scholarly journals PD-1 expression on uveal melanoma induces tumor proliferation and predicts poor patient survival

2020 ◽  
Vol 35 (3) ◽  
pp. 50-58
Author(s):  
Zhongming Jiang ◽  
Yuexiang Yan ◽  
Juan Dong ◽  
Lingling Duan

Introduction: Uveal melanoma is one of the most common primary intraocular malignant tumors with poor prognosis and limited treatments. Programmed cell death receptor-1 (PD-1) blockade represents the primary treatment strategy of immune checkpoint inhibition; however, there is a lack of studies on whether PD-1 expression in primary (ocular) uveal melanoma affects tumor progression. Methods: PD-1 expression in 82 cases of primary (ocular) uveal melanoma was detected by immunohistochemistry. The clinical significance of PD-1 expression was evaluated using univariate and multivariate analysis. PD-1 overexpression and knockdown studies were conducted in C918 and Mum-2B cell lines to analyze the effect of PD-1 expression on tumor cell proliferation and intracellular cell signaling transduction. real-time qPCR (RT-qPCR) and western blot analysis were performed to investigate the gene expression level. CCK8 assays were performed to examine the cell proliferation ability. Results: High expression of primary (ocular) intratumor PD-1 was associated with poor patient survival. Moreover, PD-1 expression was correlated with the largest tumor diameter. PD-1 expression and optic nerve invasion were independent prognostic risk factors. PD-1 overexpression in uveal melanoma cell lines promoted tumor cell proliferation, while knockdown of PD-1 inhibited cell proliferation capacity. Conclusion: Our study established the role of PD-1 in the progression of uveal melanoma and provided a new potential treatment selection for uveal melanoma.

2021 ◽  
Vol 22 (5) ◽  
pp. 2771
Author(s):  
Anna Richter ◽  
Elisabeth Fischer ◽  
Clemens Holz ◽  
Julia Schulze ◽  
Sandra Lange ◽  
...  

Aberrant PI3K/AKT signaling is a hallmark of acute B-lymphoblastic leukemia (B-ALL) resulting in increased tumor cell proliferation and apoptosis deficiency. While previous AKT inhibitors struggled with selectivity, MK-2206 promises meticulous pan-AKT targeting with proven anti-tumor activity. We herein, characterize the effect of MK-2206 on B-ALL cell lines and primary samples and investigate potential synergistic effects with BCL-2 inhibitor venetoclax to overcome limitations in apoptosis induction. MK-2206 incubation reduced AKT phosphorylation and influenced downstream signaling activity. Interestingly, after MK-2206 mono application tumor cell proliferation and metabolic activity were diminished significantly independently of basal AKT phosphorylation. Morphological changes but no induction of apoptosis was detected in the observed cell lines. In contrast, primary samples cultivated in a protective microenvironment showed a decrease in vital cells. Combined MK-2206 and venetoclax incubation resulted in partially synergistic anti-proliferative effects independently of application sequence in SEM and RS4;11 cell lines. Venetoclax-mediated apoptosis was not intensified by addition of MK-2206. Functional assessment of BCL-2 inhibition via Bax translocation assay revealed slightly increased pro-apoptotic signaling after combined MK-2206 and venetoclax incubation. In summary, we demonstrate that the pan-AKT inhibitor MK-2206 potently blocks B-ALL cell proliferation and for the first time characterize the synergistic effect of combined MK-2206 and venetoclax treatment in B-ALL.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1423-1423
Author(s):  
You Hua Yu ◽  
Na Guo ◽  
Yujing Gong ◽  
Baidong Liu ◽  
Hong Liu ◽  
...  

Abstract Abstract 1423 Patients with B cell malignaces initially respond to current treatment modalities, however, such malignances remain incurable. Many new therapeutic options have become available during the past several years but nearly all patients develop resistance to currently available therapeutic options. Ideally, a new treatment should inhibit tumor growth, improve the efficacy of other anti-tumor agents, and improve both the overal survial and the quality of life for patients. Pterostilbene is predominantly found in Rhubarb. We synthesized bipterostilbene (5-(4-(4-(3,5-dihydroxylstyryl)phenoxy)styryl)-benzene-1,3-diol) (C28H22O5) of a molecular weight of 438.48 Kda. In this study, we first examined whether bipterostilbene affects tumor cells proliferation using breast cancer, ovarian cancer, lymphoma and multiple myeloma (MM) cell lines. The results of the MTS assay demonstrated that bipterostilbene significantly inhibited tumor cell proliferation of the lymphoma cell line (Raji) and the MM cell lines (RPMI1640 and MM1s) at 48 hours (IC50: 5μM for Raji, 4μM for RPMI8226, and 2 μM for MM1s). The induction of tumor cell apoptosis was most prominent at 72 hours. The extent of the inhibition of tumor cell proliferation and the induction of apoptosis was concentration-dependent. Bipterostilbene had minimal effects on breast and ovarian cancer cell lines. Noteworthy, bipterostilbene had no detectable cytotoxic effects on normal human peripheral blood mononuclear cells (PBMCs). The molecular mechanism by which bipterostilbene mediates its effects was examined. Both the AKT and the NF-κB signaling transduction pathways have been reported to play key roles in B cell metabolism, proliferation and survival. Using RT-PCR, bipterostilbene specifically inhibited AKT1 and mTOR gene expression when Raji or RPMI8226 tumor cells were treated with the IC50 concentration of bipterostilbene for 24 hours. Analysis of downstream gene products of the AKT pathway revealed that Cyclin D1 expression was slightly reduced and P21Cip and P27 kip expressions were not changed. Bipterostilbene did not alter AKT2 or AKT3 gene expression, demonstrating that this compound is specifically targeting AKT1. We further determined whether bipterostilbene interfered with IGF1-induced AKT/mTOR activation or IL-1β –mediated NF-κB phosphorylation by Western blot. The results showed that bipterostilbene markedly inhibited IGF1-induced phosphorylation of AKT but did not interfere with IL-1β-induced NF-κB activity and IκB phosphorylation. Overall, the results of our in vitro studies demonstrate that bipterostilbene inhibits tumor cell proliferation and enhances apoptosis of B-cell malignancies via inhibition of the AKT/mTOR signaling pathway with no detectable effect on the NF-κB signaling pathway. Importantly, bipterostilbene is not cytotoxic on normal hematopoietic cells at concentrations that were highly toxic to B-cell malignancies. We propose that bipterostilbene may be better tolerated than other anti- cancer drugs that are currently being used for the treatment of B-cell malignancies. Disclosures: No relevant conflicts of interest to declare.


2004 ◽  
Vol 9 (3) ◽  
pp. 216-222 ◽  
Author(s):  
Kaumudi M. Bhawe ◽  
Robert A. Blake ◽  
Douglas O. Clary ◽  
Peter M. Flanagan

To facilitate the characterization of proteins that negatively regulate tumor cell proliferation in vitro, the authors have implemented a high-throughput functional assay that measures S-phase progression of tumor cell lines. For 2 tumor cell lines—human melanoma A375 and human lung carcinoma A549—conditions were established using the cyclin-dependent kinase inhibitor, p27kip; the tumor suppressor p53, a kinase-inactive allele of the cell cycle-regulated serine/threonine kinase Aurora2; and the G1/S drug block, aphidicolin. For screening purposes, gene libraries were delivered by adenoviral infection. Cells were fixed and labeled by immunocytochemistry, and an automated image acquisition and analysis package on a Cellomics ArrayScan®II was used to quantify the effects of these treatments on cell proliferation. The assay can be used to identify novel proteins involved in proliferation and serves as a more robust, reproducible, and sensitive alternative to enzyme-linked immunosorbent assay (ELISA)-based technologies.


2019 ◽  
Vol 25 (9) ◽  
pp. 2874-2886 ◽  
Author(s):  
Matthias A. Fink ◽  
Heiko Paland ◽  
Susann Herzog ◽  
Markus Grube ◽  
Silke Vogelgesang ◽  
...  

BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Lucy Wanjiku Macharia ◽  
Wanjiru Muriithi ◽  
Carlos Pilotto Heming ◽  
Dennis Kirii Nyaga ◽  
Veronica Aran ◽  
...  

Abstract Background Glioblastoma is a fatal brain tumour with a poor patient survival outcome. Hypoxia has been shown to reprogram cells towards a stem cell phenotype associated with self-renewal and drug resistance properties. Activation of hypoxia-inducible factors (HIFs) helps in cellular adaptation mechanisms under hypoxia. Similarly, miRNAs are known to be dysregulated in GBM have been shown to act as critical mediators of the hypoxic response and to regulate key processes involved in tumorigenesis. Methods Glioblastoma (GBM) cells were exposed to oxygen deprivation to mimic a tumour microenvironment and different cell aspects were analysed such as morphological changes and gene expression of miRNAs and survival genes known to be associated with tumorigenesis. Results It was observed that miR-128a-3p, miR-34-5p, miR-181a/b/c, were down-regulated in 6 GBM cell lines while miR-17-5p and miR-221-3p were upregulated when compared to a non-GBM control. When the same GBM cell lines were cultured under hypoxic microenvironment, a further 4–10-fold downregulation was observed for miR-34-5p, miR-128a-3p and 181a/b/c while a 3–6-fold upregulation was observed for miR-221-3p and 17-5p for most of the cells. Furthermore, there was an increased expression of SOX2 and Oct4, GLUT-1, VEGF, Bcl-2 and survivin, which are associated with a stem-like state, increased metabolism, altered angiogenesis and apoptotic escape, respectively. Conclusion This study shows that by mimicking a tumour microenvironment, miRNAs are dysregulated, stemness factors are induced and alteration of the survival genes necessary for the cells to adapt to the micro-environmental factors occurs. Collectively, these results might contribute to GBM aggressiveness.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2306-2306 ◽  
Author(s):  
Manfai Lee ◽  
Courtney Hoyt ◽  
George M Rodgers

Abstract ADAMTS13, the von Willebrand factor (vWF) cleaving protease, regulates platelet aggregation and microthrombi formation by cleaving high-molecular weight vWF multimers. It is expressed primarily in hepatic stellate cells, but is also found in endothelial cells. Recently, ADAMTS13 was reported to be expressed and regulated in astrocytes, microglial, neuroblastoma, and adult human brain endothelial cells. Previous in vitro studies by our group with human umbilical vein endothelial cells (HUVEC) showed that ADAMTS13 can promote angiogenesis via upregulating the secretion of VEGF and phosphorylation of VEGFR2, suggesting that ADAMTS13 may also be involved in physiological processes unrelated to vWF cleavage (Lee, M., et al. Microvasc Res. 2012, 84, 109-115). Herein, we report an additional possible role of ADAMTS13 secreted by brain tumor cells to modulate tumor cell angiogenesis. Using a human ADAMTS13 immunoassay, we detected ADAMTS13 in U-87 and LN-229 glioblastoma cell lysates, SW-1088 astrocytoma cell lysates, as well as in the supernatants of all cell lines (> 2.0 ng/mL). Co-incubation of U-87, LN-229, and SW-1088 tumor cell conditioned media with recombinant vWF indicated that brain tumor-secreted ADAMTS13 is biologically active in cleaving vWF multimers (measured by ELISA). Secretion of VEGF was upregulated in LN-229 and SW-1088 cell lines by ADAMTS13. 939 pg/mL and 674 pg/mL of VEGF were measured in LN-229 and SW-1088 cell lysates, respectively, after incubation with 100 ng/mL ADAMTS13. Incubation of LN-229 glioblastoma cells with 10 – 500 ng/mL rh-ADAMTS13 or 50 ng/mL VEGF165 did not affect tumor cell proliferation. No change in tumor cell proliferation was observed when LN-229 cells were incubated with a polyclonal antibody against ADAMTS13 in serum free media supplemented with 10 ng/mL ADAMTS13 or media supplemented with 5% FBS, suggesting that ADAMTS13 secreted by brain tumor cells may be involved in extracellular signaling of endothelial cells. Brain tumor cell secreted-ADAMTS13 induced HUVEC migration in a Matrigel invasion assay. Using a modified Boyden chamber fitted with a Matrigel-coated polycarbonate membrane, LN-229 glioblastoma cells increased HUVEC migration by 83%. LN-229 cells supplemented with 10 ng/mL ADAMTS13 further increased HUVEC migration by 190%, suggesting that tumor cell-secreted ADAMTS13 may modulate EC migration (Fig. 1). Combined with our previous findings suggesting that recombinant ADAMTS13 modulates EC angiogenesis, brain tumor-secreted ADAMTS13 may also be a regulator for tumor vasculature angiogenesis. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document