人类结直肠癌细胞获得性肿瘤坏死因子相关凋亡诱导配体(TRAIL)抗性与组蛋白乙酰化有关,与 HDAC 抑制剂联用可协同改善其抗性

IF 2.5 4区 医学 Q2 GASTROENTEROLOGY & HEPATOLOGY Digestive Diseases and Sciences Pub Date : 2024-08-01 DOI:10.1007/s10620-024-08569-5
Se Lim Kim, MinWoo Shin, Byung Chul Jin, SeungYoung Seo, Gi Won Ha, Sang Wook Kim
{"title":"人类结直肠癌细胞获得性肿瘤坏死因子相关凋亡诱导配体(TRAIL)抗性与组蛋白乙酰化有关,与 HDAC 抑制剂联用可协同改善其抗性","authors":"Se Lim Kim, MinWoo Shin, Byung Chul Jin, SeungYoung Seo, Gi Won Ha, Sang Wook Kim","doi":"10.1007/s10620-024-08569-5","DOIUrl":null,"url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background</h3><p>Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive target for the treatment of various malignancies; however, its therapeutic potential is limited because of the frequent occurrence of tumor cell resistance. In this study, we determined whether TRAIL resistance acquired by repeated administration could be overcome by HDAC inhibition in human colorectal cancer cells.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>TRAIL-resistant HCT116 human colorectal cancer cells (HCT116-TR) were generated by repeated treatment with 10 and 25 ng/mL TRAIL twice weekly for 28 days.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>The resulting TRAIL-resistant cells were noncross-resistant to other chemotherapeutic agents. The levels of histone acetylation-related proteins, such as ac-histone H4 and HDAC1, were altered in HCT116-TR cells compared with the parental HCT116 cell line. The combined treatment with TRAIL and HDAC inhibitors significantly increased apoptosis in HCT116-TR cells and indicated a synergistic effect. The mechanism by which HDAC inhibition sensitizes HCT116-TR cells to TRAIL is dependent on the intrinsic pathway. In addition, we found that HDAC inhibition enhanced the sensitivity of cells to TRAIL through mitogen-activated protein kinases/CCAAT/enhancer-binding protein homologs of protein-dependent upregulation of death receptor 5.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>These results suggest that histone acetylation is responsible for acquired TRAIL resistance after repeated exposure and acquired resistance to TRAIL may be overcome by combination therapies with HDAC inhibitors.</p>","PeriodicalId":11378,"journal":{"name":"Digestive Diseases and Sciences","volume":null,"pages":null},"PeriodicalIF":2.5000,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Acquired Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Resistance of Human Colorectal Cancer Cells Is Linked to Histone Acetylation and Is Synergistically Ameliorated by Combination with HDAC Inhibitors\",\"authors\":\"Se Lim Kim, MinWoo Shin, Byung Chul Jin, SeungYoung Seo, Gi Won Ha, Sang Wook Kim\",\"doi\":\"10.1007/s10620-024-08569-5\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<h3 data-test=\\\"abstract-sub-heading\\\">Background</h3><p>Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive target for the treatment of various malignancies; however, its therapeutic potential is limited because of the frequent occurrence of tumor cell resistance. In this study, we determined whether TRAIL resistance acquired by repeated administration could be overcome by HDAC inhibition in human colorectal cancer cells.</p><h3 data-test=\\\"abstract-sub-heading\\\">Methods</h3><p>TRAIL-resistant HCT116 human colorectal cancer cells (HCT116-TR) were generated by repeated treatment with 10 and 25 ng/mL TRAIL twice weekly for 28 days.</p><h3 data-test=\\\"abstract-sub-heading\\\">Results</h3><p>The resulting TRAIL-resistant cells were noncross-resistant to other chemotherapeutic agents. The levels of histone acetylation-related proteins, such as ac-histone H4 and HDAC1, were altered in HCT116-TR cells compared with the parental HCT116 cell line. The combined treatment with TRAIL and HDAC inhibitors significantly increased apoptosis in HCT116-TR cells and indicated a synergistic effect. The mechanism by which HDAC inhibition sensitizes HCT116-TR cells to TRAIL is dependent on the intrinsic pathway. In addition, we found that HDAC inhibition enhanced the sensitivity of cells to TRAIL through mitogen-activated protein kinases/CCAAT/enhancer-binding protein homologs of protein-dependent upregulation of death receptor 5.</p><h3 data-test=\\\"abstract-sub-heading\\\">Conclusion</h3><p>These results suggest that histone acetylation is responsible for acquired TRAIL resistance after repeated exposure and acquired resistance to TRAIL may be overcome by combination therapies with HDAC inhibitors.</p>\",\"PeriodicalId\":11378,\"journal\":{\"name\":\"Digestive Diseases and Sciences\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.5000,\"publicationDate\":\"2024-08-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Digestive Diseases and Sciences\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1007/s10620-024-08569-5\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"GASTROENTEROLOGY & HEPATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Digestive Diseases and Sciences","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s10620-024-08569-5","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"GASTROENTEROLOGY & HEPATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景肿瘤坏死因子相关凋亡诱导配体(TRAIL)是治疗各种恶性肿瘤的一个极具吸引力的靶点;然而,由于经常出现肿瘤细胞耐药性,其治疗潜力受到了限制。在这项研究中,我们确定了在人类结直肠癌细胞中通过重复给药获得的 TRAIL 抗性是否可以通过 HDAC 抑制来克服。方法通过每周两次连续 28 天使用 10 和 25 ng/mL TRAIL 重复处理,产生 TRAIL 抗性 HCT116 人类结直肠癌细胞(HCT116-TR)。与亲代HCT116细胞系相比,HCT116-TR细胞中组蛋白乙酰化相关蛋白(如ac-组蛋白H4和HDAC1)的水平发生了改变。TRAIL和HDAC抑制剂联合治疗可显著增加HCT116-TR细胞的凋亡,并显示出协同效应。HDAC抑制剂使HCT116-TR细胞对TRAIL敏感的机制依赖于内在途径。此外,我们还发现 HDAC 抑制通过丝裂原活化蛋白激酶/CCAAT/增强子结合蛋白同源物依赖性上调死亡受体 5 增强细胞对 TRAIL 的敏感性。
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
Acquired Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Resistance of Human Colorectal Cancer Cells Is Linked to Histone Acetylation and Is Synergistically Ameliorated by Combination with HDAC Inhibitors

Background

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive target for the treatment of various malignancies; however, its therapeutic potential is limited because of the frequent occurrence of tumor cell resistance. In this study, we determined whether TRAIL resistance acquired by repeated administration could be overcome by HDAC inhibition in human colorectal cancer cells.

Methods

TRAIL-resistant HCT116 human colorectal cancer cells (HCT116-TR) were generated by repeated treatment with 10 and 25 ng/mL TRAIL twice weekly for 28 days.

Results

The resulting TRAIL-resistant cells were noncross-resistant to other chemotherapeutic agents. The levels of histone acetylation-related proteins, such as ac-histone H4 and HDAC1, were altered in HCT116-TR cells compared with the parental HCT116 cell line. The combined treatment with TRAIL and HDAC inhibitors significantly increased apoptosis in HCT116-TR cells and indicated a synergistic effect. The mechanism by which HDAC inhibition sensitizes HCT116-TR cells to TRAIL is dependent on the intrinsic pathway. In addition, we found that HDAC inhibition enhanced the sensitivity of cells to TRAIL through mitogen-activated protein kinases/CCAAT/enhancer-binding protein homologs of protein-dependent upregulation of death receptor 5.

Conclusion

These results suggest that histone acetylation is responsible for acquired TRAIL resistance after repeated exposure and acquired resistance to TRAIL may be overcome by combination therapies with HDAC inhibitors.

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
Digestive Diseases and Sciences
Digestive Diseases and Sciences 医学-胃肠肝病学
CiteScore
6.40
自引率
3.20%
发文量
420
审稿时长
1 months
期刊介绍: Digestive Diseases and Sciences publishes high-quality, peer-reviewed, original papers addressing aspects of basic/translational and clinical research in gastroenterology, hepatology, and related fields. This well-illustrated journal features comprehensive coverage of basic pathophysiology, new technological advances, and clinical breakthroughs; insights from prominent academicians and practitioners concerning new scientific developments and practical medical issues; and discussions focusing on the latest changes in local and worldwide social, economic, and governmental policies that affect the delivery of care within the disciplines of gastroenterology and hepatology.
期刊最新文献
Endoscopic Management of Lower Gastrointestinal Tract Anastomosis Strictures: A Meta-Analysis and Systematic Review of the Literature Development of a Scoring System for Predicting the Difficulty of Bile Duct Cannulation and Selecting the Appropriate Cannulation Method Association of Race and Postoperative Outcomes in Patients with Inflammatory Bowel Disease Correction to: miR‑9‑5p Suppresses Malignant Biological Behaviors of Human Gastric Cancer Cells by Negative Regulation of TNFAIP8L3 Successful Endoscopic Resection of Multiple Colorectal Leiomyosarcomas: The First Case Report
×
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