scholarly journals Targeting Glycosylated Ptgds Displays Anti-Tumor Activities in Diffuse Large B-Cell Lymphoma through Down-Regulation of Wnt Pathway

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 13-15
Author(s):  
Shunfeng Hu ◽  
Xiangxiang Zhou ◽  
Juan Yang ◽  
Shuai Ren ◽  
Yiqing Cai ◽  
...  

Introduction: Prostaglandin D2 Synthase (PTGDS), a member of lipocalin superfamily, plays a dual role in catalyzing the conversion of PGH2 to PGD2 and transporting lipophilic substances. PTGDS protein is with different degrees of glycosylation. AT56 is a selective, competitive, and highly bioavailable inhibitor of PTGDS. However, the function and mechanism of PTGDS in diffuse large B-cell lymphoma (DLBCL) remains ill defined. Herein, we aimed to investigate the functional significance of PTGDS and proposed a novel therapeutic strategy for DLBCL. Methods: Lymph node biopsies from 53 de novo DLBCL patients and 28 reactive hyperplasia cases, and peripheral blood mononuclear cells (PBMCs) from healthy volunteers were collected with informed consents. CD19+ B cells were purified by CD19+ magnetic microbeads. The expression levels of PTGDS in DLBCL cell lines and serum were detected by western blotting and ELISA, respectively. Lentivirus vectors were transfected to stably knockdown or overexpress PTGDS. After the treatment of Tunicamycin (Tun), an N-glycosylation inhibitor, western blotting and immunofluorescence were performed to validate the molecular weight and location of PTGDS protein. Results: We first evaluated the expression of PTGDS in DLBCL. Upregulation of PTGDS mRNA in DLBCL cells was identified based on Oncomine database (Fig.1A). Then, the expression level of PTGDS protein in tumor tissue (n=53) was validated to be higher in comparison with control group (Fig.1B). Furthermore, survival analysis revealed that PTGDS high expression was associated with reduced overall survival of DLBCL patients (Fig.1C). Moreover, high level of soluble PTGDS protein was detected in the serum of DLBCL patients (n=53, Fig.1D). High expression of PTGDS was also confirmed in DLBCL cells by western blotting (Fig.2A). The biological function of PTGDS in progression of DLBCL was further verified. Gene ontology and KEGG analysis revealed that PTGDS was enriched in cellular process and biological regulation (Fig.2B). After the treatment with rhPTGDS, increased proliferation of DLBCL cell was observed in a dose-dependent manner (Fig.2D), and overexpression of PTGDS also promoted cell growth (Fig.2E). Furthermore, knockdown of PTGDS (shPTGDS) significantly restrained cell proliferation (Fig.2F), promoted cell cycle arrest (Fig.2G) and cell apoptosis (Fig.2H). AT56 suppressed the proliferation of DLBCL cells in a dose- and time-dependent manner (Fig. 3A). Additionally, with the treatment of AT56, DLBCL cells exhibited induced G0/G1 phase arrest (Fig. 3B) and increased cell apoptosis (Fig. 3C). As Bendamustine and Adriamycin were found to decrease the mRNA level of PTGDS (Fig. 3D), we further observed that AT56 enhanced sensitivity to them in cell proliferation (Fig. 3E) and cell apoptosis (Fig. 3F). Next, we explored the underlying mechanism of PTGDS in DLBCL progression. The expression of Wnt pathway molecules, such as p-LRP6, β-catenin, p-GSK3-β, Lef-1, p-STAT3, were decreased with treatment of AT56 in dose-dependent manner (Fig. 4A). Besides, STAT3 inhibitor WP1066 was found to restore the proliferation promotion (Fig. 4B) caused by PTGDS overexpression. Moreover, we validated that Wnt pathway activator Wnt3a could restore the phenotype changes caused by AT56, including cell proliferation (Fig. 4C), cell apoptosis (Fig. 4D) and cell cycle (Fig. 4E). Glycosylation, a kind of post-translational modification, could significantly alter protein function and then cellular characteristics. Protein Blast analysis indicated the potential glycosylation sites (Fig. 5A) on PTGDS protein (Ser29, Asn51 and Asn78). Furthermore, Tunicamycin was used to inhibit the N-glycosylation of protein and molecular weight of PTGDS changed from 37kD to 21kD (Fig.5B). Besides, the deglycosylation could promote the translocation of PTGDS into the nucleus (Fig.5C-D), indicating the potential role of glycosylated PTGDS in DLBCL. Conclusions : Our investigations identified for the first time the aberrant overexpression of PTGDS in DLBCL, which was associated with poor prognosis. AT56 exerted anti-tumor effect via inhibiting Wnt pathway. Besides, PTGDS protein in DLBCL cells were highly glycosylated and deglycosylation promoted its translocation into nucleus, indicating the mechanism of PTGDS in DLBCL. Further investigation is warranted to substantiate PTGDS as a promising therapeutic target in DLBCL. Disclosures No relevant conflicts of interest to declare.

Author(s):  
Yan Huang ◽  
Yuanyuan Lin ◽  
Xiangxiang Song ◽  
Depei Wu

Abstract Diffuse large B-cell lymphoma (DLBCL) remains to be a high aggressive and invasive malignancy with enigmatic etiology. Ectopic expression of long noncoding RNAs are widely involved in the progression of human cancers. We discovered that LINC00857 level was remarkably elevated in DLBCL tissues compared with non-tumor controls. High LINC00857 level predicts lower survival rate, more advanced tumor node metastasis and larger tumor size. LINC00857 overexpression promoted DLBCL cell proliferation and facilitated cell cycle as evidenced by elevated cyclinD1 and proliferating cell nuclear antigen (PCNA) accompanying with reduced p21 level. LINC00857 overexpression also suppressed DLBCL cell apoptosis as evidenced by elevated Bcl-2 protein level, reduced Bax and cleaved caspase-3 protein levels. On the contrary, LINC00857 knockdown using short hairpin RNAs inhibited DLBCL cell proliferation yet induced cell apoptosis. LINC00857 knockdown also repressed tumor growth in vivo, concomitant with decreased Ki67 level. Besides, microRNA miR-370 was down-regulated in DLBCL tissues and served as a competitive endogenous RNA (ceRNA) target of LINC00857. We further validated that chromobox homolog 3 (CBX3) served as a downstream target gene of miR-370-3p. LINC00857 level was reversely correlated with miR-370-3p level yet positively correlated with CBX3 level. In addition, CBX3 overexpression alleviated the impact of LINC00857 knockdown on DLBCL cell survival. In conclusion, our findings indicated that LINC00857 contributes to DLBCL proliferation and lymphomagenesis through regulating miR-370-3p/CBX3 axis.


2021 ◽  
Vol 11 ◽  
Author(s):  
Bing Yu ◽  
Bo Wang ◽  
Zhuman Wu ◽  
Chengnian Wu ◽  
Juan Ling ◽  
...  

Long-chain non-coding RNAs (LncRNAs) are expressed in diffuse large B-cell lymphoma (DLBCL) tissues and have played a regulatory role in DLBCL with a cancer-promoting effect. In this study, the role of LncRNA SNHG8 in the regulation of DLBCL cells is investigated, and its underlying mechanism is explored. The database of the Gene Expression Profiling Interactive Analysis (GEPIA) was searched, and the expression of SNHG8 in DLBCL and normal tissues was examined. The expression of SNHG8 was evaluated in several DLBCL cell lines and a normal lymphocyte cell line. It was found that SNHG8 was overexpressed in DLBCL tissues and cells in comparison with their normal counterparts. The short hairpin RNA (shRNA) plasmids of SNHG8 were transfected into DLBCL cells to knockdown the expression of SNHG8, followed by assays of proliferation, colony formation, apoptosis, and related protein expression. The results showed that the knockdown of SNHG8 significantly inhibited DLBCL cell proliferation and colony formation while promoting cell apoptosis. Moreover, the knockdown of SNHG8 reduced the expression of Ki-67, proliferating cell nuclear antigen (PCNA), and Bcl-2 and enhanced the expression of Bax and cleaved caspase 3/9. MiR-335-5p was predicted to be a potential target of SNHG8 by using the bioinformatics analysis, and the interaction between the two was validated by using the dual luciferase assay. In addition, the knockdown of SNHG8 increased the level of miR-335-5p, whereas miR-335-5p mimic decreased the expression of SNHG8. Finally, U2932 cells were co-transfected with or without sh-SNHG8 and miR-335-5p inhibitors, whose proliferation, colony formation, and apoptosis were determined subsequently. It was demonstrated that the presence of an miR-335-5p inhibitor partially canceled the inhibitory effects of the knockdown of SNHG8 on DLBCL cell proliferation and colony formation and the stimulating effects of the knockdown of SNHG8 on cell apoptosis. Taken together, our study suggests that lncRNA SNHG8 exerts a cancer-promoting effect on DLBCL via targeting miR-335-5p.


2020 ◽  
Vol 19 (8) ◽  
pp. 1619-1623
Author(s):  
Zhixiang Su ◽  
Bin Yu ◽  
Zhiping Deng ◽  
Haifeng Sun

Purpose: To investigate the effect of isoliquiritigenin (ISL) on diffuse large B-cell lymphoma (DLBCL) cells and its underlying mechanism of action.Methods: The DLBCL cell line OCI-Ly19 was used in this study. Cell proliferation was measured by MTT assay. Apoptosis was evaluated using flow cytometry. Phosphorylation of Akt and mTOR was assessed using Western blotting.Results: DLBCL cell proliferation was suppressed by ISL in a concentration-dependent manner. The number of apoptotic cells increased following ISL treatment in a concentration-dependent manner (p < 0.05). ISL treatment also stopped the cell cycle at the G1 phase in a concentration-dependent manner. Western blot analysis indicated that there was no significant Akt and mTOR expression in cells treated with 10, 20, or 50 μM ISL (p < 0.05). However, Akt and mTOR phosphorylation was upregulated following treatment with 10, 20, or 50 μM ISL in a concentration-dependent manner (p < 0.05).Conclusion: The results demonstrate that ISL inhibits DLBCL cell proliferation and promotes cell apoptosis by blocking the cell cycle transition from the G1 to S phase, which is mediated by the inactivation of the Akt/mTOR signaling pathway. Keywords: Isoliquiritigenin, Cell survival, Diffuse large B-cell lymphoma, Akt/mTOR signaling pathway


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 603-603
Author(s):  
Mamta Gupta ◽  
Stephen M. Ansell ◽  
Anne J. Novak ◽  
Thomas E. Witzig

Abstract Diffuse large B cell lymphoma (DLBCL) is an aggressive form of non-Hodgkin lymphoma and the treatment of these patients has improved with rituximab-based chemo-immunotherapy. However, approximately 40% of patients die of their disease and new agents with novel mechanisms of action are needed for this disease. The mammalian target of rapamycin (mTOR) has emerged as an important therapeutic target in cancer cells and rapamycin and its derivatives, which specifically inhibit mTOR, are now being actively evaluated. Recent clinical trials of mTOR inhibitors have demonstrated that a fraction of patients (35%) with relapsed DLBCL respond to single-agent temsirolimus (Smith S et al J Clin Oncol 26; May 20 Supplement abstract 8514) or everolimus (Reeder et al 2007 ASH Annual Meeting Abstracts 110 (11) abstract 121) in relapsed DLBCL, however most patients are resistant to this therapy. The mechanism of this resistance remains a subject of major therapeutic significance. Herein, we report that cells from DLBCL lines (DHL-6, Ly7 and Ly3) and primary tissues from DLBCL patients (n=10) display persistent activation of the mTOR pathway as determined by phosphorylation of mTOR targets S6 ribosomal protein (S6rp) and 4E-binding protein 1 (4E-BP1). Treatment of human DLBCL cells with various doses of rapamycin predictably demonstrated a decrease in proliferation but less than 10% reduction in overall cell survival. However, rapamycin suppressed the phosphorylation of S6rp and 4E-BP1, indicating an inhibition of raptor-mTOR (mTORC1) signaling. Paradoxically, rapamycin also concurrently increased, through a negative feedback mechanism, the phosphorylation of Akt that may contribute to drug resistance. Interestingly we found that rapamycin treatment also increased the phosphorylation of eIF4E, a survival protein downstream of mTOR, which may also be responsible for resistance of rapamycin along with Akt. To determine whether the observations found in vitro are clinically relevant, we obtained peripheral blood samples from patients with aggressive lymphoma treated with the rapamycin derivative everolimus. The levels of phosphorylated Akt and eIF4E were increased in 3 of 3 patient samples at 48 hrs and after 1 cycle compared to untreated control. Our observations therefore suggest that rapamycin derivatives potently activate Akt and eIF4E activity via activation of the mTORC2 assembly, in addition to its well-characterized ability to suppress the mTORC1 pathway. Treatment of DLBCL cells with histone deacetylase (HDAC) inhibitor LBH589 induced growth inhibition of DLBCL cells at nanomolar concentrations in a dose dependent manner in association with hyperacetylation of histones H3 and H4. LBH alone was able to inhibit the phosphorylation of S6rp and 4E-BP1, while combined treatment with rapamycin inhibited the phosphorylation of S6rp and 4EBP1 to a greater extent than either agent alone. Surprisingly LBH inhibited constitutive as well as rapamycin-induced activation of Akt and eIF4E in a time and dose dependent manner. Our co-immunoprecipitation data suggest that LBH alone was able to alter the level of intact mTORC2 by reducing the amounts of rictor bound to mTOR, which is further decreased when combined with rapamycin. In support of this model, rapamycin combined with LBH exhibited enhanced synergistic inhibitory effects on survival and proliferation of DLBCL cells. The mTOR pathway is also considered to control the translation of specific mRNA species, some of which are involved in cell cycle control and angiogenesis (e.g. cyclin D1, c-Myc and HIF-1a). LBH alone down-regulated expression of c-Myc and HIF-1a, while have no effect on cyclin D1 expression. Combination of LBH with rapamycin further decreased expression of c-Myc and HIF-1a. Overall these results indicate that pharmacological inhibition of the mTOR pathway by rapamycin and LBH interferes with essential survival and proliferating pathways in DLBCL cells. We propose that the rapamycin-induced functional blockade of Akt and eIF4E is inhibited by LBH, and the combination increased the anti tumor activity of rapamycin. In summary, our data provide a mechanistic basis for enhancing mTORtargeted cancer therapy by combining an mTOR inhibitor with a HDAC inhibitor such as LBH. A phase I/II trial of this combination is planned.


2016 ◽  
Vol 7 (12) ◽  
pp. 899-912 ◽  
Author(s):  
Qian Fan ◽  
Xiangrui Meng ◽  
Hongwei Liang ◽  
Huilai Zhang ◽  
Xianming Liu ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 808-808
Author(s):  
Xiangxiang Zhou ◽  
Juan Yang ◽  
Ya Zhang ◽  
Ying Li ◽  
Xiaosheng Fang ◽  
...  

Introduction Yes-associated protein (YAP) is an important transcriptional regulator and effector of the Hippo signaling that has emerged as a novel determinant of malignancy in several human tumors. Verteporfin (VP), a clinically used photosensitizer for treatment of macular degeneration, is recently identified as a potential inhibitor of YAP expression independent of light activation. However, the function and mechanism of Hippo-YAP in diffuse large B-cell lymphoma (DLBCL) remains ill defined. Herein, we investigated the functional significance of YAP and proposed a novel therapeutic strategy for DLBCL. Methods Lymph node biopsies from 60 de novo DLBCL patients and 30 reactive hyperplasia cases were collected with informed consents. The biological function of YAP was evaluated via RNAi-mediated knockdown and CRISPR/Cas9 mediated genomic deletion. RNA-sequencing was conducted to detect the dysregulated RNAs in DLBCL cell with YAP knockout. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analyses were used to explore the function of these differentially expressed RNAs. SCID-Beige mice were subcutaneously injected with DLBCL cells to establish xenograft model. Animal experiments were performed in accordance with the principles of the Institutional Animal Care. Results We first examined the expression of YAP in Oncomine database and discovered the upregulation of YAP mRNA in DLBCL cells (Fig.1A). High expression of YAP protein was validated in a cohort of newly diagnosed DLBCL patients (n=60). Survival analysis revealed that YAP expression was associated with aggressive disease process (p=0.014, Fig.1B). Functional enrichment analyses of YAP in DLBCL microarray profiles revealed that YAP was enriched in cellular process and biological regulation (Fig. 1C). Knockdown of YAP (shYAP) significantly suppressed cell proliferation and promoted cell cycle arrest (Fig.2A-C). VP restrained proliferation of DLBCL cells in a dose- and time-dependent manner (Fig. 2D). Treatment of VP significantly promoted cell apoptosis and cell cycle arrest in DLBCL cells (Fig. 2E). We next validated the involved mechanism of the anti-tumor effect of VP in DLBCL cells. Protein expression levels of YAP and TEAD were significantly inhibited by VP in dose-dependent manner (Fig.2F). In addition, treatment of VP remarkably restrained the mRNA expression of YAP targeted genes, including CTGF, CYR61 and NF2 (Fig. 2G). Genomic network enrichment of YAP interactions was established in STRING database (Fig. 2H). To validate the involvement of YAP in DLBCL pathogenesis, we deleted YAP expression by CRISPR/Cas9 genomic-editing system. YAP deletion (sgYAP) resulted in significantly reduction in cell proliferation and arrest of cell cycle (Fig.3A). To explore the activities of YAP inhibition in vivo, xenograft DLBCL mice model was established by LY1 cells with YAP-deletion. Compared to the control group, tumors with YAP-deletion displayed reduced growth and decreased expression levels of Ki67 and C-myc (Fig. 3B-C). To further explore the translational regulation mechanism of YAP in DLBCL, integrated analysis of mRNA and non-coding RNA (ncRNA) expression profiles were conducted by whole-transcriptome sequencing (RNA-seq). 158 miRNAs, 29 lncRNAs, 74 circRNAs and 528 mRNAs were shown to be significantly differentially expressed (DE) in DLBCL cells with YAP knockout (Fig. 4A-C). The subsequent GO and KEGG pathway analysis revealed that DE molecules mainly related to cellular metabolic process and pathways. In addition, the miRNA-circRNA network was established (Fig. 4D). Conclusion Taken together, these findings provide in vitro and in vivo pre-clinical evidence for the crucial role of YAP in lymphomagenesis and highlight that blockade of YAP represents an attractive approach in DLBCL. The clinical used photosensitizer Verteporfin exerted anti-tumor effect via disrupting YAP-TEAD complex. Further interrogation on the regulatory mechanism of YAP in DLBCL will outline a promising therapeutic option to utilize this newly identified oncogene in DLBCL therapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-15
Author(s):  
Qingqing Cai ◽  
Tianying Huang ◽  
Ning Su ◽  
Xiaolu Xu ◽  
Fengwei Wang ◽  
...  

Aims: To observe the synergistic antitumor effect of chidamide (a novel orally benzamide class of histone deacetylase inhibitors, HDACi) combined with gemcitabine, oxaliplatin or zanubrutinib (a second generation Bruton's tyrosine kinase (BTK) inhibitor) in diffuse large B-cell lymphoma cell lines and explore the possible mechanisms. Methods: We used CCK-8 assay to test the inhibitory efficacy of single agent chidamide, gemcitabine, oxaliplatin, zanubrutinib and their combinations on DLBCL cell lines, respectively. And the combination index (CI) was calculated by the CalcuSyn software. The apoptosis and the cell cycle analysis of DLBCL cell lines under different treatments were assessed by Annexin V-APC/PI double-staining and PI staining using flow cytometry, respectively. The expression levels of acetylated histone and apoptosis-related proteins were detected by Western Blot. Results:Single-agent chidamide, gemcitabine and oxaliplatin caused growth arrest of the DLBCL cell lines (OCL-LY3、OCL-LY19、U2932、DOHH2) in a dose and time dependent manner, respectively(Figure1a,1b,1c). Single-agent zanubrutinib showed antitumor activity in two activated B-cell like DLBCL (U2932 and OCI-LY3) cell lines in a dose and time dependent manner while no antitumor activity in other two GCB-DLBCL (OCI-LY19 and DOHH2) cell lines (Figure1d). Chidamide combined with gemcitabine or oxaliplatin synergistically inhibited the proliferative viability of DLBCL cell lines. Also, chidamide could enhance the inhibitory effect of zanubrutinib in the two ABC-DLBCL cell lines. And the CI values of all groups were more than 1(Fig2-1~2-8). Chidamide could significantly increase the proportion of DLBCL cells in the G0/G1 phase in a dose-dependent manner(Figure 3). And chidamide combined with gemcitabine can synergistically enhance the cell cycle arrest in U2932 cells (Figure 4). Cell apoptosis rate increased along with the ascending dosage of chidamide in the four DLBCL cell lines. The effect of cell apoptosis became more obviously when combined with oxaliplatin or zanubrutinib (Figure 5.1~5.4). Chidamide significantly up-regulated the levels of acetylated H3 (Lys 9) and cleaved-caspase 3 while down-regulated Bcl-2 (Figure 6~7). Furthermore, PD-L1 protein was upregulated in chidamide-exposed DLBCL cell lines (Figure 8). Conclusion:Chidamide combined with gemcitabine, oxaliplatin or zanubrutinib synergistically inhibited the proliferative viability and cell apoptosis rate in DLBCL cell lines. Therefore, it is worthy to further explore the value of Chidamide combination therapy in DLBCL. Figure Disclosures No relevant conflicts of interest to declare.


Gene ◽  
2017 ◽  
Vol 627 ◽  
pp. 255-262 ◽  
Author(s):  
Wenjun Xie ◽  
Meijuan Wu ◽  
Tianhong Fu ◽  
Xiaohong Li ◽  
Zhaoming Wang ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document