Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-979406

ABSTRACT

@#[摘 要] 目的:探讨碱性亮氨酸拉链转录因子ATF样蛋白3(BATF3)在肾透明细胞癌(ccRCC)组织中的表达及其调控ccRCC细胞恶性生物学行为的分子机制。方法:收集2019年3月至2022年1月间在中国人民解放军联勤保障部队第九八〇医院手术切除的64例ccRCC组织和相应癌旁组织,qPCR法检测ccRCC组织、癌旁组织和肾癌ACHN、786-O细胞中BATF3 mRNA的表达,免疫组化检测ccRCC组织、癌旁组织中BATF3蛋白的表达,并分析其与临床病理特征的关系。构建BATF3敲减及过表达质粒,分别转染786-O、ACHN细胞,通过MTS法、Transwell实验检测BATF3对786-O或ACHN细胞增殖、迁移、侵袭能力的影响,qPCR法检测敲减或过表达BATF3对786-O或ACHN细胞EMT相关基因表达的影响,CHIP、双荧光素酶报告基因实验检测BATF3是否与波形蛋白(VIM)启动子区结合并调控其转录,MTS法、Transwell实验检测同时过表达BATF3及敲减VIM对786-O细胞增殖、迁移、侵袭能力的影响。结果:与癌旁组织比较,ccRCC组织中BATF3的mRNA和蛋白均呈高表达(均P<0.01),并且BATF3 mRNA与ccRCC的分化程度和TNM分期密切关联(均P<0.01);与正常肾上皮细胞293T相比,BATF3在ACHN及786-O细胞中均呈高表达(均P<0.01)。敲减BATF3表达均能明显抑制786-O细胞的增殖、迁移、侵袭能力(均P<0.01),过表达BATF3则均能促进ACHN细胞的增殖、迁移和侵袭能力(均P<0.01),敲减或过表达BATF3能抑制786-O细胞或促进ACHN细胞的EMT相关基因的表达(均P<0.01)。BATF3可与VIM启动子区的位点结合,直接调控VIM的转录表达。同时过表达BATF3及敲减VIM可逆转过表达BATF3对786-O细胞增殖、迁移、侵袭能力的影响。结论:BATF3在ccRCC组织中呈高表达,并与其分化程度和TNM分期密切关联;BATF3通过调控VIM的表达影响ACHN、786-O细胞的恶性生物学行为,其可作为临床治疗ccRCC的潜在靶点。

2.
Open Med (Wars) ; 17(1): 1483-1494, 2022.
Article in English | MEDLINE | ID: mdl-36213440

ABSTRACT

Multiple studies have indicated that long non-coding RNAs are aberrantly expressed in cancers and are pivotal in developing various tumors. No studies have investigated the expression and function of long non-coding antisense RNA PCNA-AS1 in esophageal squamous cell carcinoma (ESCC). In this study, the expression of PCNA-AS1 was identified by qRT-PCR. Cell function assays were used to explore the potential effect of PCNA-AS1 on ESCC progression. A prediction website was utilized to discover the relationships among PCNA-AS1, miR-2467-3p and proliferating cell nuclear antigen (PCNA). Dual luciferase reporter gene and RNA immunoprecipitation (RIP) assays were executed to verify the binding activity between PCNA-AS1, miR-2467-3p and PCNA. As a result, PCNA-AS1 was highly expressed in ESCC and was associated with patient prognosis. PCNA-AS1 overexpression strongly contributed to ESCC cell proliferation, invasion and migration. PCNA-AS1 and PCNA were positively correlated in ESCC. Bioinformatics analysis, RIP and luciferase reporter gene assays revealed that PCNA-AS1 could act as a competitive endogenous RNA to sponge miR-2467-3p, thus upregulating PCNA. In conclusion, the current outcome demonstrates that PCNA-AS1 may be a star molecule in the treatment of ESCC.

3.
Kidney Blood Press Res ; 47(12): 702-710, 2022.
Article in English | MEDLINE | ID: mdl-36260980

ABSTRACT

INTRODUCTION: Renal cell carcinoma (RCC) generally has a poor prognosis because of late diagnosis and metastasis. Despite its abundance in RCC cells, the functions of kallikrein-related peptidase 4 (KLK4) in RCC cells remain unknown. The results of this investigation were examined to discover if KLK4 gene silencing influences the development of RCC cells. METHODS: The mRNA levels of KLK4 and the relationship between KLK4 and tumor stage in patients with RCC were analyzed from the GEPIA database. Real-time PCR and Western blotting were used to measure the mRNA and protein levels of KLK4. Cell Counting Kit 8 (CCK-8), colony formation, wound healing, and Transwell assays were used to examine the proliferation, invasion, and migration of RCC cells after KLK4 suppression. Finally, xenograft experiments in a mouse model helped understand the in vivo effects of KLK4 knockdown. RESULTS: Our research found that KLK4 expression was upregulated in the kidney chromophobe (KICH) specimens and cell lines. Moreover, inhibiting KLK4 growth led to a slowdown in RCC cell proliferation and colony formation. Additionally, KLK4 knockdown inhibited migration, invasion, and epithelial-mesenchymal transition (EMT) of RCC cells. AKT and ERK phosphorylation were enhanced with KLK4 silencing. In the nude mouse xenograft cancer model, KLK4 silencing also prevented the expression of Ki-67, CD105, and the growth of tumors. CONCLUSION: KLK4 accelerated KICH progression via the ERK/AKT signaling pathway, providing a novel regulatory mechanism for KICH pathogenesis.


Subject(s)
Carcinoma, Renal Cell , Kallikreins , Kidney Neoplasms , Animals , Humans , Mice , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger , Signal Transduction , Kallikreins/metabolism
4.
Front Oncol ; 12: 1041732, 2022.
Article in English | MEDLINE | ID: mdl-36313704

ABSTRACT

[This corrects the article DOI: 10.3389/fonc.2021.773864.].

5.
Mol Carcinog ; 61(9): 865-875, 2022 09.
Article in English | MEDLINE | ID: mdl-35730908

ABSTRACT

Oral squamous cell carcinoma (OSCC) is an aggressive and common malignancy in the head and neck, characterized by poor prognosis and high incidence. This study aimed to investigate the role of long noncoding RNA TFAP2A-AS1 in OSCC. The competing endogenous RNA network of TFAP2A-AS1 was constructed by bioinformatics analysis. The expressions of miR-1297, TFAP2A-AS1, and TFAP2A were measured by quantitative reverse transcription-polymerase chain reaction. The correlations of TFAP2A-AS1, miR-1297, and TFAP2A with clinicopathological characteristics of OSCC were assessed. RNA immunoprecipitation and dual-luciferase reporter assay were used to identify the target of miR-1297. Cell proliferation was measured by colony formation assay and [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay. Transwell assay and wound healing assay were performed to assess cell movement. TFAP2A-AS1 and TFAP2A were upregulated in OSCC and their expression levels were positively correlated. The levels of TFAP2A-AS1, miR-1297, and TFAP2A were also associated with lymphatic metastasis and the tumor-node-metastasis (TNM) stage of OSCC patients. TFAP2A-AS1 acted as a miR-1297 sponge. OSCC cell growth and movement were inhibited by miR-1297. Changes in the miR-1297 expression abolished the effects of TFAP2A-AS1 on OSCC cells. Additionally, TFAP2A was a target of miR-1297. TFAP2A promoted OSCC cell growth and migration/invasion, indicating that TFAP2A mediated the effects of TFAP2A-AS1 and miR-1297. TFAP2A-AS1 exerts an oncogenic effect in OSCC via the TFAP2A-AS1/miR-1297/TFAP2A axis, which may provide new targets and strategies for OSCC treatments.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , MicroRNAs , Mouth Neoplasms , RNA, Long Noncoding , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
6.
Exp Cell Res ; 416(2): 113130, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35364057

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor worldwide. Long noncoding RNAs (lncRNAs) exhibit a regulatory role in the progression of ESCC. Our research was performed to investigate the potential molecular mechanism of lncRNA GATA2-AS1 in ESCC. METHODS: The expression of GATA2-AS1 was identified by qRT-PCR. Cell function assays explored the potential effect of GATA2-AS1 on ESCC progression. The subcellular hierarchical localization method was executed to identify the subcellular localization of GATA2-AS1 in ESCC cells. A prediction website was utilized to discover the relationships among GATA2-AS1, miR-940 and PTPN12. Dual luciferase reporter gene, pull-down assays and RIP assays were executed to verify the binding activity among GATA2-AS1, miR-940 and PTPN12. Xenograft tumor experiments were performed to evaluate ESCC cell growth in vivo. RESULTS: The expression of GATA2-AS1 and PTPN12 was reduced, while miR-940 expression was enhanced in ESCC tissues and cell lines. In vivo experiments showed that GATA2-AS1 inhibited the progression of ESCC cells toward malignancy. Bioinformatics analysis, dual luciferase and RIP assays revealed that GATA2-AS1 upregulated PTPN12 expression by competitively targeting miR-940. miR-940 reversed the inhibitory effect of GATA2-AS1 on the biological behavior of ESCC cells. CONCLUSION: Our findings suggested that GATA2-AS1, expressed at low levels in ESCC, plays a crucial role in the progression of ESCC by targeting the miR-940/PTPN12 axis and could be a potential drug target to treat ESCC patients.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Protein Tyrosine Phosphatase, Non-Receptor Type 12 , RNA, Long Noncoding , Cell Line, Tumor , Cell Proliferation/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
7.
J Oncol ; 2022: 7346460, 2022.
Article in English | MEDLINE | ID: mdl-35342412

ABSTRACT

Renal cell carcinoma (RCC) serves as a prevalent malignancy of urinary system and presents severe mortality and increasing incidence. Long noncoding RNAs (lncRNAs) have demonstrated critical roles in RCC development. Here, we were interested in the function of MMP2-AS1 during RCC progression. We observed that MP2-AS1 localized in both nucleus and cytoplasm of RCC cells using fluorescent in situ hybridization (FISH). The cell viability, proliferation, invasion, and migration of RCC cells were reduced by the depletion of MMP2-AS1. The MMP2-AS1 depletion-inhibited viability, proliferation, migration, and invasion of RCC cells were rescued by the overexpression of MMP2 in vitro. Consistently, the tumor growth of RCC cells was repressed by the depletion of MMP2-AS1 in the nude mice, while the overexpression of MMP2 could reverse this effect in vivo. Mechanically, we predicted the potential interaction of miR-34c-5p with both MMP2-AS1 and MMP2. The treatment of miR-34c-5p mimic reduced the luciferase activity of MMP2-AS1 and MMP2 3'UTR. The depletion of MMP2-AS1 enhanced miR-34c-5p expression and the expression of MMP2 was inhibited by miR-34c-5p in RCC cells. The protein levels of MMP2 were downregulated by MMP2-AS1 knockdown, while the inhibitor of miR-34c-5p rescued the expression of MMP2 in the cells. The treatment of miR-34c-5p mimic attenuated the cell viability, proliferation, invasion, and migration of RCC cells, in which MMP2 overexpression restored the phenotypes. MMP2-AS1 depletion-attenuated viability, proliferation, migration, and invasion of RCC cells were reversed by miR-34c-5p inhibitor. We concluded that MMP2-AS1 contributed to progression of renal cell carcinoma by modulating miR-34c-5p/MMP2 axis.

8.
Mol Biol Rep ; 49(3): 1837-1846, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34825321

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (CCRCC) is one of the most common types of renal cell carcinoma. Accumulating evidence indicates that homeobox D10 (HOXD10) acts as a tumor suppressor or oncogene in various carcinomas. However, the regulation and potential mechanisms of HOXD10 in CCRCC remain largely unknown. PURPOSE: To explore the effect and potential mechanism of HOXD10 on the invasion and migration of CCRCC cells. METHODS: The expression of HOXD10, E-cadherin and other epithelial mesenchymal transition (EMT)-related proteins was assessed by reverse transcription-quantitative real-time PCR (qRT-PCR) and Western blots. A series of functional assays were performed in RCC cell lines to explore the function of HOXD10 in CCRCC progression. Bioinformatics analysis, ChIP assays, and dual luciferase reporter assays were utilized to identify the interaction between HOXD10 and E-cadherin. RESULTS: Low expression of HOXD10 and E-cadherin was observed in CCRCC tissues and ACHN and 786-O cells. Downregulation of HOXD10 expression was correlated with the TNM stage of CCRCC patients. Functional experiments demonstrated that malignant biological ability was significantly inhibited by HOXD10 overexpression in RCC cells. Moreover, E-cadherin was a potential target gene of HOXD10, as evidenced by a series of assays. In addition, overexpression of HOXD10 inhibited the progression of CCRCC by regulating the expression of E-cadherin, vimentin, and ß-catenin in vitro. CONCLUSION: HOXD10 acts as a tumor suppressor and suppresses invasion and migration of CCRCC cells by regulating E-cadherin and EMT processes. Thus, targeting HOXD10 may be a therapeutic strategy for CCRCC treatment.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Homeobox , Homeodomain Proteins , Humans , Kidney Neoplasms/metabolism , Transcription Factors , Up-Regulation/genetics
9.
Front Oncol ; 11: 773864, 2021.
Article in English | MEDLINE | ID: mdl-34970490

ABSTRACT

Malignant tumors are a grave threat to human health. Esophageal squamous cell carcinoma (ESCC) is a common gastrointestinal malignant tumor. China has a high incidence of ESCC, and its morbidity and mortality are higher than the global average. Increasingly, studies have shown that long noncoding RNAs (lncRNAs) play a vital function in the occurrence and development of tumors. Although the biological function of FOXP4-AS1 has been demonstrated in various tumors, the potential molecular mechanism of FOXP4-AS1 in ESCC is still poorly understood. The expression of FOXP4 and FOXP4-AS1 was detected in ESCC by quantitative real-time PCR (qRT-PCR) or SP immunohistochemistry (IHC). shRNA was used to silence gene expression. Apoptosis, cell cycle, MTS, colony formation, invasion and migration assays were employed to explore the biological functions of FOXP4 and FOXP4-AS1. The potential molecular mechanism of FOXP4-AS1 in ESCC was determined by dual-luciferase reporter, RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP). Here, we demonstrated that FOXP4-AS1 was significantly increased in ESCC tissues and cell lines, associated with lymph node metastasis and TNM staging. Cell function experiments showed that FOXP4-AS1 promoted the proliferation, invasion and migration ability of ESCC cells. The expression of FOXP4-AS1 and FOXP4 in ESCC tissues was positively correlated. Further research found that FOXP4-AS1, upregulated in ESCC, promotes FOXP4 expression by enriching MLL2 and H3K4me3 in the FOXP4 promoter through a "molecular scaffold". Moreover, FOXP4, a transcription factor of ß-catenin, promotes the transcription of ß-catenin and ultimately leads to the malignant progression of ESCC. Finally, FOXP4-AS1 may be a new therapeutic target for ESCC.

10.
Mol Carcinog ; 60(12): 859-873, 2021 12.
Article in English | MEDLINE | ID: mdl-34551139

ABSTRACT

The transcription factor forkhead box D3 (FOXD3) is an important member of the FOX family, which can maintain the pluripotent properties of cell clusters, neural crest, and trophoblastic progenitor cells in vivo. It has been shown that FOXD3 could affect proliferation, migration, and angiogenesis of various tumors and its deletion and overexpression in organisms will undoubtedly have important influence on the change of cell fate and the occurrence of tumors. However, the underlying functions and molecular mechanisms of FOXD3 in esophageal squamous cell carcinoma (ESCC) have not been fully clarified. According to the present study, the expression levels and functional roles of FOXD3 were investigated, and its prognostic value and molecular mechanisms in tumorigenesis and progression of ESCC were clarified. The expression level of FOXD3 was significantly downregulated in ESCC tissues and cell lines, and correlated with gender, family history of upper gastrointestinal cancer, TNM stage, depth of invasion, lymph node metastasis, and ESCC patients' survival. Moreover, FOXD3 inhibited cells migration and invasion as well as participated in TGF-ß1 induced epithelial-mesenchymal transition process. Furthermore, a positive correlation between FOXD3 and SMAD family member 7 (SMAD7) was explored in ESCC. FOXD3 could directly bind to promoter regions of SMAD7 gene, leading to transcriptional promotion of SMAD7 in human esophageal cancer cells. Taken together, FOXD3 may play a tumor suppressor role in ESCC and may be applied as a new therapeutic target and prognostic marker for ESCC.


Subject(s)
Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Forkhead Transcription Factors/metabolism , Smad7 Protein/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Promoter Regions, Genetic
11.
Clin Lab ; 67(7)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34258957

ABSTRACT

BACKGROUND: IncRNAs perform complex functions and play an essential role in all stages of tumor progression. However, there are few studies that discuss the function of lncRNA ZNF667-AS1in oral squamous cell carcinoma (OSCC). This study aimed at analyzing the expression and biological behavior of lncRNA ZNF667-AS1 in OSCC. METHODS: IncRNA ZNF667-AS1 expression level in OSCC tissues and cell lines was explored by real-time PCR. The role of lncRNA ZNF667-AS1 on prognosis was elucidated. Cell proliferation assay, plate colony formation assay, wound-healing assay, and transwell migration assay were used to detect cell proliferation ability, cell clone formation ability, migration ability, and invasion ability, respectively. The effect of lncRNA ZNF667-AS1 on epithelial mesenchymal transformation (EMT) process was evaluated by western blot and real-time PCR. RESULTS: The expression levels of lncRNA ZNF667-AS1 were decreased in malignant tumor tissues. The OSCC patients with high expression of lncRNA ZNF667-AS1 had a longer survival time. IncRNA ZNF667-AS1 inhibited cell proliferation, cell clone formation ability, invasion and migration. Furthermore, lncRNA ZNF667-AS1 could inhibit the EMT process by suppressing transforming growth factor-ß-1 (TGF-ß1) expression, and TGF-ß1 treatment could partially restore the inhibitory effect. CONCLUSIONS: IncRNA ZNF667-AS1 may act as an antioncogene inhibiting the ability of proliferation, cell clone formation, invasion and migration, and suppress the process of EMT by targeting TGF-ß1. IncRNA ZNF667-AS1 could be a potential therapeutic target and a new predictive biological marker of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , RNA, Long Noncoding , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Mouth Neoplasms/genetics , RNA, Long Noncoding/genetics , Squamous Cell Carcinoma of Head and Neck , Transforming Growth Factor beta1/genetics
12.
Ann Palliat Med ; 10(1): 584-589, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33474959

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) is the most common type of kidney cancer, and accounts for approximately 3% of all malignancies. Metastatic RCC (mRCC) is not sensitive to traditional radiotherapy and chemotherapy, therefore targeted therapy has become an important treatment option. In this study, the second-line targeted drug everolimus (Afinitor), a mammalian target of rapamycin (mTOR) inhibitor, was investigated for its clinical efficacy and adverse events in mRCC after failure of first-line targeted therapy, such as sorafenib, sunitinib or pazopanib. METHODS: A total of 21 patients with mRCC who had been treated with surgery or other therapies such as tyrosine kinase inhibitors (TKIs) were given oral everolimus (10 mg/day) until disease progression. Clinical efficacy was evaluated using the Response Evaluation Criteria in Solid Tumors (RECIST) 2 months after therapy, including complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD). The adverse events were observed, and timely treatment was provided. RESULTS: Everolimus extended progression-free survival (PFS) in mRCC patients from 4 to 8 months (median 6.3 months). There were 3 patients with PR, 12 with SD, and 6 with PD, and the disease control rate (DCR) was 15/21 (71.4%). Common adverse events included stomatitis, rash, and pneumonitis. CONCLUSIONS: This study provides further support that everolimus is still an important option in mRCC treatment after failure of first-line targeted therapy. However, clinical studies are still needed to further improve its therapeutic efficacy.


Subject(s)
Antineoplastic Agents , Carcinoma, Renal Cell , Kidney Neoplasms , Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/drug therapy , Everolimus/adverse effects , Humans , Kidney Neoplasms/drug therapy , Sirolimus/adverse effects , Treatment Outcome
13.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-887414

ABSTRACT

@#[摘 要] 目的:探讨叉头框蛋白D3(forkhead box protein D3,FOXD3)在胃贲门腺癌(gastric cardia adenocarcinoma,GCA)中的表达及其对SGC-7901细胞生物学行为的影响。方法:从河北医科大学第四医院生物标本库中选取2014年6月至2016年12月手术切除的49例GCA组织及相应癌旁组织标本,qRT-PCR检测FOXD3在GCA组织、癌旁组织以及在5种胃癌细胞系中(BGC-823、SGC-7901、HGC-27、MGC-803及NCI-N87)的表达。向SGC-7901细胞转染pc-DNA3.1-FOXD3或pc-DNA3.1,采用细胞增殖实验、克隆形成实验、划痕愈合实验和Transwell小室侵袭实验分别检测FOXD3过表达对SGC-7901细胞增殖、克隆形成、迁移和侵袭的影响,qRT-PCR及WB法检测细胞转染前后上皮-间质转化(epithelial-mesenchymal transition,EMT)相关分子mRNA及蛋白的表达情况,流式细胞术检测转染前后细胞周期改变。结果:GCA组织中FOXD3 mRNA的表达量明显降低,其表达水平与患者临床分期和淋巴结转移密切关联;FOXD3在胃癌细胞系中的表达均低于正常细胞(均P<0.01)。FOXD3过表达能明显抑制SGC-7901细胞的增殖、克隆形成、迁移和侵袭能力(均P<0.01),提高SGC-7901细胞中E-cadherin的表达水平,减少N-cadherin、β-catenin和vimentin的表达水平(均P<0.01),使细胞周期阻滞在G0/G1期(P<0.01)。结论: FOXD3在GCA组织中的表达明显下调,其过表达可以抑制胃癌细胞的生物学行为,FOXD3可作为抑癌基因为肿瘤治疗提供新思路。

14.
Mol Carcinog ; 59(12): 1392-1408, 2020 12.
Article in English | MEDLINE | ID: mdl-33079409

ABSTRACT

Countless evidence suggests that long noncoding RNAs (lncRNAs) are involved in human malignant cancers, including esophageal squamous cell carcinoma (ESCC), although their exact function remains unclear. In the present study, we aimed to investigate the roles and molecular mechanisms of the lncRNA LOC440173 in ESCC progression. Microarray analysis and quantitative real-time polymerase chain reaction were conducted to measure the expression levels of LOC440173 and miR-30d-5p. The biological function of this lncRNA was investigated using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, clone formation, and transwell assays, as well as flow cytometry and Western blot analysis. The function of LOC440173 was validated in vivo using tumor xenografts. The regulatory network of LOC440173/miR-30d-5p/HDAC9 was established using bioinformatic analysis and verified with dual-luciferase reporter assays, RNA immunoprecipitation assay, and rescue experiments. The expression level of LOC440173 was significantly increased in ESCC tissues and esophageal carcinoma cells. High LOC440173 expression was correlated with histological grade, tumor invasion depth, lymph node metastasis, and TNM stage. Overexpression of LOC440173 promoted esophageal cancer cell proliferation, migration, and invasion, as well as the epithelial-mesenchymal transition (EMT) process in vitro, and facilitated tumor growth in vivo. MicroRNA-30d-5p (miR-30d-5p) was downregulated in ESCC tissues and acted as a direct binding target of LOC440173 during the regulation of HDAC9 expression in esophageal carcinoma cells. In conclusion, LOC440173 exerts a promotive role in ESCC tumorigenesis by targeting the miR-30d-5p/HDAC9 axis and regulating the EMT process. LOC440173 might be a new therapeutic target for the treatment of ESCC.


Subject(s)
Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Histone Deacetylases/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Aged , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Up-Regulation
15.
ACS Omega ; 5(24): 14805-14813, 2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32596618

ABSTRACT

Designing Pt-based alloy catalysts with multicomponent composition and a controllable structure is important to improve the utilization efficiency of precious metals and catalytic activity, but it still face a lot of challenges for simple preparation. Herein, we used insulin amyloid fibrils as templates and their own one-dimensional spiral structure to synthesize Pt-Rh-Pd ternary alloy nanochains under mild conditions. The prepared Pt-Rh-Pd alloy nanochains (NCs) have uniform diameter, and the particle size is only 2 nm. This ultrafine structure increases the specific surface area of the catalyst to a certain extent, and the synergistic effect of the three metals improves the catalytic performance. Compared with commercial Pt/C and binary Pt-Rh NCs, the as-presented Pt-Rh-Pd NCs show better methanol oxidation activity ability and stability against CO poisoning. The peak current density of front sweep is 1.48 mA cm-2, which is 1.7 times higher than that of commercial Pt/C (0.89 mA cm-2) and 1.4 times higher than that of the Pt-Rh NCs (1.07 mA cm-2), indicating great application potential as high-performance electrocatalysts in fuel cells.

16.
Arch Biochem Biophys ; 686: 108371, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32325088

ABSTRACT

Understanding the role of Long non-coding RNAs (lncRNAs) in tumorigenesis in diverse human malignancies would helpful for targeted therapies, containing esophageal squamous cell carcinoma (ESCC). However, the specific role and molecular mechanisms of LINC01980 in ESCC remain unclarified. In this study, we investigated the expression level, function role, and molecular mechanisms of LINC01980 in esophageal cancer cells and ESCC tissues. The high expression of LINC01980 was detected in ESCC tissues and cells, and predicted poor prognosis. LINC01980 promoted the cell proliferation, migration, invasion ability and epithelial-mesenchymal transition (EMT) progress in ESCC cells. In addition, a negative correlation between LINC01980 and miR-190a-5p or miR-190a-5p and MYO5A was observed in ESCC. We found that miR-190a-5p could directly bind with the mRNA of LINC01980 and MYO5A, and it was detected low expression in ESCC. We further demonstrated that the downregulation of MYO5A caused by overexpressing miR-190a-5p was released via upregulation of LINC01980. Functionally, LINC01980 acted as a competitively endogenous RNA (ceRNA) to impact the expression of MYO5A by sponging miR-190a-5p in ESCC. Therefore, these findings suggest that LINC01980 may act as an oncogenic lncRNA in ESCC and LINC01980/miR-190a-5p/MYO5A pathway contributes to the development of ESCC.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , MicroRNAs/metabolism , Myosin Heavy Chains/genetics , Myosin Type V/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Down-Regulation , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Humans
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-822990

ABSTRACT

@#[Abstract] Objective: To investigate the expression of lncRNA linc01503 in esophageal squamous cell carcinoma (ESCC) tissues and cell lines, as well as its effect on the proliferation, invasion and epithelial-mesenchymal transition (EMT) of ESCC cells. Methods: A total of 119 pairs of tumor tissues and corresponding para-cancerous tissues of ESCC patients were obtained from the Fourth Affiliated Hospital of Hebei Medical University between Jan. 2012 and Dec. 2014. The expression of linc01503 in ESCC tissues and cell lines (Kyse150, Kyse170, Eca109, TE1 and TE13) was detected by qPCR. The ESCC cells were transfected with pGPU6-shRNA-linc01503 or treated with TGF-β. The expressions of EMT related genes before and after transfection as well as linc01503 expression before and after TGF-β treatment were detected with qPCR. MTS and Transwell assay were performed to assess the effect of linc01503 on proliferation and invasion of ESCC cells. Results: The expression of linc01503 was significantly elevated in ESCC tissues and cell lines (all P<0.05). High expression of linc01503 was correlated with lymph node metastasis, depth of infiltration, TNM stage and the survival of ESCC patients (all P<0.05). Treatment with TGF-β promoted EMT of ESCC cells and induced a significant up-regulation of linc01503 expression. Knockdown of linc01503 significantly inhibited proliferation and invasion ability of ESCC cells; Meanwhile, the low expression of linc01503 increased the expression of E-cadherin but decreased the expressions of N-cadherin and vimentin (all P<0.05). Conclusion: lncRNA linc01503, as one of the downstream effect genes of TGF- β, promotes the proliferation, invasion and EMT process of ESCC cells.

18.
Cell Death Dis ; 10(12): 930, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804468

ABSTRACT

Natural antisense lncRNAs can interfere with their corresponding sense transcript to elicit concordant or discordant regulation. LncRNA ZNF667-AS1 and its sense gene ZNF667 were found to be downregulated in esophageal squamous cell carcinoma (ESCC) tissues by RNA sequencing; however, the exact roles of both genes in ESCC occurrence and development have not been clarified. This study was to investigate the expression patterns, epigenetic inactivation mechanisms, function, and prognostic significance of ZNF667-AS1 and ZNF667 in ESCC tumorigenesis. Frequent downregulation of ZNF667-AS1 and ZNF667 was detected in esophageal cancer cells and ESCC tissues. The expression levels of ZNF667-AS1 and ZNF667 were significantly reversed by treatment with 5-Aza-dC and TSA in esophageal cancer cell lines. The CpG sites hypermethylation within proximal promoter influenced the binding ability of transcription factor E2F1 to the binding sites and then affected the transcription and expression of ZNF667-AS1 and ZNF667. Overexpression of ZNF667-AS1 and ZNF667 suppressed the viability, migration, and invasion of esophageal cancer cells in vitro. Overexpression of ZNF667-AS1 increased mRNA and protein expression level of ZNF667. ZNF667-AS1 interacts with and recruits TET1 to its target gene ZNF667 and E-cadherin to hydrolyze 5'-mc to 5'-hmc and further activates their expression, meanwhile, ZNF667-AS1 also interacts with UTX to decrease histone H3K27 tri-methylation to activate ZNF667 and E-cadherin expression. Furthermore, ZNF667-AS1 or ZNF667 expression and promoter methylation status were correlated with ESCC patients' survival. Thus, these findings suggest that ZNF667-AS1 and ZNF667 may act as tumor suppressors and may serve as potential targets for antitumor therapy.


Subject(s)
Carrier Proteins/genetics , Epigenesis, Genetic , Esophageal Squamous Cell Carcinoma/genetics , Oncogene Proteins/genetics , RNA, Long Noncoding/genetics , Adult , Aged , Antigens, CD/genetics , Biomarkers, Tumor/genetics , Cadherins/genetics , Cell Proliferation/genetics , DNA Methylation/genetics , Disease Progression , Disease-Free Survival , Esophageal Squamous Cell Carcinoma/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Prognosis , Promoter Regions, Genetic/genetics
19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-793113

ABSTRACT

@#Objective: To detect the expression of non-receptor protein tyrosine phosphatase 6 (PTPN6) in different esophageal squamous cell carcinoma (ESCC) cell lines, and to investigate its effect on proliferation, migration and invasion ability of Eca109 and Yes-2 cell lines. Methods: qPCR was applied to detect the mRNA expression of PTPN6 in different ESCC cell lines (TE1, Eca109, Kyse150, Kyse170 and Yes-2). pcDNA3.1-PTPN6 plasmid was transiently transfected into Eca109 and Yes-2 cells respectively. The expression of PTPN6 was detected by real-time PCR and Wb. The effects of PTPN6 over-expression on the biological behaviors of ESCC cells were detected by MTS, colony formation assay, wound healing assay and Transwell assay, respectively. Results: The mRNA expression of PTPN6 was remarkably reduced in ESCC cell lines (TE1, Eca109, Kyse150, Kyse170 and Yes-2) compared to normal esophageal epithelial cells (HEEpiC) (P<0.05). Compared to the mock cells, significant up-regulation of PTPN6 was detected in pcDNA3.1-PTPN6 transfected Eca109 and Yes-2 cells (all P<0.05 or P<0.01); PTPN6 over-expression led to a significant inhibition in migration and invasion ability of Eca109 and Yes-2 cells (all P<0.05). Conclusions: Over-expression of PTPN6 may inhibit the proliferation, migration and invasion of ESCC cells, which might be an important factor influencing the biological characteristics of ESCC cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...