Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Technol Cancer Res Treat ; 23: 15330338241262724, 2024.
Article in English | MEDLINE | ID: mdl-38860335

ABSTRACT

OBJECTIVES: Gastric cancer (GC) is one of the most prevalent malignancies worldwide, and early detection is crucial for improving patient survival rates. We aimed to identify immune infiltrating cell-related biomarkers in early gastric cancer (EGC) progression. METHODS: The GSE55696 and GSE130823 datasets with low-grade intraepithelial neoplasia (LGIN), high-grade intraepithelial neoplasia (HGIN), and EGC samples were downloaded from the Gene Expression Omnibus database to perform an observational study. Immune infiltration analysis was performed by single sample gene set enrichment analysis and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data. Weighted gene co-expression network analysis was used to explore the co-expression modules and genes, and further enrichment analysis was performed on these genes. A protein-protein interaction (PPI) network of these genes was constructed to identify biomarkers associated with EGC progression. Screened hub genes were validated by the rank sum test and reverse transcription quantitative polymerase chain reaction. RESULTS: Immune scores were significantly elevated in EGC samples compared to LGIN and HGIN samples. The green-yellow module exhibited the strongest correlation with both immune score and disease progression. The 87 genes within this module were associated with the chemokine signaling pathways, the PI3K-Akt signaling pathways, leukocyte transendothelial migration, and Ras signaling pathways. Through PPI network analysis, the hub genes identified were protein tyrosine phosphatase receptor-type C (PTPRC), pleckstrin, CD53, CD48, lymphocyte cytosolic protein 1 (LCP1), hematopoietic cell-specific Lyn substrate 1, IKAROS Family Zinc Finger 1, Bruton tyrosine kinase, and Vav guanine nucleotide exchange factor 1. Notably, CD48, LCP1, and PTPRC showed high expression levels in EGC samples, with the remaining hub genes demonstrating a similar expression trend. CONCLUSION: This study identified 9 immune cell-related biomarkers that may be actively involved in the progression of EGC and serve as potential targets for GC diagnosis and treatment.


Subject(s)
Biomarkers, Tumor , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Lymphocytes, Tumor-Infiltrating , Protein Interaction Maps , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Biomarkers, Tumor/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Computational Biology/methods , Databases, Genetic , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
2.
Cancer Sci ; 115(5): 1665-1679, 2024 May.
Article in English | MEDLINE | ID: mdl-38475675

ABSTRACT

Cholangiocarcinoma often remains undetected until advanced stages due to the lack of reliable diagnostic markers. Our goal was to identify a unique secretory protein for cholangiocarcinoma diagnosis and differentiation from other malignancies, benign hepatobiliary diseases, and chronic liver conditions. We conducted bulk RNA-seq analysis to identify genes specifically upregulated in cholangiocarcinoma but not in most other cancers, benign hepatobiliary diseases, and chronic liver diseases focusing on exocrine protein-encoding genes. Single-cell RNA sequencing examined subcellular distribution. Immunohistochemistry and enzyme-linked immunosorbent assays assessed tissue and serum expression. Diagnostic performance was evaluated via receiver-operating characteristic (ROC) analysis. Inter-alpha-trypsin inhibitor heavy chain family member five (ITIH5), a gene encoding an extracellular protein, is notably upregulated in cholangiocarcinoma. This elevation is not observed in most other cancer types, benign hepatobiliary diseases, or chronic liver disorders. It is specifically expressed by malignant cholangiocytes. ITIH5 expression in cholangiocarcinoma tissues exceeded that in nontumorous bile duct, hepatocellular carcinoma, and nontumorous hepatic tissues. Serum ITIH5 levels were elevated in cholangiocarcinoma compared with controls (hepatocellular carcinoma, benign diseases, chronic hepatitis B, and healthy individuals). ITIH5 yielded areas under the ROC curve (AUCs) from 0.839 to 0.851 distinguishing cholangiocarcinoma from controls. Combining ITIH5 with carbohydrate antigen 19-9 (CA19-9) enhanced CA19-9's diagnostic effectiveness. In conclusion, serum ITIH5 may serve as a novel noninvasive cholangiocarcinoma diagnostic marker.


Subject(s)
Bile Duct Neoplasms , Biomarkers, Tumor , Cholangiocarcinoma , Proteinase Inhibitory Proteins, Secretory , Aged , Female , Humans , Male , Middle Aged , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/blood , Bile Duct Neoplasms/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , CA-19-9 Antigen/blood , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/blood , Cholangiocarcinoma/genetics , Diagnosis, Differential , Gene Expression Regulation, Neoplastic , Liver Neoplasms/diagnosis , Liver Neoplasms/blood , Liver Neoplasms/genetics , Proteinase Inhibitory Proteins, Secretory/blood , Proteinase Inhibitory Proteins, Secretory/genetics , ROC Curve , Up-Regulation
4.
Medicine (Baltimore) ; 102(37): e35300, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37713870

ABSTRACT

This study explored the potential role of long noncoding RNA (lncRNAs) associated with genomic instability in the diagnosis and treatment of pancreatic adenocarcinoma (PAAD). Transcriptome and single-nucleotide variation data of PAAD samples were downloaded from the cancer genome atlas database to explore genomic instability-associated lncRNAs. We constructed a genomic instability-associated lncRNA prognostic signature. Then gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses were used to explore the physiological role of lncRNAs involved in genomic instability. Tumor microenvironments, immunotherapy response, immune cell infiltration, immune checkpoint, and drug sensitivity were compared between high-risk and low-risk groups. In vitro experiments were performed for external validation. Six lncRNAs associated with genomic instability were identified, capable of predicting the prognosis of PAAD. Patients were assigned to low-risk or high-risk groups using these biomarkers, with better or worse prognosis, respectively. The tumor immune score, immune cell infiltration, and efficacy of immunotherapy were worse in the high-risk group. A drug sensitivity analysis revealed the high- and low-risk groups had different half-maximal inhibitory concentrations. The expression of cancer susceptibility candidate 8 was significantly higher in tumor tissues than in normal tissues, while the expression of LYPLAL1-AS1 exhibited an opposite pattern. They may be potential diagnostic or prognostic biomarkers for patients with pancreatic cancer. Genomic instability-associated lncRNAs were explored in this study and predicted the prognosis of PAAD and stratified patients risk in PAAD. These lncRNAs also predicted the efficacy of immunotherapy and potential therapeutic targets in PAAD.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , RNA, Long Noncoding , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Long Noncoding/genetics , Genomic Instability , Tumor Microenvironment/genetics , Pancreatic Neoplasms
5.
BMC Med Genomics ; 16(1): 220, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37723519

ABSTRACT

BACKGROUND: There is still a therapeutic challenge in treating gastric cancer (GC) due to its high incidence and poor prognosis. Collagen type V alpha 2 (COL5A2) is increased in various cancers, yet it remains unclear how it contributes to the prognosis and immunity of GC. METHODS: The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets were used to download transcriptome profiling (TCGA-STAD; GSE84437), single-cell RNA sequencing (scRNA-seq) data (GSE167297) and clinical information. COL5A2 expression and its relationship with clinicopathological factors were analyzed. We conducted survival analysis and Cox regression analysis to evaluate the prognosis and independent factors of GC. Co-expressed analysis was also performed. To identify the underlying mechanism, we conducted analyses of differentially expressed genes (DEGs) and functional enrichment. The correlations between COL5A2 expression and immune cell infiltration levels and immune infiltrate gene marker sets were further explored. Additionally, we analyzed the association of COL5A2 expression with immunological checkpoint molecules. Furthermore, the relationship between COL5A2 expression and immunotherapy sensitivity was also investigated. RESULTS: COL5A2 expression was elevated in GC. More than this, the scRNA-seq analysis revealed that COL5A2 expression had a spatial gradient. The upregulated COL5A2 was associated with worse overall survival. A significant correlation was found between COL5A2 overexpression and age, T classification and clinical stage in GC. COL5A2 was found to be an independent factor for the unfortunate outcome in Cox regression analysis. The co-expressed genes of COL5A2 were associated with tumor stage or poor survival. Enrichment analysis revealed that the DEGs were mainly associated with extracellular matrix (ECM)-related processes, PI3K-AKT signaling pathway, and focal adhesion. GSEA analyses revealed that COL5A2 was associated with tumor progression-related pathways. Meanwhile, COL5A2 expression was correlated with tumor-infiltrating immune cells. Moreover, immunophenoscore (IPS) analysis and PRJEB25780 cohorts showed that patients with low COL5A2 expression were highly sensitive to immunotherapy. CONCLUSIONS: COL5A2 might act as a prognostic biomarker of GC prognosis and immune infiltration and may provide a therapeutic intervention strategy.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Prognosis , Phosphatidylinositol 3-Kinases , Transcriptome , Gene Expression Profiling , Sequence Analysis, RNA
6.
Eur J Med Chem ; 259: 115698, 2023 Nov 05.
Article in English | MEDLINE | ID: mdl-37542991

ABSTRACT

The RAS gene, also known as the mouse sarcoma virus, includes three genes (KRAS, HRAS, and NRAS) that are associated with human tumors. Among them, KRAS has the highest incidence of mutations in cancer, accounting for around 80% of cases. At the molecular level, the RAS gene plays a regulatory role in transcription and translation, while at the cellular level, it affects cell proliferation and migration, making it crucial for cancer development. In 2021, the FDA approved AMG510, the first direct inhibitor targeting the KRAS-G12C mutation, which has shown tumor regression, prolonged survival, and low off-target activity. However, with the increase of drug resistance, a single inhibitor is no longer sufficient to achieve the desired effect on tumors. Therefore, a large number of other highly efficient inhibitors are being developed at different stages. This article provides an overview of the mechanism of action targeting KRAS-G12C in the KRASGTP-KRASGDP cycle pathway, as well as the structure-activity relationship, structure optimization, and biological activity effects of inhibitors that target the upstream and downstream pathways, or combination therapy.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Animals , Mice , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Mutation , Neoplasms/pathology , Structure-Activity Relationship , Cell Proliferation
7.
Ir J Med Sci ; 192(6): 2653-2660, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37017854

ABSTRACT

OBJECTIVE: Chaperonin-containing TCP1 subunit 6A (CCT6A) facilitates several malignant cancer behaviors, but its regulation of esophageal squamous cell carcinoma (ESCC) has not been reported. This study aimed to investigate the effect of CCT6A on cell proliferation, apoptosis, invasion and epithelial-mesenchymal transition (EMT) and its interaction with the TGF-ß/Smad/c-Myc pathway in ESCC. METHODS: CCT6A expression was detected in ESCC and normal esophageal epithelial cell lines by RT‒qPCR and western blotting. Furthermore, CCT6A siRNA, negative control (NC) siRNA, CCT6A encoding plasmid and NC encoding plasmid were transfected into OE21 and TE-1 cells. Subsequently, CCT6A siRNA- and NC siRNA-transfected cells were treated with TGF-ß for rescue experiments. Cell proliferation, apoptosis, invasion, and E-cadherin/N-cadherin and p-Smad2/p-Smad3/c-Myc expression were detected. RESULTS: CCT6A expression was increased in KYSE-180, TE-1, TE-4 and OE21 cells compared with HET-1A cells. In both OE21 and TE-1 cells, CCT6A knockdown inhibited cell proliferation, invasion and N-cadherin expression while promoting cell apoptosis and E-cadherin expression; meanwhile, CCT6A overexpression had the opposite effects. Furthermore, in both OE21 and TE-1 cells, CCT6A knockdown decreased p-Smad2/Smad2, p-Smad3/Smad3 and c-Myc/GAPDH expression; CCT6A overexpression had the opposite effects. Next, TGF-ß facilitated cell proliferation, invasion, and N-cadherin, p-Smad2/Smad2, p-Smad3/Smad2 and c-Myc/GAPDH expression while repressing cell apoptosis and E-cadherin expression in OE21 and TE-1 cells; importantly, TGF-ß could compensate for the regulation of CCT6A knockdown on these activities. CONCLUSION: CCT6A facilitates ESCC malignant activities by activating the TGF-ß/Smad/c-Myc pathway, which sheds light on the identification of a possible therapeutic target in the management of ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/genetics , Cell Movement/genetics , Cell Line, Tumor , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , RNA, Small Interfering , Cadherins , Cell Proliferation , Chaperonin Containing TCP-1
9.
J Int Med Res ; 50(11): 3000605221135475, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36348613

ABSTRACT

Peroral endoscopic myotomy (POEM) has been shown to be an effective treatment for achalasia and with few adverse events. Only a few cases of delayed bleeding have been described. This current case report describes a patient with delayed bleeding in the submucosal tunnel on the eighth day after POEM. The patient was a 21-year-old woman with a 4-month history of dysphagia, vomiting and excessive weight loss. Achalasia was diagnosed according to her symptoms, barium oesophagogram, oesophageal manometry and endoscopy examination. POEM was performed by an experienced operator. On the eighth day after POEM, the patient suddenly developed continuous haematemesis presented with vomiting of fresh blood and melena. An emergency exploratory esophagogastroduodenoscopy was performed. A large number of blood clots were found at the wound and a long haematoma was found along the lining of the submucosal tunnel. Re-entry into the submucosal tunnel and exposure of the haemorrhagic site was attempted but failed because of severe submucosal tissue adhesion. An emergency angiography was undertaken and haemostasis was achieved with superselective arterial microcoil embolization.


Subject(s)
Esophageal Achalasia , Myotomy , Humans , Female , Young Adult , Adult , Esophageal Achalasia/surgery , Esophageal Achalasia/diagnosis , Manometry , Treatment Outcome , Vomiting , Esophageal Sphincter, Lower
10.
Int J Gen Med ; 15: 6387-6403, 2022.
Article in English | MEDLINE | ID: mdl-35942290

ABSTRACT

Background: Effective prognostic assessment and appropriate drug selection are important for the clinical management of pancreatic cancer (PaC). Here, we aimed to establish a pyroptosis-associated genes (PRGs) signature to predict the prognostic outcomes of PaC and guide clinical drug therapy. Methods: We identified the differentially expressed PRGs between pancreatic adenocarcinoma (n = 178) and control pancreas samples (n = 171) obtained from different databases, and performed Lasso and Cox regression analysis to create a prognosis signature. Kaplan-Meier (K-M) survival curves and time-dependent receiver operating characteristics were further constructed to assess the utility of the risk model. The International Cancer Genome Consortium (ICGC) PACA-AU cohort (n = 95) was used as a validation dataset to examine the validity of this prognostic model. The correlations of risk score (RS) with clinical features, immune cell infiltration, tumor mutation burden and half-maximal inhibitory concentrations (IC50) of chemotherapeutic drugs were analyzed, and the expression levels of PRGs in cell lines were detected. Results: A prognostic signature was constructed, which consisted of 4 PRGs (AIM2, IL18, GSMDC and PLCG1). K-M analysis demonstrated a remarkable difference in overall survival (OS) time between low-risk (LR) and high-risk (HR) groups (P < 0.001). The RS contributed to the progression of PaC, and could be a significant independent factor for prognostic prediction. The validation of the ICGC cohort confirmed the effectiveness of the proposed signature. The patients with a HR score in the TCGA cohort had higher tumor mutation burden and more sensitivity to paclitaxel, gemcitabine, 5-fluorouracil and cisplatin than those with a LR score. The differential expression levels of signature genes were verified in vitro. Conclusion: The PRGs signature can be applied for predicting the prognosis of PaC, and may provide useful information for selection of therapeutic drugs.

11.
Biomed Res Int ; 2022: 2747799, 2022.
Article in English | MEDLINE | ID: mdl-35711526

ABSTRACT

Long noncoding RNAs (lncRNAs) are closely associated with a variety of tumors, including stomach adenocarcinoma (STAD). However, the role of 5-methylcytosine- (m5C-) related lncRNAs in STAD is still uncertain. This study investigated the value of m5C-related lncRNAs in prognostic evaluation and immunotherapy of STAD. STAD transcriptome sequencing data were downloaded from The Cancer Genome Atlas (TCGA) database, and m5C-related lncRNAs were screened by Pearson correlation analysis. A prognostic m5C-related lncRNA signature (m5CRLSig) associated with STAD was established using univariate and multivariate Cox regression analysis. We constructed a prognostic risk model for STAD with six m5C-related lncRNAs. The receiver operating characteristic (ROC) curve was used to examine the predictive efficacy. Univariate and multifactorial Cox regression analysis and principal component analysis (PCA) validated m5CRLSig as an independent factor of STAD prognosis. The clinicopathological characteristics of the model showed higher risk scores for stages II-IV, grade 3, N1-3, and death status. The calibration curve of a nomogram revealed that the nomogram had an excellent predictive function for survival risk. Furthermore, the expression of six m5C-related lncRNAs in STAD and paracancerous tissues was detected by quantitative real-time PCR (qRT-PCR), which verified the feasibility of the m5CRLSig as a prognostic marker for STAD. m5C-related lncRNAs were linked to multiple immune-associated pathways, according to gene set enrichment analysis (GSEA). CIBERSORT analysis indicated that m5CRLSig was involved in immune cell infiltration. Risk score was associated with immune checkpoint gene expression, immune function scores, and chemotherapeutic drug sensitivity. Therefore, m5CRLSig can efficiently assess the prognosis of STAD patients and can be used as a biological marker for immunotherapy.


Subject(s)
Adenocarcinoma , RNA, Long Noncoding , Stomach Neoplasms , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy
12.
Biomed Res Int ; 2022: 8607671, 2022.
Article in English | MEDLINE | ID: mdl-35528160

ABSTRACT

Objective: We attempted to evaluate the effects of probiotic-assisted eradication of cytotoxin-associated gene A (cagA)+/vacuolating cytotoxin A (vacA) s1m1 Helicobacter pylori (H. pylori) on the intestinal flora, inflammatory factors, and clinical outcomes. Methods: A total of 180 patients with cagA+/vacA s1m1 H. pylori were randomly divided into two groups. Group A was treated with bismuth quadruple therapy (BQT). Group B was treated with S. boulardii in addition to BQT. The distribution of intestinal flora, serum interleukin-8 (IL-8), IL-17, tumor necrosis factor-α (TNF-α) levels, recovery time of clinical symptoms, total effective rate of clinical symptoms, H. pylori eradication rate, and adverse reactions were observed. Results: 2 weeks after treatment, the contents of Bifidobacterium, Bacteroides, and Lactobacillus in the intestinal tract of Group A decreased, while the amounts of Enterococcus and Enterobacter increased. In Group B, the contents of Bifidobacterium, Bacteroides, and Lactobacillus increased, while the amounts of Enterococcus and Enterobacter did not change significantly. Moreover, the trend of this flora change was still present at 4 weeks after treatment. Compared with Group A, Group B had lower IL-8, IL-17, and TNF-α levels, shorter recovery time of clinical symptoms, higher overall efficiency of clinical symptoms, and lower occurrence of adverse reactions. The eradication rate did not differ significantly between the two groups. Conclusion: BQT can lead to intestinal flora disorders in cagA+/vacA s1m1 H. pylori patients. S. boulardii can improve the distribution of intestinal flora, downregulate immune-inflammatory mediators, and modify clinical symptoms in patients.


Subject(s)
Gastrointestinal Microbiome , Helicobacter Infections , Helicobacter pylori , Probiotics , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bismuth/therapeutic use , Cytotoxins , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Humans , Interleukin-17/therapeutic use , Interleukin-8 , Probiotics/therapeutic use , Tumor Necrosis Factor-alpha
13.
Int J Gen Med ; 15: 3839-3849, 2022.
Article in English | MEDLINE | ID: mdl-35431569

ABSTRACT

Purpose: Knowledge on the potential association between differential gene expression and risk of gastrointestinal stromal tumors (GISTs) is currently limited. We used bioinformatics tools to identify differentially expressed genes in GIST samples and the related signaling pathways of these genes. Patients and Methods: The GSE136755 dataset was obtained from the GEO database and differentially expressed genes (CENPA, CDK1, TPX2, CCNB1, CCNA2, BUB1, AURKA, KIF11, NDC80) were screened using String and Cytoscape bioinformatics tools. Then, three groups of eight patients at high, intermediate and low risk of GIST were selected from patients diagnosed with GIST by immunohistochemistry in our hospital from October 2020 to March 2021. Differential expression of CDK1 and BUB1 was verified by comparing the amount of expressed p21-Activated kinase 4 (PAK4) protein in pathological sections. Results: SPSS26.0 analysis showed that the expression level of PAK4 in GISTs was significantly higher than in normal tissues and paratumoral tissues and there was significant difference among the three groups of patients (P < 0.01). PAK4 levels in paratumoral and normal tissues were negligible with no significant difference between the tissues. Conclusion: CENPA, CDK1, TPX2, CCNB1, CCNA2, BUB1, AURKA, KIF11 and NDC80 gene expression can be used as biomarkers to assess the risk of gastrointestinal stromal tumors whereby expression increases gradually with the increased risk of GIST formation. The genes encode proteins that regulate the division, proliferation and apoptosis of gastrointestinal stromal tumors mainly through PI3K/AKT, MARK, P53, WNT and other signaling pathways.

14.
Int J Gen Med ; 14: 5149-5157, 2021.
Article in English | MEDLINE | ID: mdl-34511997

ABSTRACT

OBJECTIVE: This study aimed to explore the clinical value of endoscopic ultrasonography (EUS) in the endoscopic resection of gastrointestinal stromal tumors (GISTs). METHODS: A retrospective study of 92 patients who were confirmed to have GISTs by endoscopic resection after EUS examination was conducted. The preoperative features of the EUS examination, ultrasound diagnosis, endoscopic resection methods, surgical procedures, complications, and complete degree of lesion resection were recorded. And 16 patients who were diagnosed by endoscopy and EUS and confirmed by surgical operation were included and analyzed in the subsequent part of the investigation (gastroscopy and EUS image analysis, EUS image and risk classification). RESULTS: The preoperative diagnosis rate of EUS and postoperative pathological diagnosis of GISTs was 78.7% (85/108), and the presence of a non-homogeneous echo and liquid anechoic zone in GISTs often indicated higher risk (P < 0.05). There was a positive correlation between tumor size and risk (P < 0.05). CONCLUSION: The endoscopic resection of GISTs is feasible and safe. EUS is of great significance for the diagnosis and risk assessment of GISTs and can assist in the endoscopic resection of GISTs.

15.
Int J Gen Med ; 14: 10439-10446, 2021.
Article in English | MEDLINE | ID: mdl-35002300

ABSTRACT

PURPOSE: We aimed to compare the histological and/or cytological diagnostic outcomes of EUS-FNA using 19G and 22G needles for solid pancreatic lesions and to evaluate the feasibility and safety of 19G needle. PATIENTS AND METHODS: Data from patients with solid pancreatic lesions, who underwent EUS-FNA, were retrospectively retrieved from a single tertiary center from June 2017 to January 2021. The sensitivity, specificity, and accuracy of diagnosis, sample adequacy, number and time of punctures, and adverse events, were compared between the 19G and 22G groups. Univariate and multivariate logistic regression analyses were used to identify optimal factors for a correct histological diagnosis. RESULTS: A total of 186 patients (19G group, n = 90; 22G group, n = 96) were analyzed in the study. The higher sensitivity and accuracy were observed in 19G group than those in the 22G group both in histological evaluation (89.3% vs 76%, p = 0.031; 91.1% vs 79.2%, p = 0.023; respectively) and in the combined histological and cytological evaluations (93.3% vs 81.3%, p = 0.027; 94.4% vs 84.3%, p = 0.027, respectively). However, there were no significant differences in specificity, positive predictive value (PPV), and negative predictive value (NPV). The number of needle passes and the puncture time were significantly lower in the 19G group than that in the 22G group (1.66 ± 0.07 vs 2.25 ± 0.08, p < 0.001; 125.4 ± 4.93s vs 169.0 ± 5.6s p < 0.001; respectively). Only 2 cases were failed in the 19G group and no serious complications occurred. Univariate and multivariate logistic analyses suggested that CA199 levels and needle types are related to the accuracy of the EUS-FNA histological diagnosis. CONCLUSION: EUS-FNA using a 19G needle is effective and safe for solid pancreatic lesions. Compared with the 22G needle, EUS-FNA with a 19G needle can obtain a better histological diagnostic accuracy of solid pancreatic lesions, and with fewer needle passes and in a shorter time.

16.
Front Genet ; 12: 774010, 2021.
Article in English | MEDLINE | ID: mdl-35003214

ABSTRACT

Long non-coding RNAs (lncRNAs) and their N6-methyladenosine (m6A) modifications play an essential role in tumorigenesis and cancer progression. This study was designed to explore the value of m6A-related lncRNAs in prognosis and therapeutic applications of immune infiltration of colon adenocarcinoma (COAD). We downloaded the COAD gene expression and clinical data from The Cancer Genome Atlas project. By co-expression analysis, Lasso Cox regression analysis, and univariate and multivariate Cox regression, we constructed an independent prognostic signature of seven m6A-related lncRNAs. The prognostic lncRNAs were divided into two clusters by consistent clustering analysis, as well as into two groups of low-high risk based on the signature. Then we identified the relationship between the different groups with clinical features and immune cell infiltration. Cluster 2 had a higher risk score with a lower survival rate. The risk score was higher in groups with advanced clinical features, such as stage III-IV, N1-3, and M1. The expression of AC156455.1 was increased in tumor tissues and cluster 2, and the lncRNA ZEB1-AS1 was notably higher in the high-risk group. Five types of immune cells showed differences in two clusters, and most were upregulated in type 2. The expression of memory B cells was positively correlated with the risk score. The prognostic model was verified by the Gene Expression Omnibus (GEO) dataset. Besides, we found that the expression of these seven lncRNAs in tumor tissues was significantly higher than that in normal tissues, which verified the feasibility of the model. Thus, the signature of seven m6A-related lncRNAs can independently predict the prognosis of COAD. This signature is also closely associated with immune cell infiltration, and new therapeutic targets can be explored from this field.

17.
Comput Biol Chem ; 88: 107360, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32841839

ABSTRACT

BACKGROUND: Anatomical subsites always harbor specific biological features in carcinogenesis. The divergent prognosis of proximal gastric cancer (PGC) and distal gastric cancer (DGC) has been reported. The current study aimed to comprehensively interpret anatomic subsite-specific genomic profiles, which may improve the effectiveness of personalized management. METHODS: Survival and genomic data from the online Surveillance, Epidemiology, and End Results (SEER) and The Cancer Genome Atlas (TCGA) databases were queried for prognostic and genetic analysis, respectively. Propensity score matching (PSM) analysis was performed to balance patient epidemiological factors. Differentially expressed genes (DEGs) were analyzed using the DESeq algorithm. Functional enrichment was performed by the clusterProfiler package. The protein-protein interaction network of DEGs was predicted by the online STRING database. RESULTS: A total of 3,955 patient pairs were assembled by PSM in SEER data with even background characteristics. Prognostic analysis indicated worse overall survival of PGC than DGC (17 vs 20 months, p = 0.0002). Genetic analysis of TCGA database identified 280 DEGs, 90 of which were upregulated in the DGC group and the remaining 190 were upregulated in the PGC group. Functional enrichment analysis indicated that kallikrein serine protease activity, ion channel (Na+/Cl-) activity, and cytoskeleton constituent might be attributed to the poor prognosis observed in PGC. Furthermore, alcohol, retinol, and lipoprotein metabolism were the features for DGC malignancy. CONCLUSION: The current study first demonstrated that PGC exerts poorer survival outcome than DGC based on the SEER database. Further bioinformatic investigation depicts the specific genetic features for PGC and DGC, which may generate differences in tumor malignancy. Our findings provide promising genetic targets for future specific and individualized gastric cancer therapy.


Subject(s)
Algorithms , Stomach Neoplasms/genetics , Aged , Computational Biology , Databases, Genetic , Female , Humans , Male , Stomach Neoplasms/diagnosis , Survival Rate
18.
Biomed Res Int ; 2020: 4673153, 2020.
Article in English | MEDLINE | ID: mdl-32685487

ABSTRACT

Stomach adenocarcinoma (STAD) is one of the most common malignancies. But the molecular mechanism is unknown. In this study, we downloaded the transcriptional profiles and clinical data of 344 STAD and 30 normal samples from The Cancer Genome Atlas (TCGA) database. Stromal and immune scores of STAD were calculated by the Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE) algorithm, and association of stromal/immune scores with tumor differentiation/T/N/M stage and survival was investigated. The differentially expressed genes (DEGs) between high and low score groups (based on media) were identified, and prognostic genes over-/underexpressed in both STAD and stromal/immune signature were retrieved. Furthermore, proportions of 22 infiltrating immune cells for the cohort from TCGA were estimated by the Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) algorithm, and association of 22 immune cells with tumor differentiation/T/N/M stage and survival was investigated. Next, coexpression analysis of 22 immune cells and intersection genes over-/underexpressed in both STAD and stromal signature was conducted. We found high stromal and immune scores and macrophage infiltration predicting poor tumor differentiation and severe local invasion, obtained a list of prognostic genes based on stromal-immune signature, and explored the interaction of collagen, chemokines such as CXCL9, CXCL10, and CXCL11, and macrophage through coexpression analysis and may provide novel prognostic biomarkers and immunotherapeutic targets for STAD.


Subject(s)
Adenocarcinoma/immunology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/immunology , Stomach Neoplasms/immunology , Tumor Microenvironment/immunology , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Humans , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology
19.
Exp Cell Res ; 389(2): 111855, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31978385

ABSTRACT

Takeda-G-protein-receptor-5 (TGR5) is a G-protein-coupled receptor (GPCR) activated by bile acids, and mortalin is a multipotent chaperone of the HSP70 family. In the present study, TGR5 was detected by immunohistochemistry (IHC) in extrahepatic cholangiocarcinoma (ECC) specimens, and TGR5 expression in ECC tissues and adjacent tissues was compared. In vitro TGR5 was overexpressed and knocked down in human intrahepatic cholangiocarcinoma (ICC) cell line RBE and human extrahepatic cholangiocarcinoma (ECC) cell line QBC-939 to observe its effects on the biological behavior of cholangiocarcinoma (CC) cells, including proliferation, apoptosis and migration. In vivo xenograft model was constructed to explore the role of TGR5 in CC growth. Proteins that interacted with TGR5 were screened using an immunoprecipitation spectrometry approach, and the identified protein was down-regulated to investigate its contribution to CC growth. The present study demonstrated that TGR5 is highly expressed in CC tissues, and strong TGR5 expression may indicate high malignancy in CC. Furthermore, TGR5 promotes CC cell proliferation, migration, and apoptosis resistance. TGR5 boosts CC growth in vivo. In addition, TGR5 combines with mortalin and regulates mortalin expression in the CC cell line. Mortalin participates in the TGR5-induced increase in CC cell proliferation. In conclusion, TGR5 is of clinical significance based on its implications for the degree of malignancy in patients with CC. Mortalin may be a downstream component regulated by TGR5, and TGR5 promotes cholangiocarcinoma at least partially by interacting with mortalin and upregulating its expression. Both TGR5 and mortalin are positive regulators, and may serve as potential therapeutic targets for CC.


Subject(s)
Bile Duct Neoplasms/pathology , Biomarkers, Tumor/metabolism , Cholangiocarcinoma/pathology , HSP70 Heat-Shock Proteins/metabolism , Mitochondrial Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Apoptosis , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Biomarkers, Tumor/genetics , Cell Proliferation , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Female , Gene Expression Regulation, Neoplastic , HSP70 Heat-Shock Proteins/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mitochondrial Proteins/genetics , Prognosis , Protein Interaction Domains and Motifs , Receptors, G-Protein-Coupled/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Front Genet ; 11: 615680, 2020.
Article in English | MEDLINE | ID: mdl-33613623

ABSTRACT

Analysis of single-cell RNA sequencing (scRNA-seq) data of immune cells from the tumor microenvironment (TME) may identify tumor progression biomarkers. This study was designed to investigate the prognostic value of differentially expressed genes (DEGs) in intrahepatic cholangiocarcinoma (ICC) using scRNA-seq. We downloaded the scRNA-seq data of 33,991 cell samples, including 17,090 ICC cell samples and 16,901 ICC adjacent tissue cell samples regarded as normal cells. scRNA-seq data were processed and classified into 20 clusters. The immune cell clusters were extracted and processed again in the same way, and each type of immune cells was divided into several subclusters. In total, 337 marker genes of macrophages and 427 marker genes of B cells were identified by comparing ICC subclusters with normal subclusters. Finally, 659 DEGs were obtained by merging B cell and macrophage marker genes. ICC sample clinical information and gene expression data were downloaded. A nine-prognosis-related-gene (PRG) signature was established by analyzing the correlation between DEGs and overall survival in ICC. The robustness and validity of the signature were verified. Functional enrichment analysis revealed that the nine PRGs were mainly involved in tumor immune mechanisms. In conclusion, we established a PRG signature based on scRNA-seq data from immune cells of patients with ICC. This PRG signature not only reflects the TME immune status but also provides new biomarkers for ICC prognosis.

SELECTION OF CITATIONS
SEARCH DETAIL