/ CD20-ABC: 240-230,546 molecules) (kindly provided by Dr. A.C.M. Martens, University Medical Center Utrecht). CAR-T cells lysed CD20-CEMs equally well from low to high level of CD20 (CD20MFI: 157/ CD20ABC: 5,172 molecules or more, 40-60% of lysis at E:T ratio of 10:1), and lysed the clone expressing the lowest level of CD20 (126/ 240 molecules, 22.8±2% of lysis). However, in rituximab-induced CDC assay against the same 30 clones, a minimum CD20 MFI of 600 (ABC of 65,000 molecule) was required to induce CDC. Next, cytokine production and proliferation upon stimulation were investigated by using four representative CD20-CEM clones out of the 30 clones that cover a wide range of CD20 expression levels; Very low (VL-CEM) (MFI: 126/ ABC:240 molecules), Low (L-CEM)(252/ 26,990 molecules), Mid (M-CEM)(826/ 91,567 molecules), High (H-CEM)(6,924/ 142,722 molecules). In intracellular IFN-gamma assay, CAR-T cells produced IFN-gamma equally well (L-CEM, 45.9%, M-CEM, 35.4% and H-CEM, 38.0%, respectively), except against VL-CEM (0.2%). In CFSE division assay, CAR-T cells proliferated upon stimulation with L-CEM, M-CEM, H-CEM (31.4%, %36.1, 37.3% at 72h and, 88.9%, 90.2%, 91.1% at 96h), except with LV-CEM (0% at 72h/ 96h). In intracellular signaling assay, phosphorylation of downstream signaling molecule, CD3-zeta and ERK were evaluated. Whereas only minimal phosphorylations of CD3-zeta and ERK were observed upon VL-CEM stimulation, other 3 CEMs induced similar strength of signals in the majority of CAR-T cells, and induced phosphorylation of proximal signaling molecule, CD3-zeta according to the CD20 expression level. Finally, we evaluated cytotoxicity against low-CD20 expressing primary tumor cells and cell lines. CAR-T cells lysed RRBL1 which is a cell line established from a patient who exhibited relapse of B-cell lymphoma with very weak CD20 expression and became resistant to rituximab (47.8±3% of lysis at E:T ratio 10:1) and primary DLBCL cells isolated from pleural effusion with low CD20 expression (32.6±2.9% of lysis). Conclusions We observed that CAR-T cells recognize CD20 antigen with considerably low expression. A minimum number of antigen molecules required to activate CAR-T cells is very low; the threshold is a few hundreds of target antigen. For the future search of a novel target of CAR-T therapy, antigens with expression above the threshold, even if that is below the range of mAb therapy, could be also considered. Conversely, antigens with off-organ expression above the threshold may induce off-organ effect, thus should be avoided. CD20-CAR therapy may be also effective for the patients with CD20 down-regulated lymphoma and refractory to CD20 mAb therapy. Disclosures: No relevant conflicts of interest to declare.


2000 ◽  
Vol 20 (24) ◽  
pp. 9138-9148 ◽  
Author(s):  
Pascale F. Dijkers ◽  
Rene H. Medema ◽  
Cornelieke Pals ◽  
Lolita Banerji ◽  
N. Shaun B. Thomas ◽  
...  

ABSTRACT Interleukin-3 (IL-3), IL-5, and granulocyte-macrophage colony-stimulating factor regulate the survival, proliferation, and differentiation of hematopoietic lineages. Phosphatidylinositol 3-kinase (PI3K) has been implicated in the regulation of these processes. Here we investigate the molecular mechanism by which PI3K regulates cytokine-mediated proliferation and survival in the murine pre-B-cell line Ba/F3. IL-3 was found to repress the expression of the cyclin-dependent kinase inhibitor p27KIP1 through activation of PI3K, and this occurs at the level of transcription. This transcriptional regulation occurs through modulation of the forkhead transcription factor FKHR-L1, and IL-3 inhibited FKHR-L1 activity in a PI3K-dependent manner. We have generated Ba/F3 cell lines expressing a tamoxifen-inducible active FKHR-L1 mutant [FKHR-L1(A3):ER*]. Tamoxifen-mediated activation of FKHR-L1(A3):ER* resulted in a striking increase in p27KIP1 promoter activity and mRNA and protein levels as well as induction of the apoptotic program. The level of p27KIP1 appears to be critical in the regulation of cell survival since mere ectopic expression of p27KIP1 was sufficient to induce Ba/F3 apoptosis. Moreover, cell survival was increased in cytokine-starved bone marrow-derived stem cells from p27KIP1 null-mutant mice compared to that in cells from wild-type mice. Taken together, these observations indicate that inhibition of p27KIP1transcription through PI3K-induced FKHR-L1 phosphorylation provides a novel mechanism of regulating cytokine-mediated survival and proliferation.


Blood ◽  
2012 ◽  
Vol 119 (18) ◽  
pp. 4174-4181 ◽  
Author(s):  
Hai Vu Nguyen ◽  
Enguerran Mouly ◽  
Karine Chemin ◽  
Romain Luinaud ◽  
Raymonde Despres ◽  
...  

Abstract In response to antigens and cytokines, mouse B cells undergo class-switch recombination (CSR) and differentiate into Ig-secreting cells. T-bet, a T-box transcription factor that is up-regulated in lymphocytes by IFN-γ or IL-27, was shown to regulate CSR to IgG2a after T cell–independent B-cell stimulations. However, the molecular mechanisms controlling this process remain unclear. In the present study, we show that inactivation of the Ets-1 transcription factor results in a severe decrease in IgG2a secretion in vivo and in vitro. No T-bet expression was observed in Ets-1–deficient (Ets-1−/−) B cells stimulated with IFN-γ and lipopolysaccharide, and forced expression of T-bet in these cells rescued IgG2a secretion. Furthermore, we identified a transcriptional enhancer in the T-bet locus with an activity in B cells that relies on ETS-binding sites. After IFN-γ stimulation of Ets-1−/− B cells, activated Stat1, which forms a complex with Ets-1 in wild-type cells, no longer binds to the T-bet enhancer or promotes histone modifications at this site. These results demonstrate that Ets-1 is critical for IgG2a CSR and acts as an essential cofactor for Stat1 in the regulation of T-bet expression in B cells.


2012 ◽  
Vol 25 (5) ◽  
pp. 684-696 ◽  
Author(s):  
Hirohisa Saga ◽  
Takumi Ogawa ◽  
Kosuke Kai ◽  
Hideyuki Suzuki ◽  
Yoshiyuki Ogata ◽  
...  

Camalexin is the major phytoalexin in Arabidopsis. An almost complete set of camalexin biosynthetic enzymes have been elucidated but only limited information is available regarding molecular mechanisms regulating camalexin biosynthesis. Here, we demonstrate that ANAC042, a member of the NAM, ATAF1/2, and CUC2 (NAC) transcription factor family genes, is involved in camalexin biosynthesis induction. T-DNA insertion mutants of ANAC042 failed to accumulate camalexin at the levels achieved in the wild type, and were highly susceptible to Alternaria brassicicola infection. The camalexin biosynthetic genes CYP71A12, CYP71A13, and CYP71B15/PAD3 were not fully induced in the mutants, indicating that the camalexin defects were at least partly a result of reduced expression levels of these P450 genes. β-Glucuronidase (GUS)-reporter assays demonstrated tissue-specific induction of ANAC042 in response to differential pathogen infections. Bacterial flagellin (Flg22) induced ANAC042 expression in the root-elongation zone, the camalexin biosynthetic site, and the induction was abolished in the presence of either a general kinase inhibitor (K252a), a Ca2+-chelator (BAPTA), or methyl jasmonate. The GUS-reporter assay revealed repression of the Flg22-dependent ANAC042 expression in the ethylene-insensitive ein2-1 background but not in sid2-2 plants defective for salicylic acid biosynthesis. We discuss ANAC042 as a key transcription factor involved in previously unknown regulatory mechanisms to induce phytoalexin biosynthesis in Arabidopsis.


2016 ◽  
Vol 77 ◽  
pp. 138
Author(s):  
Elaine C. Bellintani ◽  
Renato de Marco ◽  
Renata Fantini ◽  
Tiago Valim ◽  
Denise Macedo ◽  
...  

2020 ◽  
Vol 54 (6) ◽  
pp. 1177-1198

BACKGROUND/AIMS: Compelling evidence indicates that CK2α, which is one of the two catalytic isoforms of protein kinase CK2, is required for cell viability and plays an important role in cell proliferation and differentiation. While much is known on CK2 in the context of disease states, particularly cancer, its critical role in non-cancerous cell growth has not been extensively investigated. METHODS: In the present study, we have employed a cell line derived from rat heart with inducible down-regulation of CK2α and CK2α-knockout mouse tissue to identify CK2-mediated molecular mechanisms regulating cell growth. For this, we have performed Incucyte® live-cell analysis and applied flow cytometry, western blot, immunoprecipitation, immunohistochemistry, RT-qPCR and luciferase-based methods. RESULTS: Here, we show that lack of CK2α results in significantly delayed cell cycle progression through G1, inhibition of cyclin E-CDK2 complex, decreased phosphorylation of Rb protein at S795, and inactivation of E2F transcription factor. These events are accompanied by nuclear accumulation and up-regulation of the cyclin-dependent kinase inhibitor p27KIP1 in cells and CK2α-knockout mouse tissues. We found that increased levels of p27KIP1 are mainly attributable to post-translational modifications, namely phosphorylation at S10 and T197 amino acid residues catalyzed by Dyrk1B and AMPK, respectively, as silencing of FoxO3A transcription factor, which activates CDKN1B the gene coding for p27KIP1, does not result in markedly decreased expression levels of the corresponding protein. Interestingly, simultaneous silencing of CK2α and p27KIP1 significantly impairs cell cycle progression without increasing cell death. CONCLUSION: Taken together, our study sheds light on the molecular mechanisms controlling cell cycle progression through G1 phase when myoblasts proliferation potential is impaired by CK2α depletion. Our results suggest that elevated levels of p27KIP1,which follows CK2α depletion, contribute to delay the G1-to-S phase transition. Effects seen when p27KIP1 is down-regulated are independent of CK2α and reflect the protective role exerted by p27KIP1 under unfavorable cell growth conditions.


Sign in / Sign up

Export Citation Format

Share Document