首页 > 最新文献

Molecular Cancer最新文献

英文 中文
CircPIAS1 promotes hepatocellular carcinoma progression by inhibiting ferroptosis via the miR-455-3p/NUPR1/FTH1 axis. CircPIAS1通过miR-455-3p/NUPR1/FTH1轴抑制铁变态反应,从而促进肝细胞癌的进展。
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-28 DOI: 10.1186/s12943-024-02030-x
Xiao-Yu Zhang, Shan-Shan Li, Yu-Rong Gu, Le-Xin Xiao, Xin-Yi Ma, Xin-Ru Chen, Jia-Liang Wang, Chun-Hong Liao, Bing-Liang Lin, Yue-Hua Huang, Yi-Fan Lian

Background: The role of circRNAs in hepatocellular carcinoma (HCC) progression remains unclear. CircPIAS1 (circBase ID: hsa_circ_0007088) was identified as overexpressed in HCC cases through bioinformatics analysis. This study aimed to investigate the oncogenic properties and mechanisms of circPIAS1 in HCC development.

Methods: Functional analyses were conducted to assess circPIAS1's impact on HCC cell proliferation, migration, and ferroptosis. Xenograft mouse models were employed to evaluate circPIAS1's effects on tumor growth and pulmonary metastasis in vivo. Bioinformatics analysis, RNA immunoprecipitation, and luciferase reporter assays were utilized to elucidate the molecular pathways influenced by circPIAS1. Additional techniques, including RNA pulldown, fluorescence in situ hybridization (FISH), chromatin immunoprecipitation (ChIP), qPCR, and western blotting, were used to further explore the underlying mechanisms.

Results: CircPIAS1 expression was elevated in HCC tissues and cells. Silencing circPIAS1 suppressed HCC cell proliferation and migration both in vitro and in vivo. Mechanically, circPIAS1 overexpression inhibited ferroptosis by competitively binding to miR-455-3p, leading to upregulation of Nuclear Protein 1 (NUPR1). Furthermore, NUPR1 promoted FTH1 transcription, enhancing iron storage in HCC cells and conferring resistance to ferroptosis. Treatment with ZZW-115, an NUPR1 inhibitor, reversed the tumor-promoting effects of circPIAS1 and sensitized HCC cells to lenvatinib.

Conclusion: This study highlights the critical role of circPIAS1 in HCC progression through modulation of ferroptosis. Targeting the circPIAS1/miR-455-3p/NUPR1/FTH1 regulatory axis may represent a promising therapeutic strategy for HCC.

背景:循环RNA在肝细胞癌(HCC)进展中的作用仍不清楚。通过生物信息学分析发现,circPIAS1(circBase ID:hsa_circ_0007088)在HCC病例中过度表达。本研究旨在探讨circPIAS1在HCC发展过程中的致癌特性和机制:方法:进行功能分析以评估 circPIAS1 对 HCC 细胞增殖、迁移和铁变态反应的影响。采用异种移植小鼠模型评估 circPIAS1 对体内肿瘤生长和肺转移的影响。生物信息学分析、RNA免疫沉淀和荧光素酶报告实验被用来阐明受circPIAS1影响的分子通路。其他技术包括RNA pulldown、荧光原位杂交(FISH)、染色质免疫沉淀(ChIP)、qPCR和Western印迹,用于进一步探索其潜在机制:结果:CircPIAS1在HCC组织和细胞中表达升高。沉默 circPIAS1 可抑制 HCC 细胞在体外和体内的增殖和迁移。在机制上,circPIAS1的过表达通过与miR-455-3p竞争性结合抑制了铁凋亡,导致核蛋白1(NUPR1)上调。此外,NUPR1 还能促进 FTH1 的转录,从而增强 HCC 细胞的铁储存,并赋予其对铁凋亡的抵抗力。用NUPR1抑制剂ZZW-115治疗可逆转circPIAS1的促瘤作用,并使HCC细胞对来伐替尼敏感:本研究强调了circPIAS1通过调节铁突变在HCC进展中的关键作用。以circPIAS1/miR-455-3p/NUPR1/FTH1调控轴为靶点可能是一种很有前景的HCC治疗策略。
{"title":"CircPIAS1 promotes hepatocellular carcinoma progression by inhibiting ferroptosis via the miR-455-3p/NUPR1/FTH1 axis.","authors":"Xiao-Yu Zhang, Shan-Shan Li, Yu-Rong Gu, Le-Xin Xiao, Xin-Yi Ma, Xin-Ru Chen, Jia-Liang Wang, Chun-Hong Liao, Bing-Liang Lin, Yue-Hua Huang, Yi-Fan Lian","doi":"10.1186/s12943-024-02030-x","DOIUrl":"10.1186/s12943-024-02030-x","url":null,"abstract":"<p><strong>Background: </strong>The role of circRNAs in hepatocellular carcinoma (HCC) progression remains unclear. CircPIAS1 (circBase ID: hsa_circ_0007088) was identified as overexpressed in HCC cases through bioinformatics analysis. This study aimed to investigate the oncogenic properties and mechanisms of circPIAS1 in HCC development.</p><p><strong>Methods: </strong>Functional analyses were conducted to assess circPIAS1's impact on HCC cell proliferation, migration, and ferroptosis. Xenograft mouse models were employed to evaluate circPIAS1's effects on tumor growth and pulmonary metastasis in vivo. Bioinformatics analysis, RNA immunoprecipitation, and luciferase reporter assays were utilized to elucidate the molecular pathways influenced by circPIAS1. Additional techniques, including RNA pulldown, fluorescence in situ hybridization (FISH), chromatin immunoprecipitation (ChIP), qPCR, and western blotting, were used to further explore the underlying mechanisms.</p><p><strong>Results: </strong>CircPIAS1 expression was elevated in HCC tissues and cells. Silencing circPIAS1 suppressed HCC cell proliferation and migration both in vitro and in vivo. Mechanically, circPIAS1 overexpression inhibited ferroptosis by competitively binding to miR-455-3p, leading to upregulation of Nuclear Protein 1 (NUPR1). Furthermore, NUPR1 promoted FTH1 transcription, enhancing iron storage in HCC cells and conferring resistance to ferroptosis. Treatment with ZZW-115, an NUPR1 inhibitor, reversed the tumor-promoting effects of circPIAS1 and sensitized HCC cells to lenvatinib.</p><p><strong>Conclusion: </strong>This study highlights the critical role of circPIAS1 in HCC progression through modulation of ferroptosis. Targeting the circPIAS1/miR-455-3p/NUPR1/FTH1 regulatory axis may represent a promising therapeutic strategy for HCC.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11131253/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141158549","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regulating LIMK2b via EZH2 更正:长非编码 RNA TUG1 通过 EZH2 调节 LIMK2b 参与小细胞肺癌的细胞生长和化疗耐受性
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-25 DOI: 10.1186/s12943-024-02026-7
Yuchun Niu, Feng Ma, Weimei Huang, Shun Fang, Man Li, Ting Wei, Linlang Guo

Correction: Mol Cancer16, 5 (2017)

https://doi.org/10.1186/s12943-016-0575-6

Following publication of the original article [1], the authors noticed that two images (Figs. 2E and 5C) in the plate clone experiment section were accidentally duplicated due to personal negligence.

They fully understand that this mistake may have caused inconvenience to the readers and raised doubts about the rigor of their research. The authors deeply apologize for this oversight and have immediately taken corrective measures.

The authors hereby confirm that this error was solely caused by carelessness and does not involve any form of academic misconduct. They have sufficient original experimental images as evidence to prove that the duplication was the result of mistakenly placing the wrong images. Although this error concerns the results of the proliferation experiment, the paper also used the CCK8 proliferation assay to validate their findings. In addition, their article also proved the effect on cell proliferation through in vivo experiments, further confirming their conclusion. Therefore, this error does not affect their conclusions regarding the impact on cell proliferation or the main findings of the paper. The correct Fig. 2 is given below.

Fig. 2
figure 1

TUG1 was up-regulated in SCLC cell lines and TUG1 knockdown inhibited cell proliferation in vitro. a The expression of TUG1 was assessed in SCLC cell lines compared with the normal bronchial epithelial cell line (16HBE) by qRT-PCR. b c Inhibition of TUG1 by transfection of TUG1 siRNAs or sh RNA in H69、H69AR、H446、H446DDP cells. d CCK-8 proliferation assays were used to determine the cell viability for siTUG1 transfected SCLC cells. Experiments were performed in triplicate. e Colony formation assays were performed to determine the proliferation of shTUG1 transfected H446, H446DDP and H69AR cells. Representative photographs are shown, and the numbers of colonies were counted. *, P < 0.05; **, P < 0.001

Full size image
  1. Niu Y, Ma F, Huang W, et al. Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regul

Mol Cancer 23, 112 (2024). https://doi.org/10.1186/s12943-024-02026-7Download citationPublished: 25 May 2024DOI: https://doi.org/10.1186/s12943-024-02026-7Share this articleAnyone you share with the following link will be able to read this content:Get shareable linkSorry, a shareable link is not currently available for this article.Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative
{"title":"Correction: Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regulating LIMK2b via EZH2","authors":"Yuchun Niu, Feng Ma, Weimei Huang, Shun Fang, Man Li, Ting Wei, Linlang Guo","doi":"10.1186/s12943-024-02026-7","DOIUrl":"https://doi.org/10.1186/s12943-024-02026-7","url":null,"abstract":"<p><b>Correction:</b> <b><i>Mol Cancer</i></b><b>16, 5 (2017)</b></p><p><b>https://doi.org/10.1186/s12943-016-0575-6</b></p><p> Following publication of the original article [1], the authors noticed that two images (Figs. 2E and 5C) in the plate clone experiment section were accidentally duplicated due to personal negligence.</p><p> They fully understand that this mistake may have caused inconvenience to the readers and raised doubts about the rigor of their research. The authors deeply apologize for this oversight and have immediately taken corrective measures.</p><p> The authors hereby confirm that this error was solely caused by carelessness and does not involve any form of academic misconduct. They have sufficient original experimental images as evidence to prove that the duplication was the result of mistakenly placing the wrong images. Although this error concerns the results of the proliferation experiment, the paper also used the CCK8 proliferation assay to validate their findings. In addition, their article also proved the effect on cell proliferation through in vivo experiments, further confirming their conclusion. Therefore, this error does not affect their conclusions regarding the impact on cell proliferation or the main findings of the paper. The correct Fig. 2 is given below.</p><figure><figcaption><b data-test=\"figure-caption-text\">Fig. 2</b></figcaption><picture><source srcset=\"//media.springernature.com/lw685/springer-static/image/art%3A10.1186%2Fs12943-024-02026-7/MediaObjects/12943_2024_2026_Figa_HTML.png?as=webp\" type=\"image/webp\"/><img alt=\"figure 1\" aria-describedby=\"Fig1\" height=\"750\" loading=\"lazy\" src=\"//media.springernature.com/lw685/springer-static/image/art%3A10.1186%2Fs12943-024-02026-7/MediaObjects/12943_2024_2026_Figa_HTML.png\" width=\"685\"/></picture><p>TUG1 was up-regulated in SCLC cell lines and TUG1 knockdown inhibited cell proliferation in vitro. <b>a</b> The expression of TUG1 was assessed in SCLC cell lines compared with the normal bronchial epithelial cell line (16HBE) by qRT-PCR. <b>b c</b> Inhibition of TUG1 by transfection of TUG1 siRNAs or sh RNA in H69、H69AR、H446、H446DDP cells. <b>d</b> CCK-8 proliferation assays were used to determine the cell viability for siTUG1 transfected SCLC cells. Experiments were performed in triplicate. <b>e</b> Colony formation assays were performed to determine the proliferation of shTUG1 transfected H446, H446DDP and H69AR cells. Representative photographs are shown, and the numbers of colonies were counted. *, <i>P</i> &lt; 0.05; **, <i>P</i> &lt; 0.001</p><span>Full size image</span><svg aria-hidden=\"true\" focusable=\"false\" height=\"16\" role=\"img\" width=\"16\"><use xlink:href=\"#icon-eds-i-chevron-right-small\" xmlns:xlink=\"http://www.w3.org/1999/xlink\"></use></svg></figure><ol data-track-component=\"outbound reference\"><li data-counter=\"1.\"><p>Niu Y, Ma F, Huang W, et al. Long non-coding RNA TUG1 is involved in cell growth and chemoresistance of small cell lung cancer by regul","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141096725","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PARP1-DOT1L transcription axis drives acquired resistance to PARP inhibitor in ovarian cancer PARP1-DOT1L 转录轴驱动卵巢癌患者对 PARP 抑制剂产生获得性耐药性
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-22 DOI: 10.1186/s12943-024-02025-8
Chaohua Liu, Jiana Li, Fei Xu, Lihua Chen, Mengdong Ni, Jiangchun Wu, Haiyun Zhao, Yangjun Wu, Jiajia Li, Xiaohua Wu, Xiaojun Chen
Poly (ADP-ribose) polymerase inhibitor (PARPi) resistance poses a significant challenge in ovarian carcinoma (OC). While the role of DOT1L in cancer and chemoresistance is acknowledged, its specific role in PARPi resistance remains unclear. This study aims to elucidate the molecular mechanism of DOT1L in PARPi resistance in OC patients. This study analyzed the expression of DOT1L in PARPi-resistant cell lines compared to sensitive ones and correlated it with clinical outcomes in OC patients. Comprehensive in vitro and in vivo functional experiments were conducted using cellular and mouse models. Molecular investigations, including RNA sequencing, chromatin immunoprecipitation (ChIP) and Cleavage Under Targets and Tagmentation (CUT&Tag) assays, were employed to unravel the molecular mechanisms of DOT1L-mediated PARPi resistance. Our investigation revealed a robust correlation between DOT1L expression and clinical PARPi resistance in non-BRCA mutated OC cells. Upregulated DOT1L expression in PARPi-resistant tissues was associated with diminished survival in OC patients. Mechanistically, we identified that PARP1 directly binds to the DOT1L gene promoter, promoting transcription independently of its enzyme activity. PARP1 trapping induced by PARPi treatment amplified this binding, enhancing DOT1L transcription and contributing to drug resistance. Sequencing analysis revealed that DOT1L plays a crucial role in the transcriptional regulation of PLCG2 and ABCB1 via H3K79me2. This established the PARP1-DOT1L-PLCG2/ABCB1 axis as a key contributor to PARPi resistance. Furthermore, we discovered that combining a DOT1L inhibitor with PARPi demonstrated a synergistic effect in both cell line-derived xenograft mouse models (CDXs) and patient-derived organoids (PDOs). Our results demonstrate that DOT1L is an independent prognostic marker for OC patients. The PARP1-DOT1L/H3K79me2-PLCG2/ABCB1 axis is identified as a pivotal contributor to PARPi resistance. Targeted inhibition of DOT1L emerges as a promising therapeutic strategy for enhancing PARPi treatment outcomes in OC patients.
多聚(ADP-核糖)聚合酶抑制剂(PARPi)耐药性是卵巢癌(OC)的一大挑战。虽然 DOT1L 在癌症和化疗耐药性中的作用已得到公认,但它在 PARPi 耐药性中的具体作用仍不清楚。本研究旨在阐明DOT1L在OC患者PARPi耐药中的分子机制。本研究分析了与敏感细胞株相比,DOT1L在PARPi耐药细胞株中的表达情况,并将其与OC患者的临床预后相关联。研究利用细胞和小鼠模型进行了全面的体外和体内功能实验。为了揭示DOT1L介导的PARPi耐药的分子机制,我们采用了包括RNA测序、染色质免疫沉淀(ChIP)和靶标下裂解及标记(CUT&Tag)检测在内的分子研究方法。我们的研究发现,在非BRCA突变的OC细胞中,DOT1L的表达与临床PARPi耐药之间存在密切的相关性。PARPi耐药组织中DOT1L表达上调与OC患者存活率降低有关。从机理上讲,我们发现 PARP1 可直接与 DOT1L 基因启动子结合,从而促进转录,而与其酶活性无关。PARPi治疗诱导的PARP1捕获扩大了这种结合,增强了DOT1L的转录并导致耐药性。测序分析表明,DOT1L 通过 H3K79me2 在 PLCG2 和 ABCB1 的转录调控中起着关键作用。这就确定了 PARP1-DOT1L-PLCG2/ABCB1 轴是 PARPi 耐药性的关键因素。此外,我们发现在细胞系衍生的异种移植小鼠模型(CDXs)和患者衍生的器官组织(PDOs)中,将 DOT1L 抑制剂与 PARPi 结合使用可产生协同效应。我们的研究结果表明,DOT1L是OC患者的独立预后标志物。PARP1-DOT1L/H3K79me2-PLCG2/ABCB1轴被认为是PARPi耐药的关键因素。靶向抑制 DOT1L 是一种很有前景的治疗策略,可提高 PARPi 对 OC 患者的治疗效果。
{"title":"PARP1-DOT1L transcription axis drives acquired resistance to PARP inhibitor in ovarian cancer","authors":"Chaohua Liu, Jiana Li, Fei Xu, Lihua Chen, Mengdong Ni, Jiangchun Wu, Haiyun Zhao, Yangjun Wu, Jiajia Li, Xiaohua Wu, Xiaojun Chen","doi":"10.1186/s12943-024-02025-8","DOIUrl":"https://doi.org/10.1186/s12943-024-02025-8","url":null,"abstract":"Poly (ADP-ribose) polymerase inhibitor (PARPi) resistance poses a significant challenge in ovarian carcinoma (OC). While the role of DOT1L in cancer and chemoresistance is acknowledged, its specific role in PARPi resistance remains unclear. This study aims to elucidate the molecular mechanism of DOT1L in PARPi resistance in OC patients. This study analyzed the expression of DOT1L in PARPi-resistant cell lines compared to sensitive ones and correlated it with clinical outcomes in OC patients. Comprehensive in vitro and in vivo functional experiments were conducted using cellular and mouse models. Molecular investigations, including RNA sequencing, chromatin immunoprecipitation (ChIP) and Cleavage Under Targets and Tagmentation (CUT&Tag) assays, were employed to unravel the molecular mechanisms of DOT1L-mediated PARPi resistance. Our investigation revealed a robust correlation between DOT1L expression and clinical PARPi resistance in non-BRCA mutated OC cells. Upregulated DOT1L expression in PARPi-resistant tissues was associated with diminished survival in OC patients. Mechanistically, we identified that PARP1 directly binds to the DOT1L gene promoter, promoting transcription independently of its enzyme activity. PARP1 trapping induced by PARPi treatment amplified this binding, enhancing DOT1L transcription and contributing to drug resistance. Sequencing analysis revealed that DOT1L plays a crucial role in the transcriptional regulation of PLCG2 and ABCB1 via H3K79me2. This established the PARP1-DOT1L-PLCG2/ABCB1 axis as a key contributor to PARPi resistance. Furthermore, we discovered that combining a DOT1L inhibitor with PARPi demonstrated a synergistic effect in both cell line-derived xenograft mouse models (CDXs) and patient-derived organoids (PDOs). Our results demonstrate that DOT1L is an independent prognostic marker for OC patients. The PARP1-DOT1L/H3K79me2-PLCG2/ABCB1 axis is identified as a pivotal contributor to PARPi resistance. Targeted inhibition of DOT1L emerges as a promising therapeutic strategy for enhancing PARPi treatment outcomes in OC patients.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141079390","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
New-generation advanced PROTACs as potential therapeutic agents in cancer therapy 新一代高级 PROTACs 作为潜在的癌症治疗药物
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-21 DOI: 10.1186/s12943-024-02024-9
Chao Wang, Yujing Zhang, Wujun Chen, Yudong Wu, Dongming Xing
Proteolysis-targeting chimeras (PROTACs) technology has garnered significant attention over the last 10 years, representing a burgeoning therapeutic approach with the potential to address pathogenic proteins that have historically posed challenges for traditional small-molecule inhibitors. PROTACs exploit the endogenous E3 ubiquitin ligases to facilitate degradation of the proteins of interest (POIs) through the ubiquitin–proteasome system (UPS) in a cyclic catalytic manner. Despite recent endeavors to advance the utilization of PROTACs in clinical settings, the majority of PROTACs fail to progress beyond the preclinical phase of drug development. There are multiple factors impeding the market entry of PROTACs, with the insufficiently precise degradation of favorable POIs standing out as one of the most formidable obstacles. Recently, there has been exploration of new-generation advanced PROTACs, including small-molecule PROTAC prodrugs, biomacromolecule-PROTAC conjugates, and nano-PROTACs, to improve the in vivo efficacy of PROTACs. These improved PROTACs possess the capability to mitigate undesirable physicochemical characteristics inherent in traditional PROTACs, thereby enhancing their targetability and reducing off-target side effects. The new-generation of advanced PROTACs will mark a pivotal turning point in the realm of targeted protein degradation. In this comprehensive review, we have meticulously summarized the state-of-the-art advancements achieved by these cutting-edge PROTACs, elucidated their underlying design principles, deliberated upon the prevailing challenges encountered, and provided an insightful outlook on future prospects within this burgeoning field.
蛋白水解靶向嵌合体(PROTACs)技术在过去 10 年中备受关注,它代表了一种新兴的治疗方法,有望解决传统小分子抑制剂历来难以解决的致病蛋白问题。PROTACs 利用内源性 E3 泛素连接酶,以循环催化的方式通过泛素-蛋白酶体系统(UPS)促进相关蛋白(POIs)的降解。尽管近年来人们一直在努力推动 PROTACs 在临床环境中的应用,但大多数 PROTACs 都未能突破药物开发的临床前阶段。阻碍 PROTACs 进入市场的因素有很多,其中最严重的障碍之一就是对有利 POIs 的降解不够精确。最近,人们开始探索新一代先进的 PROTACs,包括小分子 PROTAC 原药、生物大分子-PROTAC 共轭物和纳米 PROTACs,以提高 PROTACs 的体内疗效。这些改进型 PROTACs 能够减轻传统 PROTACs 固有的不良理化特性,从而提高其靶向性并减少脱靶副作用。新一代先进的 PROTACs 将标志着靶向蛋白质降解领域的一个关键转折点。在这篇综述中,我们细致地总结了这些前沿 PROTACs 所取得的最新进展,阐明了它们的基本设计原理,讨论了目前所遇到的挑战,并对这一新兴领域的未来前景进行了深刻的展望。
{"title":"New-generation advanced PROTACs as potential therapeutic agents in cancer therapy","authors":"Chao Wang, Yujing Zhang, Wujun Chen, Yudong Wu, Dongming Xing","doi":"10.1186/s12943-024-02024-9","DOIUrl":"https://doi.org/10.1186/s12943-024-02024-9","url":null,"abstract":"Proteolysis-targeting chimeras (PROTACs) technology has garnered significant attention over the last 10 years, representing a burgeoning therapeutic approach with the potential to address pathogenic proteins that have historically posed challenges for traditional small-molecule inhibitors. PROTACs exploit the endogenous E3 ubiquitin ligases to facilitate degradation of the proteins of interest (POIs) through the ubiquitin–proteasome system (UPS) in a cyclic catalytic manner. Despite recent endeavors to advance the utilization of PROTACs in clinical settings, the majority of PROTACs fail to progress beyond the preclinical phase of drug development. There are multiple factors impeding the market entry of PROTACs, with the insufficiently precise degradation of favorable POIs standing out as one of the most formidable obstacles. Recently, there has been exploration of new-generation advanced PROTACs, including small-molecule PROTAC prodrugs, biomacromolecule-PROTAC conjugates, and nano-PROTACs, to improve the in vivo efficacy of PROTACs. These improved PROTACs possess the capability to mitigate undesirable physicochemical characteristics inherent in traditional PROTACs, thereby enhancing their targetability and reducing off-target side effects. The new-generation of advanced PROTACs will mark a pivotal turning point in the realm of targeted protein degradation. In this comprehensive review, we have meticulously summarized the state-of-the-art advancements achieved by these cutting-edge PROTACs, elucidated their underlying design principles, deliberated upon the prevailing challenges encountered, and provided an insightful outlook on future prospects within this burgeoning field.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141074141","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
KDM5 family as therapeutic targets in breast cancer: Pathogenesis and therapeutic opportunities and challenges 作为乳腺癌治疗靶点的 KDM5 家族:发病机制和治疗机遇与挑战
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-20 DOI: 10.1186/s12943-024-02011-0
Chang-Yun Li, Wanhe Wang, Chung-Hang Leung, Guan-Jun Yang, Jiong Chen
Breast cancer (BC) is the most frequent malignant cancer diagnosis and is a primary factor for cancer deaths in women. The clinical subtypes of BC include estrogen receptor (ER) positive, progesterone receptor (PR) positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative BC (TNBC). Based on the stages and subtypes of BC, various treatment methods are available with variations in the rates of progression-free disease and overall survival of patients. However, the treatment of BC still faces challenges, particularly in terms of drug resistance and recurrence. The study of epigenetics has provided new ideas for treating BC. Targeting aberrant epigenetic factors with inhibitors represents a promising anticancer strategy. The KDM5 family includes four members, KDM5A, KDM5B, KDM5C, and KDMD, all of which are Jumonji C domain-containing histone H3K4me2/3 demethylases. KDM5 proteins have been extensively studied in BC, where they are involved in suppressing or promoting BC depending on their specific upstream and downstream pathways. Several KDM5 inhibitors have shown potent BC inhibitory activity in vitro and in vivo, but challenges still exist in developing KDM5 inhibitors. In this review, we introduce the subtypes of BC and their current therapeutic options, summarize KDM5 family context-specific functions in the pathobiology of BC, and discuss the outlook and pitfalls of KDM5 inhibitors in this disease.
乳腺癌(BC)是最常见的恶性肿瘤,也是导致女性癌症死亡的主要因素。乳腺癌的临床亚型包括雌激素受体(ER)阳性、孕激素受体(PR)阳性、人表皮生长因子受体 2(HER2)阳性和三阴性乳腺癌(TNBC)。根据 BC 的分期和亚型,有多种治疗方法可供选择,患者的无疾病进展率和总生存率也各不相同。然而,BC 的治疗仍然面临挑战,尤其是在耐药性和复发方面。表观遗传学研究为治疗 BC 提供了新思路。用抑制剂靶向异常表观遗传因子是一种很有前景的抗癌策略。KDM5家族包括四个成员:KDM5A、KDM5B、KDM5C和KDMD,它们都是含Jumonji C结构域的组蛋白H3K4me2/3去甲基化酶。对 KDM5 蛋白在 BC 中的作用进行了广泛的研究,根据其特定的上游和下游途径,它们参与了抑制或促进 BC 的作用。一些 KDM5 抑制剂已在体外和体内显示出有效的 BC 抑制活性,但在开发 KDM5 抑制剂方面仍存在挑战。在这篇综述中,我们介绍了 BC 的亚型及其目前的治疗方案,总结了 KDM5 家族在 BC 病理生物学中的特异性功能,并讨论了 KDM5 抑制剂在该疾病中的前景和陷阱。
{"title":"KDM5 family as therapeutic targets in breast cancer: Pathogenesis and therapeutic opportunities and challenges","authors":"Chang-Yun Li, Wanhe Wang, Chung-Hang Leung, Guan-Jun Yang, Jiong Chen","doi":"10.1186/s12943-024-02011-0","DOIUrl":"https://doi.org/10.1186/s12943-024-02011-0","url":null,"abstract":"Breast cancer (BC) is the most frequent malignant cancer diagnosis and is a primary factor for cancer deaths in women. The clinical subtypes of BC include estrogen receptor (ER) positive, progesterone receptor (PR) positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative BC (TNBC). Based on the stages and subtypes of BC, various treatment methods are available with variations in the rates of progression-free disease and overall survival of patients. However, the treatment of BC still faces challenges, particularly in terms of drug resistance and recurrence. The study of epigenetics has provided new ideas for treating BC. Targeting aberrant epigenetic factors with inhibitors represents a promising anticancer strategy. The KDM5 family includes four members, KDM5A, KDM5B, KDM5C, and KDMD, all of which are Jumonji C domain-containing histone H3K4me2/3 demethylases. KDM5 proteins have been extensively studied in BC, where they are involved in suppressing or promoting BC depending on their specific upstream and downstream pathways. Several KDM5 inhibitors have shown potent BC inhibitory activity in vitro and in vivo, but challenges still exist in developing KDM5 inhibitors. In this review, we introduce the subtypes of BC and their current therapeutic options, summarize KDM5 family context-specific functions in the pathobiology of BC, and discuss the outlook and pitfalls of KDM5 inhibitors in this disease.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141069235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Regulatory mechanisms of PD-1/PD-L1 in cancers 癌症中 PD-1/PD-L1 的调控机制
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-18 DOI: 10.1186/s12943-024-02023-w
Xin Lin, Kuan Kang, Pan Chen, Zhaoyang Zeng, Guiyuan Li, Wei Xiong, Mei Yi, Bo Xiang
Immune evasion contributes to cancer growth and progression. Cancer cells have the ability to activate different immune checkpoint pathways that harbor immunosuppressive functions. The programmed death protein 1 (PD-1) and programmed cell death ligands (PD-Ls) are considered to be the major immune checkpoint molecules. The interaction of PD-1 and PD-L1 negatively regulates adaptive immune response mainly by inhibiting the activity of effector T cells while enhancing the function of immunosuppressive regulatory T cells (Tregs), largely contributing to the maintenance of immune homeostasis that prevents dysregulated immunity and harmful immune responses. However, cancer cells exploit the PD-1/PD-L1 axis to cause immune escape in cancer development and progression. Blockade of PD-1/PD-L1 by neutralizing antibodies restores T cells activity and enhances anti-tumor immunity, achieving remarkable success in cancer therapy. Therefore, the regulatory mechanisms of PD-1/PD-L1 in cancers have attracted an increasing attention. This article aims to provide a comprehensive review of the roles of the PD-1/PD-L1 signaling in human autoimmune diseases and cancers. We summarize all aspects of regulatory mechanisms underlying the expression and activity of PD-1 and PD-L1 in cancers, including genetic, epigenetic, post-transcriptional and post-translational regulatory mechanisms. In addition, we further summarize the progress in clinical research on the antitumor effects of targeting PD-1/PD-L1 antibodies alone and in combination with other therapeutic approaches, providing new strategies for finding new tumor markers and developing combined therapeutic approaches.
免疫逃避是癌症生长和恶化的原因之一。癌细胞能够激活不同的免疫检查点通路,这些通路具有免疫抑制功能。程序性死亡蛋白 1(PD-1)和程序性细胞死亡配体(PD-Ls)被认为是主要的免疫检查点分子。PD-1和PD-L1的相互作用主要通过抑制效应T细胞的活性,同时增强免疫抑制调节性T细胞(Tregs)的功能来负向调节适应性免疫反应,在很大程度上有助于维持免疫平衡,防止免疫失调和有害的免疫反应。然而,癌细胞会利用 PD-1/PD-L1 轴在癌症发展和恶化过程中造成免疫逃逸。通过中和抗体阻断 PD-1/PD-L1 可恢复 T 细胞活性,增强抗肿瘤免疫力,在癌症治疗中取得显著成效。因此,PD-1/PD-L1在癌症中的调控机制越来越受到关注。本文旨在全面综述 PD-1/PD-L1 信号在人类自身免疫性疾病和癌症中的作用。我们总结了 PD-1 和 PD-L1 在癌症中的表达和活性的各方面调控机制,包括遗传、表观遗传、转录后和翻译后调控机制。此外,我们还进一步总结了针对 PD-1/PD-L1 抗体单独或与其他治疗方法联合的抗肿瘤效果的临床研究进展,为寻找新的肿瘤标志物和开发联合治疗方法提供了新的策略。
{"title":"Regulatory mechanisms of PD-1/PD-L1 in cancers","authors":"Xin Lin, Kuan Kang, Pan Chen, Zhaoyang Zeng, Guiyuan Li, Wei Xiong, Mei Yi, Bo Xiang","doi":"10.1186/s12943-024-02023-w","DOIUrl":"https://doi.org/10.1186/s12943-024-02023-w","url":null,"abstract":"Immune evasion contributes to cancer growth and progression. Cancer cells have the ability to activate different immune checkpoint pathways that harbor immunosuppressive functions. The programmed death protein 1 (PD-1) and programmed cell death ligands (PD-Ls) are considered to be the major immune checkpoint molecules. The interaction of PD-1 and PD-L1 negatively regulates adaptive immune response mainly by inhibiting the activity of effector T cells while enhancing the function of immunosuppressive regulatory T cells (Tregs), largely contributing to the maintenance of immune homeostasis that prevents dysregulated immunity and harmful immune responses. However, cancer cells exploit the PD-1/PD-L1 axis to cause immune escape in cancer development and progression. Blockade of PD-1/PD-L1 by neutralizing antibodies restores T cells activity and enhances anti-tumor immunity, achieving remarkable success in cancer therapy. Therefore, the regulatory mechanisms of PD-1/PD-L1 in cancers have attracted an increasing attention. This article aims to provide a comprehensive review of the roles of the PD-1/PD-L1 signaling in human autoimmune diseases and cancers. We summarize all aspects of regulatory mechanisms underlying the expression and activity of PD-1 and PD-L1 in cancers, including genetic, epigenetic, post-transcriptional and post-translational regulatory mechanisms. In addition, we further summarize the progress in clinical research on the antitumor effects of targeting PD-1/PD-L1 antibodies alone and in combination with other therapeutic approaches, providing new strategies for finding new tumor markers and developing combined therapeutic approaches.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140953849","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Aging and cancer 衰老与癌症
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-18 DOI: 10.1186/s12943-024-02020-z
Léa Montégut, Carlos López-Otín, Guido Kroemer
Aging and cancer exhibit apparent links that we will examine in this review. The null hypothesis that aging and cancer coincide because both are driven by time, irrespective of the precise causes, can be confronted with the idea that aging and cancer share common mechanistic grounds that are referred to as ‘hallmarks’. Indeed, several hallmarks of aging also contribute to carcinogenesis and tumor progression, but some of the molecular and cellular characteristics of aging may also reduce the probability of developing lethal cancer, perhaps explaining why very old age (> 90 years) is accompanied by a reduced incidence of neoplastic diseases. We will also discuss the possibility that the aging process itself causes cancer, meaning that the time-dependent degradation of cellular and supracellular functions that accompanies aging produces cancer as a byproduct or ‘age-associated disease’. Conversely, cancer and its treatment may erode health and drive the aging process, as this has dramatically been documented for cancer survivors diagnosed during childhood, adolescence, and young adulthood. We conclude that aging and cancer are connected by common superior causes including endogenous and lifestyle factors, as well as by a bidirectional crosstalk, that together render old age not only a risk factor of cancer but also an important parameter that must be considered for therapeutic decisions.
衰老与癌症之间存在着明显的联系,我们将在本综述中对此进行研究。有人认为,衰老和癌症的发生都是由时间驱动的,无论确切的原因是什么,因此衰老和癌症的发生是一致的,但这一假设是无效的。事实上,衰老的一些特征也有助于癌变和肿瘤的发展,但衰老的一些分子和细胞特征也可能降低罹患致命癌症的几率,这或许可以解释为什么高龄(大于 90 岁)伴随着肿瘤性疾病发病率的降低。我们还将讨论衰老过程本身导致癌症的可能性,也就是说,伴随衰老而来的细胞和细胞上功能的时间依赖性退化会产生癌症这种副产品或 "年龄相关疾病"。相反,癌症及其治疗可能会侵蚀健康并推动衰老过程,这一点在童年、青春期和青壮年时期确诊的癌症幸存者身上有大量的记录。我们的结论是,衰老与癌症之间存在着共同的上位原因,包括内源性因素和生活方式因素,同时还存在着双向的相互影响,这使得老年不仅是癌症的风险因素,也是治疗决策必须考虑的一个重要参数。
{"title":"Aging and cancer","authors":"Léa Montégut, Carlos López-Otín, Guido Kroemer","doi":"10.1186/s12943-024-02020-z","DOIUrl":"https://doi.org/10.1186/s12943-024-02020-z","url":null,"abstract":"Aging and cancer exhibit apparent links that we will examine in this review. The null hypothesis that aging and cancer coincide because both are driven by time, irrespective of the precise causes, can be confronted with the idea that aging and cancer share common mechanistic grounds that are referred to as ‘hallmarks’. Indeed, several hallmarks of aging also contribute to carcinogenesis and tumor progression, but some of the molecular and cellular characteristics of aging may also reduce the probability of developing lethal cancer, perhaps explaining why very old age (> 90 years) is accompanied by a reduced incidence of neoplastic diseases. We will also discuss the possibility that the aging process itself causes cancer, meaning that the time-dependent degradation of cellular and supracellular functions that accompanies aging produces cancer as a byproduct or ‘age-associated disease’. Conversely, cancer and its treatment may erode health and drive the aging process, as this has dramatically been documented for cancer survivors diagnosed during childhood, adolescence, and young adulthood. We conclude that aging and cancer are connected by common superior causes including endogenous and lifestyle factors, as well as by a bidirectional crosstalk, that together render old age not only a risk factor of cancer but also an important parameter that must be considered for therapeutic decisions.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140953867","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neutrophils in Cancer immunotherapy: friends or foes? 癌症免疫疗法中的中性粒细胞:朋友还是敌人?
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-18 DOI: 10.1186/s12943-024-02004-z
Xueqin Huang, Eugenie Nepovimova, Vojtech Adam, Ladislav Sivak, Zbynek Heger, Marian Valko, Qinghua Wu, Kamil Kuca
Neutrophils play a Janus-faced role in the complex landscape of cancer pathogenesis and immunotherapy. As immune defense cells, neutrophils release toxic substances, including reactive oxygen species and matrix metalloproteinase 9, within the tumor microenvironment. They also modulate the expression of tumor necrosis factor-related apoptosis-inducing ligand and Fas ligand, augmenting their capacity to induce tumor cell apoptosis. Their involvement in antitumor immune regulation synergistically activates a network of immune cells, bolstering anticancer effects. Paradoxically, neutrophils can succumb to the influence of tumors, triggering signaling cascades such as JAK/STAT, which deactivate the immune system network, thereby promoting immune evasion by malignant cells. Additionally, neutrophil granular constituents, such as neutrophil elastase and vascular endothelial growth factor, intricately fuel tumor cell proliferation, metastasis, and angiogenesis. Understanding the mechanisms that guide neutrophils to collaborate with other immune cells for comprehensive tumor eradication is crucial to enhancing the efficacy of cancer therapeutics. In this review, we illuminate the underlying mechanisms governing neutrophil-mediated support or inhibition of tumor progression, with a particular focus on elucidating the internal and external factors that influence neutrophil polarization. We provide an overview of recent advances in clinical research regarding the involvement of neutrophils in cancer therapy. Moreover, the future prospects and limitations of neutrophil research are discussed, aiming to provide fresh insights for the development of innovative cancer treatment strategies targeting neutrophils.
中性粒细胞在复杂的癌症发病机制和免疫疗法中扮演着 "杰纳斯 "的角色。作为免疫防御细胞,中性粒细胞会在肿瘤微环境中释放有毒物质,包括活性氧和基质金属蛋白酶 9。它们还能调节肿瘤坏死因子相关凋亡诱导配体和 Fas 配体的表达,增强诱导肿瘤细胞凋亡的能力。中性粒细胞参与抗肿瘤免疫调节可协同激活免疫细胞网络,增强抗癌效果。矛盾的是,嗜中性粒细胞可能会屈服于肿瘤的影响,触发信号级联(如 JAK/STAT),使免疫系统网络失活,从而促进恶性细胞逃避免疫。此外,嗜中性粒细胞颗粒成分,如嗜中性粒细胞弹性蛋白酶和血管内皮生长因子,也会错综复杂地助长肿瘤细胞的增殖、转移和血管生成。了解引导中性粒细胞与其他免疫细胞合作全面消灭肿瘤的机制对于提高癌症疗法的疗效至关重要。在这篇综述中,我们将阐明中性粒细胞介导的支持或抑制肿瘤进展的潜在机制,尤其侧重于阐明影响中性粒细胞极化的内部和外部因素。我们概述了有关中性粒细胞参与癌症治疗的临床研究的最新进展。此外,我们还讨论了中性粒细胞研究的未来前景和局限性,旨在为开发针对中性粒细胞的创新癌症治疗策略提供新的见解。
{"title":"Neutrophils in Cancer immunotherapy: friends or foes?","authors":"Xueqin Huang, Eugenie Nepovimova, Vojtech Adam, Ladislav Sivak, Zbynek Heger, Marian Valko, Qinghua Wu, Kamil Kuca","doi":"10.1186/s12943-024-02004-z","DOIUrl":"https://doi.org/10.1186/s12943-024-02004-z","url":null,"abstract":"Neutrophils play a Janus-faced role in the complex landscape of cancer pathogenesis and immunotherapy. As immune defense cells, neutrophils release toxic substances, including reactive oxygen species and matrix metalloproteinase 9, within the tumor microenvironment. They also modulate the expression of tumor necrosis factor-related apoptosis-inducing ligand and Fas ligand, augmenting their capacity to induce tumor cell apoptosis. Their involvement in antitumor immune regulation synergistically activates a network of immune cells, bolstering anticancer effects. Paradoxically, neutrophils can succumb to the influence of tumors, triggering signaling cascades such as JAK/STAT, which deactivate the immune system network, thereby promoting immune evasion by malignant cells. Additionally, neutrophil granular constituents, such as neutrophil elastase and vascular endothelial growth factor, intricately fuel tumor cell proliferation, metastasis, and angiogenesis. Understanding the mechanisms that guide neutrophils to collaborate with other immune cells for comprehensive tumor eradication is crucial to enhancing the efficacy of cancer therapeutics. In this review, we illuminate the underlying mechanisms governing neutrophil-mediated support or inhibition of tumor progression, with a particular focus on elucidating the internal and external factors that influence neutrophil polarization. We provide an overview of recent advances in clinical research regarding the involvement of neutrophils in cancer therapy. Moreover, the future prospects and limitations of neutrophil research are discussed, aiming to provide fresh insights for the development of innovative cancer treatment strategies targeting neutrophils.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140953993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CircTRIM1 encodes TRIM1-269aa to promote chemoresistance and metastasis of TNBC via enhancing CaM-dependent MARCKS translocation and PI3K/AKT/mTOR activation CircTRIM1 编码 TRIM1-269aa,通过增强 CaM 依赖性 MARCKS 转位和 PI3K/AKT/mTOR 激活,促进 TNBC 的化疗抗性和转移
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-16 DOI: 10.1186/s12943-024-02019-6
Yaming Li, Zekun Wang, Jingwen Yang, Yuhan Sun, Yinqiao He, Yuping Wang, Xi Chen, Yiran Liang, Ning Zhang, Xiaolong Wang, Wenjing Zhao, Guohong Hu, Qifeng Yang
Peptides and proteins encoded by noncanonical open reading frames (ORFs) of circRNAs have recently been recognized to play important roles in disease progression, but the biological functions and mechanisms of these peptides and proteins are largely unknown. Here, we identified a potential coding circular RNA, circTRIM1, that was upregulated in doxorubicin-resistant TNBC cells by intersecting transcriptome and translatome RNA-seq data, and its expression was correlated with clinicopathological characteristics and poor prognosis in patients with TNBC. CircTRIM1 possesses a functional IRES element along with an 810 nt ORF that can be translated into a novel endogenously expressed protein termed TRIM1-269aa. Functionally, we demonstrated that TRIM1-269aa, which is involved in the biological functions of circTRIM1, promoted chemoresistance and metastasis in TNBC cells both in vitro and in vivo. In addition, we found that TRIM1-269aa can be packaged into exosomes and transmitted between TNBC cells. Mechanistically, TRIM1-269aa enhanced the interaction between MARCKS and calmodulin, thus promoting the calmodulin-dependent translocation of MARCKS, which further initiated the activation of the PI3K/AKT/mTOR pathway. Overall, circTRIM1, which encodes TRIM1-269aa, promoted TNBC chemoresistance and metastasis by enhancing MARCKS translocation and PI3K/AKT/mTOR activation. Our investigation has yielded novel insights into the roles of protein-coding circRNAs and supported circTRIM1/TRIM1-269aa as a novel promising prognostic and therapeutic target for patients with TNBC.
最近,人们认识到环状RNA的非规范开放阅读框(ORF)编码的肽和蛋白质在疾病进展中发挥着重要作用,但这些肽和蛋白质的生物学功能和机制在很大程度上是未知的。在这里,我们通过交叉转录组和转译组RNA-seq数据,发现了一种潜在的编码环状RNA--circTRIM1,它在多柔比星耐药的TNBC细胞中上调,其表达与TNBC患者的临床病理特征和不良预后相关。CircTRIM1具有一个功能性IRES元件和一个810 nt的ORF,可被翻译成一种称为TRIM1-269aa的新型内源性表达蛋白。在功能上,我们证明了TRIM1-269aa参与了circTRIM1的生物学功能,在体外和体内促进了TNBC细胞的化疗耐药性和转移。此外,我们还发现TRIM1-269aa可被包装成外泌体,并在TNBC细胞之间传播。从机制上讲,TRIM1-269aa增强了MARCKS与钙调素之间的相互作用,从而促进了钙调素依赖性的MARCKS转位,进一步启动了PI3K/AKT/mTOR通路的激活。总之,编码TRIM1-269aa的circTRIM1通过增强MARCKS的转位和PI3K/AKT/mTOR的激活,促进了TNBC的化疗耐药性和转移。我们的研究对编码蛋白的circRNA的作用有了新的认识,并支持circTRIM1/TRIM1-269aa成为TNBC患者的一个新的有希望的预后和治疗靶点。
{"title":"CircTRIM1 encodes TRIM1-269aa to promote chemoresistance and metastasis of TNBC via enhancing CaM-dependent MARCKS translocation and PI3K/AKT/mTOR activation","authors":"Yaming Li, Zekun Wang, Jingwen Yang, Yuhan Sun, Yinqiao He, Yuping Wang, Xi Chen, Yiran Liang, Ning Zhang, Xiaolong Wang, Wenjing Zhao, Guohong Hu, Qifeng Yang","doi":"10.1186/s12943-024-02019-6","DOIUrl":"https://doi.org/10.1186/s12943-024-02019-6","url":null,"abstract":"Peptides and proteins encoded by noncanonical open reading frames (ORFs) of circRNAs have recently been recognized to play important roles in disease progression, but the biological functions and mechanisms of these peptides and proteins are largely unknown. Here, we identified a potential coding circular RNA, circTRIM1, that was upregulated in doxorubicin-resistant TNBC cells by intersecting transcriptome and translatome RNA-seq data, and its expression was correlated with clinicopathological characteristics and poor prognosis in patients with TNBC. CircTRIM1 possesses a functional IRES element along with an 810 nt ORF that can be translated into a novel endogenously expressed protein termed TRIM1-269aa. Functionally, we demonstrated that TRIM1-269aa, which is involved in the biological functions of circTRIM1, promoted chemoresistance and metastasis in TNBC cells both in vitro and in vivo. In addition, we found that TRIM1-269aa can be packaged into exosomes and transmitted between TNBC cells. Mechanistically, TRIM1-269aa enhanced the interaction between MARCKS and calmodulin, thus promoting the calmodulin-dependent translocation of MARCKS, which further initiated the activation of the PI3K/AKT/mTOR pathway. Overall, circTRIM1, which encodes TRIM1-269aa, promoted TNBC chemoresistance and metastasis by enhancing MARCKS translocation and PI3K/AKT/mTOR activation. Our investigation has yielded novel insights into the roles of protein-coding circRNAs and supported circTRIM1/TRIM1-269aa as a novel promising prognostic and therapeutic target for patients with TNBC.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140949411","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NEMO/NF-κB signaling functions as a double-edged sword in PanIN formation versus progression to pancreatic cancer NEMO/NF-κB 信号在 PanIN 的形成和胰腺癌的发展过程中起着双刃剑的作用
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-16 DOI: 10.1186/s12943-024-01989-x
Miltiadis Tsesmelis, Ulrike F. G. Büttner, Melanie Gerstenlauer, Uta Manfras, Konstantinos Tsesmelis, Ziwei Du, Nadine Sperb, Stephanie Ellen Weissinger, Peter Möller, Thomas F. E. Barth, Harald J. Maier, Lap Kwan Chan, Thomas Wirth
Pancreatic ductal adenocarcinoma (PDAC) is marked by a dismal survival rate, lacking effective therapeutics due to its aggressive growth, late-stage diagnosis, and chemotherapy resistance. Despite debates on NF-κB targeting for PDAC treatment, no successful approach has emerged. To elucidate the role of NF-κB, we ablated NF-κB essential modulator (NEMO), critical for conventional NF-κB signaling, in the pancreata of mice that develop precancerous lesions (KC mouse model). Secretagogue-induced pancreatitis by cerulein injections was utilized to promote inflammation and accelerate PDAC development. NEMO deletion reduced fibrosis and inflammation in young KC mice, resulting in fewer pancreatic intraepithelial neoplasias (PanINs) at later stages. Paradoxically, however, NEMO deletion accelerated the progression of these fewer PanINs to PDAC and reduced median lifespan. Further, analysis of tissue microarrays from human PDAC sections highlighted the correlation between reduced NEMO expression in neoplastic cells and poorer prognosis, supporting our observation in mice. Mechanistically, NEMO deletion impeded oncogene-induced senescence (OIS), which is normally active in low-grade PanINs. This blockage resulted in fewer senescence-associated secretory phenotype (SASP) factors, reducing inflammation. However, blocked OIS fostered replication stress and DNA damage accumulation which accelerated PanIN progression to PDAC. Finally, treatment with the DNA damage-inducing reagent etoposide resulted in elevated cell death in NEMO-ablated PDAC cells compared to their NEMO-competent counterparts, indicative of a synthetic lethality paradigm. NEMO exhibited both oncogenic and tumor-suppressive properties during PDAC development. Caution is suggested in therapeutic interventions targeting NF-κB, which may be detrimental during PanIN progression but beneficial post-PDAC development.
胰腺导管腺癌(PDAC)的生存率很低,由于其生长凶猛、诊断较晚和化疗耐药,缺乏有效的治疗方法。尽管有关 NF-κB 靶向治疗 PDAC 的争论不绝于耳,但至今仍未出现成功的方法。为了阐明 NF-κB 的作用,我们在发生癌前病变的小鼠(KC 小鼠模型)胰腺中消减了对传统 NF-κB 信号转导至关重要的 NF-κB 重要调节因子(NEMO)。通过注射胰泌素诱发的胰腺炎可促进炎症并加速 PDAC 的发展。NEMO缺失可减少年轻KC小鼠的纤维化和炎症,从而减少后期胰腺上皮内瘤(PanIN)的发生。然而矛盾的是,NEMO缺失加速了这些较少的PanINs向PDAC的发展,并缩短了中位寿命。此外,对人类 PDAC 切片组织芯片的分析强调了肿瘤细胞中 NEMO 表达减少与预后较差之间的相关性,这也支持了我们在小鼠中的观察结果。从机理上讲,NEMO 的缺失阻碍了癌基因诱导的衰老(OIS),而这种衰老在低级别 PanINs 中通常是活跃的。这种阻断导致衰老相关分泌表型(SASP)因子减少,从而减轻了炎症。然而,OIS的阻断促进了复制应激和DNA损伤的积累,从而加速了PanIN向PDAC的发展。最后,DNA损伤诱导试剂依托泊苷的处理导致NEMO无效的PDAC细胞的细胞死亡率高于NEMO有效的细胞,这表明了一种合成致死模式。NEMO在PDAC发展过程中同时表现出致癌和抑制肿瘤的特性。针对NF-κB的治疗干预可能在PanIN发展过程中有害,但在PDAC发展后却有益,因此建议谨慎从事。
{"title":"NEMO/NF-κB signaling functions as a double-edged sword in PanIN formation versus progression to pancreatic cancer","authors":"Miltiadis Tsesmelis, Ulrike F. G. Büttner, Melanie Gerstenlauer, Uta Manfras, Konstantinos Tsesmelis, Ziwei Du, Nadine Sperb, Stephanie Ellen Weissinger, Peter Möller, Thomas F. E. Barth, Harald J. Maier, Lap Kwan Chan, Thomas Wirth","doi":"10.1186/s12943-024-01989-x","DOIUrl":"https://doi.org/10.1186/s12943-024-01989-x","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) is marked by a dismal survival rate, lacking effective therapeutics due to its aggressive growth, late-stage diagnosis, and chemotherapy resistance. Despite debates on NF-κB targeting for PDAC treatment, no successful approach has emerged. To elucidate the role of NF-κB, we ablated NF-κB essential modulator (NEMO), critical for conventional NF-κB signaling, in the pancreata of mice that develop precancerous lesions (KC mouse model). Secretagogue-induced pancreatitis by cerulein injections was utilized to promote inflammation and accelerate PDAC development. NEMO deletion reduced fibrosis and inflammation in young KC mice, resulting in fewer pancreatic intraepithelial neoplasias (PanINs) at later stages. Paradoxically, however, NEMO deletion accelerated the progression of these fewer PanINs to PDAC and reduced median lifespan. Further, analysis of tissue microarrays from human PDAC sections highlighted the correlation between reduced NEMO expression in neoplastic cells and poorer prognosis, supporting our observation in mice. Mechanistically, NEMO deletion impeded oncogene-induced senescence (OIS), which is normally active in low-grade PanINs. This blockage resulted in fewer senescence-associated secretory phenotype (SASP) factors, reducing inflammation. However, blocked OIS fostered replication stress and DNA damage accumulation which accelerated PanIN progression to PDAC. Finally, treatment with the DNA damage-inducing reagent etoposide resulted in elevated cell death in NEMO-ablated PDAC cells compared to their NEMO-competent counterparts, indicative of a synthetic lethality paradigm. NEMO exhibited both oncogenic and tumor-suppressive properties during PDAC development. Caution is suggested in therapeutic interventions targeting NF-κB, which may be detrimental during PanIN progression but beneficial post-PDAC development. ","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140949351","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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