Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Crit Rev Eukaryot Gene Expr ; 34(5): 1-13, 2024.
Article in English | MEDLINE | ID: mdl-38842200

ABSTRACT

SIAH2 function as an oncogene in various cancer. However, the roles of SIAH2 in hepatocellular carcinoma (HCC) are still unknown. This study aimed to investigate the roles of SIAH2 in HCC. Immunohistochemistry was used determine SIAH2 and ACSL4 expression in clinical samples. RT-qPCR was used to determine mRNA expression. Western blot assay was applied for determining protein expression. Ubiquitination assay was conducted for determining ubiquitination of ACSL4. Xenograft experiment was applied for determining tumor growth. Flow cytometry was applied to determine the functions of CD4+ and CD8+ T cells. SIAH2 expression was overexpressed in HCC tumors. High levels of SIAH2 predicted poor outcomes. However, SIAH2 knockdown promoted the proliferation of CD8+ T cells as well as promoted the ferroptosis of tumor cells, inhibiting tumor growth in HCC. ACSL4 is required for CD8+ T cell-mediated ferroptosis of HCC cells. However, SIAH2 induced ubiquitination of ACSL4 and inhibited its expression. SIAH2 specific inhibitor menadione promoted the immune checkpoint blockade. Taken together, SIAH2-mediated inactivation of CD8+ T cells inhibits the ferroptosis of HCC via mediating ubiquitination of ACSL4. Therefore, targeting SIAH2 may be a promising strategy for HCC.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Coenzyme A Ligases , Liver Neoplasms , Ubiquitin-Protein Ligases , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Animals , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Line, Tumor , Ubiquitination , Male , Female , Cell Proliferation , Gene Expression Regulation, Neoplastic
2.
Mol Med ; 30(1): 69, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783226

ABSTRACT

BACKGROUND: The Enoyl-CoA hydratase/isomerase family plays a crucial role in the metabolism of tumors, being crucial for maintaining the energy balance and biosynthetic needs of cancer cells. However, the enzymes within this family that are pivotal in gastric cancer (GC) remain unclear. METHODS: We employed bioinformatics techniques to identify key Enoyl-CoA hydratase/isomerase in GC. The expression of ECHDC2 and its clinical significance were validated through tissue microarray analysis. The role of ECHDC2 in GC was further assessed using colony formation assays, CCK8 assay, EDU assay, Glucose and lactic acid assay, and subcutaneous tumor experiments in nude mice. The mechanism of action of ECHDC2 was validated through Western blotting, Co-immunoprecipitation, and immunofluorescence experiments. RESULTS: Our analysis of multiple datasets indicates that low expression of ECHDC2 in GC is significantly associated with poor prognosis. Overexpression of ECHDC2 notably inhibits aerobic glycolysis and proliferation of GC cells both in vivo and in vitro. Further experiments revealed that overexpression of ECHDC2 suppresses the P38 MAPK pathway by inhibiting the protein level of MCCC2, thereby restraining glycolysis and proliferation in GC cells. Ultimately, it was discovered that ECHDC2 promotes the ubiquitination and subsequent degradation of MCCC2 protein by binding with NEDD4. CONCLUSIONS: These findings underscore the pivotal role of the ECHDC2 in regulating aerobic glycolysis and proliferation in GC cells, suggesting ECHDC2 as a potential therapeutic target in GC.


Subject(s)
Cell Proliferation , Nedd4 Ubiquitin Protein Ligases , Stomach Neoplasms , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Enoyl-CoA Hydratase/metabolism , Enoyl-CoA Hydratase/genetics , Gene Expression Regulation, Neoplastic , Glycolysis , Mice, Nude , Nedd4 Ubiquitin Protein Ligases/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Protein Binding , Proteolysis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Ubiquitination , Warburg Effect, Oncologic
3.
Cancer Cell Int ; 24(1): 184, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802855

ABSTRACT

BACKGROUND: Cancer-induced pre-metastatic niches (PMNs) play a decisive role in promoting metastasis by facilitating angiogenesis in distant sites. Evidence accumulates suggesting that microRNAs (miRNAs) exert significant influence on angiogenesis during PMN formation, yet their specific roles and regulatory mechanisms in gastric cancer (GC) remain underexplored. METHODS: miR-605-3p was identified through miRNA-seq and validated by qRT-PCR. Its correlation with the clinicopathological characteristics and prognosis was analyzed in GC. Functional assays were performed to examine angiogenesis both in vitro and in vivo. The related molecular mechanisms were elucidated using RNA-seq, immunofluorescence, transmission electron microscopy, nanoparticle tracking analysis, enzyme-linked immunosorbent assay, luciferase reporter assays and bioinformatics analysis. RESULTS: miR-605-3p was screened as a candidate miRNA that may regulate angiogenesis in GC. Low expression of miR-605-3p is associated with shorter overall survival and disease-free survival in GC. miR-605-3p-mediated GC-secreted exosomes regulate angiogenesis by regulating exosomal nitric oxide synthase 3 (NOS3) derived from GC cells. Mechanistically, miR-605-3p reduced the secretion of exosomes by inhibiting vesicle-associated membrane protein 3 (VAMP3) expression and affects the transport of multivesicular bodies to the GC cell membrane. At the same time, miR-605-3p reduces NOS3 levels in exosomes by inhibiting the expression of intracellular NOS3. Upon uptake of GC cell-derived exosomal NOS3, human umbilical vein endothelial cells exhibited increased nitric oxide levels, which induced angiogenesis, established liver PMN and ultimately promoted the occurrence of liver metastasis. Furthermore, a high level of plasma exosomal NOS3 was clinically associated with metastasis in GC patients. CONCLUSIONS: miR-605-3p may play a pivotal role in regulating VAMP3-mediated secretion of exosomal NOS3, thereby affecting the formation of GC PMN and thus inhibiting GC metastasis.

4.
Br J Cancer ; 127(2): 237-248, 2022 07.
Article in English | MEDLINE | ID: mdl-35637410

ABSTRACT

BACKGROUND: Histone deacetylases (HDACs) have been shown to be involved in tumorigenesis, but their precise role and molecular mechanisms in gastric cancer (GC) have not yet been fully elucidated. METHODS: Bioinformatics screening analysis, qRT-PCR, and immunohistochemistry (IHC) were used to identify the expression of HDAC4 in GC. In vitro and in vivo functional assays illustrated the biological function of HDAC4. RNA-seq, GSEA pathway analysis, and western blot revealed that HDAC4 activated p38 MAPK signalling. Immunofluorescence, western blot, and IHC verified the effect of HDAC4 on autophagy. ChIP and dual-luciferase reporter assays demonstrated that the transcriptional regulation mechanism of HDAC4 and ATG4B. RESULTS: HDAC4 is upregulated in GC and correlates with poor prognosis. In vitro and in vivo assays showed that HDAC4 contributes to the malignant phenotype of GC cells. HDAC4 inhibited the MEF2A-driven transcription of ATG4B and prevented MEKK3 from p62-dependent autophagic degradation, thus activating p38 MAPK signalling. Reciprocally, the downstream transcription factor USF1 enhanced HDAC4 expression by regulating HDAC4 promoter activity, forming a positive feedback loop and continuously stimulating HDAC4 expression and p38 MAPK signalling activation. CONCLUSION: HDAC4 plays an oncogenic role in GC, and HDAC4-based targeted therapy would represent a novel strategy for GC treatment.


Subject(s)
MAP Kinase Kinase Kinase 3/metabolism , MicroRNAs , Stomach Neoplasms , Autophagy/genetics , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MicroRNAs/pharmacology , Repressor Proteins/genetics , Stomach Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/genetics
5.
J Surg Res ; 260: 462-466, 2021 04.
Article in English | MEDLINE | ID: mdl-33272594

ABSTRACT

BACKGROUND: FABP3 is a member of the fatty acid-binding protein (FABP) family, whose role in various cancers has been reported in the past. However, little is known about the role that FABP3 plays in gastrointestinal stromal tumors (GISTs). METHODS: FABP3 expression was analyzed in 119 patients with GISTs using immunohistochemistry and tissue microarrays to interrogate the relationship between expression and prognosis. Kaplan-Meier analysis was used to calculate patient survival rates using complete follow-up data and to evaluate the potential prognostic value of FABP3 using Cox regression analysis. RESULTS: FABP3-positive signals were detected as brown particles located in the cytoplasm using immunohistochemistry. Among the 119 tissue samples, we observed high FABP3 expression in 64 and low or negative expression in 55. Immunohistochemical analyses suggested that FABP3 expression was significantly correlated with tumor size (P = 0.006), mitotic index (P = 0.016), gross classification (P = 0.048), and AFIP-Miettinen risk classification (P = 0.007). Multiple logistic regression analysis showed that the expression of FABP3 was significantly associated with tumor size (P = 0.021). Kaplan-Meier survival curves showed that patients with GISTs with low expression of FABP3 and classified with a very low to moderate AFIP-Miettinen risk had better prognosis. Multivariate analysis further showed that high expression of FABP3 (P = 0.017) was significantly associated with poor 5-year overall survival. CONCLUSIONS: High FABP3 expression has a prognostic value for patients with GISTs.


Subject(s)
Biomarkers, Tumor/metabolism , Fatty Acid Binding Protein 3/metabolism , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Stromal Tumors/diagnosis , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/mortality , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/mortality , Humans , Immunohistochemistry , Logistic Models , Male , Middle Aged , Prognosis , Survival Analysis , Tissue Array Analysis
6.
J Cell Mol Med ; 24(1): 98-113, 2020 01.
Article in English | MEDLINE | ID: mdl-31691494

ABSTRACT

Spondin 2 (SPON2), a member of the Mindin F-Spondin family, identifies pathogens, activates congenital immunity and promotes the growth and adhesion of neurons as well as binding to their receptors, but its role in promoting or inhibiting tumour metastasis is controversial. Here, we investigated its expression levels and mechanism of action in gastric cancer (GC). Western blotting and GC tissue arrays were used to determine the expression levels of SPON2. ELISAs were performed to measure the serum levels of SPON2 in patients with GC. Two GC cell lines expressing low levels of SPON2 were used to analyse the effects of regulating SPON2 expression on proliferation, migration, invasion, the cell cycle and apoptosis. The results revealed that SPON2 was highly expressed in GC tissues from patients with relapse or metastasis. The levels of SPON2 in sera of patients with GC were significantly higher compared with those of healthy individuals and patients with atrophic gastritis. Knockdown of SPON2 expression significantly inhibited the proliferation, migration and invasion of GC cells in vitro and in vivo. Down-regulation of SPON2 arrested the cell cycle in G1/S, accelerated apoptosis through the mitochondrial pathway and inhibited the epithelial-mesenchymal transition by blocking activation of the ERK1/2 pathway. In summary, this study suggests that SPON2 acts as an oncogene in the development of GC and may serve as a marker for the diagnosing GC as well as a new therapeutic target for GC.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Stomach Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Extracellular Matrix Proteins/genetics , Female , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Cell Mol Med ; 24(13): 7637-7651, 2020 07.
Article in English | MEDLINE | ID: mdl-32436333

ABSTRACT

The mechanism by which miR-605-3p regulates hepatocellular carcinoma (HCC) metastasis has not been clarified. In this study, we found that miR-605-3p was down-regulated in HCC and that low miR-605-3p expression was associated with tumour thrombus and tumour satellites. HCC patients with low miR-605-3p expression showed shorter overall survival and disease-free survival after surgery. Overexpression of miR-605-3p inhibited epithelial-mesenchymal transition and metastasis of HCC through NF-κB signalling by directly inhibiting expression of TRAF6, while silencing of miR-605-3p had the opposite effect. We also found that SNHG16 directly bound to miR-605-3p as a competing endogenous RNA. Mechanistically, high expression of SNHG16 promoted binding to miR-605-3p and inhibited its activity, which led to up-regulation of TRAF6 and sustained activation of the NF-κB pathway, which in turn promoted epithelial-mesenchymal transition and metastasis of HCC. TRAF6 increased SNHG16 promoter activity by activating NF-κB, thereby promoting the transcriptional expression of SNHG16 and forming a positive feedback loop that aggravated HCC malignancy. Our findings reveal a mechanism for the sustained activation of the SNHG16/miR-605-3p/TRAF6/NF-κB feedback loop in HCC and provide a potential target for a new HCC treatment strategy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Feedback, Physiological , Liver Neoplasms/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/metabolism , TNF Receptor-Associated Factor 6/metabolism , Base Sequence , Cell Line, Tumor , Cell Survival/genetics , Disease-Free Survival , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Middle Aged , Multivariate Analysis , Neoplasm Metastasis , Prognosis , RNA, Long Noncoding/genetics
8.
Exp Cell Res ; 385(2): 111691, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31678170

ABSTRACT

Rafoxanide is commonly used as anti-helminthic medicine in veterinary medicine, a main compound of salicylanilide. Previous studies have reported that rafoxanide, as an inhibitor of BRAF V600E mutant protein, inhibits the growth of colorectal cancer, multiple myeloma, and skin cancer. However, its therapeutic effect on gastric cancer (GC) and the potential mechanism has not been investigated. Here, we have found that rafoxanide inhibited the proliferation of GC cells in vitro, arrested the cell cycle in the G0/G1 phase, and promoted apoptosis and autophagy in GC cells. Treatment with specific autophagy inhibitor 3-methyladenine drastically inhibited the apoptotic cell death effect by suppressing the switch from autophagy to apoptosis. Mechanistically, we found that rafoxanide inhibited the growth of GC cells in vitro by inhibiting the activity of the PI3K/Akt/mTOR signaling pathway. This process induced autophagy, which essentially resulted in the apoptosis of GC cells. Results from subcutaneous implanted tumor models in nude mice also indicated that rafoxanide inhibited the growth of GC cells in vivo. Taken together, our findings revealed that rafoxanide inhibited the growth of GC cells both in vitro and vivo, indicating a potential drug candidate for the treatment of GC.


Subject(s)
Antineoplastic Agents/therapeutic use , Antiplatyhelmintic Agents/therapeutic use , Apoptosis , Autophagy , Rafoxanide/therapeutic use , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antiplatyhelmintic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rafoxanide/pharmacology , TOR Serine-Threonine Kinases/metabolism
9.
Exp Cell Res ; 381(1): 66-76, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31047882

ABSTRACT

The expression of HS-1-associated protein X-1 (HAX-1) plays a major role in the development of hepatocellular carcinoma (HCC). However, the function of HAX-1 in HCC metastasis is unclear. Quantitative real-time PCR and western blotting were used to examine HAX-1 expression in HCC cell lines with different metastatic potential, and in tumor tissues with or without intrahepatic metastasis. HCC tissue arrays (n = 144) were used to assess correlations between clinicopathological parameters and HAX-1 expression. We also examined the effect of HAX-1 on promoting HCC cell metastasis in vivo and in vitro. The results showed that the expression levels of HAX-1 were higher in metastatic HCC cell lines than in non-metastatic HCC cell lines. HAX-1 was also significantly upregulated in primary HCC tissues with intrahepatic metastasis compared with those without intrahepatic metastasis. HCC in patients with high HAX-1 expression is more likely to metastasize. HAX-1 expression was associated with malignant progression and poor prognosis, and HAX1 silencing inhibited HCC cell migration and invasion in vitro and decreased HCC cell lung metastasis in vivo, whereas HAX-1 overexpression had the inverse effect. Moreover, HAX-1 increased HCC cell metastasis by promoting the epithelial-mesenchymal transition (EMT) process. Finally, we revealed that HAX-1 modulated EMT in HCC cells by increasing NF-κB/p65 nuclear translocation. In conclusion, HAX-1 promotes HCC metastasis by EMT through activating the NF-κB pathway, suggesting that HAX-1 could be a potential therapeutic target for HCC treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neoplasm Metastasis , Adaptor Proteins, Signal Transducing/genetics , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Movement , Disease Progression , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Middle Aged , NF-kappa B/metabolism , Prognosis , Signal Transduction , Up-Regulation
10.
J Surg Oncol ; 119(8): 1108-1121, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30908656

ABSTRACT

BACKGROUND AND OBJECTIVES: Serine protease-3 (PRSS3) is a known contributor to the genesis and development of malignant tumors, although its role in gastric cancer (GC) is still unclear. METHODS: PRSS3 expression in GC tissue samples and its relationship with clinicopathological features were analyzed. Effects of GC cellular responses to the introduction of small interfering RNA (siRNA)-mediated and short hairpin RNA (shRNA)-mediated interference with tumor PRSS3 expression were also assessed. RESULTS: PRSS3 was significantly upregulated in GC tissues, and PRSS3 protein levels were higher in tumors that developed metastases soon after the surgery compared with those that remained metastasis-free. High expression of PRSS3 was associated with tumor N staging and independently predictive of postoperative prognosis in patients with GC. The V1 variant of PRSS3 was primarily detected in GC tissue and cell lines, the others (V2-V4) being scarcely detectable. Methylation and demethylation drugs had no impact on expression levels of any PRSS3 transcriptional variant. The downregulated PRSS3 expression suppressed GC cell growth, migration, and invasion in vitro and in vivo. CONCLUSIONS: PRSS3 appears to act as an oncogene of GC. High PRSS3 expression portends postoperative metastasis, serving as an effective biomarker of poor therapeutic outcomes.


Subject(s)
Stomach Neoplasms/enzymology , Trypsin/biosynthesis , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription, Genetic , Trypsin/genetics
11.
Am J Emerg Med ; 37(2): 254-259, 2019 02.
Article in English | MEDLINE | ID: mdl-29891121

ABSTRACT

BACKGROUND: We evaluated factors associated with mortality in patients with moderate/severe generalized tetanus. METHODS: This retrospective study included patients with moderate/severe generalized tetanus admitted to the Affiliated Hospital of Nantong University (China) between January 2005 and January 2017. Clinical data were extracted from medical records. Patients were divided into two groups based on outcome (survival or death). Factors associated with mortality were analyzed using univariate and multivariate logistic regression. RESULTS: Seventy-five patients were included (57.3% male; age, 57.9 ±â€¯18.4 years; APACHE II score, 10.6 ±â€¯3.4; severe tetanus, 49.3%; mortality, 25.3%). Multivariate analysis identified severe tetanus (odds ratio [OR], 30.364; 95% confidence interval [CI], 2.459-374.896) and APACHE II score (OR, 1.536; 95%CI, 1.051-2.243) as positively associated with mortality, whereas high-calorie nutrition (OR, 0.027; 95%CI, 0.002-0.359) and dexmedetomidine use (OR, 0.035; 95%CI, 0.003-0.467) were negatively associated with mortality (all P < 0.05). CONCLUSION: Tetanus severity and APACHE II score were associated with mortality in patients with generalized tetanus, whereas high-calorie nutrition and dexmedetomidine use reduced the odds of death. High-calorie nutrition and dexmedetomidine administration may improve prognosis in adult patients with moderate/severe generalized tetanus.


Subject(s)
Hospital Departments , Tetanus/mortality , Tetanus/therapy , APACHE , Adult , Aged , China/epidemiology , Combined Modality Therapy , Dexmedetomidine/therapeutic use , Energy Intake , Female , Hospitals, University , Humans , Hypnotics and Sedatives/therapeutic use , Male , Middle Aged , Nutritional Support , Prognosis , Retrospective Studies , Tetanus/diagnosis
12.
Tumour Biol ; 37(7): 9399-410, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26779638

ABSTRACT

The tumor-suppressing role of fibulin-1 has been described in several types of cancers. However, the expression and role of fibulin-1 in the development and progression of gastric cancer (GC) remain largely unknown. In this study, RT-PCR and immunochemistry were used to detect the fibulin-1 expression in GC samples. We have found that the fibulin-1 protein and mRNA levels were downregulated in GC. When investigating the correlation between fibulin-1 expression and clinicopathological characteristics, we have found that low fibulin-1 protein expression was associated with poor tumor differentiation and advanced N stage. Low fibulin-1 protein expression was also an independent prognostic factor for patient survival. To clarify the reason of fibulin-1 downregulation in GC, the mRNA expression and methylation status of fibulin-1 were examined in GC fresh tissue samples (n = 36). We found that the transcriptional expression of fibulin-1 was negatively associated with fibulin-1 promoter hypermethylation, and fibulin-1 hypermethylation was associated with Helicobacter pylori infection. Finally, the effects of fibulin-1 overexpression on cell proliferation and apoptosis were examined. We have found that fibulin-1 overexpression suppressed the growth of GC both in vitro and in vivo and induced apoptosis by increasing cleaved caspase-3 expression. In conclusion, fibulin-1 acts as a tumor suppressor gene, is frequently hypermethylated in GC, and can potentially serve as a useful biomarker for patient prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Calcium-Binding Proteins/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Calcium-Binding Proteins/genetics , Cell Proliferation , Female , Flow Cytometry , Follow-Up Studies , Genes, Tumor Suppressor , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Tumour Biol ; 37(8): 11249-57, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26945573

ABSTRACT

The tumor-suppressing role of Ras-association domain family 10 (RASSF10) has been described in several types of cancers. Here, we evaluated the potential use of the hypermethylation status of the RASSF10 promoter in serum as a new diagnostic and prognostic tool in gastric cancer (GC). We used bisulfite sequencing polymerase chain reaction to examine RASSF10 methylation levels in serum and/or tumor samples from 82 GC, 45 chronic atrophic gastritis (CAG), and 50 healthy control patients. In the serum of GC patients, the median level of RASSF10 methylation was higher at 47.84 % than those in the serum of CAG and healthy control patients at 11.89 and 11.35 %, respectively. The median level of RASSF10 methylation in GC tumor tissue was similarly high at 62.70 %. Furthermore, RASSF10 methylation levels were highly correlated between paired serum and tumor samples from GC patients. We performed receiver-operating characteristic curve analyses to verify that serum RASSF10 methylation levels could effectively distinguish GC from control patients. Moreover, multivariate analyses showed that high serum RASSF10 methylation levels in GC patients were associated with large tumors, lymph node metastasis, and high carcinoembryonic antigen (CEA) levels. Survival analyses showed that GC patients with high serum RASSF10 methylation levels had shorter overall and disease-free survival after D2 lymphadenectomy than those with low levels. High serum RASSF10 methylation levels were also an independent predictor of tumor recurrence and GC patient survival. In conclusion, serum RASSF10 promoter methylation levels can serve as a valuable indicator for the diagnosis and prognosis of GC in the clinic.


Subject(s)
Biomarkers, Tumor/blood , Stomach Neoplasms/diagnosis , Tumor Suppressor Proteins/blood , Tumor Suppressor Proteins/genetics , Adult , Aged , Area Under Curve , Biomarkers, Tumor/genetics , DNA Methylation , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Promoter Regions, Genetic/genetics , ROC Curve , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Stomach Neoplasms/blood , Stomach Neoplasms/genetics
14.
Hepatogastroenterology ; 59(118): 1883-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22172412

ABSTRACT

BACKGROUND/AIMS: RASSF1A hypermethylation is frequently observed in hepatocellular carcinomas (HCC). But the possible related risk factors and prognosis evaluation for this epigenetic alteration are still unknown. METHODOLOGY: Methylation status, mRNA and protein expression of RASSF1A, aflatoxin B1 (AFB1) and polycyclic aromatic hydrocarbon (PAH)-DNA adducts, were examined in 103 cases of HCC. RESULTS: The expression of RASSF1A mRNA (20/103, r=-0.665) and protein (21/103, r=-0.761) were negatively related to RASSF1A hypermethylation (82/103). Multivariate analysis indicated that RASSF1A hypermethylation was related to AFB1- (p=0.046) and PAH-DNA adducts (p=0.040). Other factors including smoking, alcohol drinking, hepatitis B virus infection and clinicopathological parameters were not significantly associated with RASSF1A methylation. No difference in overall survival was observed between patients with and without RASSF1A hypermethylation (p=0.267). CONCLUSIONS: AFB1- and PAH-DNA adducts may be associated with RASSF1A hypermethylation in hepatocarcinogenesis. RASSF1A methylation status may not be a proper predictor of overall survival for HCC.


Subject(s)
Aflatoxin B1/adverse effects , Carcinogens, Environmental/adverse effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , DNA Methylation , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Polycyclic Aromatic Hydrocarbons/adverse effects , Tumor Suppressor Proteins/genetics , Aflatoxin B1/metabolism , Carcinogens, Environmental/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Chi-Square Distribution , China , DNA Adducts/metabolism , Environmental Exposure , Female , Gene Expression Regulation, Neoplastic , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Middle Aged , Multivariate Analysis , Polycyclic Aromatic Hydrocarbons/metabolism , Prognosis , RNA, Messenger/analysis , Risk Assessment , Risk Factors , Time Factors , Tumor Suppressor Proteins/metabolism
15.
Eur J Med Res ; 27(1): 124, 2022 Jul 18.
Article in English | MEDLINE | ID: mdl-35844000

ABSTRACT

BACKGROUND: The effectiveness of laparoscopic total gastrectomy with D2 lymphadenectomy (LTGD2) remains controversial. This meta-analysis compares surgical and survival outcomes of LTGD2 and open total gastrectomy with D2 lymphadenectomy (OTGD2) for gastric cancer. METHODS: Controlled studies comparing LTGD2 and OTGD2 published before November 2021 were retrieved via database searches. We compared intraoperative outcomes, pathological data, postoperative outcomes, 5-year disease-free survival (DFS), and overall survival (OS). RESULTS: 17 studies were included, containing 4742 patients. Compared with OTGD2, the LTGD2 group had less blood loss (mean difference [MD] = - 122.48; 95% CI: - 187.60, - 57.37; P = 0.0002), fewer analgesic medication (MD = -2.48; 95% CI: - 2.69, - 2.27; P < 0.00001), earlier first flatus (MD = - 1.03; 95% CI: - 1.80, - 0.26; P = 0.009), earlier initial food intake (MD = - 0.89; 95% CI: - 1.09, - 0.68; P < 0.00001) and shorter hospital stay (MD = - 3.24; 95% CI: - 3.75, - 2.73; P < 0.00001). The LTGD2 group had lower postoperative total complication ratio (OR = 0.76; 95% CI: 0.62, 0.92; P = 0.006), incision (OR = 0.50; 95% CI:0.31, 0.79; P = 0.003) and pulmonary (OR = 0.57; 95% CI: 0.34, 0.96; P = 0.03) complication rates, but similar rates of other complications and mortality. Total number of dissected lymph nodes were similar, but the number of No. 10 dissected nodes was less with LTGD2 (MD = - 0.31; 95% CI: - 0.46, - 0.16; P < 0.0001). There was no difference in 5-year OS (P = 0.19) and DFS (P = 0.34) between LTGD2 and OTGD2 groups. CONCLUSIONS: LTGD2 produces small trauma, fast postoperative recovery and small length of hospital stays than OTGD2, and had similar long-term clinical efficacy as OTGD2. However, these results still need further high-quality prospective randomized controlled trials confirmation.


Subject(s)
Laparoscopy , Stomach Neoplasms , Gastrectomy/methods , Humans , Laparoscopy/methods , Lymph Node Excision/methods , Postoperative Complications/surgery , Prospective Studies , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Treatment Outcome
16.
J Exp Clin Cancer Res ; 40(1): 352, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34749775

ABSTRACT

BACKGROUND: Angiogenesis plays an important role in the occurrence, development and metastasis of hepatocellular carcinoma (HCC). According to previous studies, miR-378a participates in tumorigenesis and tumor metastasis, but its exact role in HCC angiogenesis remains poorly understood. METHODS: qRT-PCR was used to investigate the expression of miR-378a-3p in HCC tissues and cell lines. The effects of miR-378a-3p on HCC in vitro and in vivo were examined by Cell Counting Kit-8 (CCK-8), Transwell, tube formation and Matrigel plug assays, RNA sequencing, bioinformatics, luciferase reporter, immunofluorescence and chromatin immunoprecipitation (ChIP) assays were used to detect the molecular mechanism by which miR-378a-3p inhibits angiogenesis. RESULTS: We confirmed that miR-378a-3p expression was significantly downregulated and associated with higher microvascular density (MVD) in HCC; miR-378a-3p downregulation indicated a short survival time in HCC patients. miR-378a-3p knockdown led to a significant increase in angiogenesis in vitro and in vivo. We found that miR-378a-3p directly targeted TNF receptor associated factor 1 (TRAF1) to attenuate NF-κB signaling, and then downregulated secreted vascular endothelial growth factor. DNA methyltransferase 1 (DNMT1)-mediated hypermethylation of miR-378a-3p was responsible for downregulating miR-378a-3p. Moreover, a series of investigations indicated that p65 initiated a positive feedback loop that could upregulate DNMT1 to promote hypermethylation of the miR-378a-3p promoter. CONCLUSION: Our study indicates a novel DNMT1/miR-378a-3p/TRAF1/NF-κB positive feedback loop in HCC cells, which may become a potential therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/pathology , Down-Regulation , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Signal Transduction , Transfection
17.
J Biomed Mater Res B Appl Biomater ; 109(3): 451-462, 2021 03.
Article in English | MEDLINE | ID: mdl-32841467

ABSTRACT

The development of novel materials with effective defect-repairing properties will help avoid subtotal gastrectomy in patients with large gastric perforations. We prepared perfused decellularized gastric matrix (PDGM) and analyzed its components, spatial structure, biomechanics, cytotoxicity, and histocompatibility to validate its efficacy in the repair of gastric perforation. PDGM retained large amounts of gastric extracellular matrix, while residual glandular cells and muscle fibers were not found. The spatial structure of the tissue was well preserved, while the DNA and glycosaminoglycan contents were significantly decreased compared with normal gastric tissue (p < .01). There was no obvious deformation of the spatial structure and tissue elasticity of PDGM after sterilization by Cobalt-60 irradiation. The PDGM had good histocompatibility. PDGM was then used to repair a rat gastric perforation model. Radiography of the upper gastrointestinal tract at 24 hr postoperatively revealed no contrast agent leakage. There was evidence of early fibroblast proliferation, which was complicated by capillary regeneration. The hyperplastic gastric gland was slightly disarranged after repair. Defects of the muscular layer also healed a little with the regeneration process. PDGM is a nontoxic biocompatible biological mesh that may be useful for repairing relatively large gastric defects.


Subject(s)
Biocompatible Materials/chemistry , Decellularized Extracellular Matrix/chemistry , Stomach Rupture/surgery , Stomach/chemistry , Surgical Mesh , Tissue Scaffolds/chemistry , Animals , Male , Rats , Rats, Sprague-Dawley
18.
J Exp Clin Cancer Res ; 39(1): 123, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32605589

ABSTRACT

BACKGROUND: 3-Hydroxy butyrate dehydrogenase 2 (BDH2) is a short-chain dehydrogenase/reductase family member that plays a key role in the development and pathogenesis of human cancers. However, the role of BDH2 in gastric cancer (GC) remains largely unclear. Our study aimed to ascertain the regulatory mechanisms of BDH2 in GC, which could be used to develop new therapeutic strategies. METHODS: Western blotting, immunohistochemistry, and RT-PCR were used to investigate the expression of BDH2 in GC specimens and cell lines. Its correlation with the clinicopathological characteristics and prognosis of GC patients was analysed. Functional assays, such as CCK-8 and TUNEL assays, transmission electron microscopy, and an in vivo tumour growth assay, were performed to examine the proliferation, apoptosis, and autophagy of GC cells. Related molecular mechanisms were clarified by luciferase reporter, coimmunoprecipitation, and ubiquitination assays. RESULTS: BDH2 was markedly downregulated in GC tissues and cells, and the low expression of BDH2 was associated with poor survival of GC patients. Functionally, BDH2 overexpression significantly induced apoptosis and autophagy in vitro and in vivo. Mechanistically, BDH2 promoted Keap1 interaction with Nrf2 to increase the ubiquitination level of Nrf2. Ubiquitination/degradation of Nrf2 inhibited the activity of ARE to increase accumulation of reactive oxygen species (ROS), thereby inhibiting the phosphorylation levels of AktSer473 and mTORSer2448. CONCLUSIONS: Our study indicates that BDH2 is an important tumour suppressor in GC. BDH2 regulates intracellular ROS levels to mediate the PI3K/Akt/mTOR pathway through Keap1/Nrf2/ARE signalling, thereby inhibiting the growth of GC.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Hydroxybutyrate Dehydrogenase/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Stomach Neoplasms/pathology , Ubiquitin/metabolism , Animals , Apoptosis , Autophagy , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Female , Humans , Hydroxybutyrate Dehydrogenase/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NF-E2-Related Factor 2/genetics , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
19.
World J Gastroenterol ; 26(31): 4656-4668, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32884223

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract. Surgical resection and tyrosine kinase inhibitors are defined as the main treatments but cannot cure patients with advanced GIST, which eventually develops into recurrence and acquired drug resistance. Therefore, it is necessary to identify prognostic biomarkers and new therapeutic targets for GISTs. CC chemokine receptor type 8 (CCR8) protein participates in regulation of immune responses. Recent studies on CCR8 in non-small cell lung cancer and colorectal cancer showed that it was highly expressed in tumor-infiltrating regulatory T cells and correlated with a poor prognosis. AIM: To detect CCR8 expression in GIST tissues and analyze its relationships with clinicopathological features and prognosis in patients with GISTs. METHODS: Tissue samples were used for the tissue microarrays construction. The microarrays were then subjected to immunohistochemical analyses to detect CCR8 expression. Next, Kaplan-Meier analysis was utilized to calculate the survival rate of patients with complete follow-up data, and the potential prognostic value of CCR8 was evaluated by Cox regression analysis. Finally, a Gene Ontology/Kyoto Encyclopedia of Genes and Genomes single-gene enrichment chart of CCR8 was constructed using the STRING database. RESULTS: CCR8-positive signals were detected as brown or brown-yellow particles by immunohistochemistry located in the cytoplasm. Among 125 tissue samples, 74 had CCR8 high expression and 51 had low or negative expression. Statistical analyses suggested CCR8 was significantly correlated with tumor size, mitotic index, AFIP-Miettinen risk classification and tumor location. Kaplan-Meier and multivariate analyses showed that patients with low or negative CCR8 expression, mitotic index < 5/high-power fields (HPF) and tumor diameter < 5 cm had a better prognosis. Based on the STRING database, CCR8 was significantly enriched in biological processes such as tumor immunity, T lymphocyte chemotaxis, migration and pathways like the nuclear factor-κB and tumor necrosis factor pathways as well as intestinal immune regulation networks. CONCLUSION: CCR8 is a prognostic biomarker for malignant potential of GISTs, with high expression correlated with malignancy and poor prognosis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Lung Neoplasms , Biomarkers, Tumor/genetics , Humans , Neoplasm Recurrence, Local , Prognosis , Receptors, CCR , Receptors, CCR8
20.
Pathol Res Pract ; 216(10): 153143, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32853961

ABSTRACT

The G protein subunit gamma 13 (GNG13) plays an important role in olfaction, vision, and biological behavior. However, our knowledge of the relationship between GNG13 expression and the clinicopathological features of gastrointestinal tumors is insufficient. Therefore, we used the Oncomine database to evaluate the expression of GNG13 mRNA in gastric cancer, the result showed that there was no significant difference in the expression of GNG13 between gastric cancer and adjacent normal tissues, and GNG13 mRNA expression was assessed in 32 matched pairs of Gastrointestinal adenocarcinoma tissues and adjacent normal tissues as well as 32 matched pairs of gastrointestinal stromal tumor (GIST) and adjacent normal tissues by quantitative reverse transcription-polymerase chain reaction analysis. The results suggested that GNG13 is upregulated in gastrointestinal stromal tumors. Immunohistochemical analysis was used to detect the GNG13 in the tissues of 123 patients with GIST. High cytoplasmic expression of GNG13, which was observed in 65.85 % of GIST patients, significantly correlated with mitotic index(P = 0.036) and tumor size(P = 0.024). Multiple logistic regression analysis showed that the expression of GNG13 was significantly associated with tumor size. Kaplan-Meier analysis indicated that high GNG13 expression was associated with poor prognosis of GIST. Multivariate Cox regression analysis indicated that the expression of GNG13, mitotic index and tumor size were independent adverse prognostic factors of GIST. These findings suggest that GNG13 is associated with the malignant phenotype of GIST and may serve as a marker of poor prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Stomach Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Cytoplasm/metabolism , Cytoplasm/pathology , Disease-Free Survival , Female , Gastrointestinal Neoplasms/diagnosis , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Protein Subunits/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL