ASAP1 通过 Wnt/β-Catenin 通路促进胆管癌进展

IF 2.9 4区 医学 Q1 Medicine Journal of biomedical nanotechnology Pub Date : 2024-04-01 DOI:10.1166/jbn.2024.3814
Jiaqi He, Han Liu, Jianhua Cai, Sheng Shen, Ji-wen Wang, Houbao Liu
{"title":"ASAP1 通过 Wnt/β-Catenin 通路促进胆管癌进展","authors":"Jiaqi He, Han Liu, Jianhua Cai, Sheng Shen, Ji-wen Wang, Houbao Liu","doi":"10.1166/jbn.2024.3814","DOIUrl":null,"url":null,"abstract":"This study sought to identify the relationship between ADP-ribosylation factor GTpase-activating protein (ASAP1) expression and clinical outcomes in Cholangiocarcinoma (CC) patients. Quantitative real-time PCR (qRT-PCR), Western blotting, and immunohistochemistry were used to analyze\n the expression of ASAP1 in CC tissue samples and cell lines (IHC). The survival rate and clinicopathological characteristics were also examined. Cell counting kit-8 (CCK-8), colony formation, and 5-ethynyl-2′-deoxyuridine (EdU) assays were used to detect cell proliferation. Flow cytometry\n was used to assess the cell cycle. The terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) test and flow cytometry were used to identify cell apoptosis. Xenograft tumor development in living mice was reported. ASAP1 expression was increased and associated with\n a poor prognosis in CC tissue samples. The expression of ASAP1 was associated with the tumor’s histological grade and size in clinical specimens. In vitro and in vivo, knocking down ASAP1 expression resulted in decreased ASAP1 cell proliferation, inhibited cell cycle progression,\n and increased apoptosis. ASAP1 cholangiocarcinoma controls the Wnt/β-catenin pathway’s activity, encourages cell apoptosis, migration, and invasion in culture, and fosters tumor development in vivo. ASAP1 was crucial to the origin and growth of CC tumors, which could\n be a beneficial treatment target for CC.","PeriodicalId":15260,"journal":{"name":"Journal of biomedical nanotechnology","volume":null,"pages":null},"PeriodicalIF":2.9000,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"ASAP1 Promotes Cholangiocarcinoma Progression via Wnt/β-Catenin Pathway\",\"authors\":\"Jiaqi He, Han Liu, Jianhua Cai, Sheng Shen, Ji-wen Wang, Houbao Liu\",\"doi\":\"10.1166/jbn.2024.3814\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"This study sought to identify the relationship between ADP-ribosylation factor GTpase-activating protein (ASAP1) expression and clinical outcomes in Cholangiocarcinoma (CC) patients. Quantitative real-time PCR (qRT-PCR), Western blotting, and immunohistochemistry were used to analyze\\n the expression of ASAP1 in CC tissue samples and cell lines (IHC). The survival rate and clinicopathological characteristics were also examined. Cell counting kit-8 (CCK-8), colony formation, and 5-ethynyl-2′-deoxyuridine (EdU) assays were used to detect cell proliferation. Flow cytometry\\n was used to assess the cell cycle. The terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) test and flow cytometry were used to identify cell apoptosis. Xenograft tumor development in living mice was reported. ASAP1 expression was increased and associated with\\n a poor prognosis in CC tissue samples. The expression of ASAP1 was associated with the tumor’s histological grade and size in clinical specimens. In vitro and in vivo, knocking down ASAP1 expression resulted in decreased ASAP1 cell proliferation, inhibited cell cycle progression,\\n and increased apoptosis. ASAP1 cholangiocarcinoma controls the Wnt/β-catenin pathway’s activity, encourages cell apoptosis, migration, and invasion in culture, and fosters tumor development in vivo. ASAP1 was crucial to the origin and growth of CC tumors, which could\\n be a beneficial treatment target for CC.\",\"PeriodicalId\":15260,\"journal\":{\"name\":\"Journal of biomedical nanotechnology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.9000,\"publicationDate\":\"2024-04-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal of biomedical nanotechnology\",\"FirstCategoryId\":\"5\",\"ListUrlMain\":\"https://doi.org/10.1166/jbn.2024.3814\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"Medicine\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of biomedical nanotechnology","FirstCategoryId":"5","ListUrlMain":"https://doi.org/10.1166/jbn.2024.3814","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"Medicine","Score":null,"Total":0}
引用次数: 0

摘要

本研究旨在确定ADP-核糖基化因子GTpase-激活蛋白(ASAP1)的表达与胆管癌(CC)患者临床预后之间的关系。研究采用定量实时 PCR(qRT-PCR)、Western 印迹和免疫组织化学方法分析 ASAP1 在 CC 组织样本和细胞系(IHC)中的表达。此外,还检测了存活率和临床病理特征。细胞计数试剂盒-8(CCK-8)、菌落形成和 5-乙炔基-2′-脱氧尿苷(EdU)检测法用于检测细胞增殖。流式细胞仪用于评估细胞周期。末端脱氧核苷酸转移酶(TdT)介导的 dUTP 缺口末端标记(TUNEL)检测和流式细胞仪用于鉴定细胞凋亡。报告了活体小鼠的异种移植肿瘤发生情况。在CC组织样本中,ASAP1的表达增加并与不良预后相关。在临床样本中,ASAP1的表达与肿瘤的组织学分级和大小有关。在体外和体内,降低ASAP1的表达可减少ASAP1细胞的增殖,抑制细胞周期的进展,增加细胞凋亡。ASAP1胆管癌控制着Wnt/β-catenin通路的活性,在培养中促进细胞凋亡、迁移和侵袭,在体内促进肿瘤发展。ASAP1对CC肿瘤的起源和生长至关重要,可能成为CC的有利治疗靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
ASAP1 Promotes Cholangiocarcinoma Progression via Wnt/β-Catenin Pathway
This study sought to identify the relationship between ADP-ribosylation factor GTpase-activating protein (ASAP1) expression and clinical outcomes in Cholangiocarcinoma (CC) patients. Quantitative real-time PCR (qRT-PCR), Western blotting, and immunohistochemistry were used to analyze the expression of ASAP1 in CC tissue samples and cell lines (IHC). The survival rate and clinicopathological characteristics were also examined. Cell counting kit-8 (CCK-8), colony formation, and 5-ethynyl-2′-deoxyuridine (EdU) assays were used to detect cell proliferation. Flow cytometry was used to assess the cell cycle. The terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) test and flow cytometry were used to identify cell apoptosis. Xenograft tumor development in living mice was reported. ASAP1 expression was increased and associated with a poor prognosis in CC tissue samples. The expression of ASAP1 was associated with the tumor’s histological grade and size in clinical specimens. In vitro and in vivo, knocking down ASAP1 expression resulted in decreased ASAP1 cell proliferation, inhibited cell cycle progression, and increased apoptosis. ASAP1 cholangiocarcinoma controls the Wnt/β-catenin pathway’s activity, encourages cell apoptosis, migration, and invasion in culture, and fosters tumor development in vivo. ASAP1 was crucial to the origin and growth of CC tumors, which could be a beneficial treatment target for CC.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
4.30
自引率
17.20%
发文量
145
审稿时长
2.3 months
期刊介绍: Information not localized
期刊最新文献
Mechanism of miR-126 Loaded in Albumin Nanoparticles for Reversing the Multidrug Resistance in Breast Carcinoma Cells Application of 20(S)-Protopanaxadiol-Loaded Nanostructured Lipid Carriers for Diabetic Wound Healing and Vascular Regeneration LncRNA NEAT1 Promotes the Cancer Stem Cell-Like Properties of HCC by miR-128-3p/GP73 Axis Pterostilbene-Loaded Polydopamine Nanoparticles Down-Regulate Tumor Necrosis Factor-α and Improve Myocardial Function in Mice with Acute Myocardial Infarction Phacoemulsification Plus Intraocular Lens Implantation with Gold Nanoparticles for Complicated Cataract Secondary to Uveitis: Efficacy Analysis
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1