Impairment of rigidity sensing caused by mutant TP53 gain of function in osteosarcoma.

IF 14.3 1区 医学 Q1 CELL & TISSUE ENGINEERING
Ming Luo, Mingyang Huang, Ningning Yang, Yufan Zhu, Peng Huang, Zhujun Xu, Wengang Wang, Lin Cai
{"title":"Impairment of rigidity sensing caused by mutant TP53 gain of function in osteosarcoma.","authors":"Ming Luo,&nbsp;Mingyang Huang,&nbsp;Ningning Yang,&nbsp;Yufan Zhu,&nbsp;Peng Huang,&nbsp;Zhujun Xu,&nbsp;Wengang Wang,&nbsp;Lin Cai","doi":"10.1038/s41413-023-00265-w","DOIUrl":null,"url":null,"abstract":"<p><p>Osteosarcoma (OS) is the most common primary malignant pediatric bone tumor and is characterized by high heterogeneity. Studies have revealed a wide range of phenotypic differences among OS cell lines in terms of their in vivo tumorigenicity and in vitro colony-forming abilities. However, the underlying molecular mechanism of these discrepancies remains unclear. The potential role of mechanotransduction in tumorigenicity is of particular interest. To this end, we tested the tumorigenicity and anoikis resistance of OS cell lines both in vitro and in vivo. We utilized a sphere culture model, a soft agar assay, and soft and rigid hydrogel surface culture models to investigate the function of rigidity sensing in the tumorigenicity of OS cells. Additionally, we quantified the expression of sensor proteins, including four kinases and seven cytoskeletal proteins, in OS cell lines. The upstream core transcription factors of rigidity-sensing proteins were further investigated. We detected anoikis resistance in transformed OS cells. The mechanosensing function of transformed OS cells was also impaired, with general downregulation of rigidity-sensing components. We identified toggling between normal and transformed growth based on the expression pattern of rigidity-sensing proteins in OS cells. We further uncovered a novel TP53 mutation (R156P) in transformed OS cells, which acquired gain of function to inhibit rigidity sensing, thus sustaining transformed growth. Our findings suggest a fundamental role of rigidity-sensing components in OS tumorigenicity as mechanotransduction elements through which cells can sense their physical microenvironment. In addition, the gain of function of mutant TP53 appears to serve as an executor for such malignant programs.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":14.3000,"publicationDate":"2023-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10225464/pdf/","citationCount":"1","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Bone Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41413-023-00265-w","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CELL & TISSUE ENGINEERING","Score":null,"Total":0}
引用次数: 1

Abstract

Osteosarcoma (OS) is the most common primary malignant pediatric bone tumor and is characterized by high heterogeneity. Studies have revealed a wide range of phenotypic differences among OS cell lines in terms of their in vivo tumorigenicity and in vitro colony-forming abilities. However, the underlying molecular mechanism of these discrepancies remains unclear. The potential role of mechanotransduction in tumorigenicity is of particular interest. To this end, we tested the tumorigenicity and anoikis resistance of OS cell lines both in vitro and in vivo. We utilized a sphere culture model, a soft agar assay, and soft and rigid hydrogel surface culture models to investigate the function of rigidity sensing in the tumorigenicity of OS cells. Additionally, we quantified the expression of sensor proteins, including four kinases and seven cytoskeletal proteins, in OS cell lines. The upstream core transcription factors of rigidity-sensing proteins were further investigated. We detected anoikis resistance in transformed OS cells. The mechanosensing function of transformed OS cells was also impaired, with general downregulation of rigidity-sensing components. We identified toggling between normal and transformed growth based on the expression pattern of rigidity-sensing proteins in OS cells. We further uncovered a novel TP53 mutation (R156P) in transformed OS cells, which acquired gain of function to inhibit rigidity sensing, thus sustaining transformed growth. Our findings suggest a fundamental role of rigidity-sensing components in OS tumorigenicity as mechanotransduction elements through which cells can sense their physical microenvironment. In addition, the gain of function of mutant TP53 appears to serve as an executor for such malignant programs.

Abstract Image

Abstract Image

Abstract Image

骨肉瘤中突变型TP53功能获得引起的刚性感知损伤。
骨肉瘤(Osteosarcoma, OS)是儿童最常见的原发性恶性骨肿瘤,具有高度的异质性。研究表明,在体内致瘤性和体外集落形成能力方面,OS细胞系之间存在广泛的表型差异。然而,这些差异的潜在分子机制尚不清楚。机械转导在致瘤性中的潜在作用尤其令人感兴趣。为此,我们在体外和体内测试了OS细胞系的致瘤性和抗肿瘤性。我们采用球形培养模型、软琼脂实验和软硬水凝胶表面培养模型来研究刚性感知在骨肉瘤细胞致瘤性中的作用。此外,我们量化了传感器蛋白的表达,包括4种激酶和7种细胞骨架蛋白,在OS细胞系中。进一步研究了刚性传感蛋白上游核心转录因子。我们在转化的OS细胞中检测到anoikis抗性。转化后的OS细胞的机械感应功能也受到损害,刚性感应成分普遍下调。我们根据OS细胞中硬度感应蛋白的表达模式确定了正常生长和转化生长之间的切换。我们进一步在转化的OS细胞中发现了一种新的TP53突变(R156P),该突变获得了抑制刚性感知的功能,从而维持了转化的生长。我们的研究结果表明,在OS致瘤性中,刚性传感组分作为机械转导元件发挥了基本作用,细胞可以通过该元件感知其物理微环境。此外,突变体TP53功能的获得似乎是这些恶性程序的执行者。
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 求助全文
来源期刊
Bone Research
Bone Research CELL & TISSUE ENGINEERING-
CiteScore
20.00
自引率
4.70%
发文量
289
审稿时长
20 weeks
期刊介绍: Established in 2013, Bone Research is a newly-founded English-language periodical that centers on the basic and clinical facets of bone biology, pathophysiology, and regeneration. It is dedicated to championing key findings emerging from both basic investigations and clinical research concerning bone-related topics. The journal's objective is to globally disseminate research in bone-related physiology, pathology, diseases, and treatment, contributing to the advancement of knowledge in this field.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信