TREM-1-accentuated lung injury via miR-155 is inhibited by LP17 nanomedicine

Author:

Yuan Zhihong12,Syed Mansoor3,Panchal Dipti4,Joo Myungsoo5,Bedi Chetna1,Lim Sokbee6,Onyuksel Hayat6,Rubinstein Israel47,Colonna Marco8,Sadikot Ruxana T.12

Affiliation:

1. Department of Veterans Affairs, Atlanta Veterans Affairs Medical Center, Decatur, Georgia;

2. Division of Pulmonary and Critical Care Medicine, Emory University, Atlanta, Georgia;

3. Division of Pulmonary and Critical Medicine, Yale University, New Haven, Connecticut;

4. Division of Pulmonary and Critical Care Medicine, University of Illinois at Chicago, Chicago, Illinois;

5. Department of Immunology, Pusan University, Yangsan, Korea;

6. School of Pharmacy, University of Illinois at Chicago, Chicago, Illinois;

7. Department of Veterans Affairs, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; and

8. Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri

Abstract

Triggering receptors expressed on myeloid cell-1 (TREM-1) is a superimmunoglobulin receptor expressed on myeloid cells. Synergy between TREM-1 and Toll-like receptor amplifies the inflammatory response; however, the mechanisms by which TREM-1 accentuates inflammation are not fully understood. In this study, we investigated the role of TREM-1 in a model of LPS-induced lung injury and neutrophilic inflammation. We show that TREM-1 is induced in lungs of mice with LPS-induced acute neutrophilic inflammation. TREM-1 knockout mice showed an improved survival after lethal doses of LPS with an attenuated inflammatory response in the lungs. Deletion of TREM-1 gene resulted in significantly reduced neutrophils and proinflammatory cytokines and chemokines, particularly IL-1β, TNF-α, and IL-6. Physiologically deletion of TREM-1 conferred an immunometabolic advantage with low oxygen consumption rate (OCR) sparing the respiratory capacity of macrophages challenged with LPS. Furthermore, we show that TREM-1 deletion results in significant attenuation of expression of miR-155 in macrophages and lungs of mice treated with LPS. Experiments with antagomir-155 confirmed that TREM-1-mediated changes were indeed dependent on miR-155 and are mediated by downregulation of suppressor of cytokine signaling-1 (SOCS-1) a key miR-155 target. These data for the first time show that TREM-1 accentuates inflammatory response by inducing the expression of miR-155 in macrophages and suggest a novel mechanism by which TREM-1 signaling contributes to lung injury. Inhibition of TREM-1 using a nanomicellar approach resulted in ablation of neutrophilic inflammation suggesting that TREM-1 inhibition is a potential therapeutic target for neutrophilic lung inflammation and acute respiratory distress syndrome (ARDS).

Funder

Center for Integrated Healthcare, U.S. Department of Veterans Affairs (VISN 2 Center for Integrated Healthcare)

Publisher

American Physiological Society

Subject

Cell Biology,Physiology (medical),Pulmonary and Respiratory Medicine,Physiology

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