Clemastine and hyperthermia enhance sensitization of osteosarcoma cells for apoptosis.

IF 2.6 Q3 ONCOLOGY Molecular and Cellular Oncology Pub Date : 2024-05-14 eCollection Date: 2024-01-01 DOI:10.1080/23723556.2024.2351622
Somtochukwu Obu, Suryakant Niture, Hieu Hoang, Sashi Gadi, Vandana, Yiping He, Deepak Kumar
{"title":"Clemastine and hyperthermia enhance sensitization of osteosarcoma cells for apoptosis.","authors":"Somtochukwu Obu, Suryakant Niture, Hieu Hoang, Sashi Gadi, Vandana, Yiping He, Deepak Kumar","doi":"10.1080/23723556.2024.2351622","DOIUrl":null,"url":null,"abstract":"<p><p>Clemastine is an antagonist of histamine H1 receptor may provide benefits in the treatment of osteosarcoma (OS). In the current study, we used hyperthermia approach to sensitize OS cells to clemastine-mediated cell death. Osteosarcoma U-2 OS and Saos-2 cells were treated with clemastine at 37°C, followed by 42°C for 2 h, and released at 37°C for 6 h. The impact of clemastine and hyperthermia on OS cell survival and autophagy-mediated cell death was investigated. Exposure of U-2 OS and Saos-2 cells to clemastine and hyperthermia (42°C) inhibited dose-dependent clemastine-mediated cell survival by increasing cell apoptosis. Hyperthermia and clemastine exposure modulated inflammatory and unfolded protein response (UPR) signaling differentially in U-2 OS and Saos-2 cells. Exposure of U-2 OS and Saos-2 cells to hyperthermia and clemastine inhibited AKT/mTOR and induced expression of the autophagy biomarkers LC3B II and LC3-positive puncta formation. The inhibition of autophagy by 3-methyladenine blocked hyperthermia and clemastine-mediated induction of LC3B II, LC3-positive puncta formation, and OS cell apoptosis. These results indicate that clemastine and hyperthermia sensitize OS cell lines by inducing increased autophagic cell death. Collectively, our data suggest that hyperthermia along with antihistamine therapy may provide an improved approach for the treatment of OS.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":2.6000,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11110698/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular and Cellular Oncology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1080/23723556.2024.2351622","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q3","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Clemastine is an antagonist of histamine H1 receptor may provide benefits in the treatment of osteosarcoma (OS). In the current study, we used hyperthermia approach to sensitize OS cells to clemastine-mediated cell death. Osteosarcoma U-2 OS and Saos-2 cells were treated with clemastine at 37°C, followed by 42°C for 2 h, and released at 37°C for 6 h. The impact of clemastine and hyperthermia on OS cell survival and autophagy-mediated cell death was investigated. Exposure of U-2 OS and Saos-2 cells to clemastine and hyperthermia (42°C) inhibited dose-dependent clemastine-mediated cell survival by increasing cell apoptosis. Hyperthermia and clemastine exposure modulated inflammatory and unfolded protein response (UPR) signaling differentially in U-2 OS and Saos-2 cells. Exposure of U-2 OS and Saos-2 cells to hyperthermia and clemastine inhibited AKT/mTOR and induced expression of the autophagy biomarkers LC3B II and LC3-positive puncta formation. The inhibition of autophagy by 3-methyladenine blocked hyperthermia and clemastine-mediated induction of LC3B II, LC3-positive puncta formation, and OS cell apoptosis. These results indicate that clemastine and hyperthermia sensitize OS cell lines by inducing increased autophagic cell death. Collectively, our data suggest that hyperthermia along with antihistamine therapy may provide an improved approach for the treatment of OS.

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
氯马斯汀和热疗可增强骨肉瘤细胞对凋亡的敏感性。
氯马斯汀是组胺H1受体的拮抗剂,可用于治疗骨肉瘤(OS)。在本研究中,我们采用热疗方法使骨肉瘤细胞对氯马斯汀介导的细胞死亡敏感。骨肉瘤U-2 OS和Saos-2细胞在37摄氏度下接受氯马斯汀处理,然后在42摄氏度下处理2小时,再在37摄氏度下释放6小时,研究了氯马斯汀和高热对OS细胞存活和自噬介导的细胞死亡的影响。将U-2 OS和Saos-2细胞暴露于氯马斯汀和高热(42°C)可通过增加细胞凋亡抑制剂量依赖性氯马斯汀介导的细胞存活。高热和氯马斯汀暴露对U-2 OS和Saos-2细胞的炎症和未折叠蛋白反应(UPR)信号传导有不同的调节作用。U-2 OS和Saos-2细胞暴露于高热和氯马斯汀会抑制AKT/mTOR,并诱导自噬生物标志物LC3B II的表达和LC3阳性点的形成。3-甲基腺嘌呤对自噬的抑制阻止了高热和氯马斯汀介导的 LC3B II 诱导、LC3 阳性点形成和 OS 细胞凋亡。这些结果表明,氯马斯汀和高热可通过诱导自噬细胞死亡增加而使 OS 细胞株变得敏感。总之,我们的数据表明,热疗与抗组胺药疗法可为OS的治疗提供一种更好的方法。
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
Molecular and Cellular Oncology
Molecular and Cellular Oncology Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
3.20
自引率
0.00%
发文量
18
期刊介绍: For a long time, solid neoplasms have been viewed as relatively homogeneous entities composed for the most part of malignant cells. It is now clear that tumors are highly heterogeneous structures that evolve in the context of intimate interactions between cancer cells and endothelial, stromal as well as immune cells. During the past few years, experimental and clinical oncologists have witnessed several conceptual transitions of this type. Molecular and Cellular Oncology (MCO) emerges within this conceptual framework as a high-profile forum for the publication of fundamental, translational and clinical research on cancer. The scope of MCO is broad. Submissions dealing with all aspects of oncogenesis, tumor progression and response to therapy will be welcome, irrespective of whether they focus on solid or hematological neoplasms. MCO has gathered leading scientists with expertise in multiple areas of cancer research and other fields of investigation to constitute a large, interdisciplinary, Editorial Board that will ensure the quality of articles accepted for publication. MCO will publish Original Research Articles, Brief Reports, Reviews, Short Reviews, Commentaries, Author Views (auto-commentaries) and Meeting Reports dealing with all aspects of cancer research.
期刊最新文献
An antibody-drug conjugate for endometrioid carcinoma based on the expression of cell adhesion molecule 1. The SIRT7-nucleolus connection in cancer: ARF enters the fray. Amino acid deprivation in cancer cells with compensatory autophagy induction increases sensitivity to autophagy inhibitors. Selection forces underlying aneuploidy patterns in cancer. Clemastine and hyperthermia enhance sensitization of osteosarcoma cells for apoptosis.
×
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