首页 | 本学科首页   官方微博 | 高级检索  
     


The host inflammatory response contributes to disease severity in Crimean-Congo hemorrhagic fever virus infected mice
Authors:Joseph W. Golden  Xiankun Zeng  Curtis R. Cline  Jeffrey M. Smith  Sharon P. Daye  Brian D. Carey  Candace D. Blancett  Charles J. Shoemaker  Jun Liu  Collin J. Fitzpatrick  Christopher P. Stefan  Aura R. Garrison
Affiliation:1. Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America;2. Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America;3. Diagnostic Services Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America; CDC, UNITED STATES
Abstract:Crimean-Congo hemorrhagic fever virus (CCHFV) is an important human pathogen. In cell culture, CCHFV is sensed by the cytoplasmic RNA sensor retinoic acid-inducible gene I (RIG-I) molecule and its adaptor molecule mitochondrial antiviral signaling (MAVS) protein. MAVS initiates both type I interferon (IFN-I) and proinflammatory responses. Here, we studied the role MAVS plays in CCHFV infection in mice in both the presence and absence of IFN-I activity. MAVS-deficient mice were not susceptible to CCHFV infection when IFN-I signaling was active and showed no signs of disease. When IFN-I signaling was blocked by antibody, MAVS-deficient mice lost significant weight, but were uniformly protected from lethal disease, whereas all control mice succumbed to infection. Cytokine activity in the infected MAVS-deficient mice was markedly blunted. Subsequent investigation revealed that CCHFV infected mice lacking TNF-α receptor signaling (TNFA-R-deficient), but not IL-6 or IL-1 activity, had more limited liver injury and were largely protected from lethal outcomes. Treatment of mice with an anti-TNF-α neutralizing antibody also conferred partial protection in a post-virus exposure setting. Additionally, we found that a disease causing, but non-lethal strain of CCHFV produced more blunted inflammatory cytokine responses compared to a lethal strain in mice. Our work reveals that MAVS activation and cytokine production both contribute to CCHFV pathogenesis, potentially identifying new therapeutic targets to treat this disease.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号