1220正交CRISPR筛选鉴定人类CD8 T细胞功能的转录和表观遗传调控因子

Sean McCutcheon, Adam Swartz, Michael Brown, Alejandro Barrera, Christian McRoberts Amador, Keith Siklenka, Lucas Humayun, James Isaacs, Timothy Reddy, Smita Nair, Scott Antonia, Charles Gersbach
{"title":"1220正交CRISPR筛选鉴定人类CD8 T细胞功能的转录和表观遗传调控因子","authors":"Sean McCutcheon, Adam Swartz, Michael Brown, Alejandro Barrera, Christian McRoberts Amador, Keith Siklenka, Lucas Humayun, James Isaacs, Timothy Reddy, Smita Nair, Scott Antonia, Charles Gersbach","doi":"10.1136/jitc-2023-sitc2023.1220","DOIUrl":null,"url":null,"abstract":"<h3>Background</h3> The clinical response to adoptive T cell therapies is strongly associated with transcriptional and epigenetic state. Thus, technologies to discover regulators of T cell gene networks and their corresponding phenotypes have great potential to improve the efficacy of T cell therapies. <h3>Methods</h3> We developed pooled CRISPR screening approaches with compact epigenome editors to systematically profile the effects of activation and repression of 120 transcription factors and epigenetic modifiers on human CD8 T cell state. <h3>Results</h3> CRISPR interference and activation screens nominated known and novel regulators of T cell phenotypes with BATF3 emerging as a high confidence gene in both screens. We found that BATF3 overexpression promoted specific features of memory T cells such as increased IL7R expression and glycolytic capacity, while attenuating gene programs associated with cytotoxicity, regulatory T cell function, and T cell exhaustion. In the context of chronic antigen stimulation, BATF3 overexpression countered phenotypic and epigenetic signatures of T cell exhaustion. For example, only 13% of BATF3 engineered T cells co-expressed canonical exhaustion markers (LAG3, TIM3, TIGIT), whereas 65% of wild type T cell co-expressed all three markers after multiple rounds of antigen stimulation. CAR T cells overexpressing BATF3 significantly outperformed control CAR T cells in both in vitro and in vivo tumor models. Moreover, we found that BATF3 programmed a transcriptional profile that correlated with positive clinical response to adoptive T cell therapy. Finally, we performed CRISPR knockout screens with and without BATF3 overexpression to define co-factors and downstream factors of BATF3, as well as other therapeutic targets. <h3>Conclusions</h3> BATF3 overexpression markedly enhanced the therapeutic potential of CD8 T cells in both in vitro and in vivo tumor models. The compact size of BATF3 could seamlessly integrate into current manufacturing processes of FDA-approved adoptive T cell therapies, which all use lentivirus to deliver the CAR construct to donor T cells. To our knowledge, this work is the first example that combines overexpression of a specific transcription factor with a transcription factor wide knockout screen to dissect co-factors and downstream factors and highlights the power of this approach. These screens pointed to a model where BATF3 interacts with JUNB and IRF4 to regulate gene expression and illuminated several other novel targets for further investigation.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"1220 Orthogonal CRISPR screens to identify transcriptional and epigenetic regulators of human CD8 T cell function\",\"authors\":\"Sean McCutcheon, Adam Swartz, Michael Brown, Alejandro Barrera, Christian McRoberts Amador, Keith Siklenka, Lucas Humayun, James Isaacs, Timothy Reddy, Smita Nair, Scott Antonia, Charles Gersbach\",\"doi\":\"10.1136/jitc-2023-sitc2023.1220\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<h3>Background</h3> The clinical response to adoptive T cell therapies is strongly associated with transcriptional and epigenetic state. Thus, technologies to discover regulators of T cell gene networks and their corresponding phenotypes have great potential to improve the efficacy of T cell therapies. <h3>Methods</h3> We developed pooled CRISPR screening approaches with compact epigenome editors to systematically profile the effects of activation and repression of 120 transcription factors and epigenetic modifiers on human CD8 T cell state. <h3>Results</h3> CRISPR interference and activation screens nominated known and novel regulators of T cell phenotypes with BATF3 emerging as a high confidence gene in both screens. We found that BATF3 overexpression promoted specific features of memory T cells such as increased IL7R expression and glycolytic capacity, while attenuating gene programs associated with cytotoxicity, regulatory T cell function, and T cell exhaustion. In the context of chronic antigen stimulation, BATF3 overexpression countered phenotypic and epigenetic signatures of T cell exhaustion. For example, only 13% of BATF3 engineered T cells co-expressed canonical exhaustion markers (LAG3, TIM3, TIGIT), whereas 65% of wild type T cell co-expressed all three markers after multiple rounds of antigen stimulation. CAR T cells overexpressing BATF3 significantly outperformed control CAR T cells in both in vitro and in vivo tumor models. Moreover, we found that BATF3 programmed a transcriptional profile that correlated with positive clinical response to adoptive T cell therapy. Finally, we performed CRISPR knockout screens with and without BATF3 overexpression to define co-factors and downstream factors of BATF3, as well as other therapeutic targets. <h3>Conclusions</h3> BATF3 overexpression markedly enhanced the therapeutic potential of CD8 T cells in both in vitro and in vivo tumor models. The compact size of BATF3 could seamlessly integrate into current manufacturing processes of FDA-approved adoptive T cell therapies, which all use lentivirus to deliver the CAR construct to donor T cells. To our knowledge, this work is the first example that combines overexpression of a specific transcription factor with a transcription factor wide knockout screen to dissect co-factors and downstream factors and highlights the power of this approach. These screens pointed to a model where BATF3 interacts with JUNB and IRF4 to regulate gene expression and illuminated several other novel targets for further investigation.\",\"PeriodicalId\":500964,\"journal\":{\"name\":\"Regular and Young Investigator Award Abstracts\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2023-11-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Regular and Young Investigator Award Abstracts\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1136/jitc-2023-sitc2023.1220\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Regular and Young Investigator Award Abstracts","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1136/jitc-2023-sitc2023.1220","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

背景过继性T细胞治疗的临床反应与转录和表观遗传状态密切相关。因此,发现T细胞基因网络的调节因子及其相应表型的技术对提高T细胞治疗的疗效具有很大的潜力。方法采用紧凑的表观基因组编辑器开发了CRISPR筛选方法,系统地分析了120个转录因子和表观遗传修饰因子的激活和抑制对人CD8 T细胞状态的影响。结果CRISPR干扰和激活筛选提名了已知的和新的T细胞表型调节因子,其中BATF3在两种筛选中都作为高置信度基因出现。我们发现BATF3过表达促进了记忆性T细胞的特定特征,如IL7R表达和糖酵解能力的增加,同时减弱了与细胞毒性、调节性T细胞功能和T细胞衰竭相关的基因程序。在慢性抗原刺激的背景下,BATF3过表达对抗T细胞衰竭的表型和表观遗传特征。例如,只有13%的BATF3工程T细胞共表达典型耗竭标志物(LAG3, TIM3, TIGIT),而65%的野生型T细胞在多轮抗原刺激后共表达所有三种标志物。在体外和体内肿瘤模型中,过表达BATF3的CAR - T细胞都明显优于对照CAR - T细胞。此外,我们发现BATF3编程的转录谱与对过继T细胞治疗的阳性临床反应相关。最后,我们进行了有和没有BATF3过表达的CRISPR敲除筛选,以确定BATF3的辅助因子和下游因子,以及其他治疗靶点。结论BATF3过表达可显著增强CD8 T细胞在体内和体外肿瘤模型中的治疗潜力。BATF3的紧凑尺寸可以无缝集成到fda批准的过继性T细胞疗法的当前制造过程中,这些疗法都使用慢病毒将CAR结构体递送到供体T细胞中。据我们所知,这项工作是第一个将特定转录因子的过表达与转录因子的广泛敲除筛选相结合来解剖辅助因子和下游因子的例子,并突出了这种方法的力量。这些筛选指出了BATF3与JUNB和IRF4相互作用以调节基因表达的模型,并阐明了其他几个有待进一步研究的新靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
1220 Orthogonal CRISPR screens to identify transcriptional and epigenetic regulators of human CD8 T cell function

Background

The clinical response to adoptive T cell therapies is strongly associated with transcriptional and epigenetic state. Thus, technologies to discover regulators of T cell gene networks and their corresponding phenotypes have great potential to improve the efficacy of T cell therapies.

Methods

We developed pooled CRISPR screening approaches with compact epigenome editors to systematically profile the effects of activation and repression of 120 transcription factors and epigenetic modifiers on human CD8 T cell state.

Results

CRISPR interference and activation screens nominated known and novel regulators of T cell phenotypes with BATF3 emerging as a high confidence gene in both screens. We found that BATF3 overexpression promoted specific features of memory T cells such as increased IL7R expression and glycolytic capacity, while attenuating gene programs associated with cytotoxicity, regulatory T cell function, and T cell exhaustion. In the context of chronic antigen stimulation, BATF3 overexpression countered phenotypic and epigenetic signatures of T cell exhaustion. For example, only 13% of BATF3 engineered T cells co-expressed canonical exhaustion markers (LAG3, TIM3, TIGIT), whereas 65% of wild type T cell co-expressed all three markers after multiple rounds of antigen stimulation. CAR T cells overexpressing BATF3 significantly outperformed control CAR T cells in both in vitro and in vivo tumor models. Moreover, we found that BATF3 programmed a transcriptional profile that correlated with positive clinical response to adoptive T cell therapy. Finally, we performed CRISPR knockout screens with and without BATF3 overexpression to define co-factors and downstream factors of BATF3, as well as other therapeutic targets.

Conclusions

BATF3 overexpression markedly enhanced the therapeutic potential of CD8 T cells in both in vitro and in vivo tumor models. The compact size of BATF3 could seamlessly integrate into current manufacturing processes of FDA-approved adoptive T cell therapies, which all use lentivirus to deliver the CAR construct to donor T cells. To our knowledge, this work is the first example that combines overexpression of a specific transcription factor with a transcription factor wide knockout screen to dissect co-factors and downstream factors and highlights the power of this approach. These screens pointed to a model where BATF3 interacts with JUNB and IRF4 to regulate gene expression and illuminated several other novel targets for further investigation.
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
×
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