Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 19(5): e0302780, 2024.
Article in English | MEDLINE | ID: mdl-38713738

ABSTRACT

Reticulocalbin 1 (RCN1) is a calcium-binding protein involved in the regulation of calcium homeostasis in the endoplasmic reticulum. The aim of this study was to explore the clinical value and biological role of RCN1 in esophageal squamous cell carcinoma (ESCC). In addition, we investigated the effect of RCN1 on the polarization of tumor-associated macrophages (TAMs). The GSE53625 dataset from the Gene Expression Omnibus database was used to analyze the expression of RCN1 mRNA and its relationship with clinical value and immune cell infiltration. Immunohistochemistry was used to validate the expression of RCN1 and its correlation with clinicopathological characteristics. Subsequently, transwell and cell scratch assays were conducted to evaluate the migration and invasion abilities of ESCC cells. The expression levels of epithelial-mesenchymal transition (EMT)-related proteins were evaluated by western blot, while apoptosis was detected by flow cytometry and western blot. Additionally, qRT‒PCR was utilized to evaluate the role of RCN1 in macrophage polarization. RCN1 was significantly upregulated in ESCC tissues and was closely associated with lymphatic metastasis and a poor prognosis, and was an independent prognostic factor for ESCC in patients. Knockdown of RCN1 significantly inhibited the migration, invasion, and EMT of ESCC cells, and promoted cell apoptosis. In addition, RCN1 downregulation inhibited M2 polarization. RCN1 is upregulated in ESCC patients and is negatively correlated with patient prognosis. Knocking down RCN1 inhibits ESCC progression and M2 polarization. RCN1 can serve as a potential diagnostic and prognostic indicator for ESCC, and targeting RCN1 is a very promising therapeutic strategy.


Subject(s)
Calcium-Binding Proteins , Epithelial-Mesenchymal Transition , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Macrophages , Female , Humans , Male , Apoptosis , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Macrophages/metabolism , Prognosis , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
2.
World J Surg Oncol ; 22(1): 58, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38369496

ABSTRACT

BACKGROUND/PURPOSE: This study compared the clinical efficacy and safety of laparoscopic versus open resection for hilar cholangiocarcinoma (HCCA) and analyzed potential prognostic factors. METHODS: The study included patients who underwent HCCA resection at our center from March 2012 to February 2022. Perioperative complications and postoperative prognosis were compared between the laparoscopic surgery (LS) and open surgery (OS) groups. RESULTS: After screening 313 HCCA patients, 68 patients were eligible for the study in the LS group (n = 40) and OS group (n = 28). Kaplan-Meier survival curve analysis revealed that overall survival > 2 years and 3-year disease-free survival (DFS) were more common in the LS than OS group, but the rate of 2-year DFS was lower in the LS group than OS group. Cox multivariate regression analysis revealed age (< 65 years), radical resection, and postoperative adjuvant therapy were associated with reduced risk of death (hazard ratio [HR] = 0.380, 95% confidence interval [CI] = 0.150-0.940, P = 0.036; HR = 0.080, 95% CI = 0.010-0.710, P = 0.024 and HR = 0.380, 95% CI = 0.150-0.960, P = 0.040), whereas preoperative biliary drainage was an independent factor associated with increased risk of death (HR = 2.810, 95% CI = 1.130-6.950, P = 0.026). Perineuronal invasion was identified as an independent risk factor affecting DFS (HR = 5.180, 95% CI = 1.170-22.960, P = 0.030). CONCLUSIONS: Compared with OS, laparoscopic HCCA resection does not significantly differ in terms of clinical efficacy. Age (<65 years), radical resection, and postoperative adjuvant therapy reduce the risk of death, and preoperative biliary drainage increases the risk of death.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Klatskin Tumor , Laparoscopy , Humans , Aged , Klatskin Tumor/surgery , Klatskin Tumor/pathology , Retrospective Studies , Bile Duct Neoplasms/pathology , Treatment Outcome , Prognosis , Survival Analysis , Laparoscopy/adverse effects , Cholangiocarcinoma/pathology
3.
Int J Biochem Cell Biol ; 166: 106481, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37914022

ABSTRACT

Centromere protein L (CENPL) is involved in the mitotic process of eukaryotic cells and the development of various types of cancer. However, its role in hepatocellular carcinoma (HCC) remains unclear. This study aimed to investigate the expression and clinical significance of CENPL in HCC, and explore its involvement in regulating HCC cell proliferation, apoptosis, cell cycle, and glycolysis both in vivo and in vitro. CENPL expression was analyzed in HCC and normal liver tissues using The Cancer Genome Atlas, Gene Expression Omnibus mining, real-time quantitative polymerase chain reaction, and immunohistochemistry. Functional assays were used to assess the role of CENPL in HCC cell proliferation, apoptosis, cell cycle, and glycolysis. The potential pathways underlying the regulatory effects of CENPL, as well as the expression of mitogen-activated protein kinase (MAPK) signaling pathway-related molecules and markers of proliferation and glycolysis were investigated. CENPL was significantly upregulated in HCC tissue and associated with multiple clinicopathological features and poor patient prognosis. Univariate and multivariate analyses demonstrated that CENPL may serve as an independent prognostic factor for HCC. Upregulation of CENPL in HCC regulated tumor proliferation and glycolytic processes. Mechanistic studies revealed that differentially expressed genes between the CENPL-overexpressing and control groups were mainly concentrated in the MAPK signaling pathway. Pathway inhibition analysis indicated that CENPL activated the MEK1/2-ERK1/2 signaling pathway to promote proliferation and glycolysis in HCC cells. This study elucidated the role of CENPL in regulating cell proliferation, apoptosis, cell cycle, and glycolysis in HCC. CENPL may represent a therapeutic target and prognostic biomarker for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MAP Kinase Signaling System , Cell Line, Tumor , Cell Cycle/genetics , Cell Proliferation/genetics , Apoptosis/genetics , Glycolysis/genetics , Gene Expression Regulation, Neoplastic , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/genetics
4.
Sci Rep ; 13(1): 21488, 2023 12 06.
Article in English | MEDLINE | ID: mdl-38057406

ABSTRACT

Reticulocalbin 1 (RCN1), a calcium-binding protein located in the endoplasmic reticulum (ER) lumen, contains six conserved regions. Its main functions include maintaining intracellular homeostasis and regulating cell proliferation and apoptosis, and it plays an important role in the development of various tumours. However, the exact function of RCN1 in oral squamous cell carcinoma (OSCC) is not fully understood. Therefore, the aim of this study was to investigate the effects of RCN1 on the biological behaviour of OSCC and the regulation of tumour-associated macrophage (TAM) polarization. The expression of RCN1 in OSCC and normal oral mucosa was evaluated through bioinformatics analysis and immunohistochemical staining. The growth, migration, and invasion of OSCC cells were observed after knockdown of RCN1 using CCK-8 and Transwell assays. Apoptosis was detected by flow cytometry. The effect of tumour cell-derived RCN1 on the polarization of THP-1 macrophages was investigated by establishing a coculture model of THP-1 macrophages and OSCC cells. Additionally, changes in the expression levels of relevant proteins were detected using Western blotting. The upregulation of RCN1 in tumour tissues compared to normal oral mucosal tissues is associated with a poor prognosis and can be utilized as a prognostic indicator for OSCC. Knockdown of RCN1 inhibited the proliferation, migration, and invasion of OSCC cells. Additionally, knockdown of RCN1 in Cal-27 and SCC-25 cells resulted in inhibition of the M2 polarization of THP-1 macrophages. RCN1 knockdown inhibits OSCC progression and M2 macrophage polarization. Targeting RCN1 may be a promising approach for OSCC treatment.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Macrophages/metabolism , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology
5.
Biochem Biophys Res Commun ; 637: 161-169, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36403479

ABSTRACT

The influence of son of sevenless homolog 1 (SOS1) on invasion and metastasis of hepatocellular carcinoma (HCC) cells was investigated. HCC cells were transfected with siRNA and lentivirus to achieve SOS1 knock down/overexpression and changes in RNA and protein levels analyzed by q-PCR and Western blotting (WB). Transwell assay was utilized to assess variations in cell invasion and migration in vitro and by a lung metastasis model of liver cancer in vivo. High expression of SOS1 was observed in most human liver cancers, which indicated a worse prognosis. SOS1 knockout in HepG2 cells significantly decreased cell invasion and migration. SOS1 knockout also reduced the number of metastatic foci in a lung metastasis model of HCC established in nude mice. SOS1 knockout inhibited the epithelial-mesenchymal transition (EMT) in HepG2 cells as well as the PI3K/AKT/mTOR pathway. Overexpression of SOS1 in Huh7 cells had the opposite effect. To conclude, SOS1 may induce the EMT by the activation of the PI3K/AKT/mTOR pathway, thereby enhancing invasion, migration and metastasis of HCC cells. These findings may expose SOS1 as a new HCC therapeutic target.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , SOS1 Protein , Animals , Humans , Mice , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Mice, Nude , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , RNA, Small Interfering , SOS1 Protein/genetics , TOR Serine-Threonine Kinases
6.
Int J Gen Med ; 15: 7493-7510, 2022.
Article in English | MEDLINE | ID: mdl-36187159

ABSTRACT

Purpose: To examine the expression, clinical significance, and potential regulatory mechanism of centromere protein O (CENPO) in hepatocellular carcinoma (HCC). Methods: CENPO expression in pan-cancer was studied using the TCGA-GTEx database, in HCC and normal liver tissues using the GEO and TCGA databases, and in clinical HCC samples by RT-qPCR. The diagnostic value of CENPO was assessed using receiver operating characteristic curves. Univariate and multivariate regression analyses of factors associated with HCC prognosis were performed. CENPO function and its mechanism in HCC were explored using GO, KEGG, and GSEA analyses of differentially expressed genes (DEGs). Association of CENPO expression with immune cell infiltration and immune checkpoint-associated molecules was conducted using TCGA data and the TIMER2.0 database. Relationships between CENPO expression and DNA methylation were analyzed using the UALCAN and cBioPortal databases. CENPO expression in HCC cell lines was detected using RT-qPCR. Results: CENPO is upregulated in most cancers, including HCC and cell lines, and is a potential biomarker for HCC diagnosis (AUC = 0.936, 95% CI: 0.911-0.960). Higher CENPO expression was associated with poorer outcomes in patients with HCC (OS, p = 0.004; DSS, p = 0.002; PFI, p < 0.001), and CENPO was an independent predictor of factors influencing overall survival in HCC. DEGs between samples with high and low CENPO levels were enriched in various biological processes, including activation of the G2M checkpoint and other signaling pathways, while CENPO expression correlated with HCC immune cell infiltration and immune checkpoint-associated molecules, as well as CENPO promoter methylation (p < 0.001). Conclusion: In HCC and cell lines, CENPO is overexpressed, a potential diagnostic marker and an indicator of poor prognosis. CENPO may regulate HCC development by influencing nuclear division and tumor immune infiltration and is regulated by methylation, making it a potential target for HCC immunotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...