scholarly journals Evaluation of PAX8 Expression Promotes the Proliferation of Stomach Cancer cells

2019 ◽  
Author(s):  
Liang-Yu Bie ◽  
Ning Li ◽  
Su-Xia Luo ◽  
Wen-Ying Deng ◽  
Xiao-Yu Lu ◽  
...  

Abstract Background: PAX8 was identified as a mitosis-related regulator involved in the pathogenesis of human tumors and an indicator of the prognosis for patients. However, PAX8 function on proliferation in stomach cancer have not been studied. Aim: This study was aimed to explore the expression pattern of PAX8 in stomach cancer, and investigate the effect of PAX8 on the proliferation of stomach cancer cells. Methods: PAX8 expression pattern in stomach cancer was investigated in the “TCGA” database and compared in cancer and adjacent tissues of 36 clinical samples, in which the co-expression of SOX13 and PAX8 was confirmed by qRT-PCR. Based on the clinical record and IHC staining results, the survival curve was also mapped. Western blotting and qRT-PCR was used to analyze the effect of SOX13 on PAX8 expression in AGS and MGC803 cells, and the downstream effector Aurora B and Cyclin B1 expression. ChIP and luciferase assay were performed to explore SOX13 locate on the promoter of PAX8. Furthermore, CCK8, clone formation, EdU incorporation assay, and flow cytometry were used to detect the proliferative ability of AGS and MGC803 cell lines stably silenced PAX8. Results: PAX8 and SOX13 were identified to be synchronously upregulated in primary stomach cancer tissues and the stomach cancer TCGA datasets, and stomach cancer patients of combined high PAX8 and SOX13 expression showed worse prognosis. Furthermore, SOX13 can mediate PAX8 and its targeted genes expression, including Aurora B and Cyclin B1, in AGS and MGC803 cell lines. Flow cytometry and EdU incorporation assays showed that silencing PAX8 can block the cell cycle of stomach cancer cell in G1 phase and SOX13 expression can rescue the arrested proliferative process induced by PAX8 silenced in CCK8 and colony formation assays. Conclusion: -SOX13 participated in the elevated expression of PAX8, which promote the proliferation of stomach cancer cells, and both SOX13 and PAX8 play an oncogene role in stomach cancer.

2019 ◽  
Author(s):  
Liang-Yu Bie ◽  
Ning Li ◽  
Wen-Ying Deng ◽  
Xiao-Yu Lu ◽  
Ping Guo ◽  
...  

Abstract Background: PAX8 was identified as a mitosis-related regulator involved in the pathogenesis of human tumors and an indicator of the prognosis for patients. However, the function of PAX8 -in stomach cancer have not been studied. Aim: This study was aimed to explore the expression pattern of PAX8 in stomach cancer, and investigate the effect of PAX8 on the proliferation of stomach cancer cells. Methods: PAX8 expression pattern in stomach cancer was investigated in the “TCGA” database and compared in cancer and adjacent tissues of 36 clinical samples, in which the co-expression of SOX13 and PAX8 was confirmed by qRT-PCR. Based on the clinical record and IHC staining results, the survival curve was also mapped. Western blotting and qRT-PCR was used to analyze the effect of SOX13 on PAX8 and its downstream effector Aurora B and Cyclin B1 expression in AGS and MGC803 cells-. ChIP and luciferase assay were performed to explore SOX13 locate on the promoter of PAX8. Furthermore, CCK8, clone formation, EdU incorporation assay, and flow cytometry were used to detect the proliferative ability of AGS and MGC803 cell lines stably silenced PAX8. Results: PAX8 and SOX13 were identified to be synchronously upregulated in primary stomach cancer tissues and the TCGA stomach cancer datasets, and combined high PAX8 and SOX13 expression induced- worse prognosis. Furthermore, SOX13 can mediate PAX8 and its targeted genes expression, including Aurora B and Cyclin B1, in AGS and MGC803-. Flow cytometry and EdU incorporation assays showed that silencing PAX8 can block the cell cycle of stomach cancer cell in G1 phase and SOX13 expression can rescue the arrested proliferative process induced by PAX8 silence in CCK8 and colony formation assays. Conclusion: SOX13 participated in the elevated expression of PAX8,which promote the proliferation of stomach cancer cells, and both SOX13 and PAX8 act as an oncogene- in stomach cancer.


2019 ◽  
Author(s):  
Liang-Yu Bie ◽  
Ning Li ◽  
Wen-Ying Deng ◽  
Xiao-Yu Lu ◽  
Ping Guo ◽  
...  

Abstract Background PAX8 was not only a mitotic factor, but identified as a transcription factor involved in the prognosis of human tumor patients. Elucidating the function of PAX8 on the pathology of stomach cancer was meaningful. Results: PAX8 was found to be upregulated in primary stomach cancer tissue and the TCGA stomach cancer dataset. Interestingly, SOX13 and PAX8 showed consistent expression patterns, and the combined high PAX8 and SOX18 expression induced a worse prognosis of stomach cancer patients. SOX13 was further identified as a transcription factor of PAX8, and further affect Aurora B and Cyclin B1 expression, two cell cycle related factors of the downstream of PAX8, including. Furthermore, PAX8 depletion inducted G1-phase arrest and the decrease of EdU incorporation, cell viability and colony formation can be rescued by SOX13 overexpression. Conclusions: SOX13 participated in the elevated expression of PAX8, which promote the proliferation of stomach cancer cells. Therefore, SOX13 mediated PAX8 expression was recognized as a tumor-promoting role in stomach cancer.


2019 ◽  
Author(s):  
Liang-Yu Bie ◽  
Dan Li ◽  
Yan Wei ◽  
Ning Li ◽  
Xiao-Bing Chen ◽  
...  

Abstract PAX8 is identified as a regulator in the pathogenesis of human tumors and an indicator of the prognosis for patients. However, the role of PAX8 on proliferation in gastric cancer have not been studied. This study was aimed to explore the expression pattern of PAX8 in gastric cancer, and investigate the effect of PAX8 on the proliferation of gastric cancer cells. PAX8 and SOX13 were identified to be synchronously upregulated in primary gastric cancer in human gastric cancer tissues and the gastric cancer datasets of TCGA, and gastric cancer patients of combined high PAX8 and SOX13 expression showed poor prognosis. Furthermore, SOX13 can mediate PAX8 and its targeted genes, Aurora B and Cyclin B1, expression in AGS and MGC803 cell lines. Flow cytometry and EdU incorporation assays showed that silencing PAX8 can block the cell cycle of gastric cancer cell in G1 phase and SOX13 expression can rescue the arrested proliferative process induced by PAX8 silenced in CCK8 and colony formation assays. Thus, combined SOX13 and PAX8 expression regulate the proliferation of gastric cancer cells, and both SOX13 and PAX8 play an oncogene function in gastric cancer.


2019 ◽  
Vol 20 (1) ◽  
Author(s):  
Liang-Yu Bie ◽  
Ning Li ◽  
Wen-Ying Deng ◽  
Xiao-Yu Lu ◽  
Ping Guo ◽  
...  

Abstract Background PAX8 was not only a mitotic factor, but identified as a transcription factor involved in the prognosis of human tumor patients. Elucidating the function of PAX8 on the pathology of stomach cancer was meaningful. Results PAX8 was found to be upregulated in primary stomach cancer tissue and the TCGA stomach cancer dataset. Interestingly, SOX13 and PAX8 showed consistent expression patterns, and the combined high PAX8 and SOX18 expression induced a worse prognosis of stomach cancer patients. SOX13 was further identified as a transcription factor of PAX8, and further affect Aurora B and Cyclin B1 expression, two cell cycle related factors of the downstream of PAX8, including. Furthermore, PAX8 depletion inducted G1-phase arrest and the decrease of EdU incorporation, cell viability and colony formation can be rescued by SOX13 overexpression. Conclusions SOX13 participated in the elevated expression of PAX8, which promote the proliferation of stomach cancer cells. Therefore, SOX13 mediated PAX8 expression was recognized as a tumor-promoting role in stomach cancer.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Linwen Zhu ◽  
Zhe Li ◽  
Xiuchong Yu ◽  
Yao Ruan ◽  
Yijing Shen ◽  
...  

Abstract Background Recently, tRNA-derived fragments (tRFs) have been shown to serve important biological functions. However, the role of tRFs in gastric cancer has not been fully elucidated. This study aimed to identify the tumor suppressor role of tRF-5026a (tRF-18-79MP9P04) in gastric cancer. Methods Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was first used to detect tRF-5026a expression levels in gastric cancer tissues and patient plasma. Next, the relationship between tRF-5026a levels and clinicopathological features in gastric cancer patients was assessed. Cell lines with varying tRF-5026a levels were assessed by measuring tRF-5026a using qRT-PCR. After transfecting cell lines with a tRF-5026a mimic or inhibitor, cell proliferation, colony formation, migration, apoptosis, and cell cycle were evaluated. The expression levels of related proteins in the PTEN/PI3K/AKT pathway were also analyzed by Western blotting. Finally, the effect of tRF-5026a on tumor growth was tested using subcutaneous tumor models in nude mice. Results tRF-5026a was downregulated in gastric cancer patient tissues and plasma samples. tRF-5026a levels were closely related to tumor size, had a certain diagnostic value, and could be used to predict overall survival. tRF-5026a was also downregulated in gastric cancer cell lines. tRF-5026a inhibited the proliferation, migration, and cell cycle progression of gastric cancer cells by regulating the PTEN/PI3K/AKT signaling pathway. Animal experiments showed that upregulation of tRF-5026a effectively inhibited tumor growth. Conclusions tRF-5026a (tRF-18-79MP9P04) is a promising biomarker for gastric cancer diagnostics and has tumor suppressor effects mediated through the PTEN/PI3K/AKT signaling pathway.


2021 ◽  
Vol 2021 ◽  
pp. 1-11
Author(s):  
Yue Luo ◽  
Junhao Lin ◽  
Jiakang Zhang ◽  
Zhenghui Song ◽  
Dayong Zheng ◽  
...  

Long noncoding RNAs (lncRNAs) have been substantially reported to have critical roles in regulating tumorigenesis in recent years. However, the expression pattern and biological function of SNHG17 in hepatocellular carcinoma (HCC) remain unclear. Bioinformatics analysis and qRT-PCR were performed to detect the expression pattern of SNHG17 in HCC tissues, adjacent nontumorous tissues, and cell lines. The effect of SNHG17 on proliferation, migration, and apoptosis of HCC was investigated by knockdown and overexpressing SNHG17 in HCC cell lines. RNA sequencing was utilized to explore the underlying mechanism. Utilizing publicly available TCGA-LIHC, GSE102079 HCC datasets, and qRT-PCR, we found SNHG17 was significantly upregulated in HCC tissues and cell lines and was notably associated with larger tumor size, poorly differentiation, presence of vascular invasion, and advanced TNM stage. Furthermore, gain- and loss-of-function studies demonstrated that SNHG17 promoted cell proliferation and migration and inhibited apoptosis of HCC. By employing RNA sequencing, we found knockdown of SNHG17 caused 1037 differentially expressed genes, highly enriched in several pathways, including metabolic, PI3K-Akt, cell adhesion, regulation of cell proliferation, and apoptotic pathway; among them, 92 were overlapped with SNHG17-related genes in the TCGA-LIHC dataset. Furthermore, ERH, TBCA, TDO2, and PDK4 were successfully validated and found significantly dysregulated in HCC tissues. Moreover, HCC patients with higher SNHG17 expression had a relatively poor overall survival and disease-free survival, and ERH and PDK4 also played a marked role in the prognosis of HCC. Broadly, our findings illustrate that SNHG17 acts as a noncoding oncogene in HCC progression, suggesting its potential value as a novel target for HCC therapy.


2018 ◽  
Vol 49 (6) ◽  
pp. 2151-2162 ◽  
Author(s):  
Bo Lian ◽  
Dongxiang Yang ◽  
Yanlong Liu ◽  
Gang Shi ◽  
Jibin Li ◽  
...  

Background/Aims: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an ideal anti-tumor drug because it exhibits selective cytotoxicity against cancer cells. However, certain cancer cells are resistant to TRAIL, and the potential mechanisms are still unclear. The aim of this study was to reduce the resistance of colorectal cancer (CRC) cells to TRAIL. Methods: Quantitative real-time PCR analysis was performed to detect the expression of microRNA-128 (miR-128) in tissues from patients with CRC and CRC cell lines. MTT assays were used to evaluate the effect of miR-128 on TRAIL-induced cytotoxicity against CRC cell lines. The distribution of death receptor 5 (DR5) and the production of reactive oxygen species (ROS) were detected by flow cytometry analysis. Western blot, flow cytometry, and luciferase reporter assays were performed to evaluate the potential mechanism and pathway of miR-128-promoted apoptosis in TRAIL-treated CRC cells. Results: MiR-128 expression was downregulated in tumor tissues from patients with CRC as well as in CRC cell lines in vitro. The enforced expression of miR-128 sensitized CRC cells to TRAIL-induced cytotoxicity by inducing apoptosis. Mechanistically, bioinformatics, western blot analysis, and luciferase reporter assays showed that miR-128 directly targeted sirtuin 1 (SIRT1) in CRC cells. miR-128 overexpression suppressed SIRT1 expression, which promoted the production of ROS in TRAIL-treated CRC cells. This increase of ROS subsequently induced DR5 expression, and thus increased TRAIL-induced apoptosis in CRC cells. Conclusion: The combination of miR-128 with TRAIL may represent a novel approach for the treatment of CRC.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1581-1581
Author(s):  
Frederick Lansigan ◽  
Wilson L Davis ◽  
Nancy Kuemmerle ◽  
Leslie E Lupien ◽  
Valeriya Posternak ◽  
...  

Abstract Abstract 1581 Background It is well-recognized that de novo long chain fatty acid (FA) synthesis, driven by the key enzyme fatty acid synthase (FASN), is crucial for the growth and survival of many types of cancer cells. We and others have observed FASN protein expression in diffuse large B-cell lymphoma (DLBCL) tumors. Furthermore, we have shown that higher levels of FASN in DLBCL tumors strongly predicted inferior survival, which was independent from the international prognostic index. We also recently demonstrated that, in addition to FA synthesis, various cancer cells can acquire FA from circulating lipoproteins, using the secreted enzyme lipoprotein lipase (LPL), and that this promotes cell growth. DLBCL, however, has never been examined in this regard. In this study, we investigated the functional significance of both de novo FA synthesis via FASN and exogenous FA uptake via LPL in DLBCL. Methods Levels of FASN and LPL mRNAs in DLBCL cell lines (SUDHL4, SUDHL10, OCI-LY3, OCI-LY19) were studied using reverse transcriptase polymerase chain reaction. We determined FASN and LPL protein expression by flow cytometry using a novel anti-LPL antibody that we developed. DLBCL cell lines were cultured +/− Cerulenin (an inhibitor of FASN), Orlistat (an inhibitor of FASN and LPL), or in lipoprotein-depleted serum +/− supplementation with very low density lipoprotein (VLDL) particles. The MTT assay was used to assess cell proliferation. Results DLBCL cell lines exhibited >10-fold variation in levels of FASN mRNA. Cerulenin and Orlistat each caused dose-dependent inhibition of proliferation of each cell line. The cells were partially rescued by the addition of palmitic acid, the FA product of FASN. Surprisingly, flow cytometry revealed that SUDHL4 and OCI-LY3 cells, which did not secrete LPL or show detectable LPL activity, displayed the enzyme on the cell surface. Moreover, in stark contrast to several other cancer cell lines, DLBCL cells were exquisitely sensitive to withdrawal of lipoproteins from the culture media. Indeed, 75–95% of the cells underwent apoptosis after only 24 hours in lipoprotein-depleted serum. In complete serum, the provision of VLDL particles did not rescue DLBCL cells from FA synthesis inhibition using Cerulenin, suggesting that the serum contains sufficient lipoproteins to saturate the FA uptake system. This prediction was validated in experiments utilizing lipoprotein-depleted serum, in which add-back of VLDL particles completely rescued the cells from Cerulenin-induced demise in a dose-related manner, with full restoration at approximately 100–200mcg/ml of VLDL. Conclusions Our data demonstrate that DLBCL cells employ both de novo FA synthesis via FASN and exogenous FA uptake using LPL to satisfy their strict requirement for FA. Interference with either pathway, using FASN inhibitors or lipoprotein-depleted serum, is cytotoxic indicating that neither alone is sufficient to support proliferation. Further, DLBCL cells show a striking dependency on exogenous FA of dietary origin compared with all other cancer cells we have examined. The observation that the cell lines can be rescued by provision of VLDL particles strongly supports the functional significance of the exogenous FA uptake pathway for DLBCL. Our data thus demonstrate that the extracellular lipase LPL is critical for the growth and survival of DLBCL cells. Surprisingly, the cells deploy LPL to their surface, and we speculate that this promotes efficient FA acquisition from circulating lipoproteins. Recognition that DLBCL relies on both synthesis and uptake of FA will provide guidance for drug development and dietary modifications to effectively target the metabolic requirements of this tumor. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1821-1821
Author(s):  
Shiho Fujiwara ◽  
Naoko Wada ◽  
Yawara Kawano ◽  
Hiromichi Yuki ◽  
Yutaka Okuno ◽  
...  

Abstract Abstract 1821 Introduction It has been reported that cancer cells utilize glycolysis pathway (non-oxidative breakdown of glucose) even in the presence of adequate oxygen to provide cancer cells with energy, called the Warburg effect (aerobic glycolysis) that ultimately leads to produce lactate. We reported in the last ASH meeting that aerobic glycolysis is up-regulated in multiple myeloma (MM) cells in patients with high serum LDH levels and aerobic glycolysis itself could serve as a novel therapeutic target in MM patients. Here we report an importance of lactate transporter for the growth and survival of MM cells. Lactate, produced from pyruvate by lactate dehydrogenase A (LDHA), is known as an important energy source for solid tumor cells and is associated with tumor angiogenesis and chemo-resistance (Pinheiro, C., et al. J Bioenerg Biomembr. 44:127–139, 2012). On the other hand, LDHB converts lactate to pyruvate, thus negatively regulating lactate production. It is known that lactate is pumped out through monocarboxylate trasnporter, MCT4, while MCT1 mainly imports lactate to inside of cells. However, roles of MCT1 and MCT4 in MM cells remain to be elucidated. We here investigated the roles of these two molecules in the growth and survival of MM cells. CD147, a purported chaperone protein for MCT1, was also examined. Methods Six MM cell lines, RPMI8226, U266, KMS12BM, KMS12PE, KHM11, and KMM1 were employed. Six genes associated with glycolysis, i.e., LDHA, LDHB, MCT1-4, were examined using real time PCR analysis. Expressions of MCT1 and MCT4 were analyzed with western blotting. Expression of CD147 was investigated by flow cytometry. Lactate production into culture supernatants of MM cell lines were analyzed by using a lactate analyzer. An inhibitor of MCT1, a-cyano-4 hydroxycinnamic acid (CHC), was utilized to analyze cytotoxic effects on MM cells. AnnexinV/PI stained cells was analyzed by flow cytometry to quantify cytotoxicity. MCT1-expression was inhibited by using siRNA. Dichroloacetate (DCA), an inhibitor of PDK1, was utilized for inhibiting glycolysis. Results Accumulation of lactate was found in the supernatants of MM cell lines as cell density increased. Transporters of lactate, MCT1, MCT4 and CD147, were found in most MM cell lines at various levels, suggesting that transportation of lactate occurs through membrane of MM cells. To examine the role of lactate as a growth promotion factor, lactate was exogenously supplemented to KMS-12-PE cells. Interestingly, expressions of MCT1 and LDHB genes increased by the addition of lactate while those of MCT4 and LDHA only moderately changed (Fig. 1), suggesting that lactate was imported to cells through MCT1, then converted to pyrvate by LDHB. These results raised a possibility that lactate is utilized by MM cells as a growth factor. To examine the possibility, CHC, an inhibitor of MCT1, was supplemented to MM cell cultures. Interestingly, CHC induced apoptosis in MM cells in a dose dependent manner (Fig. 2). Moreover, inhibition of MCT1 gene by siRNA showed significant induction of apoptosis (Fig. 3), strongly suggesting that MCT1 plays a crucial role for survival of MM cells. Finally, we found a significant increase in the apoptosis of MM cells when CHC and DCA were simultaneously added in the culture (Fig.4), suggesting that MCT1 functions independently from glycolysis per se and that CHC and DCA act additively in starving lactate within MM cells. Conclusion Our results suggest that lactate is actively transported through monocarboxylate transporters. Given the results that exogenous lactate production increased MCT1 and LDHB expression, lactate should play a role as a regulator of lactate transportation and glycolysis as well as an important energy source. Because we found significant amount of lactate was produced from stromal cells obtained from MM patients, lactate may be supplied not only from MM cells themselves but also from micro-environment. Our finding that inhibition of MCT1 leads to cell death suggests that MCT1 could be a potential novel target molecule in MM therapy that could be stratified in combination with glycolysis inhibitor. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4900-4900
Author(s):  
Wenying Li ◽  
Wei Xiong ◽  
Xiaomei Chen ◽  
Shiang Huang ◽  
Li Huiyu

Abstract Objectives Microvesicles (MVs) are small vesicles that are shed from almost all cell types including cancer cells into their surroundings. These secreted MVs bear surface receptors/ligands and miRNA of the original cells and play a role in cancer. Our aim in this study was to identify MVs miRNAs that might play important roles both in solid and non-solid tumors. Methods The MVs miRNA expression from 4 kinds of cancer cell lines (SMMC-7721, K562, Nalm6 and Jurkat) and healthy peripheral blood cells were analyzed by miRNA microarray. Quantitative real-time polymerase chain reaction (qRT-PCR) was further performed to verify the expression of 4 dysregulated miRNAs. Results The miRNA microarray revealed aberrantly expressed miRNAs of MVs between the four types of cancer cells and normal cells. In the MVs from cancer groups, 73 miRNAs were up-regulated and 148 were down-regulated in all the four types of cancer cell lines, indicating that solid and non-solid tumors shared some dysregulated MVs miRNAs. qRT-PCR verified statistically consistent expression of four selected miRNAs with microarray analysis. The top six shared upregulated MVs miRNAs in the four cancer cells lines were miR-1290, miR-1268, miR-1246, miR-125a-3p, miR-1305, miR-1226*. The top seven shared downregulated MVs miRNAs in the four cancer cells lines were miR-335, miR-1, miR-363, miR-122, miR-223, miR-338-3p and miR-340. The general roles of the upregulated MVs miRNAs were oncomiRNAs in multicancer. MiR-1290 and its potential targets are associated with characteristics of estrogen receptor α-positive breast cancer. Up-regulations of miR-1290 impaired cytokinesis and affected the reprogramming of colon cancer cells. miR-1246 might serve as a likely link of the p53 family with Down syndrome and proposed a new p53-miR-1246-DYRK1A-NFAT pathway in cancer. The functional roles of the downregulated MVs miRNAs were tumor suppressors in many kinds of cancer. MiR-335 acted as a candidate tumor suppressor and inhibited tumor reinitiation and represented an invasion suppressor gene by targeting Bcl-w in many kinds of cancer. HBx regulated miR-338-3p in HCC and miR-338-3p inhibited proliferation by regulating CyclinD1, and miR-338-3p suppressed the invasion of liver cancer cell by targeting smoothened gene in liver cancer. miR-340 inhibited of breast cancer cell migrations and invasions through targeting of oncoprotein c-Met and may play an important role in breast cancer progression. Conclusion Our study identifies oncomiRNAs and tumor suppressors in MVs from four types of cancer. These MVs miRNAs may contribute to the developments of solid and non-solid tumors and should be further evaluated as a novel biomarker for cancers and may provide potential targets for novel therapeutic strategies. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document