1466 .半乳糖凝集素-1协调了肿瘤到间质细胞外囊泡系统的分级,促进免疫抑制

Camila A Bach, Ada Blidner, Ramiro Perrotta, Joaquin P Merlo, Mariana Salatino, Gabriel Rabinovich
{"title":"1466 .半乳糖凝集素-1协调了肿瘤到间质细胞外囊泡系统的分级,促进免疫抑制","authors":"Camila A Bach, Ada Blidner, Ramiro Perrotta, Joaquin P Merlo, Mariana Salatino, Gabriel Rabinovich","doi":"10.1136/jitc-2023-sitc2023.1466","DOIUrl":null,"url":null,"abstract":"<h3>Background</h3> Extracellular vesicles (EVs) suppress effector cells and activate immunosuppressive cells in the ‘escape phase’ of tumor immunoediting. Tumor derived EVs contribute to the differentiation and expansion of immunosuppressive cell subsets. Bone marrow-derived cells (BMDCs) can uptake EVs released by 4T1 -a triple-negative breast cancer cell line- and induce myeloid-derived suppressor cells (MDSCs).<sup>1</sup> Immunosuppressive myeloid cells expansion can also be promoted by their own EV in an autocrine manner. Moreover, EVs secreted by immunosuppressive myeloid cells, may inherit their parental functions.<sup>2</sup> Nevertheless, the molecular circuits and mediators of tumor microenvironment EV release remain uncertain. <h3>Methods</h3> We generated immunosuppressive myeloid cells by culturing mouse BMDCs with GM-CSF for three days in the presence of Gal1. Additionally, we cultured 4T1 WT and <i>Lgals1</i> knocked out cells. We purified Small EVs by size exclusion chromatography from conditioned medium of immunosuppressive myeloid cells (control and Gal1-treated) or 4T1 cells (WT or Gal1 KO). We further purified 4T1 EVs with CD63+ beads, a tumoral EV marker. ELISA, flow cytometry, miRNA sequencing, proteomic and metabolomic analyses verified EV identity. We performed functional assays by co-culturing tumoral or myeloid EVs with BMDC, activated T or B cells. <h3>Results</h3> When exposed to EVs from 4T1 cells <i>in vitro</i>, BMDCs switched their differentiation pathway to a M-MDSCs phenotype (CD11b<sup>+</sup>Ly6G<sup>-</sup>Ly6C<sup>hi</sup>) and showed higher immune checkpoint molecules expression, including PD-L1 (p=0.005) and VISTA (p=0.003). However, when exposed to tumoral EVs lacking Gal1, BMDCs reinforced a PMN-MDSCs phenotype (CD11b<sup>+</sup>Ly6G<sup>+</sup>Ly6C<sup>lo</sup>). In contrast with Gal1<sup>+</sup> EVs, tumoral EVs lacking Gal1 failed to inhibit CD4<sup>+</sup> and CD8<sup>+</sup> T and B cell proliferation and activation. Gal1 is both in the cargo and corona of 4T1 EVs as revealed by flow cytometry and ELISA (p=0.0032). Moreover, EV production inhibition with GW4869 in 4T1 cells decreased Gal1 levels secreted (p=0.037). To analyze EV-derived Gal1 contribution as an MDSC autocrine signal, we added EVs from Gal1-treated immunosuppressive myeloid cells to new BMDCs and observed CD11b<sup>+</sup> cells expansion and higher VISTA expression on cells with M-MDSCs phenotype. Moreover, these EVs showed greater T cell-suppressive capacity and has different protein, metabolic, and miRNA cargo compared to control EVs. <h3>Conclusions</h3> Here, we propose that Gal1<sup>+</sup> EVs released by tumoral cells interact with myeloid cells and potentiate their immunosuppressive properties, including the release of EV with autocrine and paracrine functions. Targeting Gal1 as a molecular mechanism of EV-mediated tumor development may help overcome therapy resistance. <h3>References</h3> Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, Grizzle WE, Moble J, Zhang HG. Induction of myeloid-derived suppressor cells by tumor exosomes. <i>Int J Cancer</i>. 2009;<i><b>124</b></i>:2621–2633. Rashid MH, Borin TF. <i>Rep.</i> Critical immunosuppressive effect of MDSC-derived exosomes in the tumor microenvironment.2021;<i><b>45</b></i>:1171–1181.","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\":\"1466 Galectin-1 orchestrates a hierarchical tumor to stromal extracellular vesicles system that fosters immune suppression\",\"authors\":\"Camila A Bach, Ada Blidner, Ramiro Perrotta, Joaquin P Merlo, Mariana Salatino, Gabriel Rabinovich\",\"doi\":\"10.1136/jitc-2023-sitc2023.1466\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<h3>Background</h3> Extracellular vesicles (EVs) suppress effector cells and activate immunosuppressive cells in the ‘escape phase’ of tumor immunoediting. Tumor derived EVs contribute to the differentiation and expansion of immunosuppressive cell subsets. Bone marrow-derived cells (BMDCs) can uptake EVs released by 4T1 -a triple-negative breast cancer cell line- and induce myeloid-derived suppressor cells (MDSCs).<sup>1</sup> Immunosuppressive myeloid cells expansion can also be promoted by their own EV in an autocrine manner. Moreover, EVs secreted by immunosuppressive myeloid cells, may inherit their parental functions.<sup>2</sup> Nevertheless, the molecular circuits and mediators of tumor microenvironment EV release remain uncertain. <h3>Methods</h3> We generated immunosuppressive myeloid cells by culturing mouse BMDCs with GM-CSF for three days in the presence of Gal1. Additionally, we cultured 4T1 WT and <i>Lgals1</i> knocked out cells. We purified Small EVs by size exclusion chromatography from conditioned medium of immunosuppressive myeloid cells (control and Gal1-treated) or 4T1 cells (WT or Gal1 KO). We further purified 4T1 EVs with CD63+ beads, a tumoral EV marker. ELISA, flow cytometry, miRNA sequencing, proteomic and metabolomic analyses verified EV identity. We performed functional assays by co-culturing tumoral or myeloid EVs with BMDC, activated T or B cells. <h3>Results</h3> When exposed to EVs from 4T1 cells <i>in vitro</i>, BMDCs switched their differentiation pathway to a M-MDSCs phenotype (CD11b<sup>+</sup>Ly6G<sup>-</sup>Ly6C<sup>hi</sup>) and showed higher immune checkpoint molecules expression, including PD-L1 (p=0.005) and VISTA (p=0.003). However, when exposed to tumoral EVs lacking Gal1, BMDCs reinforced a PMN-MDSCs phenotype (CD11b<sup>+</sup>Ly6G<sup>+</sup>Ly6C<sup>lo</sup>). In contrast with Gal1<sup>+</sup> EVs, tumoral EVs lacking Gal1 failed to inhibit CD4<sup>+</sup> and CD8<sup>+</sup> T and B cell proliferation and activation. Gal1 is both in the cargo and corona of 4T1 EVs as revealed by flow cytometry and ELISA (p=0.0032). Moreover, EV production inhibition with GW4869 in 4T1 cells decreased Gal1 levels secreted (p=0.037). To analyze EV-derived Gal1 contribution as an MDSC autocrine signal, we added EVs from Gal1-treated immunosuppressive myeloid cells to new BMDCs and observed CD11b<sup>+</sup> cells expansion and higher VISTA expression on cells with M-MDSCs phenotype. Moreover, these EVs showed greater T cell-suppressive capacity and has different protein, metabolic, and miRNA cargo compared to control EVs. <h3>Conclusions</h3> Here, we propose that Gal1<sup>+</sup> EVs released by tumoral cells interact with myeloid cells and potentiate their immunosuppressive properties, including the release of EV with autocrine and paracrine functions. Targeting Gal1 as a molecular mechanism of EV-mediated tumor development may help overcome therapy resistance. <h3>References</h3> Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, Grizzle WE, Moble J, Zhang HG. Induction of myeloid-derived suppressor cells by tumor exosomes. <i>Int J Cancer</i>. 2009;<i><b>124</b></i>:2621–2633. Rashid MH, Borin TF. <i>Rep.</i> Critical immunosuppressive effect of MDSC-derived exosomes in the tumor microenvironment.2021;<i><b>45</b></i>:1171–1181.\",\"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-sitc2023.1466\",\"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-sitc2023.1466","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

细胞外囊泡(EVs)在肿瘤免疫编辑的“逃逸期”抑制效应细胞并激活免疫抑制细胞。肿瘤源性ev有助于免疫抑制细胞亚群的分化和扩增。骨髓源性细胞(bmdc)可以摄取由4T1(三阴性乳腺癌细胞系)释放的ev,并诱导髓源性抑制细胞(MDSCs) 1免疫抑制性骨髓细胞的扩张也可以通过自身的EV以自分泌的方式促进。此外,免疫抑制髓细胞分泌的ev可能继承了亲代的功能然而,肿瘤微环境EV释放的分子通路和介质仍不确定。方法在Gal1存在下,用GM-CSF培养小鼠BMDCs 3 d,产生免疫抑制的骨髓细胞。此外,我们还培养了4T1 WT和Lgals1敲除的细胞。我们从免疫抑制骨髓细胞(对照和Gal1处理)或4T1细胞(WT或Gal1 KO)的条件培养基中纯化了小ev。我们进一步用CD63+珠(一种肿瘤EV标记物)纯化了4T1 EV。ELISA、流式细胞术、miRNA测序、蛋白质组学和代谢组学分析证实了EV的身份。我们通过将肿瘤或髓系EVs与BMDC、活化的T细胞或B细胞共培养进行了功能测定。结果当体外暴露于4T1细胞的EVs时,bmdscs将其分化途径转换为M-MDSCs表型(CD11b+Ly6G-Ly6Chi),并表现出更高的免疫检查点分子表达,包括PD-L1 (p=0.005)和VISTA (p=0.003)。然而,当暴露于缺乏Gal1的肿瘤ev时,BMDCs增强了PMN-MDSCs表型(CD11b+Ly6G+Ly6Clo)。与Gal1+ ev相比,缺乏Gal1的肿瘤ev不能抑制CD4+和CD8+ T细胞和B细胞的增殖和活化。流式细胞术和酶联免疫吸附试验显示,4T1型EVs的货物和冠状细胞中均存在Gal1 (p=0.0032)。此外,GW4869抑制4T1细胞的EV产生可降低Gal1分泌水平(p=0.037)。为了分析ev来源的Gal1作为MDSC自分泌信号的贡献,我们将来自Gal1处理的免疫抑制骨髓细胞的ev添加到新的bmscs中,观察到CD11b+细胞在M-MDSCs表型细胞上的扩增和更高的VISTA表达。此外,与对照电动汽车相比,这些电动汽车表现出更强的T细胞抑制能力,并且具有不同的蛋白质、代谢和miRNA载货。结论:肿瘤细胞释放的Gal1+ EV与髓系细胞相互作用,增强其免疫抑制特性,包括释放具有自分泌和旁分泌功能的EV。靶向Gal1作为ev介导的肿瘤发展的分子机制可能有助于克服治疗耐药性。向祥,Poliakov A,刘超,刘勇,邓志斌,王军,程忠,沙世伟,王广军,张磊,Grizzle WE, Moble J,张洪柱。肿瘤外泌体诱导髓源性抑制细胞的研究。中华医学杂志,2009;24(4):391 - 391。Rashid MH, Borin TF。mscs来源的外泌体在肿瘤微环境中的免疫抑制作用。2021;45:1171-1181。
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
1466 Galectin-1 orchestrates a hierarchical tumor to stromal extracellular vesicles system that fosters immune suppression

Background

Extracellular vesicles (EVs) suppress effector cells and activate immunosuppressive cells in the ‘escape phase’ of tumor immunoediting. Tumor derived EVs contribute to the differentiation and expansion of immunosuppressive cell subsets. Bone marrow-derived cells (BMDCs) can uptake EVs released by 4T1 -a triple-negative breast cancer cell line- and induce myeloid-derived suppressor cells (MDSCs).1 Immunosuppressive myeloid cells expansion can also be promoted by their own EV in an autocrine manner. Moreover, EVs secreted by immunosuppressive myeloid cells, may inherit their parental functions.2 Nevertheless, the molecular circuits and mediators of tumor microenvironment EV release remain uncertain.

Methods

We generated immunosuppressive myeloid cells by culturing mouse BMDCs with GM-CSF for three days in the presence of Gal1. Additionally, we cultured 4T1 WT and Lgals1 knocked out cells. We purified Small EVs by size exclusion chromatography from conditioned medium of immunosuppressive myeloid cells (control and Gal1-treated) or 4T1 cells (WT or Gal1 KO). We further purified 4T1 EVs with CD63+ beads, a tumoral EV marker. ELISA, flow cytometry, miRNA sequencing, proteomic and metabolomic analyses verified EV identity. We performed functional assays by co-culturing tumoral or myeloid EVs with BMDC, activated T or B cells.

Results

When exposed to EVs from 4T1 cells in vitro, BMDCs switched their differentiation pathway to a M-MDSCs phenotype (CD11b+Ly6G-Ly6Chi) and showed higher immune checkpoint molecules expression, including PD-L1 (p=0.005) and VISTA (p=0.003). However, when exposed to tumoral EVs lacking Gal1, BMDCs reinforced a PMN-MDSCs phenotype (CD11b+Ly6G+Ly6Clo). In contrast with Gal1+ EVs, tumoral EVs lacking Gal1 failed to inhibit CD4+ and CD8+ T and B cell proliferation and activation. Gal1 is both in the cargo and corona of 4T1 EVs as revealed by flow cytometry and ELISA (p=0.0032). Moreover, EV production inhibition with GW4869 in 4T1 cells decreased Gal1 levels secreted (p=0.037). To analyze EV-derived Gal1 contribution as an MDSC autocrine signal, we added EVs from Gal1-treated immunosuppressive myeloid cells to new BMDCs and observed CD11b+ cells expansion and higher VISTA expression on cells with M-MDSCs phenotype. Moreover, these EVs showed greater T cell-suppressive capacity and has different protein, metabolic, and miRNA cargo compared to control EVs.

Conclusions

Here, we propose that Gal1+ EVs released by tumoral cells interact with myeloid cells and potentiate their immunosuppressive properties, including the release of EV with autocrine and paracrine functions. Targeting Gal1 as a molecular mechanism of EV-mediated tumor development may help overcome therapy resistance.

References

Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, Grizzle WE, Moble J, Zhang HG. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124:2621–2633. Rashid MH, Borin TF. Rep. Critical immunosuppressive effect of MDSC-derived exosomes in the tumor microenvironment.2021;45:1171–1181.
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
1455 Characterization of gene expression signatures of tumor immunogenicity and cellular proliferation from murine cancer models grownin vitroandin vivo 1193 Synergistic targeting of multiple activating pathways with natural killer cell engagers 1413 Immunogenicity of SARS-CoV-2 mRNA vaccines in individuals with thymic epithelial tumors 1150 DM005, an EGFR × MET bispecific antibody-drug conjugate, showed robust anti-tumor activity in PDX models 1274 Infliximab for decompensated diabetes following immune checkpoint inhibitor therapy
×
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