84
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lectin-Dependent Enhancement of Ebola Virus Infection via Soluble and Transmembrane C-type Lectin Receptors

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Mannose-binding lectin (MBL) is a key soluble effector of the innate immune system that recognizes pathogen-specific surface glycans. Surprisingly, low-producing MBL genetic variants that may predispose children and immunocompromised individuals to infectious diseases are more common than would be expected in human populations. Since certain immune defense molecules, such as immunoglobulins, can be exploited by invasive pathogens, we hypothesized that MBL might also enhance infections in some circumstances. Consequently, the low and intermediate MBL levels commonly found in human populations might be the result of balancing selection. Using model infection systems with pseudotyped and authentic glycosylated viruses, we demonstrated that MBL indeed enhances infection of Ebola, Hendra, Nipah and West Nile viruses in low complement conditions. Mechanistic studies with Ebola virus (EBOV) glycoprotein pseudotyped lentiviruses confirmed that MBL binds to N-linked glycan epitopes on viral surfaces in a specific manner via the MBL carbohydrate recognition domain, which is necessary for enhanced infection. MBL mediates lipid-raft-dependent macropinocytosis of EBOV via a pathway that appears to require less actin or early endosomal processing compared with the filovirus canonical endocytic pathway. Using a validated RNA interference screen, we identified C1QBP (gC1qR) as a candidate surface receptor that mediates MBL-dependent enhancement of EBOV infection. We also identified dectin-2 (CLEC6A) as a potentially novel candidate attachment factor for EBOV. Our findings support the concept of an innate immune haplotype that represents critical interactions between MBL and complement component C4 genes and that may modify susceptibility or resistance to certain glycosylated pathogens. Therefore, higher levels of native or exogenous MBL could be deleterious in the setting of relative hypocomplementemia which can occur genetically or because of immunodepletion during active infections. Our findings confirm our hypothesis that the pressure of infectious diseases may have contributed in part to evolutionary selection of MBL mutant haplotypes.

          Related collections

          Most cited references80

          • Record: found
          • Abstract: found
          • Article: not found

          Virus entry by endocytosis.

          Although viruses are simple in structure and composition, their interactions with host cells are complex. Merely to gain entry, animal viruses make use of a repertoire of cellular processes that involve hundreds of cellular proteins. Although some viruses have the capacity to penetrate into the cytosol directly through the plasma membrane, most depend on endocytic uptake, vesicular transport through the cytoplasm, and delivery to endosomes and other intracellular organelles. The internalization may involve clathrin-mediated endocytosis (CME), macropinocytosis, caveolar/lipid raft-mediated endocytosis, or a variety of other still poorly characterized mechanisms. This review focuses on the cell biology of virus entry and the different strategies and endocytic mechanisms used by animal viruses.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections.

            Susceptibility to tuberculosis is historically ascribed to an inadequate immune response that fails to control infecting mycobacteria. In zebrafish, we find that susceptibility to Mycobacterium marinum can result from either inadequate or excessive acute inflammation. Modulation of the leukotriene A(4) hydrolase (LTA4H) locus, which controls the balance of pro- and anti-inflammatory eicosanoids, reveals two distinct molecular routes to mycobacterial susceptibility converging on dysregulated TNF levels: inadequate inflammation caused by excess lipoxins and hyperinflammation driven by excess leukotriene B(4). We identify therapies that specifically target each of these extremes. In humans, we identify a single nucleotide polymorphism in the LTA4H promoter that regulates its transcriptional activity. In tuberculous meningitis, the polymorphism is associated with inflammatory cell recruitment, patient survival and response to adjunctive anti-inflammatory therapy. Together, our findings suggest that host-directed therapies tailored to patient LTA4H genotypes may counter detrimental effects of either extreme of inflammation. Copyright © 2012 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Role of endosomal cathepsins in entry mediated by the Ebola virus glycoprotein.

              Using chemical inhibitors and small interfering RNA (siRNA), we have confirmed roles for cathepsin B (CatB) and cathepsin L (CatL) in Ebola virus glycoprotein (GP)-mediated infection. Treatment of Ebola virus GP pseudovirions with CatB and CatL converts GP1 from a 130-kDa to a 19-kDa species. Virus with 19-kDa GP1 displays significantly enhanced infection and is largely resistant to the effects of the CatB inhibitor and siRNA, but it still requires a low-pH-dependent endosomal/lysosomal function. These and other results support a model in which CatB and CatL prime GP by generating a 19-kDa intermediate that can be acted upon by an as yet unidentified endosomal/lysosomal enzyme to trigger fusion.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS One
                PLoS ONE
                plos
                plosone
                PLoS ONE
                Public Library of Science (San Francisco, USA )
                1932-6203
                2013
                2 April 2013
                3 April 2013
                : 8
                : 4
                : e60838
                Affiliations
                [1 ]Programs of Developmental Immunology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
                [2 ]Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
                [3 ]Department of Immunology and Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
                [4 ]Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, Victoria, Australia
                [5 ]Australian Animal Health Laboratory, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Livestock Industries, Geelong, Victoria, Australia
                [6 ]Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
                [7 ]New England Primate Research Center, Southborough, Massachusetts, United States of America
                [8 ]CETRI, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
                [9 ]Harvard Medical School, Boston, Massachusetts, United States of America
                Metabiota, United States of America
                Author notes

                Competing Interests: The authors have declared that no competing interests exist.

                Conceived and designed the experiments: CL BAM ABE GTS GGO EVS ICM. Performed the experiments: MB MK CL LC LMY CS A. Sarraju A. Sokolovska MRZ BAM AO IH ICM. Analyzed the data: EVS ICM. Contributed reagents/materials/analysis tools: DPE DNK MF KT LS GLS GTS GGO ICM. Wrote the paper: MB MK EVS ICM.

                [¤a]

                Current address: Alpert Medical School of Brown University, Providence, Rhode Island, United States of America

                [¤b]

                Current address: Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California, United States of America

                [¤c]

                Current address: Department of Infectious Diseases, The Scripps Research Institute Florida Campus, Jupiter, Florida, United States of America

                Article
                PONE-D-11-10768
                10.1371/journal.pone.0060838
                3614905
                23573288
                9d8da5ba-d114-44da-9266-7de7c55ce1e5
                Copyright @ 2013

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 14 June 2011
                : 5 March 2013
                Page count
                Pages: 19
                Funding
                This work was supported by National Institutes of Health Grant U01 AI070330 (to EVS) and by United States Department of Defense, Defense Threat Reduction Agency Biological Therapeutics Program Grants 4.10006 and 4.10007 (to GGO). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology
                Anatomy and Physiology
                Immune Physiology
                Complement System
                Immunology
                Immune System
                Complement System
                Microbiology
                Immunity
                Innate Immunity
                Virology
                Viral Transmission and Infection
                Coreceptors
                Medicine
                Clinical Immunology
                Immunity
                Innate Immunity
                Immunomodulation
                Infectious Diseases
                Viral Diseases
                Ebola Hemorrhagic Fever
                West Nile fever
                Zoonoses
                Hendra Virus
                Nipah Fever

                Uncategorized
                Uncategorized

                Comments

                Comment on this article