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










Publication year range
1.
Front Immunol ; 14: 1228331, 2023.
Article in English | MEDLINE | ID: mdl-37671150

ABSTRACT

Cancer immunotherapy has exhibited promising antitumor effects in various tumors. Infiltrated regulatory T cells (Tregs) in the tumor microenvironment (TME) restrict protective immune surveillance, impede effective antitumor immune responses, and contribute to the formation of an immunosuppressive microenvironment. Selective depletion or functional attenuation of tumor-infiltrating Tregs, while eliciting effective T-cell responses, represents a potential approach for anti-tumor immunity. Furthermore, it does not disrupt the Treg-dependent immune homeostasis in healthy organs and does not induce autoimmunity. Yet, the shared cell surface molecules and signaling pathways between Tregs and multiple immune cell types pose challenges in this process. Noncoding RNAs (ncRNAs), including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), regulate both cancer and immune cells and thus can potentially improve antitumor responses. Here, we review recent advances in research of tumor-infiltrating Tregs, with a focus on the functional roles of immune checkpoint and inhibitory Tregs receptors and the regulatory mechanisms of ncRNAs in Treg plasticity and functionality.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , RNA, Untranslated , Homeostasis , Immunotherapy , Tumor Microenvironment
2.
J Exp Clin Cancer Res ; 41(1): 174, 2022 May 14.
Article in English | MEDLINE | ID: mdl-35562774

ABSTRACT

BACKGROUND: Hypoxia and inflammation tumor microenvironment (TME) play a crucial role in tumor development and progression. Although increased understanding of TME contributed to gastric cancer (GC) progression and prognosis, the direct interaction between macrophage and GC cells was not fully understood. METHODS: Hypoxia and normoxia macrophage microarrays of GEO database was analyzed. The peripheral blood mononuclear cell acquired from the healthy volunteers. The expression of C-X-C Motif Chemokine Ligand 8 (CXCL8) in GC tissues and cell lines was detected by quantitative reverse transcription PCR (qRT-PCR), western-blot, Elisa and immunofluorescence. Cell proliferation, migration, and invasion were evaluated by cell counting kit 8 (CCK8), colony formation, real-time imaging of cell migration and transwell. Flow Cytometers was applied to identify the source of cytokines. Luciferase reporter assays and chromatin immunoprecipitation were used to identify the interaction between transcription factor and target gene. Especially, a series of truncated and mutation reporter genes were applied to identify precise binding sites. The corresponding functions were verified in the complementation test and in vivo animal experiment. RESULTS: Our results revealed that hypoxia triggered macrophage secreted CXCL8, which induced GC invasion and proliferation. This macrophage-induced GC progression was CXCL8 activated C-X-C Motif Chemokine Receptor 1/2 (CXCR1/2) on the GC cell membrane subsequently hyperactivated Janus kinase 1/ Signal transducer and activator of transcription 1 (JAK/STAT1) signaling pathway. Then, the transcription factor STAT1 directly led to the overexpression and secretion of Interleukin 10 (IL-10). Correspondingly, IL-10 induced the M2-type polarization of macrophages and continued to increase the expression and secretion of CXCL8. It suggested a positive feedback loop between macrophage and GC. In clinical GC samples, increased CXCL8 predicted a patient's pessimistic outcome. CONCLUSION: Our work identified a positive feedback loop governing cancer cells and macrophage in GC that contributed to tumor progression and patient outcome.


Subject(s)
MicroRNAs , Stomach Neoplasms , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , MicroRNAs/genetics , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages
3.
Cancer Chemother Pharmacol ; 89(4): 441-449, 2022 04.
Article in English | MEDLINE | ID: mdl-35195773

ABSTRACT

AIM: Cetuximab is an essential drug for the treatment of wild-type K-RAS colorectal cancer (CRC). It improves the overall survival of patients. However, acquired resistance prevents its clinical efficacy. Tumor heterogeneity may be a nonnegligible reason for cetuximab resistance. We attempted to explore the corresponding molecular mechanism. METHODS: Cetuximab-resistant CRC cell RKO and cetuximab-sensitive CRC cell Caco-2 were applied in this study. Cells were centrifuged to determine the concentration in the culture supernatant (CS). MTT, EdU, and colony formation assays were utilized to evaluate cell survival and proliferation. Chromatin immunoprecipitation (ChIP) and promoter-luciferase reporter assays were employed to confirm the direct binding of transcription factors. Western blot and reverse transcription-polymerase chain reaction (RT-PCR) assays were used to detect the expression of molecular markers in the pathway. RESULTS: Hepatocyte growth factor (HGF) was up-regulated in RKO cell culture supernatant and induced cetuximab resistance in Caco-2 cells. SRY-Box Transcription Factor 8 (SOX8) bound to the promoter sequence of HGF. HGF activated the HGF/MET bypass pathway and induced cetuximab resistance in Caco-2 cells. CONCLUSION: The SOX8/HGF/MET axis played a crucial role in the communication between cetuximab-resistant cells and cetuximab-sensitive cells, inducing treatment resistance.


Subject(s)
Colorectal Neoplasms , Hepatocyte Growth Factor , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/pharmacology , Hepatocyte Growth Factor/therapeutic use , Humans , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , SOXE Transcription Factors
4.
Gastric Cancer ; 25(1): 44-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34247316

ABSTRACT

Gastric cancer (GC) is a common malignant solid tumor that is characterized by high hypoxia. The transcription of genes associated with hypoxia affects tumor occurrence and development. Long non-coding RNAs (lncRNAs) have been reported to play important roles in cancer development. In this study, we screened for differentially expressed ncRNAs (non-coding RNA) and mRNAs between hypoxia-inducible factor-1 (HIF-1α) knockdown GC cells and scrambled GC cells. Microarray data revealed that HIF-1α regulated the expression of LINC01355 (Hypoxia Yield Proliferation Associated LncRNA, HYPAL). HYPAL was found to be significantly upregulated in GC cells and tissues and was correlated with poor GC prognosis. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays revealed that HIF-1α promoted HYPAL expression by binding the promoter region. A regulatory network for the competing endogenous RNA (ceRNA) was constructed using bioinformatics tools. Mechanistic studies revealed that HYPAL acted as a ceRNA of miR-431-5p to regulate CDK14 expression. Carcinogenic effects of HYPAL were evaluated in vitro and in vivo. The HIF-1α/HYPAL/miR-431-5p/CDK14 (Cyclin-dependent kinase 14) axis activated the Wnt/ß-catenin signaling pathway and induced GC cell proliferation while inhibiting apoptosis. In conclusion, HYPAL is a potential molecular target for GC therapy.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Stomach Neoplasms , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stomach Neoplasms/pathology
5.
J Cancer ; 12(8): 2336-2350, 2021.
Article in English | MEDLINE | ID: mdl-33758610

ABSTRACT

Background: Disrupted gene levels are intimately correlated with the occurrence and prognosis of gastric cancer (GC). As genes do not function in isolation, we set out to investigate the possible relationship among mRNA and non-coding RNAs (ncRNAs). Materials and methods: RNA sequencing from 406 cases of GC was acquired through the TCGA database. R packages were utilized to assess differential RNA expression. The competing endogenous RNA (ceRNA) network was predicted using miRcode, miRDB, mirTarBase, Target Scan and constructed by Cytoscape 3.6.1. GO enrichment analysis, KEGG pathway analysis, GSEA, and WGCNA were applied for pathway analysis. The expression of select candidate molecules was confirmed using western blot and RT-PCR in GC cells and tissues. CCK-8, EdU staining, and Transwell assays were conducted to assess the influence of candidate molecules on proliferation and invasion. The gain and loss-of-function were achieved by co-culture with sh-lncRNA, mimics and sh-mRNA. Luciferase reporters were created using the psiCHECK2 vector, and the relative luciferase activity was calculated. Results: Using data from TCGA, we determined differentially expressed RNAs and created a ceRNA regulatory network. Interestingly, we identified a regulatory complex surrounding ANGPT2. We detected that ANGPT2 was highly expressed in GC, which correlated with a worse prognosis. Our findings indicated that ANGPT2 encourages growth, invasion, and epithelial-mesenchymal transition (EMT) in GC. Importantly, miR-145 inhibits ANGPT2 and abrogates its effects. Furthermore, LINC00184, a ceRNA, blocks miR-145, thereby improving ANGPT2-mediated carcinogenesis. Conclusions: Our findings indicate that the LINC00184/miR-145/ANGPT2 pathway has a crucial function in the development of GC and can act as a possible biomarker and targets for GC therapy.

6.
World J Gastroenterol ; 26(44): 6929-6944, 2020 Nov 28.
Article in English | MEDLINE | ID: mdl-33311941

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the most frequently diagnosed gastrointestinal cancers throughout the world. Novel prognostic biomarkers are required to predict the prognosis of GC. AIM: To identify a multi-long noncoding RNA (lncRNA) prognostic model for GC. METHODS: Transcriptome data and clinical data were downloaded from The Cancer Genome Atlas. COX and least absolute shrinkage and selection operator regression analyses were performed to screen for prognosis associated lncRNAs. Receiver operating characteristic curve and Kaplan-Meier survival analyses were applied to evaluate the effectiveness of the model. RESULTS: The prediction model was established based on the expression of AC007991.4, AC079385.3, and AL109615.2 Based on the model, GC patients were divided into "high risk" and "low risk" groups to compare the differences in survival. The model was re-evaluated with the clinical data of our center. CONCLUSION: The 3-lncRNA combination model is an independent prognostic factor for GC.


Subject(s)
RNA, Long Noncoding , Stomach Neoplasms , Biomarkers, Tumor/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Prognosis , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics
7.
J Cancer ; 11(24): 7320-7328, 2020.
Article in English | MEDLINE | ID: mdl-33193896

ABSTRACT

Background: Electrolyte disturbance and systemic inflammation contributes to poor prognosis of cancer patients. Levels of serum sodium and globulin can reflect electrolyte homeostasis and inflammatory state, respectively, therefore have potential as prognostic factors for cancer patients. In this study, we hypothesized that sodium to globulin ratio (SGR) could have superior accuracy in predicting cancer patient survival, than sodium and globulin alone. We therefore sought to investigate its efficacy in prognosis of patients with advanced gastric cancer (GC) receiving first-line chemotherapy. Methods: A total of 265 patients, with advanced GC, were recruited in this retrospective study from January 2014 to January 2019. We first determined SGR cut-off values using the receiver operating characteristic (ROC) analysis, then analyzed the relationship between pretreatment SGR and clinicopathological features and the effect of chemotherapy. Finally, we evaluated progression-free survival (PFS) and overall survival (OS) rates of the entire and subgroup populations using univariate and multivariate logistic regressions. Results: SGR recorded a cut-off value of 5.54, and had a significantly higher area under the curve (AUC) value (0.619, p = 0.001) than fibrinogen (0.575, p = 0.034) and albumin (0.610, p = 0.002) alone. Organ metastasis, and peritoneal invasion ratios, as well as neutrophil and CA72-4 levels varied significantly between the low-SGR (SGR≤ 5.54) and high SGR (SGR> 5.54) groups (all p < 0.05). Specifically, patients in the low-SGR group exhibited significantly lower disease control rates (83.4%) than those in the high-SGR group (97.2%) (p < 0.001). Results from multivariate analysis indicated that high-SGR was an independent risk factor for PFS (Hazard ratio [HR]: 0.539, p < 0.001) and OS (HR: 0.574, p < 0.001). Moreover, patients in the low-SGR group exhibited significantly worse PFS (134 vs. 221 days, p < 0.001) and OS (311 vs. 420 days, p < 0.001) than those in the high-SGR group. Furthermore, subgroup analysis revealed that SGR was still a powerful prognostic indicator in GC patients with good prognosis or normal biochemical indexes, including no peritoneal infiltration, normal neutrophil counts, and normal serum sodium and globulin levels (all p < 0.001). Conclusions: Overall, our findings indicate that SGR is a novel and promising prognostic factor for GC patients. It has superior accuracy, to sodium and globulin alone, hence it is a powerful tool for evaluating effects of treatment, PFS, and OS in patients with advanced GC, who receive first-line chemotherapy.

8.
Onco Targets Ther ; 13: 10785-10795, 2020.
Article in English | MEDLINE | ID: mdl-33122917

ABSTRACT

BACKGROUND: Gastric cancer (GC) accounts for high mortality. RNA methylation has recently gained interest as markers in specific tumors. This study aimed to uncover the function of the roles of 25 RNA methylation regulators in GC. METHODS: RNA sequence and clinical data were downloaded from The Cancer Genome Atlas (TCGA) database. "STRING" and R were performed to analyze the correlation among the methylase. COX and LASSO were performed to screen for prognostic associated RNA methylation regulators. A prognostic model was established based on the expression of methylase. RT-PCR and immunohistochemistry detected the expression of methylase in GC cells and tissue. Kaplan-Meier curve and Cox analysis were applied to evaluate the effectiveness of the model. RESULTS: The prediction model was established based on the expression of m6A RNA methylation regulators FTO (fat mass and obesity-associated) and RBM15 (RNA binding motif protein 15). Based on the model, GC patients were divided into "high risk" and "low risk" groups to compare the differences in survival. The model was re-evaluated with the clinical data of our center. CONCLUSION: The two-methylase combination model was an independent prognostic factor of GC.

9.
Oncol Lett ; 20(4): 10, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32774483

ABSTRACT

The fibrinogen-to-albumin ratio (FAR), reflecting the systemic coagulation, nutritional and inflammation status of patients, has matured into a prognostic marker for several tumor types. However, only a few studies have assessed the utility of the FAR as a prognostic indicator in patients with advanced gastric cancer (GC) receiving first-line chemotherapy. In the present study, 273 patients with advanced GC who received first-line chemotherapy between January 2014 and January 2019 at the Cancer Hospital of China Medical University (Shenyang, China) were retrospectively analyzed. Using the cut-off values determined by receiver operating characteristic (ROC) analysis, the patients were divided into low-FAR (≤10.03) and high-FAR (>10.03), low-fibrinogen (<3.8 g/l) and high-fibrinogen (≥3.8 g/l), and low-albumin (<40.55 g/l) and high-albumin (≥40.55 g/l) groups. The associations of the pretreatment FAR and clinicopathological characteristics with progression-free survival (PFS) and overall survival (OS) were evaluated. In order to estimate the prognostic value of the FAR for patients with poor prognosis or normal fibrinogen and albumin levels, subgroup analyses were performed. The FAR had a higher area under the ROC curve (0.690; 95% CI: 0.628-0.752; P<0.001) compared with either fibrinogen or albumin alone, which are common indicators of coagulation, nutritional and inflammatory indices. A high FAR was significantly associated with a more advanced stage, peritoneal metastasis, increased CA72-4 levels and anemia (all P<0.05). On survival analysis, a low FAR was associated with a longer PFS and OS compared with a high FAR (202 vs. 130 days and 376 vs. 270 days, respectively; both P<0.001), while the hazard ratio (HR) and P-values of the FAR were lower compared with those of fibrinogen and albumin alone on multivariate analysis (PFS: HR=0.638, 95% CI: 0.436-0.932, P=0.020; OS: HR=0.568, 95% CI: 0.394-0.819, P=0.002). Subgroup analysis indicated that among patients with poor prognosis, including multiple metastases, TNM stage IV and abnormal CA72-4 levels, the FAR may be used as an accurate prognostic marker (all P<0.05), and may also reliably identify patients with poor prognosis among those with normal fibrinogen and albumin levels (all P<0.001). The FAR was indicated to be a valuable marker for predicting PFS and OS in patients with advanced GC receiving first-line chemotherapy and is superior to either fibrinogen or albumin alone.

10.
J Immunol Res ; 2020: 6813176, 2020.
Article in English | MEDLINE | ID: mdl-32832571

ABSTRACT

Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer-related mortality worldwide. Inflammation and the nutritional status of patients with GC are important factors affecting the therapeutic effect and prognosis. Inflammatory and nutrition-related markers have been shown to be prognostic factors for patients with GC. However, few studies have investigated the relationship of the prealbumin-to-globulin ratio (PGR) with the prognosis of GC patients. The objective of the present study was to examine whether pretreatment PGR is related to the prognosis and chemotherapy outcomes of in-patients with advanced GC undergoing first-line chemotherapy. We retrospectively reviewed the data of 281 patients with unresectable GC from January 2013 to January 2018. The receiver operating characteristic curve analysis determined the cut-off values for the PGR. The relationship between the PGR and chemotherapy effectiveness was evaluated using the chi-square test. Kaplan-Meier's method was used to plot progression-free survival (PFS) and overall survival (OS) curves, using multivariable Cox regression analysis to identify promising predictors of mortality. The cut-off value for the PGR was 7.21. The high-PGR (≥7.21) group had a higher disease control rate than that of the low-PGR group (93.66% vs. 78.42%, p < 0.001). Kaplan-Meier's analysis showed significantly higher median PFS (189 vs. 125 days, p < 0.001) and OS (350 vs. 288 days, p < 0.001) in the high-PGR group. The multivariate analyses revealed that a high PGR is an independent protective factor in patients with advanced GC, both in terms of PFS (hazard ratio [HR]: 0.672; 95% confidence interval [CI]: 0.527-0.857; p < 0.001) and OS (HR: 0.675; 95% CI: 0.530-0.861; p = 0.002). In conclusion, the prechemotherapy PGR can accurately predict the chemotherapy outcome, PFS, and OS of patients with advanced GC. Therefore, medical practitioners can utilize the PGR as a novel dependable prognostic tool to weigh the prognosis of patients with GC.


Subject(s)
Biomarkers/blood , Prealbumin , Serum Globulins , Stomach Neoplasms/blood , Stomach Neoplasms/mortality , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , ROC Curve , Reference Values , Stomach Neoplasms/diagnosis , Stomach Neoplasms/drug therapy , Treatment Outcome
11.
Exp Mol Pathol ; 116: 104487, 2020 10.
Article in English | MEDLINE | ID: mdl-32622013

ABSTRACT

AIM: Aberrantly expressed long non-coding RNAs (lncRNAs) are critical instigators of gastric cancer (GC) progression and metastasis. The ceRNA (competing endogenous RNAs) network is well-known in modulating tumor pathological and physiological processes. This research aims to determine the more effective molecular mechanisms of lncRNA PCGEM1 (prostate cancer gene expression marker 1). METHODS: Bioinformatics database and Ago2-RIP were performed to predict and verify the potential targets of lncRNA PCGEM1. Both gain- and loss-of-function experiments were carried out to dissect the biological functions of RNAs. Fluorescence in situ hybridization, dual-luciferase reporter assays, western blot, and real-time PCR (RT-PCR) experiments were utilized to determine the pathophysiological pathways of competitive endogenous RNAs (ceRNAs). RESULTS: GC cells expressed high levels of cytoplasmic PCGEM1. Loss-of-function experiments displayed that the silencing of PCGEM1 suppressed metastatic and invasive cell qualities. PCGEM1 was also found to have associations with miR-129-5p. Subsequently, luciferase reporter and RIP experiments, together with RT-PCR, verified that PCGEM1 functioned as a ceRNA of P4HA2 (Prolyl 4-Hydroxylase Subunit Alpha 2) via sponging miR-129-5p to up-regulate P4HA2 expression. Finally, the rescue assays determined that P4HA2 overexpression rescued the inhibited cell invasion and metastasis caused by PCGEM1 down-regulation. CONCLUSION: These findings found that an over-expression of PCGEM1 in GC acts as a miR-129-5p sponge, leading to higher levels of P4HA2. The PCGEM1/miR-129-5p/P4HA2 axis was confirmed to possess a crucial role in GC metastasis and invasion, suggesting its utility as a potential diagnostic and therapeutic biomarker.


Subject(s)
MicroRNAs/genetics , Prolyl Hydroxylases/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , In Situ Hybridization, Fluorescence , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Stomach Neoplasms/pathology
12.
Mol Cancer ; 19(1): 95, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32443980

ABSTRACT

BACKGROUND: Increasing evidence supports the role of small nucleolar RNAs (snoRNAs) and long non-coding RNAs (lncRNAs) as master gene regulators at the epigenetic modification level. However, the underlying mechanism of these functional ncRNAs in colorectal cancer (CRC) has not been well investigated. METHODS: The dysregulated expression profiling of lncRNAs-snoRNAs-mRNAs and their correlations and co-expression enrichment were assessed by GeneChip microarray analysis. The candidate lncRNAs, snoRNAs, and target genes were detected by in situ hybridization (ISH), RT-PCR, qPCR and immunofluorescence (IF) assays. The biological functions of these factors were investigated using in vitro and in vivo studies that included CCK8, trans-well, cell apoptosis, IF assay, western blot method, and the xenograft mice models. rRNA 2'-O-methylation (Me) activities were determined by the RTL-P assay and a novel double-stranded primer based on the single-stranded toehold (DPBST) assay. The underlying molecular mechanisms were explored by bioinformatics and RNA stability, RNA fluorescence ISH, RNA pull-down and translation inhibition assays. RESULTS: To demonstrate the involvement of lncRNA and snoRNAs in 2'-O-Me modification during tumorigenesis, we uncovered a previously unreported mechanism linking the snoRNPs NOP58 regulated by ZFAS1 in control of SNORD12C, SNORD78 mediated rRNA 2'-O-Me activities in CRC initiation and development. Specifically, ZFAS1 exerts its oncogenic functions and significantly up-regulated accompanied by elevated NOP58, SNORD12C/78 expression in CRC cells and tissues. ZFAS1 knockdown suppressed CRC cell proliferation, migration, and increased cell apoptosis, and this inhibitory effect could be reversed by NOP58 overexpression in vitro and in vivo. Mechanistically, the NOP58 protein could be recognized by the specific motif (AAGA or CAGA) of ZFAS1. This event accelerates the assembly of SNORD12C/78 to allow for further guiding of 2'-O-Me at the corresponding Gm3878 and Gm4593 sites. Importantly, silencing SNORD12C or 78 reduced the rRNAs 2'-O-Me activities, which could be rescued by overexpression ZFAS1, and this subsequently inhibits the RNA stability and translation activity of their downstream targets (e.g., EIF4A3 and LAMC2). CONCLUSION: The novel ZFAS1-NOP58-SNORD12C/78-EIF4A3/LAMC2 signaling axis that functions in CRC tumorigenesis provides a better understanding regarding the role of lncRNA-snoRNP-mediated rRNAs 2'-O-Me activities for the prevention and treatment of CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Nuclear Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/genetics , RNA Stability , RNA, Small Nucleolar/chemistry , Ribonucleoproteins, Small Nucleolar/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Onco Targets Ther ; 13: 1365-1374, 2020.
Article in English | MEDLINE | ID: mdl-32110038

ABSTRACT

AIM: Traditional non-invasive diagnostic markers for gastric cancer (GC) exhibit insufficient sensitivity and specificity. Circulating exosomes are clinically useful non-invasive biomarkers for tumor diagnosis. In addition to their potential role in cancer biology, circulating long non-coding RNAs (lncRNAs) are a new class of promising cancer biomarkers. In the present study, we aimed to identify lncRNAs in circulating exosomes with potential as biomarkers for GC detection. METHODS: We compared the expression of CEBPA-AS1 between GC cells and gastric epithelial cells. The biological function of exosomal CEBPA-AS1 was determined by cell phenotype experiments and rescue assays. We also compared the expression of CEBPA-AS1 in cancerous tissue from GC patients and corresponding adjacent normal tissues, as well as the expression of CEBPA-AS1 in plasma exosomes of GC patients and healthy controls. Diagnostic accuracy was assessed by the receiver operating characteristic (ROC) curve and area under the curve (AUC). RESULTS: CEBPA-AS1 was highly expressed in both GC cells and in exosomes secreted by GC cells. In addition, CEBPA-AS1-containing exosomes secreted by GC cells could promote cell proliferation and inhibit apoptosis, thereby inducing the malignant behavior of GC cells. The level of CEBPA-AS1 was also significantly increased in tissues and plasma exosomes of GC patients. Stability tests showed that most plasma CEBPA-AS1 was encased in exosomes, thus avoiding degradation by RNases. We evaluated the diagnostic accuracy of exosome-derived CEBPA-AS1. The AUC value of CEBPA-AS1 in discriminating GC patients from healthy controls was 0.824, which was higher than the diagnostic accuracy of other traditional tumor biomarkers. CONCLUSION: CEBPA-AS1-containing exosomes secreted from GC cells could promote cell proliferation, inhibit apoptosis, and induce GC progression, indicating that exosomal CEBPA-AS1 is involved in cell-to-cell communication in GC carcinogenesis. Exosomal CEBPA-AS1 is a promising new biomarker for clinical diagnosis of GC.

14.
Int J Clin Oncol ; 25(4): 570-583, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31894433

ABSTRACT

BACKGROUND: Gastric cancer (GC) is the most common and aggressive cancer of the digestive system and poses a serious threat to human health. Since genes do not work alone, our aim was to elucidate the potential network of mRNAs and noncoding RNAs (ncRNAs) in this study. METHODS: Transcriptome data of GC were obtained from TCGA. R and Perl were used to obtain the differentially expressed RNAs and construct a competing endogenous RNA (ceRNA) regulatory network. To investigate the biological functions of differentially expressed RNAs, loss-of-function and gain-of-function experiments were performed. Real-time PCR (RT-qPCR), western blot analysis, dual-luciferase reporter assays and fluorescence in situ hybridization were conducted to explore the underlying mechanisms of competitive endogenous RNAs (ceRNAs). RESULTS: Based on TCGA data and bioinformatics analysis, we identified the LINC00163/miR-183/A-Kinase Anchoring Protein 12 (AKAP12) axis. We observed that AKAP12 was weakly expressed in GC and suppressed invasion and metastasis in GC cells, which could be abolished by miR-183. In addition, LINC00163 can be used as a ceRNA to inhibit the expression of miR-183, thus enhancing the anticancer effect of AKAP12. CONCLUSION: Our results demonstrated that weak LINC00163 expression in GC can sponge miR-183 to promote AKAP12. We established that the LINC00163/miR-183/AKAP12 axis plays an important role in GC invasion and metastasis and may be a potential biomarker and target for GC treatment.


Subject(s)
A Kinase Anchor Proteins/genetics , Cell Cycle Proteins/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Stomach Neoplasms/pathology
15.
Int J Cancer ; 146(1): 169-180, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31090062

ABSTRACT

Our previous study demonstrated that heterogeneous nuclear ribonucleoprotein AB (HNRNPAB) is a key gene that facilitates metastasis of hepatocellular carcinoma (HCC). However, the molecular mechanisms behind this relationship are not fully understood. In our study, we utilized long-noncoding RNA (lncRNA) microarrays to identify a HNRNPAB-regulated lncRNA named lnc-ELF209. Our findings from chromatin immunoprecipitation assays indicate that HNRNPAB represses lnc-ELF209 transcription by directly binding to its promoter region. We also analyzed clinical samples from HCC patients and cell lines with quantitative real-time polymerase chain reactions, RNA in situ hybridization and immunohistochemistry, and found that there is a negative relationship between HNRNPAB and lnc-ELF209 expression. Up/downregulation assays and rescue assays indicate that lnc-ELF209 inhibits cell migration, invasion and epithelial-mesenchymal transition regulated by HNRNPAB. This suggests a new regulatory mechanism for HNRNPAB-promoted HCC progression. RNA pull-down and LC-MS/MS were used to determine triosephosphate isomerase, heat shock protein 90-beta and vimentin may be involved in the tumor-suppressed function of lnc-ELF209. Furthermore, we found lnc-ELF209 could stabilize TPI protein expression. We also found that lnc-ELF209 overexpression in HCCLM3 cell resulted in a lower rate of lung metastatic, which suggested a less aggressive HCC phenotype. Collectively, these findings offer new insights into the regulatory mechanisms that underlie HNRNPAB cancer-promoting activities and demonstrate that lnc-ELF209 is a HNRNPAB-regulated lncRNA that may play an important role in the inhibition of HCC progression.


Subject(s)
Carcinoma, Hepatocellular/pathology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/physiology , Liver Neoplasms/pathology , RNA, Long Noncoding/physiology , Animals , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics
16.
World J Gastroenterol ; 25(44): 6508-6526, 2019 Nov 28.
Article in English | MEDLINE | ID: mdl-31802831

ABSTRACT

BACKGROUND: Long noncoding RNAs (lncRNAs) are aberrant and play critical roles in gastric cancer (GC) progression and metastasis. Searching for coexpressed lncRNA clusters or representative biomarkers related to malignant phenotypes of GC may help to elucidate the mechanism of tumor development and predict the prognosis of GC. AIM: To investigate the prognostic value of NOTCH1 associated with lncRNA in T cell acute lymphoblastic leukemia 1 (NALT1) in GC and the mechanism of its involvement in GC invasion and metastasis. METHODS: RNA sequencing and corresponding clinical data were downloaded from The Cancer Genome Atlas database. The significance module was studied by weighted gene coexpression network analysis. A total of 336 clinical samples were included in the study. Gene silencing, reverse transcription polymerase chain reaction, western blotting, scrape motility assay, and Transwell migration assay were used to assess the function of hub-lncRNAs. RESULTS: At the transcriptome level, 3339 differentially expressed lncRNAs were obtained. weighted gene coexpression network analysis was used to obtain 15 lncRNA clusters and observe their coexpression. Pearson's correlation showed that blue module was correlated with tumor grade and survival. NALT1 was the hub-lncRNA of blue module and was an independent risk factor for GC prognosis. NALT1 was overexpressed in GC and its expression was closely related to invasion and metastasis. The mechanism may involve NALT1 regulation of NOTCH1, which is associated with lncRNA in T cell acute lymphoblastic leukemia, through cis regulation, thereby affecting the expression of the NOTCH signaling pathway. CONCLUSION: NALT1 is overexpressed and promotes invasion and metastasis of GC. The mechanism may be related to regulation of NOTCH1 by NALT1 and its effect on NOTCH signaling pathway expression.


Subject(s)
Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/metabolism , Receptor, Notch1/genetics , Signal Transduction/genetics , Stomach Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Datasets as Topic , Disease-Free Survival , Female , Follow-Up Studies , Gastrectomy , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Prognosis , RNA-Seq , Receptor, Notch1/metabolism , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Analysis
17.
Pathol Res Pract ; 215(10): 152569, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31421977

ABSTRACT

AIM: Previous studies have confirmed that overexpression of the long non-coding RNA prostate cancer gene expression marker 1 (PCGEM1) contributes to the invasion and metastasis of gastric cancer (GC) cells. However, the expression of circulating PCGEM1 in the plasma of GC patients and its clinical value remain unclear. METHODS: A total of 317 patients with GC and 100 healthy subjects were enrolled in this study. Circulating PCGEM1 was detected by reverse transcription-polymerase chain reaction. The diagnostic value of plasma PCGEM1 was evaluated by receiver operating characteristic curves and the area under the curve (AUC) value. RESULTS: The expression level of PCGEM1 in the GC group was significantly higher than that in the healthy control subjects. In addition, the PCGEM1 expression level was associated with tumor differentiation and TNM stage. The AUC value of PCGEM1 was higher than that of other conventional tumor markers (CEA, CA12-5, CA72-4, AFP, and CA19-9), although the combination of all markers showed the highest predictive value. CONCLUSION: Plasma PCGEM1 may be a potential novel circulating biomarker for GC diagnosis and prognosis.


Subject(s)
RNA, Long Noncoding/blood , Stomach Neoplasms/diagnosis , Biomarkers, Tumor/blood , Case-Control Studies , Female , Humans , Male , Middle Aged , Stomach/pathology , Stomach Neoplasms/blood , Stomach Neoplasms/pathology
18.
J Physiol Biochem ; 75(3): 379-389, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31290116

ABSTRACT

N6-Methyladenosine (m6A) is the most common posttranscriptional modification of RNA and plays critical roles in cancer pathogenesis. However, the biological function of long noncoding RNA (lncRNA) methylation remains unclear. As a demethylase, ALKBH5 (alkylation repair homolog protein 5) is involved in mediating methylation reversal. The purpose of this study was to investigate lncRNA m6A modification and its role in gastric cancer (GC). Bioinformatics predicted interactions of ALKBH5 with lncRNAs. Five methods were employed to assess the function of nuclear paraspeckle assembly transcript 1 (NEAT1), including gene silencing, RT-PCR, separation of nuclear and cytoplasmic fractions, scrape motility assays, and transwell migration assays. Then, m6A RNA immunoprecipitation and immunofluorescence were used to detect methylated NEAT1 in GC cells. Rescue assays were performed to define the relationship between NEAT1 and ALKBH5. NEAT1 is a potential binding lncRNA of ALKBH5. NEAT1 was overexpressed in GC cells and tissue. Additional experiments confirmed that knockdown of NEAT1 significantly repressed invasion and metastasis of GC cells. ALKBH5 affected the m6A level of NEAT1. The binding of ALKBH5 and NEAT1 influences the expression of EZH2 (a subunit of the polycomb repressive complex) and thus affects GC invasion and metastasis. Our findings indicate a novel mechanism by which ALKBH5 promotes GC invasion and metastasis by demethylating the lncRNA NEAT1. They may be potential therapeutic targets for GC.


Subject(s)
AlkB Homolog 5, RNA Demethylase/physiology , RNA, Long Noncoding/physiology , Stomach Neoplasms , Cell Line, Tumor , Cell Movement , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Humans , Methylation , Neoplasm Invasiveness , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
19.
J Clin Lab Anal ; 33(8): e22958, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31218745

ABSTRACT

OBJECTIVES: The purpose of this study was to explore whether pretreatment potential prognostic factors are related to chemotherapeutic outcomes and the prognosis of inpatients with metastatic colorectal cancer (mCRC) undergoing chemotherapy. MATERIALS AND METHODS: Data from 71 patients with mCRC were analyzed retrospectively. The relationship between the potential prognostic factors before first-line chemotherapy and the clinicopathological characteristics and chemotherapy response of the patients was calculated using Fisher's exact test and the chi-square test. The prognostic factors were analyzed using univariate and multivariate analyses. We analyzed the subgroups using the Mann-Whitney U test. RESULTS: Four factors were eventually used as prognostic factors, namely the albumin-to-globulin ratio (AGR), the fibrinogen-to-albumin ratio (FAR), the prealbumin-to-globulin ratio (PGR), and the fibrinogen-to-prealbumin ratio (FPR); the cutoff values of the four potential prognostic factors were 1.40, 10.63, 5.44, and 18.49, respectively. The high AGR and PGR groups had a higher response rate than that of the low groups. Patients in the low FAR and FPR groups showed a higher objective response rate than the high FAR and FPR groups. Patients with low FPR were associated with a higher disease control rate than patients with high FPR. Higher progression-free survival (PFS) was observed in the high AGR and PGR and low FAR and FPR groups. The AGR, FAR, PGR, and FPR were considered reliable prognostic factors for PFS in a univariate analysis. CONCLUSIONS: The prechemotherapy AGR, FAR, PGR, and FPR were good prognostic factors to predict the chemotherapy response and PFS in patients with mCRC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Colorectal Neoplasms/pathology , Fibrinogen/analysis , Globulins/analysis , Serum Albumin/analysis , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Retrospective Studies , Survival Rate
20.
Mol Med Rep ; 18(3): 3280-3288, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30066932

ABSTRACT

Pancreatic cancer (PCC) is one of the most dangerous types of tumor as it is very difficult to treat and its 5­year survival rate is <6%. To date, there have been no effective therapeutic strategies to treat PCC, thus, novel effective therapeutic methods are required. Tetraspanin 1 (Tspan1) is a novel member of the tetraspanins superfamily and is highly expressed in a variety of types of cancer, including gastric, hepatocellular and colonic carcinomas. However, the detailed functional role of Tspan1 in pancreatic cancer cells is still unclear and further investigation is required to uncover its therapeutic potential for the treatment of different tumor types. The purpose of the present study was to investigate the expression of Tspan1 in human PCC tissues and cells, and explore the effect of Tspan1 silencing on invasion, migration, cell survival and apoptosis in human PCC to clarify its function. Expression levels of Tspan1 were analyzed in human pancreatic cancer tissues and the cell lines Capan­2 and SW1990 using immunohistochemistry staining, reverse transcription­quantitative polymerase chain reaction and western blotting. The effects of downregulation of Tspan1 expression on cell survival, apoptosis, invasion and migration were investigated viaTspan1­small interfering (si)RNA transfection into human PCC cell lines. The results indicated that Tspan1 expression was increased in human PCC tissues compared with the adjacent normal pancreatic tissues. Tspan1 was highly expressed in the human PCC cell lines Capan­2 and SW1990 when compared with the normal pancreatic cell line HPC­Y5. In addition, transfection with siRNA­targeting Tspan1 significantly reduced cell migration and invasion, and increased the cell apoptosis of Capan­2 and SW1990. The present findings highlighted the important role of Tspan1 in human PCC cell migration, invasion and apoptosis. Thus, Tspan1 RNA interference may serve as a potential therapeutic strategy to treat human PCC.


Subject(s)
Apoptosis/genetics , Gene Silencing , Pancreatic Neoplasms/genetics , Tetraspanins/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression , Gene Knockout Techniques , Humans , Immunohistochemistry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Small Interfering/genetics , Tetraspanins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...