Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Mol Cancer ; 23(1): 124, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849840

ABSTRACT

BACKGROUND: Intestinal metaplasia (IM) is classified into complete intestinal metaplasia (CIM) and incomplete intestinal metaplasia (IIM). Patients diagnosed with IIM face an elevated susceptibility to the development of gastric cancer, underscoring the critical need for early screening measures. In addition to the complexities associated with diagnosis, the exact mechanisms driving the progression of gastric cancer in IIM patients remain poorly understood. OLFM4 is overexpressed in several types of tumors, including colorectal, gastric, pancreatic, and ovarian cancers, and its expression has been associated with tumor progression. METHODS: In this study, we used pathological sections from two clinical centers, biopsies of IM tissues, precancerous lesions of gastric cancer (PLGC) cell models, animal models, and organoids to explore the role of OLFM4 in IIM. RESULTS: Our results show that OLFM4 expression is highly increased in IIM, with superior diagnostic accuracy of IIM when compared to CDX2 and MUC2. OLFM4, along with MYH9, was overexpressed in IM organoids and PLGC animal models. Furthermore, OLFM4, in combination with Myosin heavy chain 9 (MYH9), accelerated the ubiquitination of GSK3ß and resulted in increased ß-catenin levels through the Wnt signaling pathway, promoting the proliferation and invasion abilities of PLGC cells. CONCLUSIONS: OLFM4 represents a novel biomarker for IIM and could be utilized as an important auxiliary means to delimit the key population for early gastric cancer screening. Finally, our study identifies cell signaling pathways involved in the progression of IM.


Subject(s)
Disease Progression , Glycogen Synthase Kinase 3 beta , Metaplasia , Myosin Heavy Chains , beta Catenin , Humans , Metaplasia/metabolism , Metaplasia/pathology , Metaplasia/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Animals , beta Catenin/metabolism , beta Catenin/genetics , Mice , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Female , Wnt Signaling Pathway , Cell Proliferation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Disease Models, Animal , Male , Organoids/metabolism , Organoids/pathology
2.
Mol Cancer ; 23(1): 49, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38459596

ABSTRACT

Circular RNAs (circRNAs) play important roles in gastric cancer progression but the regulatory role of circRNAs in controlling macrophage function remains elusive. Exosomes serve as cargo for circRNAs and play a crucial role as mediators in facilitating communication between cancer cells and the tumor microenvironment. In this study, we found that circATP8A1, a previously unreported circular RNA, is highly expressed in both gastric cancer tissues and exosomes derived from plasma. Increased circATP8A1 was associated with advanced TNM stage and worse prognosis in patients with gastric cancer. We showed that  the circATP8A1 knockdown significantly inhibited gastric cancer proliferation and invasion in vitro and in vivo. Functionally, exosome circATP8A1 induced the M2 polarization of macrophages through the STAT6 pathway instead of the STAT3 pathway. Mechanistically, circATP8A1 was shown to activate the STAT6 pathway through competitive binding to miR-1-3p, as confirmed by Fluorescence In Situ Hybridization (FISH), RNA immunoprecipitation, RNA pulldown, and Luciferase reporter assays. The reversal of circATP8A1-induced STAT6 pathway activation and macrophage polarization was observed upon blocking miR-1-3p. Macrophages treated with exosomes from gastric cancer cells overexpressing circATP8A1 were able to promote gastric cancer migration, while knockdown of circATP8A1 reversed these effects in vivo. In summary, exosome-derived circATP8A1 from gastric cancer cells induce macrophages M2 polarization via the circATP8A1/miR-1-3p/STAT6 axis, and tumor progression. Our results highlight circATP8A1 as a potential prognostic biomarker and therapeutic target in gastric cancer.


Subject(s)
Exosomes , MicroRNAs , Stomach Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Exosomes/genetics , In Situ Hybridization, Fluorescence , Macrophages , MicroRNAs/genetics , RNA, Circular/genetics , STAT6 Transcription Factor/genetics , Stomach Neoplasms/genetics , Tumor Microenvironment
3.
Cancers (Basel) ; 15(24)2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38136340

ABSTRACT

Tumor-associated macrophages (TAMs) play a pivotal role in shaping the tumor microenvironment. Lactic acid (LA) has been identified as an influential factor in promoting immune escape and tumor progression. However, the mechanisms through which LA modulates TAMs in colorectal cancer (CRC) remain poorly understood. We used qRT-PCR to quantify the expression of LA-related genes (LDHA and LAMP2) in CRC tumor tissues and adjacent nontumor tissues (n = 64). The biological effects and mechanisms of LA on macrophages and tumors were evaluated via qRT-PCR, Western blot, RNA-seq, wound healing assay, colony formation assay in vitro, and allograft mouse tumor models in vivo. We found the expression of LDHA and LAMP2 was highly elevated in the tumor regions and positively associated with a poor clinical stage of CRC. A high concentration of LA was generated under hypoxia; it could promote tumor progression and metastasis with the involvement of macrophages. The inhibition of LA release impaired this protumor phenomenon. Mechanically, LA induced M2 macrophages through the AKT/ERK signaling pathway; subsequently, M2 macrophages secreted CCL8 and facilitated the proliferation and metastasis of CRC cells by activating the CCL8/CCR5/mTORC1 axis. This effect was inhibited by the antagonist or knockdown of CCR5. In conclusion, lactate-induced CCL8 in TAMs accelerated CRC proliferation and metastasis through the CCL8/CCR5/mTORC1 axis.

4.
Front Mol Biosci ; 10: 1163977, 2023.
Article in English | MEDLINE | ID: mdl-37255541

ABSTRACT

Introduction: Gastric cancer (GC) is the fifth frequent malignancy and is responsible for the third leading cause of cancer-related deaths. Gastric cancer is an aging-related disease, with incidence and mortality rates increasing with aging. The development of GC is affected by lncRNAs, miRNAs, and mRNAs at the transcriptional and posttranscriptional levels. This study aimed to establish a prognostic panel for GC based on competing endogenous RNA (ceRNA) networks. Methods: RNA sequences were obtained from the TCGA database. Different expressions of RNAs were scrutinized with the EdgeR package. The ceRNA network was built using the starBase database and the Cytoscape. The prognostic panel was constituted with the LASSO algorithm. We developed a nomogram comprising clinical characteristic and risk score. The receiver operating characteristic (ROC) was used to evaluate the accuracy of the nomogram prediction. Hub RNAs expressions were detected by qPCR, immunohistochemistry and western blot respectively. Clinical relevance and survival analyses were analyzed. The relationship between RNAs and immune infiltrations, as well as immune checkpoints, was analyzed and evaluated using the CIBERSORT, TIMER and TISIDB databases. Results: Four DElncRNAs, 21 DEmiRNAs and 45 DEmRNAs were included in the ceRNA network. A 3-element panel (comprising lncRNA PVT1, hsa-miR-130a-3p and RECK) with poor overall survival (OS) was established and qPCR was applied to validate the expressions of hub RNAs. Hub RNAs were firmly associated with T, M, and N stage. The CIBERSORT database showed that the high lassoScore group exhibited a significantly high ratio of resting memory CD4+ T cells, M2 macrophages and a significantly low ratio of activated memory CD4+ T cells and M1 macrophages. According to the TIMER database, this panel was linked to immune infiltrations and immune cell gene markers. TISIDB database indicated that RECK was positively correlated with immune checkpoints (including CD160, CD244, PDCD1, and TGFBR1). Discussion: A novel triple prognostic panel of GC constructed based on the ceRNA network was associated with clinical prognostic, clinicopathological features, immune infiltrations, immune checkpoints and immune gene markers. This panel might provide potential therapeutic targets for GC and more experimental verification research is needed.

5.
Front Oncol ; 13: 976854, 2023.
Article in English | MEDLINE | ID: mdl-36824130

ABSTRACT

Background: Gastric cancer (GC) is a serious threat to human health. The clinical GC characteristics in China may be impacted by changes in people's lifestyles and the promotion of early GC (EGC) screening. The present study aims to evaluate the recent trends of GC characteristics in South China and search for hazardous factors limiting the survival time of GC patients. Methods: Data on GC patients that were hospitalized in the Department of Digestive Center, the First Affiliated Hospital, Sun Yat-sen University, from 1994 to 2019 were collected and divided into two categories according to the time when the EGC screening began in China: the PRE group (previous 13 years, 1994-2006) and the PAS group (past 13 years, 2007-2019). Results: We found that, although the 5-year survival rate increased in the PAS group compared with the PRE group (P < 0.0001), patients with age ≥60 years or Borrmann type IV still had a worse prognosis. In the PAS group, the larger percentages of elderly patients and patients with Borrmann type IV in the lymphatic metastases (N1) group (41.0% vs. 51.1%, P = 0.0014) and stage IV subgroup (20.7% vs. 32.2%, P = 0.016), respectively, when compared with the PRE group, may have contributed to the poor outcome of GC. By comparing the odds ratio (OR) of 5-year overall survival (OS) in the two 13-year periods, female sex and T2 turned into risk factors because of a greater proportion of Borrmann type IV or elderly patients in the PAS group (OR = 0.983, 95% CI = 0.723-1.336 vs. OR = 1.277, 95% CI = 1.028-1.586 and OR = 1.545, 95% CI = 0.499-4.775 vs. OR = 2.227, 95% CI = 1.124-4.271, respectively). Conclusions: Despite the GC epidemiology changes, the overall prognosis of GC patients has improved in South China. However, old age and Borrmann type IV are still the major restrictions affecting the survival of GC patients, a situation which calls for additional attention.

6.
J Cancer Res Clin Oncol ; 149(9): 5871-5884, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36592213

ABSTRACT

BACKGROUND: According to the guidelines, PD-L1 expression is a critical indicator for guiding immunotherapy application. According to certain studies, regardless of PD-L1 expression, immunotherapy could be advantageous for individuals with gastric cancer. Therefore, new scoring systems or biomarkers are required to enhance treatment strategies. METHODS: Mass spectrometry and machine learning were used to search for strongly related PD-L1 genes, and the NMF approach was then used to separate gastric cancer patients into two categories. Differentially expressed genes (DEGs) between the two subtypes identified in this investigation were utilized to develop the UBscore predictive model, which was verified by the Gene Expression Omnibus (GEO) database. Coimmunoprecipitation, protein expression, and natural killing (NK) cell coculture experiments were conducted to validate the findings. RESULTS: A total of 123 proteins were identified as PD-L1 interactors that are substantially enriched in the proteasome complex at the mRNA level. Using random forest, 30 UPS genes were discovered in the GSE66229 cohort, and ANAPC7 was experimentally verified as one of 123 PD-L1 interactors. Depending on the expression of PD-L1 and ANAPC7, patients were separated into two subgroups with vastly distinct immune infiltration. Low UBscore was related to increased tumor mutation burden (TMB) and microsatellite instability-high (MSI-H). In addition, chemotherapy medications were more effective in individuals with a low UBscore. Finally, we discovered that ANAPC7 might lead to the incidence of immunological escape when cocultured with NK-92 cells. CONCLUSION: According to our analysis of the PD-L1-related signature in GC, the UBscore played a crucial role in prognosis and had a strong relationship with TMB, MSI, and chemotherapeutic drug sensitivity. This research lays the groundwork for improving GC patient prognosis and treatment response.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , Stomach Neoplasms/pathology , B7-H1 Antigen , Apc7 Subunit, Anaphase-Promoting Complex-Cyclosome , Prognosis , Mass Spectrometry , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Microsatellite Instability
7.
Cancers (Basel) ; 15(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36612024

ABSTRACT

Syntaxin-6 (STX6), a vesicular transport protein, is a direct target of the tumor suppressor gene P53, supporting cancer growth dependent on P53. However, STX6's function in the tumor microenvironment has yet to be reported. In this research, we comprehensively explored the role of the oncogene STX6 in pan-cancer by combining data from several databases, including the Cancer Genome Atlas, CPTAC, cBioPortal, and TIMER. Then, we verified the carcinogenic effect of STX6 in hepatocellular carcinoma (HCC) and colorectal cancer (CRC) through a series of experiments in vitro and in vivo. Bioinformatics analysis demonstrated that STX6 is an oncogene for several cancers and is mainly involved in the cell cycle, epithelial-mesenchymal transition, oxidative phosphorylation, and tumor immune modulation, especially for tumor-associated fibroblasts (CAFs) and NKT cells. Additionally, a high level of STX6 could indicate patients' resistance to immunotherapy. Our own data indicated that the STX6 level was upregulated in HCC and CRC. Knockdown of the STX6 levels could arrest the cell cycle and restrain cell proliferation, migration, and invasion. RNA-seq indicated that STX6 was significantly involved in pathways for cancer, such as the MAPK signal pathway. In a mouse model, knockdown of STX6 inhibited tumor growth and potentiated anti-PD-1 efficacy. In light of the essential roles STX6 plays in carcinogenesis and cancer immunology, it has the potential to be a predictive biomarker and a target for cancer immunotherapy.

8.
Hum Cell ; 33(3): 630-640, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32130678

ABSTRACT

Aberrant expression of miR-1256 has been reported to be closely associated with the development and progression of tumors, including colon cancer and lung cancer. However, study of its expression pattern and functional role in papillary thyroid cancer (PTC) is rare. Using quantitative real time PCR analysis, we found miR-1256 was significantly down-regulated in PTC tissues and cell lines. The correlation of miR-1256 expression with clinicopathological features was statistically analyzed. The results showed miR-1256 expression was significantly correlated with tumor size (p = 0.0124) and TNM stage (p = 0.0032). Restoring miR-1256 expression significantly inhibited proliferation and cell cycle progression of PTC cells demonstrated by CCK-8 and flow cytometry assays. Luciferase reporter assay and biotin-avidin pull-down assay showed miR-1256 can directly target 5-hydroxytryptamine receptor 3A (HTR3A) in PTC cells. The expression of miR-1256 was inversely correlated with HTR3A expression in PTC tissues. Knockdown of HTR3A imitated the suppressive effects of miR-1256 in PTC cells. Ectopic expression of HTR3A can antagonize the effects of miR-1256 on PTC cells. Furthermore, the suppressive effects of miR-1256 on the expression of PCNA, CDK4, Cyclin D1, and p21 were partially reversed by HTR3A overexpression in PTC cells. In summary, our data suggested that miR-1256 could suppress PTC cellular function by targeting HTR3A, which might be a potential therapeutic target for patients with PTC.


Subject(s)
Cell Cycle/genetics , Cell Proliferation/genetics , MicroRNAs/physiology , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Humans , Molecular Targeted Therapy , Thyroid Cancer, Papillary/therapy , Thyroid Neoplasms/therapy , Tumor Cells, Cultured
9.
Biomed Pharmacother ; 95: 1868-1875, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28968944

ABSTRACT

The purpose of the present study was to evaluate the effects of cordycepin (CA) on N-nitrosodiethylamine (NDEA)-induced hepatocellular carcinomas (HCC) and explore its potential mechanisms. Mice were randomly assigned to four groups: control group, NDEA group, NDEA+CA (20mg/kg) group, NDEA+CA (40mg/kg) group. The animal of each group were given NDEA (100ppm) in drinking water. One hour later, CA, which was dissolved in PBS, were intragastrically administered for continuous seven days. The results showed that CA reduced the activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in liver and serum. CA also reduced the levels of interleukin-6 (IL-6), IL-1ß, tumor necrosis factor-α (TNF-α), methane dicarboxylic aldehyde (MDA), and stored the activity of superoxygen dehydrogenises (SOD) in serum. CA could obviously attenuate the hepatic pathological alteration. Furthermore, CA effectively inhibited the phosphorylations of phosphatidylinositol 3 kinase(PI3K), protein kinase B (Akt), mammalian target of rapamycin (mTOR). In conclusion, our research suggested that CA exhibited protective effects on NDEA-induced hepatocellular carcinomas via the PI3K/Akt/mTOR pathway.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Hepatocellular/prevention & control , Deoxyadenosines/pharmacology , Liver Neoplasms, Experimental/prevention & control , Animals , Anticarcinogenic Agents/administration & dosage , Deoxyadenosines/administration & dosage , Diethylnitrosamine/toxicity , Dose-Response Relationship, Drug , Heme Oxygenase-1/metabolism , Male , Mice , Mice, Inbred AKR , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
10.
Medicine (Baltimore) ; 95(4): e2589, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26825905

ABSTRACT

The clinical value of a prominent metastasis suppressor, nonmetastatic protein 23 (NM23), remains controversial. In this study, we examined the correlation between NM23 protein levels and the clinicopathologic features of colorectal cancers (CRC), and assessed the overall prognostic value of NM23 for CRC. Embase, PubMed, Web of Science, and other scientific literature databases were exhaustively searched to identify relevant studies published prior to June 31, 2015. The methodological qualities of selected studies were scored based on the critical appraisal skills program (CASP) criteria, as independently assessed by 2 reviewers. NM23 protein levels in tumor tissues of CRC patients were examined in relation to Dukes stage, differentiation grade, T-stage, lymph node metastasis status, and overall survival (OS). STATA software version 12.0 (Stata Corp, College Station, TX) was used for statistical analysis of data pooled from selected studies. Nineteen cohort studies met the inclusion criteria for present study and contained a combined total of 2148 study subjects. Pooled odd ratios (ORs) for NM23 expression revealed that reduced NM23 protein levels in CRC tumor tissues correlated with Dukes stage C and D (OR = 1.89, 95% CI: 1.06-3.39, P = 0.032), poor differentiation grades (OR = 1.41, 95% CI: 1.03-1.94, P = 0.032), and positive lymph node metastasis status (OR = 3.21, 95% CI: 1.95-5.29, P < 0.001). On the other hand, no such correlations were evident with T-stage T3-4 (OR = 1.56, 95% CI: 0.60-4.06, P = 0.367) or OS (OR = 0.79, 95% CI: 0.58-1.08, P = 0.138). Our analysis of pooled data found that NM23 expression is reduced in CRC tissues and low NM23 levels tightly correlate with higher Dukes stages, poorer differentiation grade, and positive lymph node metastases. However, NM23 levels did not influence the OS in CRC patients.


Subject(s)
Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , NM23 Nucleoside Diphosphate Kinases/analysis , Humans , Lymphatic Metastasis , Neoplasm Grading , Neoplasm Staging , Survival Rate
11.
Endocr J ; 61(11): 1087-92, 2014.
Article in English | MEDLINE | ID: mdl-25100151

ABSTRACT

Graves' ophthalmopathy (GO) is a common autoimmune disease that is difficult to deal with due to limited clinical evaluation methods. Recently miR-146a and Interleukin-17 (IL-17) have been found to be involved in autoimmune disorders and correlated with disease activity. However, it is unclear whether they are involved in Graves' ophthalmopathy (GO). The aim of this study is to investigate the correlation of circulating levels of miR-146a and IL-17 with clinical activity in GO patients. Fifty-seven study subjects were enrolled in four groups according to the corresponding criteria: active-GO, inactive-GO, Graves disease (GD) without ophthalmopathy, and healthy control group. The circulating levels of miR-146a and IL-17 were determined by qRT-PCR and ELISA, respectively. Serum IL-17 levels of GD, inactive-GO, and active-GO groups were all significantly higher than that of control (all P < 0.001). Active-GO group had significantly higher IL-17 level than inactive-GO and GD groups (P = 0.024 and P = 0.001, respectively). Active-GO and inactive-GO group had significantly lower miR-146a expressions than control (P < 0.05). Active-GO group had significantly lower miR-146a than inactive-GO group (P < 0.05). Serum levels of IL-17 and miR-146a were both significantly correlated with clinical activity score (CAS) in GO patients (P < 0.001, P < 0.001, respectively). There was a significant negative correlation of circulating miR-146a expression with serum IL-17 levels (P < 0.01). These findings indicated that circulating levels of miR-146a and IL-17 may be potential biomarkers of active GO, and may play a key role in the progression of GO.


Subject(s)
Graves Ophthalmopathy/blood , Interleukin-17/blood , MicroRNAs/blood , Adult , Female , Graves Disease/blood , Graves Ophthalmopathy/physiopathology , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...