1.Down-regulation of DNA key protein-FEN1 inhibits OSCC growth by affecting immunosuppressive phenotypes via IFN-γ/JAK/STAT-1.
Shimeng WANG ; Xiangjian WANG ; Jun SUN ; Jin YANG ; Deyang WU ; Fanglong WU ; Hongmei ZHOU
International Journal of Oral Science 2023;15(1):17-17
		                        		
		                        			
		                        			Oral squamous cell carcinoma (OSCC) escape from the immune system is mediated through several immunosuppressive phenotypes that are critical to the initiation and progression of tumors. As a hallmark of cancer, DNA damage repair is closely related to changes in the immunophenotypes of tumor cells. Although flap endonuclease-1 (FEN1), a pivotal DNA-related enzyme is involved in DNA base excision repair to maintain the stability of the cell genome, the correlation between FEN1 and tumor immunity has been unexplored. In the current study, by analyzing the clinicopathological characteristics of FEN1, we demonstrated that FEN1 overexpressed and that an inhibitory immune microenvironment was established in OSCC. In addition, we found that downregulating FEN1 inhibited the growth of OSCC tumors. In vitro studies provided evidence that FEN1 knockdown inhibited the biological behaviors of OSCC and caused DNA damage. Performing multiplex immunohistochemistry (mIHC), we directly observed that the acquisition of critical immunosuppressive phenotypes was correlated with the expression of FEN1. More importantly, FEN1 directly or indirectly regulated two typical immunosuppressive phenotype-related proteins human leukocyte antigen (HLA-DR) and programmed death receptor ligand 1 (PD-L1), through the interferon-gamma (IFN-γ)/janus kinase (JAK)/signal transducer and activator transcription 1 (STAT1) pathway. Our study highlights a new perspective on FEN1 action for the first time, providing theoretical evidence that it may be a potential immunotherapy target for OSCC.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/pathology*
		                        			;
		                        		
		                        			DNA
		                        			;
		                        		
		                        			Down-Regulation
		                        			;
		                        		
		                        			Flap Endonucleases/metabolism*
		                        			;
		                        		
		                        			Head and Neck Neoplasms
		                        			;
		                        		
		                        			Interferon-gamma/metabolism*
		                        			;
		                        		
		                        			Mouth Neoplasms/pathology*
		                        			;
		                        		
		                        			Phenotype
		                        			;
		                        		
		                        			Squamous Cell Carcinoma of Head and Neck
		                        			;
		                        		
		                        			Tumor Microenvironment
		                        			;
		                        		
		                        			Janus Kinases/metabolism*
		                        			
		                        		
		                        	
2.LIMP-2 enhances cancer stem-like cell properties by promoting autophagy-induced GSK3β degradation in head and neck squamous cell carcinoma.
Yuantong LIU ; Shujin LI ; Shuo WANG ; Qichao YANG ; Zhizhong WU ; Mengjie ZHANG ; Lei CHEN ; Zhijun SUN
International Journal of Oral Science 2023;15(1):24-24
		                        		
		                        			
		                        			Cancer stem cell-like cells (CSCs) play an integral role in the heterogeneity, metastasis, and treatment resistance of head and neck squamous cell carcinoma (HNSCC) due to their high tumor initiation capacity and plasticity. Here, we identified a candidate gene named LIMP-2 as a novel therapeutic target regulating HNSCC progression and CSC properties. The high expression of LIMP-2 in HNSCC patients suggested a poor prognosis and potential immunotherapy resistance. Functionally, LIMP-2 can facilitate autolysosome formation to promote autophagic flux. LIMP-2 knockdown inhibits autophagic flux and reduces the tumorigenic ability of HNSCC. Further mechanistic studies suggest that enhanced autophagy helps HNSCC maintain stemness and promotes degradation of GSK3β, which in turn facilitates nuclear translocation of β-catenin and transcription of downstream target genes. In conclusion, this study reveals LIMP-2 as a novel prospective therapeutic target for HNSCC and provides evidence for a link between autophagy, CSC, and immunotherapy resistance.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Autophagy
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/pathology*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Glycogen Synthase Kinase 3 beta/metabolism*
		                        			;
		                        		
		                        			Head and Neck Neoplasms/pathology*
		                        			;
		                        		
		                        			Neoplastic Stem Cells/pathology*
		                        			;
		                        		
		                        			Squamous Cell Carcinoma of Head and Neck/pathology*
		                        			;
		                        		
		                        			Lysosome-Associated Membrane Glycoproteins
		                        			
		                        		
		                        	
3.Clinicopathological features of patients with RET fusion-positive non-small cell lung cancer.
Qi TAN ; Yu JI ; Xiao Li WANG ; Zhen Wei WANG ; Xiao Wei QI ; Yan Kui LIU
Chinese Journal of Pathology 2023;52(2):124-128
		                        		
		                        			
		                        			Objective: To investigate the clinicopathological features, treatment and prognosis of patients with RET fusion positive non-small cell lung cancer (NSCLC). Methods: A total of 1 089 NSCLCs were retrieved at Affiliated Hospital of Jiangnan University from August 2018 to April 2020. In all cases, multiple gene fusion detection kits (fluorescent PCR method) were used to detect the gene status of RET, EGFR, ALK, ROS1, KRAS, BRAF and HER2; and immunohistochemical method was used to detect the expression of PD-L1 and mismatch repair related proteins. The correlation between RET-fusion and patients' age, gender, smoking history, tumor stage, grade, pathologic type, and PD-L1, mismatch repair related protein expression was analyzed. Results: There were 22 cases (2.02%) detected with RET fusion-positive in 1 089 NSCLC patients, in which 11 males and 11 females; and the median age was 63.5 years. There were 20 adenocarcinomas, including 11 acinar predominant adenocarcinoma (APA), five solid predominant adenocarcinoma (SPA) and four lepidic predominant adenocarcinoma (LPA); There were one case each of squamous cell carcinoma (non-keratinizing type) and sarcomatoid carcinoma (pleomorphic carcinoma). There were 6 and 16 patients with RET fusion-positive who were in stage Ⅰ-Ⅱ and Ⅲ-Ⅳ respectively, and 16 cases with lymph node metastasis, 11 cases with distant metastasis. Among RET fusion-positive cases, one was detected with HER2 co-mutation. The tumor proportion score of PD-L1≥1% in patients with RET fusion positive lung cancer was 54.5% (12/22). Defects in mismatch repair protein expression were not found in patients with RET fusion positive NSCLC. Four patients with RET fusions positive (two cases of APA and two cases of SPA) received pratinib-targeted therapy, and two showed benefits from this targeted therapy. Conclusions: The histological subtypes of RET fusions positive NSCLC are more likely to be APA or SPA. RET fusion-positive NSCLC patients are associated with advanced clinical stage, lymph node metastases, and they may benefit from targeted therapy with RET-specific inhibitors.
		                        		
		                        		
		                        		
		                        			Male
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Middle Aged
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/pathology*
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			;
		                        		
		                        			B7-H1 Antigen/genetics*
		                        			;
		                        		
		                        			Protein-Tyrosine Kinases/genetics*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-ret/metabolism*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins/genetics*
		                        			;
		                        		
		                        			Adenocarcinoma/pathology*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/genetics*
		                        			;
		                        		
		                        			Mutation
		                        			
		                        		
		                        	
4.Lung Squamous Cell Carcinoma with EML4-ALK Fusion and TP53 Co-mutation Treated with Ensartinib: A Case Report and Literature Review.
Donglai LV ; Chunwei XU ; Chong WANG ; Qiuju SANG
Chinese Journal of Lung Cancer 2023;26(1):78-82
		                        		
		                        			
		                        			Lung squamous cell carcinoma (LSCC) accounts for approximately 30% of non-small cell lung cancer (NSCLC) cases and is the second most common histological type of lung cancer. Anaplastic lymphoma kinase (ALK)-positive NSCLC accounts for only 2%-5% of all NSCLC cases, and is almost exclusively detected in patients with lung adenocarcinoma. Thus, ALK testing is not routinely performed in the LSCC population, and the efficacy of such treatment for ALK-rearranged LSCC remains unknown. Echinoderm microtubule associated protein like 4 (EML4)-ALK (V1) and TP53 co-mutations were identified by next generation sequencing (NGS) in this patient with advanced LSCC. On December 3, 2020, Ensatinib was taken orally and the efficacy was evaluated as partial response (PR). The progression-free survival (PFS) was 19 months. When the disease progressed, the medication was changed to Loratinib. To our knowledge, Enshatinib created the longest PFS of ALK-mutant LSCC patients treated with targeted therapy since literature review. Herein, we described one case treated by Enshatinib involving a patient with both EML4-ALK and TP53 positive LSCC, and the relevant literatures were reviewed for discussing the treatment of this rare disease.
.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/drug therapy*
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			;
		                        		
		                        			Anaplastic Lymphoma Kinase/metabolism*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/genetics*
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			Cytoskeletal Proteins/genetics*
		                        			;
		                        		
		                        			Lung/pathology*
		                        			;
		                        		
		                        			Oncogene Proteins, Fusion/genetics*
		                        			;
		                        		
		                        			Protein Kinase Inhibitors/therapeutic use*
		                        			;
		                        		
		                        			Tumor Suppressor Protein p53/genetics*
		                        			
		                        		
		                        	
5.Mechanism Research of lncRNA miR143HG on Regulating the Biological Behavior of Lung Squamous Cell Carcinoma H520 Cells.
Longfei GOU ; Yayuan HE ; Pengcheng QIU ; Bo HUANG
Chinese Journal of Lung Cancer 2023;26(10):741-752
		                        		
		                        			BACKGROUND:
		                        			There is a high morbidity, mortality, and poor clinical prognosis of lung squamous cell carcinoma (LUSC). However, there is currently no effective targeted treatment plan for LUSC. As a long non-coding RNA (lncRNA), lncRNA miR143HG has been proven to play an important role in the occurrence and development of various tumors. However, the biological role played by lncRNA miR143HG in LUSC cells is still unclear. Therefore, this study aimed to investigate the mechanism of lncRNA miR143HG on regulating the biological behavior of LUSC H520 cells.
		                        		
		                        			METHODS:
		                        			Pan-cancer analysis and differential expression analysis of lncRNA miR143HG were performed based on The Cancer Genome Atlas (TCGA) database. The predictive effect of lncRNA miR143HG on the diagnosis and prognosis of LUSC was evaluated by adopting the receiver operating characteristic (ROC) curve and timeROC curve. The enrichment degree of each pathway to lncRNA miR143HG was determined. The expression of lncRNA miR143HG and miR-155 in BEAS-2B cells and H520 cells was detected using quantitative real-time polymerase chain reaction (qRT-PCR). H520 cells were randomly divided into blank control group (without any treatment), negative control group (transfected with lncRNA-NC), lncRNA miR143HG group (transfected with lncRNA miR143HG), and lncRNA miR143HG+miR-155 group (co-transfected with lncRNA miR143HG and miR-155). The approaches of CCK-8, wound healing test, Transwell assay, flow cytometry, qRT-PCR, and Western blot were respectively employed to detect the cell proliferation ability, cell migration ability, cell invasion ability, cell apoptosis rate, and expression level of related genes and proteins of the Wnt/β-Catenin pathway.
		                        		
		                        			RESULTS:
		                        			The results of pan-cancer analysis and differential analysis collectively showed that except for renal clear cell carcinoma, the expression of lncRNA miR143HG in other cancer tissues was higher than that in healthy tissues, and the differences were significant in LUSC. The evaluation results of the ROC curve and timeROC curve suggested that lncRNA miR143HG was of great significance in the prediction of diagnosis and prognosis of LUSC. The pathways enriched in high expression of lncRNA miR143HG mainly included focal adhesion, vascular smooth muscle contraction, calcium signaling pathways, and so on; the pathways enriched in the low expression of lncRNA miR143HG embraced oxidative phosphorylation, cell cycle, basic transcription factors, etc. The qRT-PCR results showed that lncRNA miR143HG was low expressed but miR-155 was highly expressed in H520 cells when compared to BEAS-2B cells (P<0.05). Compared with the negative control group, the expression levels of the gene of lncRNA miR143HG, the gene and protein of Wnt, as well as the gene and protein of β-Catenin were significantly increased, while the gene expression of miR-155, the ability of cell proliferation, cell migration, and cell invasion were significantly reduced, but the cell apoptosis rate was dominantly elevated in cells of lncRNA miR143HG group (P<0.05). In addition, compared with the lncRNA miR143HG group, overexpression of miR-155 could reverse the biological behavior mediated by lncRNA miR143HG, and the difference was statistically significant (P<0.05).
		                        		
		                        			CONCLUSIONS
		                        			LncRNA miR143HG was of great significance for the biological behavior of H520 cells. LncRNA miR143HG inhibited the ability of proliferation, migration, and invasion, as well as enhanced the apoptosis of H520 cells by downregulating miR-155 expression, which may be related to the Wnt/β-Catenin pathway.
.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			RNA, Long Noncoding/genetics*
		                        			;
		                        		
		                        			beta Catenin/metabolism*
		                        			;
		                        		
		                        			Lung Neoplasms/genetics*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/genetics*
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/genetics*
		                        			;
		                        		
		                        			MicroRNAs/genetics*
		                        			;
		                        		
		                        			Lung/pathology*
		                        			;
		                        		
		                        			Cell Proliferation/genetics*
		                        			;
		                        		
		                        			Cell Movement/genetics*
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			
		                        		
		                        	
6.Luteolin suppresses oral carcinoma 3 (OC3) cell growth and migration via modulating polo-like kinase 1 (PLK1) expression and cellular energy metabolism.
Pengfei GAO ; Wentao ZHANG ; Yujie LIN ; Ruijie LU ; Zijian LOU ; Gang LU ; Ruolang PAN ; Yunfang CHEN
Journal of Zhejiang University. Science. B 2023;24(12):1151-1158
		                        		
		                        			
		                        			Oral squamous cell carcinoma (OSCC) is a prevalent malignant tumor affecting the head and neck region (Leemans et al., 2018). It is often diagnosed at a later stage, leading to a poor prognosis (Muzaffar et al., 2021; Li et al., 2023). Despite advances in OSCC treatment, the overall 5-year survival rate of OSCC patients remains alarmingly low, falling below 50% (Jehn et al., 2019; Johnson et al., 2020). According to statistics, only 50% of patients with oral cancer can be treated with surgery. Once discovered, it is more frequently at an advanced stage. In addition, owing to the aggressively invasive and metastatic characteristics of OSCC, most patients die within one year of diagnosis. Hence, the pursuit of novel therapeutic drugs and treatments to improve the response of oral cancer to medication, along with a deeper understanding of their effects, remains crucial objectives in oral cancer research (Johnson et al., 2020; Bhat et al., 2021; Chen et al., 2023; Ruffin et al., 2023).
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mouth Neoplasms/pathology*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/metabolism*
		                        			;
		                        		
		                        			Luteolin/therapeutic use*
		                        			;
		                        		
		                        			Squamous Cell Carcinoma of Head and Neck/drug therapy*
		                        			;
		                        		
		                        			Head and Neck Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			
		                        		
		                        	
7.Carcinoma-associated fibroblast-derived lysyl oxidase-rich extracellular vesicles mediate collagen crosslinking and promote epithelial-mesenchymal transition via p-FAK/p-paxillin/YAP signaling.
Xue LIU ; Jiao LI ; Xuesong YANG ; Xiaojie LI ; Jing KONG ; Dongyuan QI ; Fuyin ZHANG ; Bo SUN ; Yuehua LIU ; Tingjiao LIU
International Journal of Oral Science 2023;15(1):32-32
		                        		
		                        			
		                        			Carcinoma-associated fibroblasts (CAFs) are the main cellular components of the tumor microenvironment and promote cancer progression by modifying the extracellular matrix (ECM). The tumor-associated ECM is characterized by collagen crosslinking catalyzed by lysyl oxidase (LOX). Small extracellular vesicles (sEVs) mediate cell-cell communication. However, the interactions between sEVs and the ECM remain unclear. Here, we demonstrated that sEVs released from oral squamous cell carcinoma (OSCC)-derived CAFs induce collagen crosslinking, thereby promoting epithelial-mesenchymal transition (EMT). CAF sEVs preferably bound to the ECM rather than being taken up by fibroblasts and induced collagen crosslinking, and a LOX inhibitor or blocking antibody suppressed this effect. Active LOX (αLOX), but not the LOX precursor, was enriched in CAF sEVs and interacted with periostin, fibronectin, and bone morphogenetic protein-1 on the surface of sEVs. CAF sEV-associated integrin α2β1 mediated the binding of CAF sEVs to collagen I, and blocking integrin α2β1 inhibited collagen crosslinking by interfering with CAF sEV binding to collagen I. CAF sEV-induced collagen crosslinking promoted the EMT of OSCC through FAK/paxillin/YAP pathway. Taken together, these findings reveal a novel role of CAF sEVs in tumor ECM remodeling, suggesting a critical mechanism for CAF-induced EMT of cancer cells.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Paxillin/metabolism*
		                        			;
		                        		
		                        			Protein-Lysine 6-Oxidase/metabolism*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/pathology*
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition
		                        			;
		                        		
		                        			Integrin alpha2beta1/metabolism*
		                        			;
		                        		
		                        			Mouth Neoplasms/pathology*
		                        			;
		                        		
		                        			Collagen/metabolism*
		                        			;
		                        		
		                        			Fibroblasts
		                        			;
		                        		
		                        			Extracellular Vesicles/metabolism*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Tumor Microenvironment
		                        			
		                        		
		                        	
8.The mechanism of S100A7 inducing the migration and invasion in cervical cancer cells.
Tian TIAN ; Zhen HUA ; Yan KONG ; Ling Zhi WANG ; Xiang Yu LIU ; Yi HAN ; Xue Min ZHOU ; Zhu Mei CUI
Chinese Journal of Oncology 2023;45(5):375-381
		                        		
		                        			
		                        			Objective: To investigate the mechanism of S100A7 inducing the migration and invasion in cervical cancers. Methods: Tissue samples of 5 cases of cervical squamous cell carcinoma and 3 cases of adenocarcinoma were collected from May 2007 to December 2007 in the Department of Gynecology of the Affiliated Hospital of Qingdao University. Immunohistochemistry was performed to evaluate the expression of S100A7 in cervical carcinoma tissues. S100A7-overexpressing HeLa and C33A cells were established with lentiviral systems as the experimental group. Immunofluorescence assay was performed to observe the cell morphology. Transwell assay was taken to detect the effect of S100A7-overexpression on the migration and invasion of cervical cancer cells. Reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) was used to examine the mRNA expressions of E-cadherin, N-cadherin, vimentin and fibronectin. The expression of extracellular S100A7 in conditioned medium of cervical cancer cell was detected by western blot. Conditioned medium was added into Transwell lower compartment to detect cell motility. Exosomes were isolated and extracted from the culture supernatant of cervical cancer cell, the expressions of S100A7, CD81 and TSG101 were detected by western blot. Transwell assay was taken to detect the effect of exosomes on the migration and invasion of cervical cancer cells. Results: S100A7 expression was positively expressed in cervical squamous carcinoma and negative expression in adenocarcinoma. Stable S100A7-overexpressing HeLa and C33A cells were successfully constructed. C33A cells in the experimental group were spindle shaped while those in the control group tended to be polygonal epithelioid cells. The number of S100A7-overexpressed HeLa cells passing through the Transwell membrane assay was increased significantly in migration and invasion assay (152.00±39.22 vs 105.13±15.75, P<0.05; 115.38±34.57 vs 79.50±13.68, P<0.05). RT-qPCR indicated that the mRNA expressions of E-cadherin in S100A7-overexpressed HeLa and C33A cells decreased (P<0.05) while the mRNA expressions of N-cadherin and fibronectin in HeLa cells and fibronectin in C33A cells increased (P<0.05). Western blot showed that extracellular S100A7 was detected in culture supernatant of cervical cancer cells. HeLa cells of the experimental group passing through transwell membrane in migration and invasion assays were increased significantly (192.60±24.41 vs 98.80±47.24, P<0.05; 105.40±27.38 vs 84.50±13.51, P<0.05) when the conditional medium was added into the lower compartment of Transwell. Exosomes from C33A cell culture supernatant were extracted successfully, and S100A7 expression was positive. The number of transmembrane C33A cells incubated with exosomes extracted from cells of the experimental group was increased significantly (251.00±49.82 vs 143.00±30.85, P<0.05; 524.60±52.74 vs 389.00±63.23, P<0.05). Conclusion: S100A7 may promote the migration and invasion of cervical cancer cells by epithelial-mesenchymal transition and exosome secretion.
		                        		
		                        		
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Uterine Cervical Neoplasms/pathology*
		                        			;
		                        		
		                        			HeLa Cells
		                        			;
		                        		
		                        			Fibronectins/metabolism*
		                        			;
		                        		
		                        			Culture Media, Conditioned
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/metabolism*
		                        			;
		                        		
		                        			Adenocarcinoma
		                        			;
		                        		
		                        			Cadherins/metabolism*
		                        			;
		                        		
		                        			RNA, Messenger/metabolism*
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition/genetics*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			S100 Calcium Binding Protein A7/metabolism*
		                        			
		                        		
		                        	
9.Knockdown of ACC1 promotes migration of esophageal cancer cell.
He QIAN ; Cheng Wei GU ; Yu Zhen LIU ; Bao Sheng ZHAO
Chinese Journal of Oncology 2023;45(6):482-489
		                        		
		                        			
		                        			Objective: To investigate the effect of acetyl-CoA carboxylase 1 (ACC1) knockdown on the migration of esophageal squamous cell carcinoma (ESCC) KYSE-450 cell and underlying mechanism. Methods: Lentiviral transfection was conducted to establish sh-NC control cell and ACC1 knocking down cell (sh-ACC1). Human siRNA HSP27 and control were transfected by Lipo2000 to get si-HSP27 and si-NC. The selective acetyltransferase P300/CBP inhibitor C646 was used to inhibit histone acetylation and DMSO was used as vehicle control. Transwell assay was performed to detect cell migration. The expression of HSP27 mRNA was examined by reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) and the expressions of ACC1, H3K9ac, HSP27 and epithelial-mesenchymal transition-related proteins E-cadherin and Vimentin were detected by western blot. Results: The expression level of ACC1 in sh-NC group was higher than that in sh-ACC1 group (P<0.01). The number of cell migration in sh-NC group was (159.00±24.38), lower than (361.80±26.81) in sh-ACC1 group (P<0.01). The protein expression levels of E-cadherin and Vimentin in sh-NC group were statistically significant compared with sh-AAC1 group (P<0.05). The migrated cell number in sh-NC+ si-NC group was (189.20±16.02), lower than (371.60±38.40) in sh-ACC1+ si-NC group (P<0.01). The migrated cell number in sh-NC+ si-NC group was higher than that in sh-NC+ si-HSP27 group (152.40±24.30, P<0.01), and the migrated cell number in sh-ACC1+ si-NC group was higher than that in sh-ACC1+ si-HSP27 group (P<0.01). The protein expression levels of E-cadherin and Vimentin in sh-NC+ si-NC group were significantly different from those in sh-ACC1+ si-NC and sh-NC+ si-HSP27 groups (P<0.01). The protein expression levels of E-cadherin and Vimentin in sh-ACC1+ si-NC group were significantly different from those in sh-ACC1+ si-HSP27 group (P<0.01). After 24 h treatment with C646 at 20 μmmo/L, the migrated cell number in sh-NC+ DMSO group was (190.80±11.95), lower than (395.80±17.10) in sh-ACC1+ DMSO group (P<0.01). The migrated cell number in sh-NC+ DMSO group was lower than that in sh-NC+ C646 group (256.20±23.32, P<0.01). The migrated cell number in sh-ACC1+ DMSO group was higher than that in sh-ACC1+ C646 group (87.80±11.23, P<0.01). The protein expressions of H3K9ac, HSP27, E-cadherin and Vimentin in sh-NC+ DMSO group were significantly different from those in sh-ACC1+ DMSO group and sh-NC+ C646 group (P<0.01). The protein expression levels of H3K9ac, HSP27, E-cadherin and Vimentin in sh-ACC1+ DMSO group were significantly different from those in sh-ACC1+ C646 group (P<0.01). Conclusion: Knockdown of ACC1 promotes the migration of KYSE-450 cell by up-regulating HSP27 and increasing histone acetylation.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Esophageal Neoplasms/pathology*
		                        			;
		                        		
		                        			Esophageal Squamous Cell Carcinoma/genetics*
		                        			;
		                        		
		                        			Vimentin/metabolism*
		                        			;
		                        		
		                        			Dimethyl Sulfoxide
		                        			;
		                        		
		                        			HSP27 Heat-Shock Proteins/metabolism*
		                        			;
		                        		
		                        			Histones/metabolism*
		                        			;
		                        		
		                        			Cadherins/metabolism*
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation/genetics*
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition/genetics*
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			
		                        		
		                        	
10.Interferon-γ induces immunosuppression in salivary adenoid cystic carcinoma by regulating programmed death ligand 1 secretion.
Qiuyun FU ; Xingchi LIU ; Houfu XIA ; Yicun LI ; Zili YU ; Bing LIU ; Xuepeng XIONG ; Gang CHEN
International Journal of Oral Science 2022;14(1):47-47
		                        		
		                        			
		                        			Interferon-γ (IFN-γ), a key effector molecule in anti-tumor immune response, has been well documented to correlate with the intratumoral infiltration of immune cells. Of interest, however, a high level of IFN-γ has been reported in salivary adenoid cystic carcinoma (SACC), which is actually a type of immunologically cold cancer with few infiltrated immune cells. Investigating the functional significance of IFN-γ in SACC would help to explain such a paradoxical phenomenon. In the present study, we revealed that, compared to oral squamous cell carcinoma cells (a type of immunologically hot cancer), SACC cells were less sensitive to the growth-inhibition effect of IFN-γ. Moreover, the migration and invasion abilities of SACC cells were obviously enhanced upon IFN-γ treatment. In addition, our results revealed that exposure to IFN-γ significantly up-regulated the level of programmed death ligand 1 (PD-L1) on SACC cell-derived small extracellular vesicles (sEVs), which subsequently induced the apoptosis of CD8+ T cells through antagonizing PD-1. Importantly, it was also found that SACC patients with higher levels of plasma IFN-γ also had higher levels of circulating sEVs that carried PD-L1 on their surface. Our study unveils a mechanism that IFN-γ induces immunosuppression in SACC via sEV PD-L1, which would account for the scarce immune cell infiltration and insensitivity to immunotherapy.
		                        		
		                        		
		                        		
		                        			B7-H1 Antigen/metabolism*
		                        			;
		                        		
		                        			CD8-Positive T-Lymphocytes/pathology*
		                        			;
		                        		
		                        			Carcinoma, Adenoid Cystic/pathology*
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/pathology*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Immunosuppression Therapy
		                        			;
		                        		
		                        			Interferon-gamma/pharmacology*
		                        			;
		                        		
		                        			Mouth Neoplasms/metabolism*
		                        			;
		                        		
		                        			Programmed Cell Death 1 Receptor/metabolism*
		                        			;
		                        		
		                        			Salivary Gland Neoplasms/pathology*
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail