TNFα and IFNγ cooperate for efficient pro- to anti-inflammatory transition of macrophages during muscle regeneration

Author:

Babaeijandaghi Farshad1ORCID,Paiero Adrianna12,Long Reece13,Tung Lin Wei1,Smith Shannon Percival1,Cheng Ryan1ORCID,Smandych Joshua1ORCID,Kajabadi Nasim1,Chang Chih-Kai1,Ghassemi Amirhossein1,Kennedy William D. M.14,Soliman Hesham156,Schutz Peter W.7,Rossi Fabio M. V.1ORCID

Affiliation:

1. Biomedical Research Centre, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada

2. Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada

3. Department of Integrative Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada

4. Department of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada

5. Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt

6. Aspect Biosystems, Vancouver, BC, V6P 6P2, Canada

7. Department of Pathology, University of British Columbia and Vancouver General Hospital, Vancouver, BC, V6T 2B5, Canada

Abstract

IFNγ is traditionally known as a proinflammatory cytokine with diverse roles in antimicrobial and antitumor immunity. Yet, findings regarding its sources and functions during the regeneration process following a sterile injury are conflicting. Here, we show that natural killer (NK) cells are the main source of IFNγ in regenerating muscle. Beyond this cell population, IFNγ production is limited to a small population of T cells. We further show that NK cells do not play a major role in muscle regeneration following an acute injury or in dystrophic mice. Surprisingly, the absence of IFNγ per se also has no effect on muscle regeneration following an acute injury. However, the role of IFNγ is partially unmasked when TNFα is also neutralized, suggesting a compensatory mechanism. Using transgenic mice, we showed that conditional inhibition of IFNGR1 signaling in muscle stem cells or fibro-adipogenic progenitors does not play a major role in muscle regeneration. In contrast to common belief, we found that IFNγ is not present in the early inflammatory phase of the regeneration process but rather peaks when macrophages are acquiring an anti-inflammatory phenotype. Further transcriptomic analysis suggests that IFNγ cooperates with TNFα to regulate the transition of macrophages from pro- to anti-inflammatory states. The absence of the cooperative effect of these cytokines on macrophages, however, does not result in significant regeneration impairment likely due to the presence of other compensatory mechanisms. Our findings support the arising view of IFNγ as a pleiotropic inflammatory regulator rather than an inducer of the inflammatory response.

Funder

Gouvernement du Canada | Canadian Institutes of Health Research

Publisher

Proceedings of the National Academy of Sciences

Subject

Multidisciplinary

Cited by 6 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3