髓系细胞中的 ATG16L1 通过减少炎症小体的激活,限制了 ApcMin/+ 小鼠感染产大肠杆菌后大肠肿瘤的生长。

Autophagy Pub Date : 2024-10-01 Epub Date: 2024-06-10 DOI:10.1080/15548627.2024.2359770
Laurène Salesse, Angéline Duval, Pierre Sauvanet, Alison Da Silva, Nicolas Barnich, Catherine Godfraind, Guillaume Dalmasso, Hang Thi Thu Nguyen
{"title":"髓系细胞中的 ATG16L1 通过减少炎症小体的激活,限制了 ApcMin/+ 小鼠感染产大肠杆菌后大肠肿瘤的生长。","authors":"Laurène Salesse, Angéline Duval, Pierre Sauvanet, Alison Da Silva, Nicolas Barnich, Catherine Godfraind, Guillaume Dalmasso, Hang Thi Thu Nguyen","doi":"10.1080/15548627.2024.2359770","DOIUrl":null,"url":null,"abstract":"<p><p><i>Escherichia coli</i> strains producing the genotoxin colibactin, designated as CoPEC (colibactin-producing <i>E. coli</i>), have emerged as an important player in the etiology of colorectal cancer (CRC). Here, we investigated the role of macroautophagy/autophagy in myeloid cells, an important component of the tumor microenvironment, in the tumorigenesis of a susceptible mouse model infected with CoPEC. For that, a preclinical mouse model of CRC, the <i>Apc</i><sup><i>Min/+</i></sup> mice, with <i>Atg16l1</i> deficiency specifically in myeloid cells (<i>Apc</i><sup><i>Min/+</i></sup>/<i>Atg16l1[∆MC]</i>) and the corresponding control mice (<i>Apc</i><sup><i>Min/+</i></sup>), were infected with a clinical CoPEC strain 11G5 or its isogenic mutant 11G5<i>∆clbQ</i> that does not produce colibactin. We showed that myeloid cell-specific <i>Atg16l1</i> deficiency led to an increase in the volume of colonic tumors in <i>Apc</i><sup><i>Min/+</i></sup> mice under infection with 11G5, but not with 11G5<i>∆clbQ</i>. This was accompanied by increased colonocyte proliferation, enhanced inflammasome activation and IL1B/IL-1β secretion, increased neutrophil number and decreased total T cell and cytotoxic CD8<sup>+</sup> T cell numbers in the colonic mucosa and tumors. In bone marrow-derived macrophages (BMDMs), compared to uninfected and 11G5∆<i>clbQ</i>-infected conditions, 11G5 infection increased inflammasome activation and IL1B secretion, and this was further enhanced by autophagy deficiency. These data indicate that ATG16L1 in myeloid cells was necessary to inhibit colonic tumor growth in CoPEC-infected <i>Apc</i><sup><i>Min/+</i></sup> mice <i>via</i> inhibiting colibactin-induced inflammasome activation and modulating immune cell response in the tumor microenvironment. <b>Abbreviation</b>: AOM, azoxymethane; APC, APC regulator of WNT signaling pathway; ATG, autophagy related; <i>Atg16l1[∆MC]</i> mice, mice deficient for <i>Atg16l1</i> specifically in myeloid cells; CASP1, caspase 1; BMDM, bone marrow-derived macrophage; CFU, colony-forming unit; CoPEC, colibactin-producing <i>Escherichia coli</i>; CRC, colorectal cancer; CXCL1/KC, C-X-C motif chemokine ligand 1; ELISA, enzyme-linked immunosorbent assay; IL, interleukin; MC, myeloid cell; MOI, multiplicity of infection; PBS, phosphate-buffered saline; <i>pks</i>, polyketide synthase; qRT-PCR, quantitative real-time reverse-transcription polymerase chain reaction; siRNA, small interfering RNA; TME, tumor microenvironment; TNF/TNF-α, tumor necrosis factor.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11423662/pdf/","citationCount":"0","resultStr":"{\"title\":\"ATG16L1 in myeloid cells limits colorectal tumor growth in <i>Apc<sup>Min/+</sup></i> mice infected with colibactin-producing <i>Escherichia coli</i> via decreasing inflammasome activation.\",\"authors\":\"Laurène Salesse, Angéline Duval, Pierre Sauvanet, Alison Da Silva, Nicolas Barnich, Catherine Godfraind, Guillaume Dalmasso, Hang Thi Thu Nguyen\",\"doi\":\"10.1080/15548627.2024.2359770\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p><i>Escherichia coli</i> strains producing the genotoxin colibactin, designated as CoPEC (colibactin-producing <i>E. coli</i>), have emerged as an important player in the etiology of colorectal cancer (CRC). Here, we investigated the role of macroautophagy/autophagy in myeloid cells, an important component of the tumor microenvironment, in the tumorigenesis of a susceptible mouse model infected with CoPEC. For that, a preclinical mouse model of CRC, the <i>Apc</i><sup><i>Min/+</i></sup> mice, with <i>Atg16l1</i> deficiency specifically in myeloid cells (<i>Apc</i><sup><i>Min/+</i></sup>/<i>Atg16l1[∆MC]</i>) and the corresponding control mice (<i>Apc</i><sup><i>Min/+</i></sup>), were infected with a clinical CoPEC strain 11G5 or its isogenic mutant 11G5<i>∆clbQ</i> that does not produce colibactin. We showed that myeloid cell-specific <i>Atg16l1</i> deficiency led to an increase in the volume of colonic tumors in <i>Apc</i><sup><i>Min/+</i></sup> mice under infection with 11G5, but not with 11G5<i>∆clbQ</i>. This was accompanied by increased colonocyte proliferation, enhanced inflammasome activation and IL1B/IL-1β secretion, increased neutrophil number and decreased total T cell and cytotoxic CD8<sup>+</sup> T cell numbers in the colonic mucosa and tumors. In bone marrow-derived macrophages (BMDMs), compared to uninfected and 11G5∆<i>clbQ</i>-infected conditions, 11G5 infection increased inflammasome activation and IL1B secretion, and this was further enhanced by autophagy deficiency. These data indicate that ATG16L1 in myeloid cells was necessary to inhibit colonic tumor growth in CoPEC-infected <i>Apc</i><sup><i>Min/+</i></sup> mice <i>via</i> inhibiting colibactin-induced inflammasome activation and modulating immune cell response in the tumor microenvironment. <b>Abbreviation</b>: AOM, azoxymethane; APC, APC regulator of WNT signaling pathway; ATG, autophagy related; <i>Atg16l1[∆MC]</i> mice, mice deficient for <i>Atg16l1</i> specifically in myeloid cells; CASP1, caspase 1; BMDM, bone marrow-derived macrophage; CFU, colony-forming unit; CoPEC, colibactin-producing <i>Escherichia coli</i>; CRC, colorectal cancer; CXCL1/KC, C-X-C motif chemokine ligand 1; ELISA, enzyme-linked immunosorbent assay; IL, interleukin; MC, myeloid cell; MOI, multiplicity of infection; PBS, phosphate-buffered saline; <i>pks</i>, polyketide synthase; qRT-PCR, quantitative real-time reverse-transcription polymerase chain reaction; siRNA, small interfering RNA; TME, tumor microenvironment; TNF/TNF-α, tumor necrosis factor.</p>\",\"PeriodicalId\":93893,\"journal\":{\"name\":\"Autophagy\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-10-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11423662/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Autophagy\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1080/15548627.2024.2359770\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/6/10 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Autophagy","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1080/15548627.2024.2359770","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/6/10 0:00:00","PubModel":"Epub","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

大肠杆菌菌株可产生基因毒素大肠杆菌素,被称为 CoPEC(产大肠杆菌素大肠杆菌),已成为结直肠癌(CRC)病因中的一个重要角色。在此,我们研究了肿瘤微环境的重要组成部分--髓系细胞中的大自噬/自噬在感染 CoPEC 的易感小鼠模型的肿瘤发生中的作用。为此,我们用临床CoPEC菌株11G5或其不产生colibactin的同源突变体11G5ΔclbQ感染了CRC临床前小鼠模型ApcMin/+(髓样细胞中特异性缺乏Atg16l1(ApcMin/+/Atg16l1[ΔMC])和相应的对照小鼠(ApcMin/+)。我们发现,髓系细胞特异性 Atg16l1 缺乏会导致 ApcMin/+ 小鼠感染 11G5 后结肠肿瘤体积增大,而感染 11G5∆clbQ 则不会。与此同时,结肠粘膜和肿瘤中的结肠细胞增殖增加,炎性体活化和 IL1B/IL-1β 分泌增强,中性粒细胞数量增加,T 细胞总数和细胞毒性 CD8+ T 细胞数量减少。在骨髓源性巨噬细胞(BMDMs)中,与未感染和11G5∆clbQ感染的情况相比,11G5感染增加了炎性体的激活和IL1B的分泌,而自噬功能的缺乏又进一步增强了炎性体的激活和IL1B的分泌。这些数据表明,髓系细胞中的ATG16L1是抑制CoPEC感染的ApcMin/+小鼠结肠肿瘤生长的必要条件,它通过抑制colibactin诱导的炎症小体活化和调节肿瘤微环境中的免疫细胞反应来实现。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
ATG16L1 in myeloid cells limits colorectal tumor growth in ApcMin/+ mice infected with colibactin-producing Escherichia coli via decreasing inflammasome activation.

Escherichia coli strains producing the genotoxin colibactin, designated as CoPEC (colibactin-producing E. coli), have emerged as an important player in the etiology of colorectal cancer (CRC). Here, we investigated the role of macroautophagy/autophagy in myeloid cells, an important component of the tumor microenvironment, in the tumorigenesis of a susceptible mouse model infected with CoPEC. For that, a preclinical mouse model of CRC, the ApcMin/+ mice, with Atg16l1 deficiency specifically in myeloid cells (ApcMin/+/Atg16l1[∆MC]) and the corresponding control mice (ApcMin/+), were infected with a clinical CoPEC strain 11G5 or its isogenic mutant 11G5∆clbQ that does not produce colibactin. We showed that myeloid cell-specific Atg16l1 deficiency led to an increase in the volume of colonic tumors in ApcMin/+ mice under infection with 11G5, but not with 11G5∆clbQ. This was accompanied by increased colonocyte proliferation, enhanced inflammasome activation and IL1B/IL-1β secretion, increased neutrophil number and decreased total T cell and cytotoxic CD8+ T cell numbers in the colonic mucosa and tumors. In bone marrow-derived macrophages (BMDMs), compared to uninfected and 11G5∆clbQ-infected conditions, 11G5 infection increased inflammasome activation and IL1B secretion, and this was further enhanced by autophagy deficiency. These data indicate that ATG16L1 in myeloid cells was necessary to inhibit colonic tumor growth in CoPEC-infected ApcMin/+ mice via inhibiting colibactin-induced inflammasome activation and modulating immune cell response in the tumor microenvironment. Abbreviation: AOM, azoxymethane; APC, APC regulator of WNT signaling pathway; ATG, autophagy related; Atg16l1[∆MC] mice, mice deficient for Atg16l1 specifically in myeloid cells; CASP1, caspase 1; BMDM, bone marrow-derived macrophage; CFU, colony-forming unit; CoPEC, colibactin-producing Escherichia coli; CRC, colorectal cancer; CXCL1/KC, C-X-C motif chemokine ligand 1; ELISA, enzyme-linked immunosorbent assay; IL, interleukin; MC, myeloid cell; MOI, multiplicity of infection; PBS, phosphate-buffered saline; pks, polyketide synthase; qRT-PCR, quantitative real-time reverse-transcription polymerase chain reaction; siRNA, small interfering RNA; TME, tumor microenvironment; TNF/TNF-α, tumor necrosis factor.

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