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.
Front Microbiol ; 14: 1249289, 2023.
Article in English | MEDLINE | ID: mdl-37808305

ABSTRACT

Background: Glioma is the most common central nervous malignancy. Due to its poor survival outcomes, it is essential to identify novel individualized therapy. Oncolytic virus (OV) treatment is a key therapy regulating tumor microenvironment in malignant glioma. Herein, we aim to identify the key genes after OV infection and its role in glioma. Methods: Performing an RNA-seq analysis, the differentially expressed genes (DEGs) between EV-A71-infection and mock group were screened with GFold values. DAVID online analysis was performed to identify the functional classification. Overall survival (OS) or disease-free survival (DFS) was evaluated to analyze the relation between PTBP1 expression levels and prognosis of glioma patients. Additionally, the ssGSEA and TIMER algorithms were applied for evaluating immune cell infiltration in glioma. Results: Following EV-A71 infection in glioma cells, PTBP1, one of the downregulated DEGs, was found to be associated with multiple categories of GO and KEGG enrichment analysis. We observed elevated expression levels of PTBP1 across various tumor grades of glioma in comparison to normal brain samples. High PTBP1 expression had a notable impact on the OS of patients with low-grade glioma (LGG). Furthermore, we observed an obvious association between PTBP1 levels and immune cell infiltration in LGG. Notably, PTBP1 was regarded as an essential prognostic biomarker in immune cells of LGG. Conclusion: Our research uncovered a critical role of PTBP1 in outcomes and immune cell infiltration of glioma patients, particularly in those with LGG.

2.
Cell Death Discov ; 8(1): 229, 2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35474040

ABSTRACT

Glioma is an intracranial malignant tumor and remains largely incurable. Circular RNAs are prominent modulators in glioma progression. This study investigated the function of circular RNA DLC1 (circDLC1) in the malignant proliferation of glioma cells. circDLC1 expression in glioma tissues and cells was determined using RT-qPCR. The effect of circDLC1 on the malignant proliferation of glioma cells was analyzed using CCK-8, colony formation, and EdU staining assays. METTL3, miR-671-5p, and CTNNBIP1 expressions were determined. N6 methyladenosine (m6A) level of circDLC1 was analyzed using MeRIP. The binding relationship between miR-671-5p and circDLC1 or CTNNBIP1 was verified using RNA pull-down and dual-luciferase assays. A xenograft tumor model was established in nude mice to verify the effect of METTL3-mediated circDLC1 on glioma in vivo. circDLC1 was poorly expressed in glioma. circDLC1 overexpression suppressed glioma cell proliferation. Mechanically, METTL3-mediated m6A modification enhanced circDLC1 stability and upregulated circDLC1 expression in glioma. circDLC1 upregulated CTNNBIP1 transcription by competitively binding to miR-671-5p. METTL3 overexpression repressed the malignant proliferation of glioma via circDLC1/miR-671-5p/CTNNBIP1 in vivo. Collectively, METTL3-mediated m6A modification upregulated circDLC1 expression, and circDLC1 promoted CTNNBIP1 transcription by sponging miR-671-5p, thus repressing the malignant proliferation of glioma.

3.
Biomed Eng Online ; 21(1): 26, 2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35436915

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most malignant grade of glioma. Highly aggressive characteristics of GBM and poor prognosis cause GBM-related deaths. The potential prognostic biomarkers remain to be demonstrated. This research builds up predictive gene targets of expression alterations in GBM utilizing bioinformatics analysis. METHODS AND RESULTS: The microarray datasets (GSE15824 and GSE16011) associated with GBM were obtained from Gene Expression Omnibus (GEO) database to identify the differentially expressed genes (DEGs) between GBM and non-tumor tissues. In total, 719 DEGs were obtained and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for function enrichment analysis. Furthermore, we constructed protein-protein Interaction (PPI) network among DEGs utilizing Search Tool for the Retrieval of Interacting Genes (STRING) online tool and Cytoscape software. The DEGs of degree > 10 was selected as hub genes, including 73 upregulated genes and 21 downregulated genes. Moreover, MCODE application in Cytoscape software was employed to identify three key modules involved in GBM development and prognosis. Additionally, we used the Gene expression profiling and interactive analyses (GEPIA) online tool to further confirm four genes involving in poor prognosis of GBM patients, including interferon-gamma-inducible protein 30 (IFI30), major histocompatibility complex class II-DM alpha (HLA-DMA), Prolyl 4-hydroxylase beta polypeptide (P4HB) and reticulocalbin-1 (RCN1). Furthermore, the correlation analysis indicated that the expression of IFI30, an acknowledged biomarker in glioma, was positively correlated with HLA-DMA, P4HB and RCN1. RCN1 expression was positively correlated with P4HB and HLA-DMA. Moreover, qRT-PCR and immunohistochemistry analysis further validated the upregulation of four prognostic markers in GBM tissues. CONCLUSIONS: Analysis of multiple datasets combined with global network information and experimental verification presents a successful approach to uncover the risk hub genes and prognostic markers of GBM. Our study identified four risk- and prognostic-related gene signatures, including IFI30, HLA-DMA, P4HB and RCN1. This gene sets contribute a new perspective to improve the diagnostic, prognostic, and therapeutic outcomes of GBM.


Subject(s)
Glioblastoma , Glioma , Biomarkers, Tumor/genetics , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/genetics , Glioblastoma/metabolism , Glioma/genetics , Humans , Prognosis
4.
PLoS One ; 16(12): e0260864, 2021.
Article in English | MEDLINE | ID: mdl-34852024

ABSTRACT

Secreted frizzled-related protein 2 (SFRP2) is a glycoprotein with frizzled-like cysteine-rich domain that binds with Wnt ligands or frizzled receptors to regulate Wnt signaling. SFRP2 is frequently hypermethylated in glioma patients, and analysis of TCGA data indicates that SFRP2 is one of the most downregulated genes in radiotherapy treated glioma patients. In the present study, we aimed to explore the potential function of SFRP2 in tumorigenesis and radioresistance of glioma. The RNA sequencing data of TCGA glioma samples were downloaded and analyzed. SFRP2 expression in 166 glioma patients was evaluated by qRT-PCR. The potential functions of SFRP2 in glioma were evaluated by loss-of-function assays and gain-of-function assays in glioma cell lines. We found that SFRP2 was downregulated in radiotherapy-treated glioma patients, and low SFRP2 expression was correlated with advanced tumor stage and poor prognosis. CRISP/Cas9-meidated SFRP2 knockdown promoted soft agar colony formation, cancer stemness and radioresistance of glioma cells, while enforced SFRP2 expression exhibited opposite effects. Moreover, Wnt/ß-catenin signaling was activated in radiotherapy treated glioma patients. SFRP2 knockdown activated Wnt/ß-catenin signaling in glioma cell lines, while overexpression of SFRP2 inhibited Wnt/ß-catenin activation. Besides, pharmacological inhibition of Wnt/ß-catenin signaling by XAV-939 abrogated the effects of SFRP2 knockdown on cancer stemness and radioresistance of glioma cells. Our data for the first time demonstrated a role of SFRP2 in radioresistance of glioma cells, and suggested that inhibition of Wnt/ß-catenin signaling might be a potential strategy for increasing radiosensitivity of glioma patients.


Subject(s)
Brain Neoplasms/pathology , Down-Regulation , Gene Expression Profiling/methods , Glioma/pathology , Membrane Proteins/genetics , Neoplastic Stem Cells/metabolism , Radiation Tolerance , Animals , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioma/genetics , Glioma/radiotherapy , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Male , Mice , Neoplasm Grading , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Prognosis , Radiation Tolerance/drug effects , Sequence Analysis, RNA , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/radiation effects
5.
J Clin Lab Anal ; 35(3): e23697, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33458871

ABSTRACT

BACKGROUND: This study aimed to explore the association of long non-coding RNA urothelial carcinoma-associated 1 (lncRNA UCA1) expression with disease severity, inflammation, and prognosis in acute ischemic stroke (AIS) patients. METHODS: The lncRNA UCA1 expression of blood CD4+ T cells from 160 first-episode AIS patients and 160 non-AIS patients with high-stroke-risk factors (as controls) was detected by reverse transcription quantitative polymerase chain reaction. For AIS patients, interleukin (IL)-6, IL-17, and intracellular adhesion molecule-1 (ICAM1) were determined by enzyme-linked immunosorbent assay; Th17 cell ratio in CD4+ T cells was detected by flow cytometry. Their follow-up data were recorded up to 36 months, recurrence of stroke or death. The recurrence-free survival (RFS) analysis was assessed according to the follow-up data. RESULTS: LncRNA UCA1 expression was higher in AIS patients compared to controls (p < 0.001), and it was positively correlated to national institute of health stroke scale score (r = 0.436, p < 0.001), Th17 cell ratio (r = 0.398, p < 0.001), IL-6 (r = 0.204, p = 0.010), IL-17 (r = 0.326, p < 0.001), and ICAM1 (r = 0.276, p < 0.001) in AIS patients. Regarding prognosis, lncRNA UCA1 expression was elevated in 2-year recurrence/death AIS patients compared to those patients without recurrence or death within 2 years (p = 0.033), also increased in 3-year recurrence/death AIS patients compared to those patients without recurrence or death within 3 years (p = 0.008). Furthermore, high lncRNA UCA1 expression was associated with worse accumulating RFS (p = 0.017) in AIS patients. CONCLUSION: LncRNA UCA1 might sever as a candidate prognostic biomarker in AIS patients, suggesting its potency for AIS management.


Subject(s)
Inflammation/etiology , Ischemic Stroke/etiology , Ischemic Stroke/mortality , RNA, Long Noncoding/blood , Th17 Cells/pathology , Aged , Case-Control Studies , Female , Gene Expression , Humans , Inflammation/genetics , Intercellular Adhesion Molecule-1/blood , Interleukin-17/blood , Interleukin-6/blood , Ischemic Stroke/blood , Male , Middle Aged , Prognosis , Severity of Illness Index
6.
Cancer Biol Ther ; 16(9): 1308-15, 2015.
Article in English | MEDLINE | ID: mdl-26176806

ABSTRACT

Glioma is the most common malignant brain tumors with poor prognosis. The molecular events involved in the development and progression of glioma remain unclear. In this study, the expression levels of miR-302c-3p were examined in glioma tissues by qRT-PCR. The in vitro and in vivo functional effects of miR-302c-3p were examined further. Luciferase reporter assays were conducted to confirm the targeting associations. Results showed that the expression level of miR-302c-3p in glioma tissues was significantly lower than those in normal brain tissues (P < 0.001). The decreased expression of mi-302c-3p in glioma was positively associated with WHO grade (P < 0.001). Up-regulation of MTDH was also detected in glioma tumors compared with normal brain tissues (P = 0.0027) and is inversely correlated with miR-302c-3p expression (P = 0.003, R(2) = 0.4065). MTDH mRNA is a direct target of miR-302c-3p, whose ectopic expression decreases MTDH expression through binding to its 3'-untranslated region. Overexpression of miR-302c-3p results in a dramatic inhibition of glioma cells proliferation and invasion in vitro and in vivo. These data suggest that miR-302c-3p play a pivotal role in the progression of glioma by targeting MTDH and is a potential inhibitor in glioma treatment.


Subject(s)
Brain Neoplasms/metabolism , Cell Adhesion Molecules/genetics , Glioma/metabolism , MicroRNAs/genetics , Adolescent , Adult , Aged , Animals , Base Sequence , Binding Sites , Brain Neoplasms/pathology , Carcinogenesis , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Child , Child, Preschool , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans , Male , Membrane Proteins , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , RNA Interference , RNA-Binding Proteins , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...