Marburg and Ebola Virus Infections Elicit a Complex, Muted Inflammatory State in Bats

Author:

Jayaprakash Anitha D.1,Ronk Adam J.23ORCID,Prasad Abhishek N.23ORCID,Covington Michael F.4,Stein Kathryn R.5,Schwarz Toni M.6,Hekmaty Saboor5ORCID,Fenton Karla A.37,Geisbert Thomas W.37ORCID,Basler Christopher F.6ORCID,Bukreyev Alexander237,Sachidanandam Ravi5ORCID

Affiliation:

1. Girihlet Inc., Oakland, CA 94609, USA

2. Department of Pathology, the University Texas Medical Branch, Galveston, TX 77555, USA

3. Galveston National Laboratory, the University of Texas Medical Branch, Galveston, TX 77555, USA

4. Amaryllis Nucleics, Oakland, CA 94609, USA

5. Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

6. Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

7. Department Microbiology & Immunology, the University of Texas Medical Branch, Galveston, TX 77555, USA

Abstract

The Marburg and Ebola filoviruses cause a severe, often fatal, disease in humans and nonhuman primates but have only subclinical effects in bats, including Egyptian rousettes, which are a natural reservoir of Marburg virus. A fundamental question is why these viruses are highly pathogenic in humans but fail to cause disease in bats. To address this question, we infected one cohort of Egyptian rousette bats with Marburg virus and another cohort with Ebola virus and harvested multiple tissues for mRNA expression analysis. While virus transcripts were found primarily in the liver, principal component analysis (PCA) revealed coordinated changes across multiple tissues. Gene signatures in kidney and liver pointed at induction of vasodilation, reduction in coagulation, and changes in the regulation of iron metabolism. Signatures of immune response detected in spleen and liver indicated a robust anti-inflammatory state signified by macrophages in the M2 state and an active T cell response. The evolutionary divergence between bats and humans of many responsive genes might provide a framework for understanding the differing outcomes upon infection by filoviruses. In this study, we outline multiple interconnected pathways that respond to infection by MARV and EBOV, providing insights into the complexity of the mechanisms that enable bats to resist the disease caused by filoviral infections. The results have the potential to aid in the development of new strategies to effectively mitigate and treat the disease caused by these viruses in humans.

Funder

Defense Threat Reduction Agency

NIH/NIAID

Publisher

MDPI AG

Subject

Virology,Infectious Diseases

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