Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Technol Cancer Res Treat ; 23: 15330338241262724, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38860335

RESUMO

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.


Assuntos
Biomarcadores Tumorais , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Linfócitos do Interstício Tumoral , Mapas de Interação de Proteínas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Biomarcadores Tumorais/genética , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Biologia Computacional/métodos , Bases de Dados Genéticas , Prognóstico , Microambiente Tumoral/imunologia , Microambiente Tumoral/genética
2.
Cancer Sci ; 115(5): 1665-1679, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38475675

RESUMO

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.


Assuntos
Neoplasias dos Ductos Biliares , Biomarcadores Tumorais , Colangiocarcinoma , Proteínas Secretadas Inibidoras de Proteinases , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias dos Ductos Biliares/diagnóstico , Neoplasias dos Ductos Biliares/sangue , Neoplasias dos Ductos Biliares/genética , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Antígeno CA-19-9/sangue , Colangiocarcinoma/diagnóstico , Colangiocarcinoma/sangue , Colangiocarcinoma/genética , Diagnóstico Diferencial , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/genética , Proteínas Secretadas Inibidoras de Proteinases/sangue , Proteínas Secretadas Inibidoras de Proteinases/genética , Curva ROC , Regulação para Cima
3.
Medicine (Baltimore) ; 102(37): e35300, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37713870

RESUMO

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.


Assuntos
Adenocarcinoma , Neoplasias Pancreáticas , RNA Longo não Codificante , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , RNA Longo não Codificante/genética , Instabilidade Genômica , Microambiente Tumoral/genética , Neoplasias Pancreáticas
4.
BMC Med Genomics ; 16(1): 220, 2023 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-37723519

RESUMO

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.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Prognóstico , Fosfatidilinositol 3-Quinases , Transcriptoma , Perfilação da Expressão Gênica , Análise de Sequência de RNA
5.
Eur J Med Chem ; 259: 115698, 2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37542991

RESUMO

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.


Assuntos
Neoplasias , Proteínas Proto-Oncogênicas p21(ras) , Animais , Camundongos , Humanos , Proteínas Proto-Oncogênicas p21(ras)/genética , Mutação , Neoplasias/patologia , Relação Estrutura-Atividade , Proliferação de Células
6.
Ir J Med Sci ; 192(6): 2653-2660, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37017854

RESUMO

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.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Humanos , Transição Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/genética , Movimento Celular/genética , Linhagem Celular Tumoral , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , RNA Interferente Pequeno , Caderinas , Proliferação de Células , Chaperonina com TCP-1
8.
Int J Gen Med ; 15: 6387-6403, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35942290

RESUMO

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.

9.
Biomed Res Int ; 2022: 2747799, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35711526

RESUMO

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.


Assuntos
Adenocarcinoma , RNA Longo não Codificante , Neoplasias Gástricas , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/terapia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia , Prognóstico , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia
10.
Biomed Res Int ; 2022: 8607671, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35528160

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Infecções por Helicobacter , Helicobacter pylori , Probióticos , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Bismuto/uso terapêutico , Citotoxinas , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Humanos , Interleucina-17/uso terapêutico , Interleucina-8 , Probióticos/uso terapêutico , Fator de Necrose Tumoral alfa
11.
Int J Gen Med ; 15: 3839-3849, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35431569

RESUMO

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.

12.
Int J Gen Med ; 14: 5149-5157, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34511997

RESUMO

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.

13.
Front Genet ; 12: 774010, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35003214

RESUMO

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.

14.
Comput Biol Chem ; 88: 107360, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32841839

RESUMO

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.


Assuntos
Algoritmos , Neoplasias Gástricas/genética , Idoso , Biologia Computacional , Bases de Dados Genéticas , Feminino , Humanos , Masculino , Neoplasias Gástricas/diagnóstico , Taxa de Sobrevida
15.
Biomed Res Int ; 2020: 4673153, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32685487

RESUMO

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.


Assuntos
Adenocarcinoma/imunologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/imunologia , Neoplasias Gástricas/imunologia , Microambiente Tumoral/imunologia , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Humanos , Prognóstico , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia
16.
Exp Cell Res ; 389(2): 111855, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-31978385

RESUMO

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.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Biomarcadores Tumorais/metabolismo , Colangiocarcinoma/patologia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas Mitocondriais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Apoptose , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Biomarcadores Tumorais/genética , Proliferação de Células , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteínas Mitocondriais/genética , Prognóstico , Domínios e Motivos de Interação entre Proteínas , Receptores Acoplados a Proteínas G/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Front Genet ; 11: 615680, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33613623

RESUMO

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.

18.
Clin Res Hepatol Gastroenterol ; 43(4): 446-450, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30683632

RESUMO

INTRODUCTION: HBV and/or HCV infection is the main cause of hepatocellular carcinoma (HCC), but the molecular mechanisms by which HBV promotes HCC are not clear. In 2011, the result of a GWAS revealed a common variant of DEPDC5 affected HCC susceptibility in patient with chronic HCV infection in Japan. This study investigated the correlation between DEPDC5 polymorphism and HBV-related HCC. MATERIALS AND METHODS: 1289 samples of Han population were involved in northern China and peripheral blood samples were obtained, including 506 healthy controls, 217 Hepatitis B chronic (CHB) and 258 liver cirrhosis (LC), and 308 HBV-related HCC patients. SNPs in the DEPDC5 rs1012068 were detected by MALDI-TOF-MS. RESULTS: After controlling for the influence of sex, smoking and drinking, this study showed a significant relationship between the polymorphism of DEPDC5 rs1012068 and HBV-related HCC. Healthy participants with CC genotype showed 2.008 (95% CI = 1.145, 3.520; P = 0.015) times more likely to develop HCC; CHB cases with CC genotype showed 2.241 (95% CI = 1.226, 4.461; P = 0.022) times more likely to develop HCC; LC cases with CC genotype showed 2.706 (95% CI = 1.371, 5.340; P = 0.004) times more likely to develop HCC; and individuals with AC genotype showed 1.615 (95% CI = 1.110, 2.352; P = 0.012) times more likely to develop HCC. CONCLUSIONS: There was a significant correlation between DEPDC5 rs1012068A/C and HBV-related HCC in the Han Chinese population. A to C mutation increased the risk of the developing of HBV-related HCC.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas Ativadoras de GTPase/genética , Hepatite B Crônica/genética , Cirrose Hepática/genética , Neoplasias Hepáticas/genética , Polimorfismo de Nucleotídeo Único , Adulto , Alelos , Povo Asiático/genética , Estudos de Casos e Controles , China/etnologia , Feminino , Genótipo , Hepatite B Crônica/complicações , Humanos , Cirrose Hepática/complicações , Masculino , Pessoa de Meia-Idade , Análise Multivariada
19.
Saudi J Gastroenterol ; 24(4): 242-248, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29806597

RESUMO

Background/Aim: : We studied the expression of interleukin-17 and interleukin-22 in the serum and the lower esophageal sphincter (LES) in healthy individuals and in patients diagnosed with achalasia (AC) to gain a better understanding of the etiopathogenesis of AC. Patients and Methods: Our study comprised 14 randomly selected patients with AC who underwent peroral endoscopic myotomy and 14 randomly selected healthy individuals who served as controls. Venous blood samples were evaluated in all study subjects to detect the expression of interleukin-17 and interleukin-22 in the serum using an enzyme-linked immunosorbent assay. Immunohistochemistry studies were performed to evaluate LES myofilaments obtained from both groups, as well as from 12 patients diagnosed with a subendothelial non-invasive tumor and who had undergone submucosal tunneling endoscopic resection, to assess the expression of interleukin-17 and interleukin-22 in LES myofilaments. Results: Compared with that in the control group, the expression of interleukin-17 and interleukin-22 in the serum and LES, in patients with AC, was significantly increased and was positively correlated. Conclusion: Interleukin-17 and interleukin-22 are upregulated in the serum and LES in patients with AC, suggesting that both interleukin-17 and interleukin-22 are involved in the pathogenesis of AC, and that AC may be an immune-mediated inflammatory disease.


Assuntos
Acalasia Esofágica/sangue , Esfíncter Esofágico Inferior/metabolismo , Interleucina-17/biossíntese , Interleucinas/biossíntese , Adulto , Endoscopia/métodos , Endoscopia Gastrointestinal/métodos , Acalasia Esofágica/diagnóstico , Acalasia Esofágica/fisiopatologia , Acalasia Esofágica/cirurgia , Esfíncter Esofágico Inferior/cirurgia , Feminino , Humanos , Incidência , Interleucina-17/sangue , Interleucina-17/metabolismo , Interleucinas/sangue , Interleucinas/metabolismo , Masculino , Manometria/métodos , Pessoa de Meia-Idade , Miotomia/métodos , Interleucina 22
20.
Transl Res ; 182: 88-102, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28034761

RESUMO

Bile acids stimulate intestinal epithelial proliferation in vitro. We sought to investigate the role of the bile acid receptor TGR5 in the protection of intestinal epithelial proliferation in obstructive jaundice. Intestinal tissues and serum samples were obtained from patients with malignant obstructive jaundice and from bile duct ligation (BDL) rats. Intestinal permeability and morphological changes in the intestinal mucosa were observed. The functions of TGR5 in cell proliferation in intestinal epithelial injury were determined by overexpression or knockdown studies in Caco-2 and FHs 74 Int cells pretreated with lipopolysaccharide (LPS). Internal biliary drainage was superior to external biliary drainage in recovering intestinal permeability and mucosal histology in patients with obstructive jaundice. In BDL rats, feeding of chenodeoxycholic acid (CDCA) decreased intestinal mucosa injury. The levels of PCNA, a marker of proliferation, increased in response to CDCA feeding and were paralleled by elevated TGR5 expression. CDCA upregulated TGR5 expression and promoted proliferation in Caco-2 and FHs 74 Int cells pretreated with LPS. Overexpression of TGR5 resulted in increased PCNA, cell viability, EdU incorporation, and the proportion of cells in S phase, whereas knockdown of TGR5 had the opposite effect. Our data indicate that bile acids promote intestinal epithelial cell proliferation and decrease mucosal injury by upregulating TGR5 expression in obstructive jaundice.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/patologia , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Icterícia Obstrutiva/metabolismo , Icterícia Obstrutiva/patologia , Receptores Acoplados a Proteínas G/metabolismo , Idoso , Animais , Ductos Biliares/efeitos dos fármacos , Ductos Biliares/patologia , Biomarcadores/sangue , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Ácido Quenodesoxicólico/farmacologia , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Icterícia Obstrutiva/sangue , Ligadura , Lipopolissacarídeos , Masculino , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA