Cargando…

Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites

Plasmodium falciparum malaria is a deadly pathogen. The invasion of red blood cells (RBCs) by merozoites is a target for vaccine development. Although anti-merozoite antibodies can block invasion in vitro, there is no efficacy in vivo. To explain this discrepancy we hypothesized that complement acti...

Descripción completa

Detalles Bibliográficos
Autores principales: Biryukov, Sergei, Angov, Evelina, Landmesser, Mary E., Spring, Michele D., Ockenhouse, Christian F., Stoute, José A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4972486/
https://www.ncbi.nlm.nih.gov/pubmed/27333049
http://dx.doi.org/10.1016/j.ebiom.2016.05.015
_version_ 1782446252921716736
author Biryukov, Sergei
Angov, Evelina
Landmesser, Mary E.
Spring, Michele D.
Ockenhouse, Christian F.
Stoute, José A.
author_facet Biryukov, Sergei
Angov, Evelina
Landmesser, Mary E.
Spring, Michele D.
Ockenhouse, Christian F.
Stoute, José A.
author_sort Biryukov, Sergei
collection PubMed
description Plasmodium falciparum malaria is a deadly pathogen. The invasion of red blood cells (RBCs) by merozoites is a target for vaccine development. Although anti-merozoite antibodies can block invasion in vitro, there is no efficacy in vivo. To explain this discrepancy we hypothesized that complement activation could enhance RBC invasion by binding to the complement receptor 1 (CR1). Here we show that a monoclonal antibody directed against the merozoite and human polyclonal IgG from merozoite vaccine recipients enhanced RBC invasion in a complement-dependent manner and that soluble CR1 inhibited this enhancement. Sialic acid-independent strains, that presumably are able to bind to CR1 via a native ligand, showed less complement-dependent enhancement of RBC invasion than sialic acid-dependent strains that do not utilize native CR1 ligands. Confocal fluorescent microscopy revealed that complement-dependent invasion resulted in aggregation of CR1 at the RBC surface in contact with the merozoite. Finally, total anti-P. berghei IgG enhanced parasite growth and C3 deficiency decreased parasite growth in mice. These results demonstrate, contrary to current views, that complement activation in conjunction with antibodies can paradoxically aid parasites invade RBCs and should be considered in future design and testing of merozoite vaccines.
format Online
Article
Text
id pubmed-4972486
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-49724862016-08-10 Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites Biryukov, Sergei Angov, Evelina Landmesser, Mary E. Spring, Michele D. Ockenhouse, Christian F. Stoute, José A. EBioMedicine Research Paper Plasmodium falciparum malaria is a deadly pathogen. The invasion of red blood cells (RBCs) by merozoites is a target for vaccine development. Although anti-merozoite antibodies can block invasion in vitro, there is no efficacy in vivo. To explain this discrepancy we hypothesized that complement activation could enhance RBC invasion by binding to the complement receptor 1 (CR1). Here we show that a monoclonal antibody directed against the merozoite and human polyclonal IgG from merozoite vaccine recipients enhanced RBC invasion in a complement-dependent manner and that soluble CR1 inhibited this enhancement. Sialic acid-independent strains, that presumably are able to bind to CR1 via a native ligand, showed less complement-dependent enhancement of RBC invasion than sialic acid-dependent strains that do not utilize native CR1 ligands. Confocal fluorescent microscopy revealed that complement-dependent invasion resulted in aggregation of CR1 at the RBC surface in contact with the merozoite. Finally, total anti-P. berghei IgG enhanced parasite growth and C3 deficiency decreased parasite growth in mice. These results demonstrate, contrary to current views, that complement activation in conjunction with antibodies can paradoxically aid parasites invade RBCs and should be considered in future design and testing of merozoite vaccines. Elsevier 2016-05-14 /pmc/articles/PMC4972486/ /pubmed/27333049 http://dx.doi.org/10.1016/j.ebiom.2016.05.015 Text en © 2016 The Authors http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Research Paper
Biryukov, Sergei
Angov, Evelina
Landmesser, Mary E.
Spring, Michele D.
Ockenhouse, Christian F.
Stoute, José A.
Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites
title Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites
title_full Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites
title_fullStr Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites
title_full_unstemmed Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites
title_short Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites
title_sort complement and antibody-mediated enhancement of red blood cell invasion and growth of malaria parasites
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4972486/
https://www.ncbi.nlm.nih.gov/pubmed/27333049
http://dx.doi.org/10.1016/j.ebiom.2016.05.015
work_keys_str_mv AT biryukovsergei complementandantibodymediatedenhancementofredbloodcellinvasionandgrowthofmalariaparasites
AT angovevelina complementandantibodymediatedenhancementofredbloodcellinvasionandgrowthofmalariaparasites
AT landmessermarye complementandantibodymediatedenhancementofredbloodcellinvasionandgrowthofmalariaparasites
AT springmicheled complementandantibodymediatedenhancementofredbloodcellinvasionandgrowthofmalariaparasites
AT ockenhousechristianf complementandantibodymediatedenhancementofredbloodcellinvasionandgrowthofmalariaparasites
AT stoutejosea complementandantibodymediatedenhancementofredbloodcellinvasionandgrowthofmalariaparasites