Mantle Cell Lymphoma Cells Express B7 Family Molecules and B7-H1 Expression Are up-Regulated After LPS Exposure Via MEK-Dependent and PI3K/Akt-Dependent Pathways In Mantle Cell Lymphoma Cells

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3114-3114
Author(s):  
Lijuan Wang ◽  
Hanying Bao ◽  
Yang Yang ◽  
Yi Zhao ◽  
Donghua He ◽  
...  

Abstract Abstract 3114 Mantle cell lymphoma (MCL) is a distinct subtype of B-cell non-Hodgkin lymphomas characterized by a specific t(11;14) (q13;q32) translocation, causing over-expression of cyclin D1. Recent studies demonstrated that B7 family molecules were not only expressed on antigen presenting cells but also on various hematopoietic malignancies, solid tumors and infiltrating immune cells and may play important roles in tumor immunology. Many cytokines could upregulate the expression of B7 molecules, however, the molecular mechanism of regulating expressions of B7 molecules is still unknown. In this study, we analyzed B7 family molecule expression on cultured mantle cell lymphoma cells and primary MCL cells by RT-PCR. We confirmed that B7 family molecules were detected in mantle cell lymphoma cell lines SP53, Jeko-1 and Mino and primary MCL cells. We next used LPS to activate TLR4 signaling pathway. Although B7 family molecules were detected in mantle cell lymphoma cells, only B7-H1 expression was greatly enhanced after LPS exposure. B7-H1 mRNA was constitutively expressed on MCL cells and was further up-regulated after LPS stimulation in dose and time dependent manners. Western blot also indicated that total and surface B7-H1 protein expression were up-regulated after stimulated by LPS. Flow cytometry demonstrated that surface B7-H1 protein expression were up-regulated after stimulated by LPS. When we knockdown TLR4, LPS stimulation did not up-regulate B7-H1 expression. To clarify the signaling pathways of LPS induced B7-H1 up-regulation in MCL cells, we incubated the SP53 and Jeko-1 cells with LPS after pretreatment with different signal transduction pathway inhibitors for 1h and detect the activity of each inhibitor. Pretreatment and coincubation of SP53 and Jeko-1 cells with the MEK1/2 inhibitor UO126 and PI3K/Akt inhibitor LY294002 strongly reduced LPS induced B7-H1 expression, indicating that the MEK1/2 and PI3K/Akt pathway are crucial for B7-H1 expression in MCL cells. We also tested whether JNK or p38, respectively, are involved in controlling the expression of B7-H1 in MCL cells. Blocking of JNK and p38 MAPK with specific inhibitors reduced B7-H1 mRNA expression, but B7-H1 protein expression was not obvious, which may be regulated after transcriptional factors. To confirm that LPS induced B7-H1 expression through a MEK and PI3K/Akt pathway pathways in MCL cells, we stimulated SP53 and Jeko-1 with LPS and analyzed the phosphorylation of ERK1/2, p38, and JNK at different time. ERK1/2, p38, and JNK phosphorylation were significantly up-regulated following LPS treatment. We confirmed that pretreatment of the cells with these specific inhibitors inhibited LPS-induced phosphorylation of ERK1/2, p38 MAPK, and JNK, respectively. In conclusion, our study demonstrated that mantle cell lymphoma cells express B7 family molecules. B7-H1 expression were up-regulated after LPS exposure via MEK-dependent and PI3K/Akt-dependent in mantle cell lymphoma cells. Disclosures: No relevant conflicts of interest to declare.

Haematologica ◽  
2015 ◽  
Vol 100 (11) ◽  
pp. e458-e461 ◽  
Author(s):  
M. Rudelius ◽  
H. Rauert-Wunderlich ◽  
E. Hartmann ◽  
E. Hoster ◽  
M. Dreyling ◽  
...  

2014 ◽  
Vol 142 (suppl_1) ◽  
pp. A099-A099
Author(s):  
Jindong Wang ◽  
Viren Patel ◽  
Siddharth Bhattacharyya ◽  
Ashish Bains

Leukemia ◽  
2019 ◽  
Vol 33 (7) ◽  
pp. 1675-1686 ◽  
Author(s):  
Pratikkumar H. Vekaria ◽  
Amar Kumar ◽  
Dharmalingam Subramaniam ◽  
Neil Dunavin ◽  
Anusha Vallurupalli ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (13) ◽  
pp. 1565-1574 ◽  
Author(s):  
Baohua Sun ◽  
Bhavin Shah ◽  
Warren Fiskus ◽  
Jun Qi ◽  
Kimal Rajapakshe ◽  
...  

Key Points BA reduces MYC, CDK4/6, nuclear RelA, and BTK expression and is synergistically lethal with ibrutinib in MCL cells. Cotreatment with BA and inhibitor of BCL2, CDK4/6, or histone deacetylases is synergistically lethal against ibrutinib-resistant MCL cells.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3184-3184
Author(s):  
Robert W. Chen ◽  
Lynne Bemis ◽  
Carol Amato ◽  
Birks Diane ◽  
Myint Han ◽  
...  

Abstract Mantle Cell Lymphoma (MCL) represents only 5–10% of all non-Hodgkins lymphomas, making it an uncommon but difficult form of lymphoma to treat. It has a poor prognosis among the B cell lymphomas with median survival of three years. The genetic hallmark of MCL is the t(11,14) translocation causing amplification of cyclin D1 (CCND1), a known cell cycle regulator which is overexpressed in many other cancers. MicroRNAs (miRNA) are a new class of abundant small RNAs that play important regulatory roles at the post transcriptional level. They act by binding to the 3′ untranslated region (UTR) of mRNAs and block either their translation or initiate their degradation. Recent reports have shown truncations in the CCND1 3′ UTR occur in MCL and indicate a worse prognosis. We hypothesized that truncations in 3′ UTR of CCND1 alter it’s regulation by microRNAs. Based on bioinformatics, we identified microRNA 16 with putative docking sites in the 3′UTR of CCND1. Mir-16 has been implicated as a cell cycle regulator. We identified 2 cell lines (Jeko-1 and Z138) with truncations in CCND1 3′ UTR and demonstrated increased CCND1 mRNA expression by qRT-PCR, increased protein expression by western blot, and higher proliferative potential by cell cycle. We prepared a reporter construct by ligating the full length 3′ UTR of CCND1 to GFP. We then co-transfected this construct with mimics of mir-16 into a cancer cell line and demonstrated downregulation of CCND1 protein expression by flow cytometry. In the MCL cell line Granta-519 with non-truncated CCND1, transfection with mimics of mir-16 deminstrated decreased expression of CCND1 mRNA. These studies suggest that the overexpression of CCDN1 In MCL may result from altered regulation of gene expression from loss of a miRNA regulatory site and may give new clues into the patho-biology of this disease and insights into possible new therapies.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1692-1692 ◽  
Author(s):  
Daniela Buglio ◽  
Sangeetha Palakurthi ◽  
Katharine F. Byth ◽  
Anas Younes

Abstract Abstract 1692 Poster Board I-718 Transforming growth factor-b-activated kinase 1 (TAK1) is a key regulator of NF-kB activation. TAK1 can be activated by a variety of pro-inflammatory cytokines and T and B cell receptors. Recent experiments demonstrated that deletion of TAK1 results in inactivation of both JNK and NF-kB signaling resulting in massive apoptotic death of hematopoietic cells in mice. In this study, we examined the expression pattern of TAK1 and its role as a potential therapeutic target for lymphoma. First, we examined TAK1 expression in a panel of lymphoid cell lines by western blot, and found it to be highly expressed in mantle cell lymphoma cell lines (Mino, SP53, and Jeko-1). These lines expressed relatively low levels of the tumor suppressor protein A20. Mino and SP53 expressed high level of p-p38. Subsequently, we investigated the in vitro activity of the novel TAK1 small molecule inhibitor AZ-Tak1 in these cell lines. AZ-Tak1 is a potent and a relatively selective inhibitor of TAK1 kinase activity, with an IC50 of 0.009 mM. It also inhibits Jak2 but at a much higher concentration (IC50=0.18 mM). AZ-Tak1 treatment decreased the level of p38 and ERK in mantle cell lymphoma cells, and induced apoptosis in a dose and time dependent manner, with an IC50 of 0.1-0.5 mM. Using the annexin-V and PI staining and FACS analysis, After 48 hours of incubation, AZ-Tak1 (0.1 mM) induced apoptosis in 28%, 34% and 86% of Mino, SP53, and Jeko cells, respectively, which was increased to 32%, 42%, and 86% when 0.5 mM concentration was used. Similar activity was also observed when primary mantle cell lymphoma cells were examined. Using pathway-specific protein arrays focusing on apoptosis, kinases, and transcription factors, AZ-Tak1 (0.5 mM) altered the level of several proteins that regulate cell growth and survival, especially members of the inhibitors of apoptosis (IAP) family. Specifically, AZ-Tak1 decreased the level of SMAC/DIABOLO and cytochrome –C in the mitochondria, which was associated with a decrease in the level of the anti-apoptotic protein X-linked IAP (XIAP) and activation of the intrinsic apoptotic pathway as evident by activation of caspase 9, cleavage of caspase 3, and induction of apoptosis. Furthermore, and consistant with its ability to inhibit Jak2 activity, AZ-Tak1 reduced STAT2 and STAT6 levels. AZ-Tak1 demonstrated no significant effect on bcl-2 family members. Finally, co-treatment with HDAC inhibitors demonstrated synergistic effect with low concentrations of AZ-Tak1. Collectively, our data demonstrate that targeting TAK1 by the small molecule inhibitor AZ-Tak1 induces cell death in mantle cell lymphoma by activating the intrinsic apoptosis pathway, suggesting that targeting TAK1 may have a therapeutic value for the treatment of mantle cell lymphoma. Disclosures Palakurthi: Astra Zeneca: Employment. Byth:Astra Zeneca : Employment.


2011 ◽  
Vol 35 (3) ◽  
pp. 380-386 ◽  
Author(s):  
Zhengzi Qian ◽  
Liang Zhang ◽  
Zhen Cai ◽  
Luhong Sun ◽  
Huaqing Wang ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document