用于生成肿瘤免疫微环境模型的器官组织的显微切割工具

IF 7.3 1区 工程技术 Q1 INSTRUMENTS & INSTRUMENTATION Microsystems & Nanoengineering Pub Date : 2024-09-10 DOI:10.1038/s41378-024-00756-8
Seth C. Cordts, Kanako Yuki, Maria F. Henao Echeverri, Balasubramanian Narasimhan, Calvin J. Kuo, Sindy K. Y. Tang
{"title":"用于生成肿瘤免疫微环境模型的器官组织的显微切割工具","authors":"Seth C. Cordts, Kanako Yuki, Maria F. Henao Echeverri, Balasubramanian Narasimhan, Calvin J. Kuo, Sindy K. Y. Tang","doi":"10.1038/s41378-024-00756-8","DOIUrl":null,"url":null,"abstract":"<p>Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.</p><figure></figure>","PeriodicalId":18560,"journal":{"name":"Microsystems & Nanoengineering","volume":null,"pages":null},"PeriodicalIF":7.3000,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Microdissection tools to generate organoids for modeling the tumor immune microenvironment\",\"authors\":\"Seth C. Cordts, Kanako Yuki, Maria F. Henao Echeverri, Balasubramanian Narasimhan, Calvin J. Kuo, Sindy K. Y. Tang\",\"doi\":\"10.1038/s41378-024-00756-8\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.</p><figure></figure>\",\"PeriodicalId\":18560,\"journal\":{\"name\":\"Microsystems & Nanoengineering\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":7.3000,\"publicationDate\":\"2024-09-10\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Microsystems & Nanoengineering\",\"FirstCategoryId\":\"5\",\"ListUrlMain\":\"https://doi.org/10.1038/s41378-024-00756-8\",\"RegionNum\":1,\"RegionCategory\":\"工程技术\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"INSTRUMENTS & INSTRUMENTATION\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Microsystems & Nanoengineering","FirstCategoryId":"5","ListUrlMain":"https://doi.org/10.1038/s41378-024-00756-8","RegionNum":1,"RegionCategory":"工程技术","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"INSTRUMENTS & INSTRUMENTATION","Score":null,"Total":0}
引用次数: 0

摘要

源自患者的肿瘤器官组织已成为预测癌症治疗中个性化药物反应的有前途的模型,但它们通常缺乏免疫成分。保留肿瘤细胞与内源性免疫细胞之间的体内关联对于准确测试癌症免疫疗法至关重要。将肿瘤标本机械解剖成肿瘤碎片,而不是酶解成单细胞,对于保持这些原生肿瘤-免疫细胞的空间关系至关重要。然而,传统的机械解剖依赖于人工碾碎,既费时又不可重复。本研究介绍了µDicer和µGrater这两种显微切割设备,它们有助于从小鼠B16黑色素瘤(一种常见的人类黑色素瘤模型)中生成完整的肿瘤片段。用µDicer和µGrater切割的肿瘤片段生成的气液界面(ALI)类器官无需人工重组即可保留肿瘤细胞和浸润免疫亚群。µDicer由六角形硅微刀片阵列组成,用于研究类器官大小的影响。ALI类器官免疫细胞的存活率似乎对超过400微米的类器官大小不敏感,但在200微米的类器官中则会降低。µGrater由不锈钢上的亚毫米孔阵列组成,用于加速解剖。对于所研究的样本,µGrater 的速度是人工切割的 4.5 倍。与人工切割相比,µGrater 生成的 ALI 器官组织显示出相似的存活率、免疫细胞组成以及对抗 PD-1 免疫疗法的反应。通过进一步优化,µGrater 有潜力整合到临床工作流程中,支持个性化癌症免疫疗法的发展。
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
Microdissection tools to generate organoids for modeling the tumor immune microenvironment

Patient-derived tumor organoids have emerged as promising models for predicting personalized drug responses in cancer therapy, but they typically lack immune components. Preserving the in vivo association between tumor cells and endogenous immune cells is critical for accurate testing of cancer immunotherapies. Mechanical dissection of tumor specimens into tumor fragments, as opposed to enzymatic digestion into single cells, is essential for maintaining these native tumor-immune cell spatial relationships. However, conventional mechanical dissection relying on manual mincing is time-consuming and irreproducible. This study describes two microdissection devices, the µDicer and µGrater, to facilitate the generation of intact tumor fragments from mouse B16 melanoma, a common model of human melanoma. The µDicer- and µGrater-cut tumor fragments were used to generate air‒liquid interface (ALI) organoids that copreserve tumor cells with infiltrating immune subsets without artificial reconstitution. The µDicer, consisting of a hexagonal array of silicon microblades, was employed to investigate the effect of organoid size. The viability of ALI organoid immune cells appeared insensitive to organoid sizes exceeding ~400 µm but diminished in organoids ~200 µm in size. The µGrater, consisting of an array of submillimeter holes in stainless steel, was employed to accelerate dissection. For the samples studied, the µGrater was 4.5 times faster than manual mincing. Compared with those generated by manual mincing, ALI organoids generated by the µGrater demonstrated similar viability, immune cell composition, and responses to anti-PD-1 immunotherapy. With further optimization, the µGrater holds potential for integration into clinical workflows to support the advancement of personalized cancer immunotherapy.

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
Microsystems & Nanoengineering
Microsystems & Nanoengineering Materials Science-Materials Science (miscellaneous)
CiteScore
12.00
自引率
3.80%
发文量
123
审稿时长
20 weeks
期刊介绍: Microsystems & Nanoengineering is a comprehensive online journal that focuses on the field of Micro and Nano Electro Mechanical Systems (MEMS and NEMS). It provides a platform for researchers to share their original research findings and review articles in this area. The journal covers a wide range of topics, from fundamental research to practical applications. Published by Springer Nature, in collaboration with the Aerospace Information Research Institute, Chinese Academy of Sciences, and with the support of the State Key Laboratory of Transducer Technology, it is an esteemed publication in the field. As an open access journal, it offers free access to its content, allowing readers from around the world to benefit from the latest developments in MEMS and NEMS.
期刊最新文献
Designing magnetic microcapsules for cultivation and differentiation of stem cell spheroids A low-voltage-driven MEMS ultrasonic phased-array transducer for fast 3D volumetric imaging Microdissection tools to generate organoids for modeling the tumor immune microenvironment Sustainability inspired fabrication of next generation neurostimulation and cardiac rhythm management electrodes via reactive hierarchical surface restructuring Noncontact excitation of multi-GHz lithium niobate electromechanical resonators.
×
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