Caspases 在肝癌发生过程中影响 SLU7 和 UPF1 的稳定性和 NMD 活性

IF 9.5 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY JHEP Reports Pub Date : 2024-05-09 DOI:10.1016/j.jhepr.2024.101118
Carla Rojo , María Gárate-Rascón , Miriam Recalde , Ane Álava , María Elizalde , María Azkona , Iratxe Aldabe , Elisabet Guruceaga , Amaya López-Pascual , M Ujue Latasa , Bruno Sangro , Maite G. Fernández-Barrena , Matías A. Ávila , María Arechederra , Carmen Berasain
{"title":"Caspases 在肝癌发生过程中影响 SLU7 和 UPF1 的稳定性和 NMD 活性","authors":"Carla Rojo ,&nbsp;María Gárate-Rascón ,&nbsp;Miriam Recalde ,&nbsp;Ane Álava ,&nbsp;María Elizalde ,&nbsp;María Azkona ,&nbsp;Iratxe Aldabe ,&nbsp;Elisabet Guruceaga ,&nbsp;Amaya López-Pascual ,&nbsp;M Ujue Latasa ,&nbsp;Bruno Sangro ,&nbsp;Maite G. Fernández-Barrena ,&nbsp;Matías A. Ávila ,&nbsp;María Arechederra ,&nbsp;Carmen Berasain","doi":"10.1016/j.jhepr.2024.101118","DOIUrl":null,"url":null,"abstract":"<div><h3>Background &amp; Aims</h3><p>The homeostasis of the cellular transcriptome depends on transcription and splicing mechanisms. Moreover, the fidelity of gene expression, essential to preserve cellular identity and function is secured by different quality control mechanisms including nonsense-mediated RNA decay (NMD). In this context, alternative splicing is coupled to NMD, and several alterations in these mechanisms leading to the accumulation of aberrant gene isoforms are known to be involved in human disease including cancer.</p></div><div><h3>Methods</h3><p>RNA sequencing, western blotting, qPCR and co-immunoprecipitation were performed in multiple silenced culture cell lines (replicates n ≥4), primary hepatocytes and samples of animal models (Jo2, APAP, <em>Mdr2</em><sup>-/-</sup> mice, n ≥3).</p></div><div><h3>Results</h3><p>Here we show that in animal models of liver injury and in human HCC (TCGA, non-tumoral = 50 <em>vs</em>. HCC = 374), the process of NMD is inhibited. Moreover, we demonstrate that the splicing factor SLU7 interacts with and preserves the levels of the NMD effector UPF1, and that SLU7 is required for correct NMD. Our previous findings demonstrated that SLU7 expression is reduced in the diseased liver, contributing to hepatocellular dedifferentiation and genome instability during disease progression. Here we build on this by providing evidence that caspases activated during liver damage are responsible for the cleavage and degradation of SLU7.</p></div><div><h3>Conclusions</h3><p>Here we identify the downregulation of UPF1 and the inhibition of NMD as a new molecular pathway contributing to the malignant reshaping of the liver transcriptome. Moreover, and importantly, we uncover caspase activation as the mechanism responsible for the downregulation of SLU7 expression during liver disease progression, which is a new link between apoptosis and hepatocarcinogenesis.</p></div><div><h3>Impact and implications:</h3><p>The mechanisms involved in reshaping the hepatocellular transcriptome and thereby driving the progressive loss of cell identity and function in liver disease are not completely understood. In this context, we provide evidence on the impairment of a key mRNA surveillance mechanism known as nonsense-mediated mRNA decay (NMD). Mechanistically, we uncover a novel role for the splicing factor SLU7 in the regulation of NMD, including its ability to interact and preserve the levels of the key NMD factor UPF1. Moreover, we demonstrate that the activation of caspases during liver damage mediates SLU7 and UPF1 protein degradation and NMD inhibition. Our findings identify potential new markers of liver disease progression, and SLU7 as a novel therapeutic target to prevent the functional decay of the chronically injured organ.</p></div>","PeriodicalId":14764,"journal":{"name":"JHEP Reports","volume":null,"pages":null},"PeriodicalIF":9.5000,"publicationDate":"2024-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2589555924001228/pdfft?md5=aa09618a4a3e9536787d7fb6616e445a&pid=1-s2.0-S2589555924001228-main.pdf","citationCount":"0","resultStr":"{\"title\":\"Caspases compromise SLU7 and UPF1 stability and NMD activity during hepatocarcinogenesis\",\"authors\":\"Carla Rojo ,&nbsp;María Gárate-Rascón ,&nbsp;Miriam Recalde ,&nbsp;Ane Álava ,&nbsp;María Elizalde ,&nbsp;María Azkona ,&nbsp;Iratxe Aldabe ,&nbsp;Elisabet Guruceaga ,&nbsp;Amaya López-Pascual ,&nbsp;M Ujue Latasa ,&nbsp;Bruno Sangro ,&nbsp;Maite G. Fernández-Barrena ,&nbsp;Matías A. Ávila ,&nbsp;María Arechederra ,&nbsp;Carmen Berasain\",\"doi\":\"10.1016/j.jhepr.2024.101118\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Background &amp; Aims</h3><p>The homeostasis of the cellular transcriptome depends on transcription and splicing mechanisms. Moreover, the fidelity of gene expression, essential to preserve cellular identity and function is secured by different quality control mechanisms including nonsense-mediated RNA decay (NMD). In this context, alternative splicing is coupled to NMD, and several alterations in these mechanisms leading to the accumulation of aberrant gene isoforms are known to be involved in human disease including cancer.</p></div><div><h3>Methods</h3><p>RNA sequencing, western blotting, qPCR and co-immunoprecipitation were performed in multiple silenced culture cell lines (replicates n ≥4), primary hepatocytes and samples of animal models (Jo2, APAP, <em>Mdr2</em><sup>-/-</sup> mice, n ≥3).</p></div><div><h3>Results</h3><p>Here we show that in animal models of liver injury and in human HCC (TCGA, non-tumoral = 50 <em>vs</em>. HCC = 374), the process of NMD is inhibited. Moreover, we demonstrate that the splicing factor SLU7 interacts with and preserves the levels of the NMD effector UPF1, and that SLU7 is required for correct NMD. Our previous findings demonstrated that SLU7 expression is reduced in the diseased liver, contributing to hepatocellular dedifferentiation and genome instability during disease progression. Here we build on this by providing evidence that caspases activated during liver damage are responsible for the cleavage and degradation of SLU7.</p></div><div><h3>Conclusions</h3><p>Here we identify the downregulation of UPF1 and the inhibition of NMD as a new molecular pathway contributing to the malignant reshaping of the liver transcriptome. Moreover, and importantly, we uncover caspase activation as the mechanism responsible for the downregulation of SLU7 expression during liver disease progression, which is a new link between apoptosis and hepatocarcinogenesis.</p></div><div><h3>Impact and implications:</h3><p>The mechanisms involved in reshaping the hepatocellular transcriptome and thereby driving the progressive loss of cell identity and function in liver disease are not completely understood. In this context, we provide evidence on the impairment of a key mRNA surveillance mechanism known as nonsense-mediated mRNA decay (NMD). Mechanistically, we uncover a novel role for the splicing factor SLU7 in the regulation of NMD, including its ability to interact and preserve the levels of the key NMD factor UPF1. Moreover, we demonstrate that the activation of caspases during liver damage mediates SLU7 and UPF1 protein degradation and NMD inhibition. Our findings identify potential new markers of liver disease progression, and SLU7 as a novel therapeutic target to prevent the functional decay of the chronically injured organ.</p></div>\",\"PeriodicalId\":14764,\"journal\":{\"name\":\"JHEP Reports\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":9.5000,\"publicationDate\":\"2024-05-09\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.sciencedirect.com/science/article/pii/S2589555924001228/pdfft?md5=aa09618a4a3e9536787d7fb6616e445a&pid=1-s2.0-S2589555924001228-main.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"JHEP Reports\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S2589555924001228\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"GASTROENTEROLOGY & HEPATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"JHEP Reports","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2589555924001228","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GASTROENTEROLOGY & HEPATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景& 目的细胞转录组的平衡取决于转录和剪接机制。此外,不同的质量控制机制(包括无义介导的 RNA 衰变(NMD))确保了基因表达的保真度,这对保持细胞特性和功能至关重要。在这种情况下,替代剪接与 NMD 相结合,这些机制的一些改变导致了异常基因异构体的积累,已知与包括癌症在内的人类疾病有关。结果我们发现,在肝损伤动物模型和人类 HCC(TCGA,非肿瘤 = 50 vs. HCC = 374)中,NMD 过程受到抑制。此外,我们还证明了剪接因子 SLU7 与 NMD 效应物 UPF1 相互作用并保持其水平,而且 SLU7 是正确 NMD 的必要条件。我们之前的研究结果表明,SLU7 在病变肝脏中的表达量减少,在疾病进展过程中导致肝细胞去分化和基因组不稳定。在此基础上,我们提供了证据,证明在肝脏损伤过程中激活的 Caspases 对 SLU7 的裂解和降解负责。结论在此,我们发现 UPF1 的下调和 NMD 的抑制是导致肝脏转录组恶性重塑的新分子途径。此外,重要的是,我们发现在肝病进展过程中,Caspase 激活是导致 SLU7 表达下调的机制,这是细胞凋亡与肝癌发生之间的新联系。在此背景下,我们提供了有关无义介导的mRNA衰变(NMD)这一关键mRNA监控机制受损的证据。从机理上讲,我们发现了剪接因子 SLU7 在 NMD 调控中的新作用,包括其与关键 NMD 因子 UPF1 相互作用并保持其水平的能力。此外,我们还证明了肝损伤过程中 Caspases 的激活介导了 SLU7 和 UPF1 蛋白的降解以及 NMD 的抑制。我们的研究结果确定了肝病进展的潜在新标志物,并将 SLU7 作为一种新的治疗靶点,以防止慢性损伤器官的功能衰退。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Caspases compromise SLU7 and UPF1 stability and NMD activity during hepatocarcinogenesis

Background & Aims

The homeostasis of the cellular transcriptome depends on transcription and splicing mechanisms. Moreover, the fidelity of gene expression, essential to preserve cellular identity and function is secured by different quality control mechanisms including nonsense-mediated RNA decay (NMD). In this context, alternative splicing is coupled to NMD, and several alterations in these mechanisms leading to the accumulation of aberrant gene isoforms are known to be involved in human disease including cancer.

Methods

RNA sequencing, western blotting, qPCR and co-immunoprecipitation were performed in multiple silenced culture cell lines (replicates n ≥4), primary hepatocytes and samples of animal models (Jo2, APAP, Mdr2-/- mice, n ≥3).

Results

Here we show that in animal models of liver injury and in human HCC (TCGA, non-tumoral = 50 vs. HCC = 374), the process of NMD is inhibited. Moreover, we demonstrate that the splicing factor SLU7 interacts with and preserves the levels of the NMD effector UPF1, and that SLU7 is required for correct NMD. Our previous findings demonstrated that SLU7 expression is reduced in the diseased liver, contributing to hepatocellular dedifferentiation and genome instability during disease progression. Here we build on this by providing evidence that caspases activated during liver damage are responsible for the cleavage and degradation of SLU7.

Conclusions

Here we identify the downregulation of UPF1 and the inhibition of NMD as a new molecular pathway contributing to the malignant reshaping of the liver transcriptome. Moreover, and importantly, we uncover caspase activation as the mechanism responsible for the downregulation of SLU7 expression during liver disease progression, which is a new link between apoptosis and hepatocarcinogenesis.

Impact and implications:

The mechanisms involved in reshaping the hepatocellular transcriptome and thereby driving the progressive loss of cell identity and function in liver disease are not completely understood. In this context, we provide evidence on the impairment of a key mRNA surveillance mechanism known as nonsense-mediated mRNA decay (NMD). Mechanistically, we uncover a novel role for the splicing factor SLU7 in the regulation of NMD, including its ability to interact and preserve the levels of the key NMD factor UPF1. Moreover, we demonstrate that the activation of caspases during liver damage mediates SLU7 and UPF1 protein degradation and NMD inhibition. Our findings identify potential new markers of liver disease progression, and SLU7 as a novel therapeutic target to prevent the functional decay of the chronically injured organ.

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
JHEP Reports
JHEP Reports GASTROENTEROLOGY & HEPATOLOGY-
CiteScore
12.40
自引率
2.40%
发文量
161
审稿时长
36 days
期刊介绍: JHEP Reports is an open access journal that is affiliated with the European Association for the Study of the Liver (EASL). It serves as a companion journal to the highly respected Journal of Hepatology. The primary objective of JHEP Reports is to publish original papers and reviews that contribute to the advancement of knowledge in the field of liver diseases. The journal covers a wide range of topics, including basic, translational, and clinical research. It also focuses on global issues in hepatology, with particular emphasis on areas such as clinical trials, novel diagnostics, precision medicine and therapeutics, cancer research, cellular and molecular studies, artificial intelligence, microbiome research, epidemiology, and cutting-edge technologies. In summary, JHEP Reports is dedicated to promoting scientific discoveries and innovations in liver diseases through the publication of high-quality research papers and reviews covering various aspects of hepatology.
期刊最新文献
Contents Editorial Board page Copyright and information Contents Metabolomics biomarkers of hepatocellular carcinoma in a prospective cohort of patients with cirrhosis
×
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