Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Sci Rep ; 14(1): 12090, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802444

ABSTRACT

Micropeptides hidden in long non-coding RNAs (lncRNAs) have been uncovered to program various cell-biological changes associated with malignant transformation-glioblastoma (GBM) cascade. Here, we identified and characterized a novel hidden micropeptide implicated in GBM. We screened potential candidate lncRNAs by establishing a workflow involving ribosome-bound lncRNAs, publicly available MS/MS data, and prognosis-related lncRNAs. Micropeptide expression was detected by western blot (WB), immunofluorescence (IF), and immunohistochemistry (IHC). Cell proliferation rate was assessed by calcein/PI staining and EdU assay. Proteins interacted with the micropeptide were analyzed by proteomics after co-immunoprecipitation (Co-IP). We discovered that lncRNA AF127577.4 indeed encoded an endogenous micropeptide, named AF127577.4-ORF. AF127577.4-ORF was associated with GBM clinical grade. In vitro, AF127577.4-ORF could suppress GBM cell proliferation. Moreover, AF127577.4-ORF reduced m6A methylation level of GBM cells. Mechanistically, AF127577.4-ORF diminished ERK2 interaction with m6A reader methyltransferase like 3 (METTL3) and downregulated phosphorylated ERK (p-ERK) level. The ERK inhibitor reduced p-ERK level and downregulated METTL3 protein expression. AF127577.4-ORF weakened the stability of METTL3 protein by ERK. Also, AF127577.4-ORF suppressed GBM cell proliferation via METTL3. Our study identifies a novel micropeptide AF127577.4-ORF hidden in a lncRNA, with a potent anti-proliferating function in GBM by diminishing METTL3 protein stability by reducing the ERK2/METTL3 interaction. This micropeptide may be beneficial for development of therapeutic strategies against GBM.


Subject(s)
Cell Proliferation , Glioblastoma , Methyltransferases , Mitogen-Activated Protein Kinase 1 , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Cell Line, Tumor , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Gene Expression Regulation, Neoplastic , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Peptides/metabolism
2.
Brain Res ; 1838: 149009, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38763504

ABSTRACT

Intracranial aneurysms (IAs) often go undetected until rupture, leading to significant morbidity and mortality. Identifying biomarkers for early detection of IAs is crucial. The current study attempted to identify core genes linked with IAs and determine their relevance through Mendelian randomization. Limma helped identify differentially expressed genes between IAs and control superficial temporal artery samples. WGCNA was utilized to find IA-related modules and associated genes, which were further evaluated using KEGG and GO analyses to ascertain their potential roles. Five highly associated genes were screened with the CytoHubba plugin of Cytoscape software. ROC curves assessed the diagnostic efficacy of these genes. A two-sample Mendelian randomization evaluated the causal relationship between the core gene PTRPC and IAs, along with its correlation with immune infiltration. WGCNA and differential expression analysis depicted 584 related genes involved in cellular metabolism and chemokine activity. PTPRC was among the top highly associated genes identified through Cytoscape. It showed significant diagnostic value for IAs. Moreover, mendelian randomization depicted that PTPRC in CD4+ T cells is related to IA risk, with an OR of 0.63538 (95 % CI = 0.41636-0.96959, p = 0.03545). No reverse causal relationship was observed between PTPRC and IAs, with an OR of 0.99947 (95 % CI = 0.99719-1.00176, p = 0.65022). Additionally, immune cell infiltration results indicated a positive correlation between PTPRC in IAs with neutrophils and unactivated dendritic cells and a negative association with regulatory T cells (Tregs). PTPRC was identified as a core gene linked with IAs, providing evidence for IA diagnosis and studying molecular mechanisms.


Subject(s)
Computational Biology , Intracranial Aneurysm , Mendelian Randomization Analysis , Intracranial Aneurysm/genetics , Humans , Mendelian Randomization Analysis/methods , Computational Biology/methods , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics
3.
J Genet Genomics ; 50(3): 192-203, 2023 03.
Article in English | MEDLINE | ID: mdl-35940521

ABSTRACT

Many circular RNAs (circRNAs) are reported to be abnormally expressed during the progression of various tumors, and these circRNAs can be used as anti-tumor targets. Therefore, it is important to identify circRNAs that can be used effectively for the clinical diagnosis and treatment of colorectal cancer (CRC). Here, we report that hsa_Circ_0000826 (Circ_0000826), a circRNA with significantly reduced expression level in CRC tissues, is associated with a poor prognosis in patients. The silencing of Circ_0000826 promotes the proliferation of CRC cells. Conversely, the overexpression of Circ_0000826 restricted CRC cell proliferation both in vitro and in vivo. Furthermore, Circ_0000826 could target AU-rich element RNA-binding protein 1 (AUF1). AUF1, known as heterogeneous nuclear ribonucleoprotein D (hnRNP D), could bind to the c-MYC 3'-UTR and promote c-MYC expression. When Circ_0000826 binds to AUF1, it competitively inhibits the binding of AUF1 to the c-MYC 3'-UTR, which inhibits the c-MYC expression and cell proliferation. These results provide novel insights into the functional mechanism of Circ_0000826 action in CRC progression and indicate its potential use as a therapeutic target in CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , RNA, Circular/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , MicroRNAs/genetics
4.
Cell Death Dis ; 13(11): 941, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36347844

ABSTRACT

FMR1, a new m6A reader, is known to be involved in the regulation of cancer progression. However, its role, regulatory mechanism, and clinical significance in colorectal cancer (CRC) are elusive. Here, we showed that FMR1 was upregulated in CRC, and it promoted proliferation and metastasis of CRC cells in vitro and in vivo. Mechanically, FMR1 recognized the m6A-modification site in EGFR mRNA, a key molecule in cancer occurrence and targeted therapy, sustained its stability and maintained its expression in an m6A-dependent manner, thereby promoting the tumorigenesis and metastasis of CRC. And the effect of FMR1 knockdown in CRC cells could be abolished by METTL3. Furthermore, FMR1 shRNA plasmid carried by attenuated Salmonella has an effective anti-tumor effect in vivo. Collectively, we identified the METTL3/FMR1/EGFR axis in the progression of CRC. This novel mechanism indicated that the METTL3/FMR1/EGFR axis is a potential target for early therapeutic intervention in CRC progression.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Humans , RNA, Messenger/genetics , Colorectal Neoplasms/pathology , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Cell Proliferation/genetics , Methyltransferases/metabolism , Fragile X Mental Retardation Protein/genetics
5.
Front Genet ; 13: 891270, 2022.
Article in English | MEDLINE | ID: mdl-35646079

ABSTRACT

Tumor-infiltrating immune cells are associated with prognosis and immunotherapy targets in colorectal cancer (CRC). The recently developed CIBERSORT method allows immune cell analysis by deconvolution of high-throughput data onto gene expression. In this study, we analyzed the relative proportions of immune cells in GEO (94 samples) and TCGA (522 samples) CRC data based on the CIBERSORT method. A total of 22 types of tumor-infiltrating immune cells were evaluated. Combined with GEO and TCGA data, it was found that naive B cells, M2 macrophages, and resting mast cells were highly expressed in normal tissues, while M0 macrophages, M1 macrophages, activated mast cells, and neutrophils were highly expressed in tumors. Moreover, we constructed a prognostic model by infiltrating immune cells that showed high specificity and sensitivity in both the training (AUC of 5-year survival = 0.699) and validation (AUC of 5-year survival = 0.844) sets. This provides another basis for clinical prognosis. The results of multiple immunofluorescence detection showed that there were differences in the results of bioinformatics analysis. Neutrophils were highly expressed in normal tissues, and M2 macrophages were highly expressed in tumor tissues. Collectively, our data suggested that infiltrating immune cells in CRC may be an important determinant of prognosis and immunotherapy.

6.
Chin Neurosurg J ; 8(1): 7, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35361282

ABSTRACT

BACKGROUND: Glioma is a common malignant brain tumor. The purpose of this study was to investigate the role of the transcription factor SPI1 in glioma. METHODS: SPI1 expression in glioma was identified using qRT-PCR and Western blotting. Cell proliferation was assessed using the CCK8 assay. Transwell and wound healing assays were utilized to evaluate cell migration. Additionally, cell cycle and apoptosis were detected using flow cytometry. RESULTS: We observed that the expression level of SPI1 was up-regulated in glioma tissues, compared to normal tissues. Furthermore, we found that SPI1 is able to promote proliferation and migration of glioma cells in vitro. Flow cytometry results demonstrate that, compared to si-NC cells, si-SPI1 cells stagnated in the G1 phase, and down-regulation of SPI1 expression is able to increase rates of apoptosis. Double luciferase activity and chromatin immunoprecipitation assay results indicated that SPI1 can bind to the promoter sites and promote the proliferation and migration of glioma cells by regulating the expression of oncogenic PAICS. CONCLUSIONS: Our results suggest that SPI1 can promote proliferation and migration of glioma. Furthermore, SPI1 can be utilized as a potential diagnostic marker and therapeutic target for glioma.

7.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(2): 171-175, 2022 Feb 10.
Article in Chinese | MEDLINE | ID: mdl-35076913

ABSTRACT

OBJECTIVE: To explore the effect and mechanism of down-regulating lncRNA TTTY15 targeting miR-4500 on the proliferation, apoptosis, migration and invasion of A172 glioma cells. METHODS: The difference in TTTY15 expression between the glioma cells and tissue was determined with a qRT-PCR method. Complementary binding sites of TTTY15 and miR-4500 were predicted with Starbase software, and the targeting relationship was validated with a luciferase reporter system. A172 glioma cells were divided into Control, si-NC (transfected with control siRNA), si-TTTY15 (transfected with TTTY15 siRNA), si-TTTY15+Anti-miR-NC (co-transfected with TTTY15 siRNA and inhibitor control) and si-TTTY15+Anti-miR-4500 (co-transfected with TTTY15 siRNA and miR-4500 inhibitor) groups. Proliferation, apoptosis, migration and invasion, and the expression of Bax, Bcl-2, MMP-2 and MMP-9 proteins of the A172 glioma cells were respectively detected with CCK-8, flow cytometry, Transwell chamber and Western blotting assays. RESULTS: The expression of TTTY15 in glioma cells and glioma tissues have both increased. The expression levels of TTTY15 and miR-4500 in glioma tissues were inversely correlated. TTTY15 and miR-4500 are mutually targeted. Compared with those of the Control and si-NC groups, the glioma cells in the si-TTTY15 group showed increased level of miR-4500, decreased survival rate, increased apoptosis rate, enhanced cell migration and invasion, increased expression of Bax protein, and decreased expression of Bcl-2, MMP-2 and MMP-9 proteins (P<0.05). Compared with those of the si-TTTY15+Anti-miR-NC group, the A172 glioma cells in the si-TTTY15+Anti-miR-4500 group showed decreased level of miR-4500, increased cell survival rate, decreased apoptosis rate, enhanced cell migration and invasion, decreased expression of Bax protein, and increased expression of Bcl-2, MMP-2, and MMP-9 proteins (P<0.05). CONCLUSION: Down-regulating TTTY15 targeting miR-4500 can inhibit the proliferation, migration, invasion and induce apoptosis of the A172 glioma cells.


Subject(s)
Glioma , MicroRNAs , RNA, Long Noncoding , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , MicroRNAs/genetics
8.
RSC Adv ; 13(1): 141-154, 2022 Dec 19.
Article in English | MEDLINE | ID: mdl-36605636

ABSTRACT

Plasma characteristic models were established in cylindrical coordinates according to the plasma expansion characteristics of pulsed laser processing of metal materials, mainly including plasma expansion characteristic models and a change rate model for the collisional ionization effect. The plasma characteristics (expansion dimension, expansion velocity, electron density and collision rate) for the pulsed laser machining of a bronze grinding wheel were obtained by using the plasma characteristic models. The results show that the expansion velocity direction can be changed after plasma collision, resulting in particles returning and depositing onto the processed material surface. Plasma spectrum measurements for the pulsed laser machining of a bronze grinding wheel and grinding tests were carried out. Based on the measured spectral data, the plasma electron temperature and plasma electron density were calculated, and the topography of the machined grinding wheel surface was observed, which confirms that black particles can return to cover the grinding wheel surface. Through grinding experiments, it is verified that the returning particles reduce the height of the abrasive protruding binder and block the chip space around the abrasive particles, resulting in reduced grinding performance. The experimental calculation data and numerical simulation results are basically consistent with each other, which not only verifies the correctness and feasibility of the plasma characteristic models but also provides theoretical guidance and process optimization for subsequent research into laser machining of materials.

9.
Front Genet ; 12: 647152, 2021.
Article in English | MEDLINE | ID: mdl-34589110

ABSTRACT

Colorectal cancer (CRC) is one of the most prevalent malignant tumors worldwide. Colon adenocarcinoma (COAD) is the most common pathological type of CRC and several biomarkers related to survival have been confirmed. Yet, the predictive effect of a single gene biomarker is not enough. The tricarboxylic acid (TCA) cycle and carbon metabolism play an important role in tumors. Thus, we aimed to identify new gene signatures from the TCA cycle and carbon metabolism to better predict the survival of COAD. This study performed mRNA expression profiling in large COAD cohorts (n = 417) from The Cancer Genome Atlas (TCGA) database. Univariate Cox regression and multivariate Cox regression analysis were performed, and receiver operating characteristic (ROC) curve was used to screen the variable combinations model which is most relevant to patient prognosis survival mostly. Univariable or multivariate analysis results showed that SUCLG2, SUCLG1, ACLY, SUCLG2P2, ATIC and ACO2 have associations with survival in COAD. Combined with clinical variables, we confirmed model 1 (AUC = 0.82505), most relevant to patient prognosis survival. Model 1 contains three genes: SUCLG2P2, SUCLG2 and ATIC, in which SUCLG2P2 and SUCLG2 were low-expressed in COAD, however, ATIC was highly expressed, and the expressions above are related to stages of CRC. Pearson analysis showed that SUCLG2P2, SUCLG2 and ATIC were correlated in normal COAD tissues, while only SUCLG2P2 and SUCLG2 were correlated in tumor tissues. Finally, we verified the expressions of these three genes in COAD samples. Our study revealed a possible connection between the TCA cycle and carbon metabolism and prognosis and showed a TCA cycle and carbon metabolism related gene signature which could better predict survival in COAD patients.

10.
PLoS One ; 16(8): e0255489, 2021.
Article in English | MEDLINE | ID: mdl-34343214

ABSTRACT

MACC1 gene is a newly discovered gene and plays an important role in the metastasis of colorectal cancer (CRC). The objective of this study was to investigate whether MACC1 is an independent factor associated with lymphatic metastasis in CRC patients. We analyzed the association between MACC1 expression and lymphatic metastasis in a nested case-control study including 99 cases and 198 matched controls in CRC patients, assessed from August 2001 to March 2015. Cases were defined as lymphatic metastasis and non-lymphatic metastasis according to AJCC TNM stages; for each case, two age-matched control without lymphatic and distant metastasis was randomly selected from the study participants. Demographic, variables about metastasis and MACC1 expression were collected. In multivariate analysis, the OR (95% CI) of MACC1 expression was 1.5 (1.1 to 2.0) in patients with lymphatic metastasis versus non-lymphatic metastasis after adjusting all variables. After adjustment for all variables and age stratification, MACC1 expression was found to be an independent risk factor for lymph node metastasis in the middle-aged group (OR 2.1, 95%CI 1.1-4.0). A nonlinear relationship between MACC1 expression and 64-75 age group was observed. The probability of metastasis slightly increased with the MACC1 level lower than turning point 1.4. At the same time, the probability of lymphatic metastasis was obviously increased even after adjusting all variables when MACC1 level higher than 1.4 (OR 11.2, 95% CI 1.5-81.5; p = 0.017) in the middle age group. The expression of MACC1 was not associated with lymphatic metastasis in populations younger than 64 or older than 75. The results demonstrates that increased MACC1 level in 64-75 age group might be associated with lymphatic metastasis in CRC patients.


Subject(s)
Colorectal Neoplasms/pathology , Lymphatic Metastasis/pathology , Neoplasm Recurrence, Local/pathology , Trans-Activators/metabolism , Aged , Biomarkers, Tumor , Case-Control Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Combined Modality Therapy , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis/therapy , Male , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/therapy , Prognosis , Trans-Activators/genetics
11.
J Cell Mol Med ; 25(16): 7720-7733, 2021 08.
Article in English | MEDLINE | ID: mdl-34173716

ABSTRACT

Glioma is a common malignant tumour of the brain. In this study, we aimed to investigate diagnostic biomarkers and its role in glioma. Weighted gene co-expression network analysis (WGCNA) and Cytoscape software were used to screen the marker genes in glioma. RT-qPCR and Western blotting methods were performed to determine the expression of PAICS, ERCC1 and XPA genes in glioma tissues. Expression level of PAICS in different grades of glioma was examined by immunohistochemistry. CCK8 and Colony formation assays were used to detect cell proliferation. Cell adhesion assay was used to detect adhesion ability. Wound healing and transwell tests were used to detect cell migration ability. Flow cytometry was used to detect cell cycle and apoptosis. According to the predicted co-expression network, we identified the hub gene PAICS. Furthermore, we observed that PAICS expression level was up-regulated in glioma tissues compared with normal tissues, and the expression level was correlated with the grade of glioma. Moreover, we found PAICS can promote glioma cells proliferation and migration in vitro. Flow cytometry results showed that si-PAICS cells were stalled at the G1 phase compared with the si-NC cells and knocking down PAICS expression can increase apoptotic rate. PAICS can regulate the mRNA and protein levels of nucleotide excision repair pathway core genes ERCC1 and XPA. l-aspartic acid can affect the expression of PAICS and then inhibit glioma cell proliferation. Our results indicated that PAICS can promote glioma proliferation and migration. PAICS may act as a potential diagnostic marker and a therapeutic target for glioma.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioma/pathology , Peptide Synthases/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Computational Biology/methods , Databases, Genetic , Glioma/genetics , Glioma/metabolism , Humans , Neoplasm Invasiveness , Peptide Synthases/metabolism , Signal Transduction
12.
Front Immunol ; 12: 672356, 2021.
Article in English | MEDLINE | ID: mdl-33936118

ABSTRACT

Cancer immunotherapy works by stimulating and strengthening the body's anti-tumor immune response to eliminate cancer cells. Over the past few decades, immunotherapy has shown remarkable efficacy in the treatment of cancer, particularly the success of immune checkpoint blockade targeting CTLA-4, PD-1 and PDL1, which has led to a breakthrough in tumor immunotherapy. Tumor neoantigens, a new approach to tumor immunotherapy, include antigens produced by tumor viruses integrated into the genome and antigens produced by mutant proteins, which are abundantly expressed only in tumor cells and have strong immunogenicity and tumor heterogeneity. A growing number of studies have highlighted the relationship between neoantigens and T cells' recognition of cancer cells. Vaccines developed against neoantigens are now being used in clinical trials in various solid tumors. In this review, we summarized the latest advances in the classification of immunotherapy and the process of classification, identification and synthesis of tumor-specific neoantigens, as well as their role in current cancer immunotherapy. Finally, the application prospects and existing problems of neoantigens were discussed.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy/methods , Neoplasms/immunology , Antigens, Neoplasm/pharmacology , Cancer Vaccines/immunology , Humans , Neoplasms/therapy
13.
Front Oncol ; 11: 657650, 2021.
Article in English | MEDLINE | ID: mdl-33937069

ABSTRACT

BACKGROUND: We previously reported that the long non-coding RNA (lncRNA) CASC11 promotes colorectal cancer (CRC) progression as an oncogene by binding to HNRNPK. However, it remains unknown whether CASC11 can act as a competitive endogenous RNA (ceRNA) in CRC. In this study, we focused on the role of CASC11 as a ceRNA in CRC by regulating miR-646 and miR-381-3p targeting of RAB11FIP2. METHODS: We identified the target microRNAs (miRNAs) of CASC11 and the target genes of miR-646 and miR-381-3p using bioinformatic methods. A dual-luciferase reporter assay was performed to validate the target relationship. Quantitative real-time PCR (qRT-PCR), western blotting (WB), and immunohistochemistry (IHC) were used to measure the RNA and protein expression levels. Rescue experiments in vitro and in vivo were performed to investigate the influence of the CASC11/miR-646 and miR-381-3p/RAB11FIP2 axis on CRC progression. RESULTS: We found that CASC11 binds to miR-646 and miR-381-3p in the cytoplasm of CRC cells. Moreover, miR-646 and miR-381-3p inhibitors reversed the suppressive effect of CASC11 silencing on CRC growth and metastasis in vitro and in vivo. We further confirmed that RAB11FIP2 is a mutual target of miR-646 and miR-381-3p. The expression levels of CASC11 and RAB11FIP2 in CRC were positively correlated and reciprocally regulated. Further study showed that CASC11 played an important role in regulating PI3K/AKT pathway by miR-646 and miR-381-3p/RAB11FIP2 axis. CONCLUSION: Our study showed that CASC11 promotes the progression of CRC as a ceRNA by sponging miR-646 and miR-381-3p. Thus, CASC11 is a potential biomarker and a therapeutic target of CRC.

14.
Life Sci ; 263: 118597, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33075373

ABSTRACT

AIMS: To explore the biological function and mechanism of Syntaxin2 (STX2) in Colorectal cancer (CRC) proliferation. MAIN METHODS: A series of gain- and loss-of-function analysis were conducted the to explore the biological function of STX2 in CRC proliferation in vivo and in vitro. Western blot, Co-immunoprecipitation (Co-IP) and the functional analyses were taken to analyze the regulative role of STX2 on Exosome Complex 4 (EXOSC4) in CRC proliferation; Immunohistochemistry (IHC) and Real-time quantitative polymerase chain reaction (qPCR) were used to further verify the relationship between the expression of STX2 and EXOSC4 in human CRC samples. KEY FINDINGS: Our study revealed that the over-expression of STX2 promoted CRC proliferation, while knockdown of STX2 repressed CRC proliferation; STX2 promoted CRC proliferation via increasing EXOSC4 protein; There was a positive correlation between STX2 and EXOSC4 expression. SIGNIFICANCE: The current data verify that STX2 drives the proliferation of CRC via increasing the expression of EXOSC4.


Subject(s)
Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Exosome Multienzyme Ribonuclease Complex/genetics , RNA-Binding Proteins/genetics , Syntaxin 1/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Up-Regulation
15.
Gland Surg ; 9(6): 2144-2154, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33447565

ABSTRACT

BACKGROUND: This study aims to investigate the mechanism through which Caveolin-1 (CAV-1) regulates the expression of micro ribonucleic acid (miR)-183 in invasive pituitary adenoma (IPA) tissues and GH3 cells, and explore the effects of CAV-1 and miR-183 on the invasion and migration ability of GH3 cells. METHODS: Western blotting was used to detect the expression level of CAV-1, early growth response 1 (EGR1) and Krueppel-like factor 5 (KLF5). Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression of miR-183. The mechanisms of interaction between CAV-1, EGR1, and KLF5 were studied by immunoprecipitation experiments. Transwell and cell scratch tests were used to determine the invasion and migration ability of GH3 cells. The dual-luciferase reporter gene experiment was used to detect the effects of EGR1 and KLF5 on miR-183 luciferase activity and verify the targeting relationship between miR-183 and ezrin. RESULTS: The expression of CAV-1 was up-regulated. However, following the knockdown of CAV-1, the invasion and migration ability of GH3 cells was significantly inhibited (P<0.05). The expression of miR-183 was down-regulated, but the expression level of miR-183 was markedly increased following the knockdown of CAV-1 (P<0.05). The knockdown of CAV-1 inhibited the nuclear ectopic of the EGR1 protein in GH3 cells. At the same time, the interaction between EGR1 and KLF5 in GH3 cells was significantly inhibited (P<0.05). The luciferase activity of miR-183 increased significantly after overexpression of KLF5 while overexpression of EGR1 and KLF5 had no significant effect on intracellular luciferase activity. Overexpression of miR-183 markedly inhibited the luciferase activity of wild-type EZR and the expression of the EZR protein in GH3 cells. Furthermore, the overexpression of miR-183 or the inhibition of EZR can reduce the invasion and migration ability of GH3 cells. The simultaneous overexpression or inhibition of miR-183 and EZR expression has no obvious effect on the invasion and migration ability of GH3 cells. CONCLUSIONS: CAV-1 up-regulates the expression of miR-183 by inhibiting the nuclear ectopic of EGR1 and the interaction between EGR1 and KLF5 in GH3 cells. Also, miR-183 negatively regulates the expression of EZR and inhibits the invasion and migration of GH3 cells.

16.
Front Oncol ; 9: 1140, 2019.
Article in English | MEDLINE | ID: mdl-31750242

ABSTRACT

Breast cancer is still the most common and leading cause of cancer-related deaths in women worldwide. Long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) have shown key regulator roles in various cancers. Previous reports have identified miR-125b as a critical tumor suppressor in breast cancer. However, the role of lncRNAs in breast cancer is far from well-characterized. In this study, we identified a novel lncRNA LINC01787, which specifically binds pre-miR-125b, inhibits the binding between DICER and pre-miR-125b, represses the processing of pre-miR-125b by DICER, and therefore induces pre-miR-125b accumulation and represses mature miR-125b generation. Functional assays showed that LINC01787 promotes breast cancer cell proliferation and migration and breast cancer xenograft growth in vivo, which is abolished by the mutation of pre-miR-125b binding sites on LINC01787 or overexpression of miR-125b. Furthermore, LINC01787 is up-regulated in breast cancer tissues and is associated with advanced stages and poor survival. The expression of LINC01787 is inversely associated with that of miR-125b in breast cancer tissues. In conclusion, our findings identified a novel up-regulated and oncogenic lncRNA LINC01787 in breast cancer, which binds pre-miR-125b and represses mature miR-125b generation. Our data suggests LINC01787 as a potential prognostic biomarker and therapeutic target for breast cancer.

17.
Onco Targets Ther ; 12: 8105-8115, 2019.
Article in English | MEDLINE | ID: mdl-31632063

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignancies worldwide. Studies have demonstrated that epigenetic modifications play essential roles in the development of CRC. ADHFE1 is a differentially expressed gene that has been reported to be hypermethylated in CRC. However, the role and mechanism of ADHFE1 in the proliferation of CRC remain unclear. MATERIALS AND METHODS: ADHFE1 expression was analyzed in CRC tissues by IHC and qRT-PCR, and the relationship between ADHFE1 expression and the clinicopathological parameters was analyzed. Cell proliferation were assessed by the in vitro and in vivo experimental models. GSEA assay was performed to explore the mechanism of ADHFE1 in the proliferation of CRC. Flow cytometry and Western blot were used to detect the activation of the cell cycle signaling. Bisulfite genomic sequence (BSP) assay was used to test the methylation degree of ADHFE1 gene promoter in CRC tissues. RESULTS: Here, we verified that ADHFE1 was down-regulated and hypermethylated in CRC tissues. The down-regulation of ADHFE1 was correlated with poor differentiation and advanced TNM stage of CRC patients. And ADHFE1 expression restored when the CRC cell line SW620 was treated with the demethylating agent 5-Aza-CdR. Overexpression of ADHFE1 inhibited the proliferation of CRC, while ADHFE1 knockdown promoted the proliferation of CRC cells in vitro and in vivo. Moreover, ADHFE1 overexpression could induce a significant G1-S cell cycle arrest in CRC cells and vice versa. CONCLUSION: Hypermethylation of ADHFE1 might promote cell proliferation by modulating cell cycle progression in CRC, potentially providing a new therapeutic target for CRC patients.

18.
Am J Cancer Res ; 9(5): 1061-1073, 2019.
Article in English | MEDLINE | ID: mdl-31218112

ABSTRACT

Slingshot phosphatase 3 (SSH3) is a member of the SSH phosphatase family that regulates actin filament dynamics. However, its role in cancer metastasis is relatively unclear compared to that of SSH1. Here, we showed that SSH3 was upregulated in colorectal cancer (CRC). Of note, SSH3 was upregulated in the tumor thrombus and lymph node metastasis compared with that in paired primary CRC tissues. High SSH3 expression was associated with the aggressive phenotype of CRC and may be an independent prognostic factor for the poor survival of patients with CRC. SSH3 significantly enhanced the invasion and metastasis of CRC cells in vitro and in vivo. Moreover, SSH3 regulated the remodeling of actin, which is involved in the cytoskeleton signaling pathway, through its interaction with LIMK1/Rac1 and subsequently promoted CRC cell invasion and metastasis. Our data elucidate an important role for SSH3 in the progression of CRC, and SSH3 may be considered a potential therapeutic target for CRC.

19.
Onco Targets Ther ; 12: 4331-4340, 2019.
Article in English | MEDLINE | ID: mdl-31213848

ABSTRACT

Background: The incidence and mortality of colorectal cancer (CRC) are rising worldwide. Long-noncoding RNAs (lncRNAs) are known to play key roles in the development of human cancers, including CRC. However, the function and underlying mechanism of long intergenic noncoding RNA 00707 (LINC00707) in the development of CRC are unknown. Materials and methods: The expression of LINC00707 and miR-206 in tissue samples or cell lines was measured by quantitative reverse transcription PCR (qRT-PCR). The protein expression of neurogenic locus notch homolog protein 3 (NOTCH3) and transmembrane 4 L6 family member 1 (TM4SF1) was assessed by Western blotting. Cell proliferation, migration, and invasion were assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and transwell assays. Luciferase reporter assay and biotin-coupled miRNA capture assay were used to explore the relationship between LINC00707 and miR-206 expression. Results: The expression of LINC00707 was significantly upregulated in CRC tissues as compared with the adjacent non-CRC tissues. LINC00707 expression was significantly correlated with tumor size, lymphatic metastasis, and distant metastasis, but not significantly correlated with age and gender. Knockdown of LINC00707 expression significantly inhibited LoVo and HCT116 cell proliferation, migration, and invasion. LINC00707 acted as a molecular sponge by competing for miR-206 and indirectly modulating the expression of its targets, NOTCH3 and TM4SF1. Conclusion: LINC00707 promotes CRC cell proliferation and metastasis by sponging miR-206, suggestive of its potential application for CRC treatment.

20.
J Cell Biochem ; 120(4): 6409-6419, 2019 04.
Article in English | MEDLINE | ID: mdl-30320923

ABSTRACT

Circular RNAs (circRNAs) play an important regulatory role in tumorigenesis. The aim of the present study was to analyze the circRNA expression network and elucidate its potential implications in colorectal cancer (CRC). The circRNA expression profile was analyzed in CRC tissues by RNA sequencing, and the functions of differentially expressed genes were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The circRNA network was predicted with bioinformatics. On the basis of the results, we identified 23 differentially expressed circRNAs in CRC; GO and KEGG analyses demonstrated that the changes in circRNAs were mainly associated with regulation of biological and metabolic processes through binding to other molecules. In addition, based on the predicted coexpression network, we identified a hub circRNA, hsa_circ_0009022. Subsequently, the results of sequencing were confirmed by reverse transcription-quantitative polymerase chain reaction, and hsa_circ_0000826 was found to be downregulated in CRC. Taken together, these findings indicate a set of differentially expressed circRNAs that may serve as a candidate diagnostic biomarker and a promising therapeutic target in CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , MicroRNAs/genetics , RNA, Circular/genetics , RNA, Messenger/genetics , Adult , Aged , Colorectal Neoplasms/pathology , Computational Biology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Lymphatic Metastasis , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL