Carbonic anhydrases reduce the acidity of the tumor microenvironment, promote immune infiltration, decelerate tumor growth, and improve survival in ErbB2/HER2-enriched breast cancer.

Soojung Lee, Nicolai J Toft, Trine V Axelsen, Maria Sofia Espejo, Tina M Pedersen, Marco Mele, Helene L Pedersen, Eva Balling, Tonje Johansen, Mark Burton, Mads Thomassen, Pernille Vahl, Peer Christiansen, Ebbe Boedtkjer
{"title":"Carbonic anhydrases reduce the acidity of the tumor microenvironment, promote immune infiltration, decelerate tumor growth, and improve survival in ErbB2/HER2-enriched breast cancer.","authors":"Soojung Lee,&nbsp;Nicolai J Toft,&nbsp;Trine V Axelsen,&nbsp;Maria Sofia Espejo,&nbsp;Tina M Pedersen,&nbsp;Marco Mele,&nbsp;Helene L Pedersen,&nbsp;Eva Balling,&nbsp;Tonje Johansen,&nbsp;Mark Burton,&nbsp;Mads Thomassen,&nbsp;Pernille Vahl,&nbsp;Peer Christiansen,&nbsp;Ebbe Boedtkjer","doi":"10.1186/s13058-023-01644-1","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Carbonic anhydrases catalyze CO<sub>2</sub>/HCO<sub>3</sub><sup>-</sup> buffer reactions with implications for effective H<sup>+</sup> mobility, pH dynamics, and cellular acid-base sensing. Yet, the integrated consequences of carbonic anhydrases for cancer and stromal cell functions, their interactions, and patient prognosis are not yet clear.</p><p><strong>Methods: </strong>We combine (a) bioinformatic analyses of human proteomic data and bulk and single-cell transcriptomic data coupled to clinicopathologic and prognostic information; (b) ex vivo experimental studies of gene expression in breast tissue based on quantitative reverse transcription and polymerase chain reactions, intracellular and extracellular pH recordings based on fluorescence confocal microscopy, and immunohistochemical protein identification in human and murine breast cancer biopsies; and (c) in vivo tumor size measurements, pH-sensitive microelectrode recordings, and microdialysis-based metabolite analyses in mice with experimentally induced breast carcinomas.</p><p><strong>Results: </strong>Carbonic anhydrases-particularly the extracellular isoforms CA4, CA6, CA9, CA12, and CA14-undergo potent expression changes during human and murine breast carcinogenesis. In patients with basal-like/triple-negative breast cancer, elevated expression of the extracellular carbonic anhydrases negatively predicts survival, whereas, surprisingly, the extracellular carbonic anhydrases positively predict patient survival in HER2/ErbB2-enriched breast cancer. Carbonic anhydrase inhibition attenuates cellular net acid extrusion and extracellular H<sup>+</sup> elimination from diffusion-restricted to peripheral and well-perfused regions of human and murine breast cancer tissue. Supplied in vivo, the carbonic anhydrase inhibitor acetazolamide acidifies the microenvironment of ErbB2-induced murine breast carcinomas, limits tumor immune infiltration (CD3<sup>+</sup> T cells, CD19<sup>+</sup> B cells, F4/80<sup>+</sup> macrophages), lowers inflammatory cytokine (Il1a, Il1b, Il6) and transcription factor (Nfkb1) expression, and accelerates tumor growth. Supporting the immunomodulatory influences of carbonic anhydrases, patient survival benefits associated with high extracellular carbonic anhydrase expression in HER2-enriched breast carcinomas depend on the tumor inflammatory profile. Acetazolamide lowers lactate levels in breast tissue and blood without influencing breast tumor perfusion, suggesting that carbonic anhydrase inhibition lowers fermentative glycolysis.</p><p><strong>Conclusions: </strong>We conclude that carbonic anhydrases (a) elevate pH in breast carcinomas by accelerating net H<sup>+</sup> elimination from cancer cells and across the interstitial space and (b) raise immune infiltration and inflammation in ErbB2/HER2-driven breast carcinomas, restricting tumor growth and improving patient survival.</p>","PeriodicalId":9283,"journal":{"name":"Breast Cancer Research : BCR","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10127511/pdf/","citationCount":"2","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Breast Cancer Research : BCR","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s13058-023-01644-1","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 2

Abstract

Background: Carbonic anhydrases catalyze CO2/HCO3- buffer reactions with implications for effective H+ mobility, pH dynamics, and cellular acid-base sensing. Yet, the integrated consequences of carbonic anhydrases for cancer and stromal cell functions, their interactions, and patient prognosis are not yet clear.

Methods: We combine (a) bioinformatic analyses of human proteomic data and bulk and single-cell transcriptomic data coupled to clinicopathologic and prognostic information; (b) ex vivo experimental studies of gene expression in breast tissue based on quantitative reverse transcription and polymerase chain reactions, intracellular and extracellular pH recordings based on fluorescence confocal microscopy, and immunohistochemical protein identification in human and murine breast cancer biopsies; and (c) in vivo tumor size measurements, pH-sensitive microelectrode recordings, and microdialysis-based metabolite analyses in mice with experimentally induced breast carcinomas.

Results: Carbonic anhydrases-particularly the extracellular isoforms CA4, CA6, CA9, CA12, and CA14-undergo potent expression changes during human and murine breast carcinogenesis. In patients with basal-like/triple-negative breast cancer, elevated expression of the extracellular carbonic anhydrases negatively predicts survival, whereas, surprisingly, the extracellular carbonic anhydrases positively predict patient survival in HER2/ErbB2-enriched breast cancer. Carbonic anhydrase inhibition attenuates cellular net acid extrusion and extracellular H+ elimination from diffusion-restricted to peripheral and well-perfused regions of human and murine breast cancer tissue. Supplied in vivo, the carbonic anhydrase inhibitor acetazolamide acidifies the microenvironment of ErbB2-induced murine breast carcinomas, limits tumor immune infiltration (CD3+ T cells, CD19+ B cells, F4/80+ macrophages), lowers inflammatory cytokine (Il1a, Il1b, Il6) and transcription factor (Nfkb1) expression, and accelerates tumor growth. Supporting the immunomodulatory influences of carbonic anhydrases, patient survival benefits associated with high extracellular carbonic anhydrase expression in HER2-enriched breast carcinomas depend on the tumor inflammatory profile. Acetazolamide lowers lactate levels in breast tissue and blood without influencing breast tumor perfusion, suggesting that carbonic anhydrase inhibition lowers fermentative glycolysis.

Conclusions: We conclude that carbonic anhydrases (a) elevate pH in breast carcinomas by accelerating net H+ elimination from cancer cells and across the interstitial space and (b) raise immune infiltration and inflammation in ErbB2/HER2-driven breast carcinomas, restricting tumor growth and improving patient survival.

Abstract Image

Abstract Image

Abstract Image

在富含ErbB2/ her2的乳腺癌中,碳酸酐酶降低肿瘤微环境的酸度,促进免疫浸润,减缓肿瘤生长,提高生存率。
背景:碳酸酐酶催化CO2/HCO3-缓冲反应,具有有效的H+迁移率,pH动力学和细胞酸碱感应的意义。然而,碳酸酐酶对癌症和基质细胞功能的综合影响、它们的相互作用和患者预后尚不清楚。方法:我们结合了(a)人类蛋白质组学数据的生物信息学分析以及与临床病理和预后信息相结合的大量和单细胞转录组学数据;(b)基于定量逆转录和聚合酶链反应的乳腺组织基因表达的离体实验研究,基于荧光共聚焦显微镜的细胞内和细胞外pH记录,以及人类和小鼠乳腺癌活检的免疫组织化学蛋白鉴定;(c)实验诱导乳腺癌小鼠体内肿瘤大小测量、ph敏感微电极记录和基于微透析的代谢物分析。结果:碳酸酐酶,特别是细胞外异构体CA4、CA6、CA9、CA12和ca14,在人类和小鼠乳腺癌发生过程中发生了显著的表达变化。在基底样/三阴性乳腺癌患者中,细胞外碳酸酐酶的表达升高负预测生存率,然而,令人惊讶的是,细胞外碳酸酐酶正预测HER2/ erbb2富集乳腺癌患者的生存率。碳酸酐酶抑制抑制细胞净酸挤压和细胞外H+消除扩散,限制在人类和小鼠乳腺癌组织的外周和灌注良好的区域。在体内,碳酸酸酶抑制剂乙酰唑胺酸化erbb2诱导的小鼠乳腺癌微环境,限制肿瘤免疫浸润(CD3+ T细胞,CD19+ B细胞,F4/80+巨噬细胞),降低炎症细胞因子(Il1a, Il1b, Il6)和转录因子(Nfkb1)的表达,并加速肿瘤生长。支持碳酸酐酶的免疫调节作用,在her2富集的乳腺癌中,与细胞外碳酸酐酶高表达相关的患者生存益处取决于肿瘤炎症谱。乙酰唑胺降低乳腺组织和血液中的乳酸水平,但不影响乳腺肿瘤灌注,提示碳酸酐酶抑制可降低发酵糖酵解。结论:我们得出结论,碳酸酐酶(a)通过加速癌细胞和间质间隙的净H+消除来提高乳腺癌中的pH值,(b)在ErbB2/ her2驱动的乳腺癌中提高免疫浸润和炎症,限制肿瘤生长并提高患者生存率。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
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