Matrix Stiffness Enhances VEGFR-2 Internalization, Signaling, and Proliferation in Endothelial Cells.

Convergent science physical oncology Pub Date : 2017-01-01 Epub Date: 2017-11-29 DOI:10.1088/2057-1739/aa9263
Danielle J LaValley, Matthew R Zanotelli, Francois Bordeleau, Wenjun Wang, Samantha C Schwager, Cynthia A Reinhart-King
{"title":"Matrix Stiffness Enhances VEGFR-2 Internalization, Signaling, and Proliferation in Endothelial Cells.","authors":"Danielle J LaValley,&nbsp;Matthew R Zanotelli,&nbsp;Francois Bordeleau,&nbsp;Wenjun Wang,&nbsp;Samantha C Schwager,&nbsp;Cynthia A Reinhart-King","doi":"10.1088/2057-1739/aa9263","DOIUrl":null,"url":null,"abstract":"<p><p>Vascular endothelial growth factor (VEGF) can mediate endothelial cell migration, proliferation, and angiogenesis. During cancer progression, VEGF production is often increased to stimulate the growth of new blood vessels to supply growing tumors with the additional oxygen and nutrients they require. Extracellular matrix stiffening also occurs during tumor progression, however, the crosstalk between tumor mechanics and VEGF signaling remains poorly understood. Here, we show that matrix stiffness heightens downstream endothelial cell response to VEGF by altering VEGF receptor-2 (VEGFR-2) internalization, and this effect is influenced by cell confluency. In sub-confluent endothelial monolayers, VEGFR-2 levels, but not VEGFR-2 phosphorylation, are influenced by matrix rigidity. Interestingly, more compliant matrices correlated with increased expression and clustering of VEGFR-2; however, stiffer matrices induced increased VEGFR-2 internalization. These effects are most likely due to actin-mediated contractility, as inhibiting ROCK on stiff substrates increased VEGFR-2 clustering and decreased internalization. Additionally, increasing matrix stiffness elevates ERK 1/2 phosphorylation, resulting in increased cell proliferation. Moreover, cells on stiff matrices generate more actin stress fibers than on compliant substrates, and the addition of VEGF stimulates an increase in fiber formation regardless of stiffness. In contrast, once endothelial cells reached confluency, stiffness-enhanced VEGF signaling was no longer observed. Together, these data show a complex effect of VEGF and matrix mechanics on VEGF-induced signaling, receptor dynamics, and cell proliferation that is mediated by cell confluency.</p>","PeriodicalId":91466,"journal":{"name":"Convergent science physical oncology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2017-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1088/2057-1739/aa9263","citationCount":"53","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Convergent science physical oncology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1088/2057-1739/aa9263","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2017/11/29 0:00:00","PubModel":"Epub","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 53

Abstract

Vascular endothelial growth factor (VEGF) can mediate endothelial cell migration, proliferation, and angiogenesis. During cancer progression, VEGF production is often increased to stimulate the growth of new blood vessels to supply growing tumors with the additional oxygen and nutrients they require. Extracellular matrix stiffening also occurs during tumor progression, however, the crosstalk between tumor mechanics and VEGF signaling remains poorly understood. Here, we show that matrix stiffness heightens downstream endothelial cell response to VEGF by altering VEGF receptor-2 (VEGFR-2) internalization, and this effect is influenced by cell confluency. In sub-confluent endothelial monolayers, VEGFR-2 levels, but not VEGFR-2 phosphorylation, are influenced by matrix rigidity. Interestingly, more compliant matrices correlated with increased expression and clustering of VEGFR-2; however, stiffer matrices induced increased VEGFR-2 internalization. These effects are most likely due to actin-mediated contractility, as inhibiting ROCK on stiff substrates increased VEGFR-2 clustering and decreased internalization. Additionally, increasing matrix stiffness elevates ERK 1/2 phosphorylation, resulting in increased cell proliferation. Moreover, cells on stiff matrices generate more actin stress fibers than on compliant substrates, and the addition of VEGF stimulates an increase in fiber formation regardless of stiffness. In contrast, once endothelial cells reached confluency, stiffness-enhanced VEGF signaling was no longer observed. Together, these data show a complex effect of VEGF and matrix mechanics on VEGF-induced signaling, receptor dynamics, and cell proliferation that is mediated by cell confluency.

Abstract Image

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
基质硬度增强内皮细胞VEGFR-2内化、信号传导和增殖
血管内皮生长因子(VEGF)可以介导内皮细胞的迁移、增殖和血管生成。在癌症进展过程中,VEGF的产生通常会增加,以刺激新血管的生长,为生长中的肿瘤提供所需的额外氧气和营养。细胞外基质硬化也发生在肿瘤进展过程中,然而,肿瘤机制和VEGF信号之间的相互作用仍然知之甚少。在这里,我们发现基质刚度通过改变VEGF受体-2 (VEGFR-2)内化来增强下游内皮细胞对VEGF的反应,这种作用受到细胞融合的影响。在亚融合内皮单层中,VEGFR-2水平受基质刚度影响,而VEGFR-2磷酸化不受其影响。有趣的是,更柔顺的基质与VEGFR-2的表达和聚集增加相关;然而,更硬的基质诱导VEGFR-2内化增加。这些影响很可能是由于肌动蛋白介导的收缩性,因为抑制坚硬底物上的ROCK会增加VEGFR-2聚集并减少内化。此外,增加基质硬度会提高ERK 1/2磷酸化,导致细胞增殖增加。此外,坚硬基质上的细胞比柔顺基质上的细胞产生更多的肌动蛋白应力纤维,VEGF的加入刺激了纤维形成的增加,而不管硬度如何。相反,一旦内皮细胞达到融合,僵硬增强的VEGF信号不再被观察到。总之,这些数据显示了VEGF和基质力学对VEGF诱导的信号、受体动力学和由细胞融合介导的细胞增殖的复杂影响。
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
自引率
0.00%
发文量
0
期刊最新文献
Time for a change: considering the rights of study participants to ownership of their personal research-grade genomic data Meeting report: The physics of life—merging clinical, biological and physical sciences approaches for cancer research Changing cell mechanics—a precondition for malignant transformation of oral squamous carcinoma cells Heterogeneous radiotherapy dose-outcomes response in parotid glands Secondary use of electronic medical records for clinical research: Challenges and Opportunities.
×
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