Peptidyl-prolyl isomerase F as a prognostic biomarker associated with immune infiltrates and mitophagy in lung adenocarcinoma.

IF 4 2区 医学 Q2 ONCOLOGY Translational lung cancer research Pub Date : 2024-06-30 Epub Date: 2024-06-20 DOI:10.21037/tlcr-24-344
Zitong Feng, Lin Yuan, Luyuan Ma, Wenhao Yu, Fayez Kheir, Lukas Käsmann, Wolfgang M Brueckl, Kai Jin, Dingxin Wang, Yi Shen, Rongyang Li, Hui Tian
{"title":"Peptidyl-prolyl isomerase F as a prognostic biomarker associated with immune infiltrates and mitophagy in lung adenocarcinoma.","authors":"Zitong Feng, Lin Yuan, Luyuan Ma, Wenhao Yu, Fayez Kheir, Lukas Käsmann, Wolfgang M Brueckl, Kai Jin, Dingxin Wang, Yi Shen, Rongyang Li, Hui Tian","doi":"10.21037/tlcr-24-344","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Lung adenocarcinoma (LUAD) is among the most prevalent malignancies worldwide, with unfavorable treatment outcomes. Peptidyl-prolyl isomerase F (<i>PPIF</i>) is known to influence the malignancy traits of tumor progression by modulating the bioenergetics and mitochondrial permeability in cancer cells; however, its role in LUAD remains unclear. Our study seeks to investigate the clinical significance, tumor proliferation, and immune regulatory functions of <i>PPIF</i> in LUAD.</p><p><strong>Methods: </strong>The expression of <i>PPIF</i> in LUAD tissues and cells was assessed using bioinformatics analysis, immunohistochemistry (IHC), and Western blotting. Survival curve analysis was conducted to examine the prognostic association between <i>PPIF</i> expression and LUAD. The immunomodulatory role of <i>PPIF</i> in LUAD was assessed through the analysis of <i>PPIF</i> expression and immune cell infiltration. A series of gain- and loss-of-function experiments were conducted on <i>PPIF</i> to investigate its biological functions in LUAD both <i>in vitro</i> and <i>in vivo</i>. The mechanisms underlying <i>PPIF</i>'s effects on LUAD were delineated through functional enrichment analysis and Western blotting assays.</p><p><strong>Results: </strong><i>PPIF</i> exhibited overexpression in LUAD tissues compared to normal controls. Survival curve analysis revealed that patients with LUAD exhibiting higher <i>PPIF</i> expression demonstrated decreased overall survival and a shorter progression-free interval. <i>PPIF</i> was implicated in modulating immune cell infiltration, particularly in regulating the T helper 1-T helper 2 cell balance. Functionally, <i>PPIF</i> was discovered to promote tumor cell proliferation and advance cell-cycle progression. Furthermore, <i>PPIF</i> could impede mitophagy by targeting the FOXO3a/PINK1-Parkin signaling pathway.</p><p><strong>Conclusions: </strong>The findings of this study indicate that the prognosis-related gene <i>PPIF</i> may have a significant role in the regulation of LUAD cell proliferation, tumor-associated immune cell infiltration, and mitophagy, and thus <i>PPIF</i> may be a promising therapeutic target of LUAD.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0000,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11225036/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Translational lung cancer research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.21037/tlcr-24-344","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/6/20 0:00:00","PubModel":"Epub","JCR":"Q2","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Lung adenocarcinoma (LUAD) is among the most prevalent malignancies worldwide, with unfavorable treatment outcomes. Peptidyl-prolyl isomerase F (PPIF) is known to influence the malignancy traits of tumor progression by modulating the bioenergetics and mitochondrial permeability in cancer cells; however, its role in LUAD remains unclear. Our study seeks to investigate the clinical significance, tumor proliferation, and immune regulatory functions of PPIF in LUAD.

Methods: The expression of PPIF in LUAD tissues and cells was assessed using bioinformatics analysis, immunohistochemistry (IHC), and Western blotting. Survival curve analysis was conducted to examine the prognostic association between PPIF expression and LUAD. The immunomodulatory role of PPIF in LUAD was assessed through the analysis of PPIF expression and immune cell infiltration. A series of gain- and loss-of-function experiments were conducted on PPIF to investigate its biological functions in LUAD both in vitro and in vivo. The mechanisms underlying PPIF's effects on LUAD were delineated through functional enrichment analysis and Western blotting assays.

Results: PPIF exhibited overexpression in LUAD tissues compared to normal controls. Survival curve analysis revealed that patients with LUAD exhibiting higher PPIF expression demonstrated decreased overall survival and a shorter progression-free interval. PPIF was implicated in modulating immune cell infiltration, particularly in regulating the T helper 1-T helper 2 cell balance. Functionally, PPIF was discovered to promote tumor cell proliferation and advance cell-cycle progression. Furthermore, PPIF could impede mitophagy by targeting the FOXO3a/PINK1-Parkin signaling pathway.

Conclusions: The findings of this study indicate that the prognosis-related gene PPIF may have a significant role in the regulation of LUAD cell proliferation, tumor-associated immune cell infiltration, and mitophagy, and thus PPIF may be a promising therapeutic target of LUAD.

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
肽基脯氨酰异构酶F是与肺腺癌免疫浸润和有丝分裂相关的预后生物标志物
背景:肺腺癌(LUAD)是全球发病率最高的恶性肿瘤之一,治疗效果不佳。众所周知,肽基脯氨酰异构酶 F(PPIF)可通过调节癌细胞的生物能和线粒体通透性来影响肿瘤进展的恶性特征;然而,它在肺腺癌中的作用仍不清楚。我们的研究旨在探讨 PPIF 在 LUAD 中的临床意义、肿瘤增殖和免疫调节功能:方法:采用生物信息学分析、免疫组织化学(IHC)和 Western 印迹法评估 PPIF 在 LUAD 组织和细胞中的表达。研究人员对PPIF的表达与LUAD的预后关系进行了生存曲线分析。通过分析 PPIF 表达和免疫细胞浸润,评估了 PPIF 在 LUAD 中的免疫调节作用。对PPIF进行了一系列功能增益和功能缺失实验,以研究其在体外和体内LUAD中的生物学功能。通过功能富集分析和Western印迹分析,研究了PPIF对LUAD的影响机制:结果:与正常对照组相比,PPIF在LUAD组织中呈现过表达。生存曲线分析表明,PPIF表达较高的LUAD患者总生存率下降,无进展间隔时间缩短。PPIF 参与调节免疫细胞浸润,尤其是调节 T 辅助细胞 1-T 辅助细胞 2 的平衡。在功能上,研究发现 PPIF 能促进肿瘤细胞增殖并推进细胞周期的进展。此外,PPIF还能通过靶向FOXO3a/PINK1-Parkin信号通路阻碍有丝分裂:本研究结果表明,预后相关基因PPIF可能在LUAD细胞增殖、肿瘤相关免疫细胞浸润和有丝分裂的调控中发挥重要作用,因此PPIF可能是LUAD的一个有前景的治疗靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
7.20
自引率
2.50%
发文量
137
期刊介绍: Translational Lung Cancer Research(TLCR, Transl Lung Cancer Res, Print ISSN 2218-6751; Online ISSN 2226-4477) is an international, peer-reviewed, open-access journal, which was founded in March 2012. TLCR is indexed by PubMed/PubMed Central and the Chemical Abstracts Service (CAS) Databases. It is published quarterly the first year, and published bimonthly since February 2013. It provides practical up-to-date information on prevention, early detection, diagnosis, and treatment of lung cancer. Specific areas of its interest include, but not limited to, multimodality therapy, markers, imaging, tumor biology, pathology, chemoprevention, and technical advances related to lung cancer.
期刊最新文献
LungPath: artificial intelligence-driven histologic pattern recognition for improved diagnosis of early-stage invasive lung adenocarcinoma. A nomogram predicting the risk of extrathoracic metastasis at initial diagnosis of T≤3cmN0 lung cancer. Are PD-1T TILs merely an expensive and unuseful whim as biomarker? Assessing the transportability of radiomic models for lung cancer diagnosis: commercial vs. open-source feature extractors. Breaking barriers: patient-derived xenograft (PDX) models in lung cancer drug development-are we close to the finish line?
×
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