细胞内运输与肿瘤学关系中的非对称拥挤器和膜形态学

Kshitiz Parihar , Seung-Hyun B. Ko , Ryan P. Bradley , Phillip Taylor , N. Ramakrishnan , Tobias Baumgart , Wei Guo , Valerie M. Weaver , Paul A. Janmey , Ravi Radhakrishnan
{"title":"细胞内运输与肿瘤学关系中的非对称拥挤器和膜形态学","authors":"Kshitiz Parihar ,&nbsp;Seung-Hyun B. Ko ,&nbsp;Ryan P. Bradley ,&nbsp;Phillip Taylor ,&nbsp;N. Ramakrishnan ,&nbsp;Tobias Baumgart ,&nbsp;Wei Guo ,&nbsp;Valerie M. Weaver ,&nbsp;Paul A. Janmey ,&nbsp;Ravi Radhakrishnan","doi":"10.1016/j.mbm.2024.100071","DOIUrl":null,"url":null,"abstract":"<div><p>A definitive understanding of the interplay between protein binding/migration and membrane curvature evolution is emerging but needs further study. The mechanisms defining such phenomena are critical to intracellular transport and trafficking of proteins. Among trafficking modalities, exosomes have drawn attention in cancer research as these nano-sized naturally occurring vehicles are implicated in intercellular communication in the tumor microenvironment, suppressing anti-tumor immunity and preparing the metastatic niche for progression. A significant question in the field is how the release and composition of tumor exosomes are regulated. In this perspective article, we explore how physical factors such as geometry and tissue mechanics regulate cell cortical tension to influence exosome production by co-opting the biophysics as well as the signaling dynamics of intracellular trafficking pathways and how these exosomes contribute to the suppression of anti-tumor immunity and promote metastasis. We describe a multiscale modeling approach whose impact goes beyond the fundamental investigation of specific cellular processes toward actual clinical translation. Exosomal mechanisms are critical to developing and approving liquid biopsy technologies, poised to transform future non-invasive, longitudinal profiling of evolving tumors and resistance to cancer therapies to bring us one step closer to the promise of personalized medicine.</p></div>","PeriodicalId":100900,"journal":{"name":"Mechanobiology in Medicine","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2949907024000342/pdfft?md5=82f74f64557cd0e9b08dee1c94f412b1&pid=1-s2.0-S2949907024000342-main.pdf","citationCount":"0","resultStr":"{\"title\":\"Asymmetric crowders and membrane morphology at the nexus of intracellular trafficking and oncology\",\"authors\":\"Kshitiz Parihar ,&nbsp;Seung-Hyun B. Ko ,&nbsp;Ryan P. Bradley ,&nbsp;Phillip Taylor ,&nbsp;N. Ramakrishnan ,&nbsp;Tobias Baumgart ,&nbsp;Wei Guo ,&nbsp;Valerie M. Weaver ,&nbsp;Paul A. Janmey ,&nbsp;Ravi Radhakrishnan\",\"doi\":\"10.1016/j.mbm.2024.100071\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><p>A definitive understanding of the interplay between protein binding/migration and membrane curvature evolution is emerging but needs further study. The mechanisms defining such phenomena are critical to intracellular transport and trafficking of proteins. Among trafficking modalities, exosomes have drawn attention in cancer research as these nano-sized naturally occurring vehicles are implicated in intercellular communication in the tumor microenvironment, suppressing anti-tumor immunity and preparing the metastatic niche for progression. A significant question in the field is how the release and composition of tumor exosomes are regulated. In this perspective article, we explore how physical factors such as geometry and tissue mechanics regulate cell cortical tension to influence exosome production by co-opting the biophysics as well as the signaling dynamics of intracellular trafficking pathways and how these exosomes contribute to the suppression of anti-tumor immunity and promote metastasis. We describe a multiscale modeling approach whose impact goes beyond the fundamental investigation of specific cellular processes toward actual clinical translation. Exosomal mechanisms are critical to developing and approving liquid biopsy technologies, poised to transform future non-invasive, longitudinal profiling of evolving tumors and resistance to cancer therapies to bring us one step closer to the promise of personalized medicine.</p></div>\",\"PeriodicalId\":100900,\"journal\":{\"name\":\"Mechanobiology in Medicine\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-05-03\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.sciencedirect.com/science/article/pii/S2949907024000342/pdfft?md5=82f74f64557cd0e9b08dee1c94f412b1&pid=1-s2.0-S2949907024000342-main.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Mechanobiology in Medicine\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S2949907024000342\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Mechanobiology in Medicine","FirstCategoryId":"1085","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2949907024000342","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

对蛋白质结合/迁移与膜曲率演变之间相互作用的明确认识正在形成,但还需要进一步研究。定义这种现象的机制对于蛋白质的细胞内运输和迁移至关重要。在各种转运方式中,外泌体在癌症研究中备受关注,因为这些纳米级的天然载体参与了肿瘤微环境中的细胞间交流,抑制了抗肿瘤免疫,并为转移龛的进展做好了准备。该领域的一个重要问题是如何调控肿瘤外泌体的释放和组成。在这篇视角文章中,我们将探讨几何和组织力学等物理因素如何通过共同作用于生物物理学以及细胞内转运途径的信号动力学来调节细胞皮质张力,从而影响外泌体的产生,以及这些外泌体如何有助于抑制抗肿瘤免疫并促进转移。我们描述了一种多尺度建模方法,它的影响超出了对特定细胞过程的基础研究,而是走向实际的临床转化。外泌体机制对于开发和批准液体活检技术至关重要,它将改变未来对不断发展的肿瘤和癌症疗法耐药性的非侵入性纵向剖析,使我们离个性化医疗的承诺更近一步。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Asymmetric crowders and membrane morphology at the nexus of intracellular trafficking and oncology

A definitive understanding of the interplay between protein binding/migration and membrane curvature evolution is emerging but needs further study. The mechanisms defining such phenomena are critical to intracellular transport and trafficking of proteins. Among trafficking modalities, exosomes have drawn attention in cancer research as these nano-sized naturally occurring vehicles are implicated in intercellular communication in the tumor microenvironment, suppressing anti-tumor immunity and preparing the metastatic niche for progression. A significant question in the field is how the release and composition of tumor exosomes are regulated. In this perspective article, we explore how physical factors such as geometry and tissue mechanics regulate cell cortical tension to influence exosome production by co-opting the biophysics as well as the signaling dynamics of intracellular trafficking pathways and how these exosomes contribute to the suppression of anti-tumor immunity and promote metastasis. We describe a multiscale modeling approach whose impact goes beyond the fundamental investigation of specific cellular processes toward actual clinical translation. Exosomal mechanisms are critical to developing and approving liquid biopsy technologies, poised to transform future non-invasive, longitudinal profiling of evolving tumors and resistance to cancer therapies to bring us one step closer to the promise of personalized medicine.

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
自引率
0.00%
发文量
0
期刊最新文献
Strain and hyaluronic acid interact to regulate ovarian cancer cell proliferation, migration, and drug resistance In vivo analysis of hybrid hydrogels containing dual growth factor combinations, and skeletal stem cells under mechanical stimulation for bone repair Low-magnitude high-frequency vibration reduces prostate cancer growth and extravasation in vitro Application of biomechanics in tumor epigenetic research YAP/TAZ as mechanobiological signaling pathway in cardiovascular physiological regulation and pathogenesis
×
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