Cargando…

Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1

The gp41 membrane-proximal external region (MPER) is a target for broadly neutralizing antibody responses against human immunodeficiency virus type 1 (HIV-1). However, replication-defective virus vaccines currently under evaluation in clinical trials do not efficiently elicit MPER-specific antibodie...

Descripción completa

Detalles Bibliográficos
Autores principales: Boehme, Karl W., Ikizler, Mine', Iskarpatyoti, Jason A., Wetzel, J. Denise, Willis, Jordan, Crowe, James E., LaBranche, Celia C., Montefiori, David C., Wilson, Gregory J., Dermody, Terence S.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Microbiology 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4888892/
https://www.ncbi.nlm.nih.gov/pubmed/27303748
http://dx.doi.org/10.1128/mSphere.00086-16
_version_ 1782434919142653952
author Boehme, Karl W.
Ikizler, Mine'
Iskarpatyoti, Jason A.
Wetzel, J. Denise
Willis, Jordan
Crowe, James E.
LaBranche, Celia C.
Montefiori, David C.
Wilson, Gregory J.
Dermody, Terence S.
author_facet Boehme, Karl W.
Ikizler, Mine'
Iskarpatyoti, Jason A.
Wetzel, J. Denise
Willis, Jordan
Crowe, James E.
LaBranche, Celia C.
Montefiori, David C.
Wilson, Gregory J.
Dermody, Terence S.
author_sort Boehme, Karl W.
collection PubMed
description The gp41 membrane-proximal external region (MPER) is a target for broadly neutralizing antibody responses against human immunodeficiency virus type 1 (HIV-1). However, replication-defective virus vaccines currently under evaluation in clinical trials do not efficiently elicit MPER-specific antibodies. Structural modeling suggests that the MPER forms an α-helical coiled coil that is required for function and immunogenicity. To maintain the native MPER conformation, we used reverse genetics to engineer replication-competent reovirus vectors that displayed MPER sequences in the α-helical coiled-coil tail domain of viral attachment protein σ1. Sequences in reovirus strain type 1 Lang (T1L) σ1 were exchanged with sequences encoding HIV-1 strain Ba-L MPER epitope 2F5 or the entire MPER. Individual 2F5 or MPER substitutions were introduced at virion-proximal or virion-distal sites in the σ1 tail. Recombinant reoviruses containing heterologous HIV-1 sequences were viable and produced progeny yields comparable to those with wild-type virus. HIV-1 sequences were retained following 10 serial passages in cell culture, indicating that the substitutions were genetically stable. Recombinant viruses engineered to display the 2F5 epitope or full-length MPER in σ1 were recognized by purified 2F5 antibody. Inoculation of mice with 2F5-containing vectors or rabbits with 2F5- or MPER-containing vectors elicited anti-reovirus antibodies, but HIV-1-specific antibodies were not detected. Together, these findings indicate that heterologous sequences that form α-helices can functionally replace native sequences in the α-helical tail domain of reovirus attachment protein σ1. However, although these vectors retain native antigenicity, they were not immunogenic, illustrating the difficulty of experimentally inducing immune responses to this essential region of HIV-1. IMPORTANCE Vaccines to protect against HIV-1, the causative agent of AIDS, are not approved for use. Antibodies that neutralize genetically diverse strains of HIV-1 bind to discrete regions of the envelope glycoproteins, including the gp41 MPER. We engineered recombinant reoviruses that displayed MPER epitopes in attachment protein σ1 (REO-MPER vectors). The REO-MPER vectors replicated with wild-type efficiency, were genetically stable, and retained native antigenicity. However, we did not detect HIV-1-specific immune responses following inoculation of the REO-MPER vectors into small animals. This work provides proof of principle for engineering reovirus to express antigenic epitopes and illustrates the difficulty in eliciting MPER-specific immune responses.
format Online
Article
Text
id pubmed-4888892
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher American Society for Microbiology
record_format MEDLINE/PubMed
spelling pubmed-48888922016-06-14 Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1 Boehme, Karl W. Ikizler, Mine' Iskarpatyoti, Jason A. Wetzel, J. Denise Willis, Jordan Crowe, James E. LaBranche, Celia C. Montefiori, David C. Wilson, Gregory J. Dermody, Terence S. mSphere Research Article The gp41 membrane-proximal external region (MPER) is a target for broadly neutralizing antibody responses against human immunodeficiency virus type 1 (HIV-1). However, replication-defective virus vaccines currently under evaluation in clinical trials do not efficiently elicit MPER-specific antibodies. Structural modeling suggests that the MPER forms an α-helical coiled coil that is required for function and immunogenicity. To maintain the native MPER conformation, we used reverse genetics to engineer replication-competent reovirus vectors that displayed MPER sequences in the α-helical coiled-coil tail domain of viral attachment protein σ1. Sequences in reovirus strain type 1 Lang (T1L) σ1 were exchanged with sequences encoding HIV-1 strain Ba-L MPER epitope 2F5 or the entire MPER. Individual 2F5 or MPER substitutions were introduced at virion-proximal or virion-distal sites in the σ1 tail. Recombinant reoviruses containing heterologous HIV-1 sequences were viable and produced progeny yields comparable to those with wild-type virus. HIV-1 sequences were retained following 10 serial passages in cell culture, indicating that the substitutions were genetically stable. Recombinant viruses engineered to display the 2F5 epitope or full-length MPER in σ1 were recognized by purified 2F5 antibody. Inoculation of mice with 2F5-containing vectors or rabbits with 2F5- or MPER-containing vectors elicited anti-reovirus antibodies, but HIV-1-specific antibodies were not detected. Together, these findings indicate that heterologous sequences that form α-helices can functionally replace native sequences in the α-helical tail domain of reovirus attachment protein σ1. However, although these vectors retain native antigenicity, they were not immunogenic, illustrating the difficulty of experimentally inducing immune responses to this essential region of HIV-1. IMPORTANCE Vaccines to protect against HIV-1, the causative agent of AIDS, are not approved for use. Antibodies that neutralize genetically diverse strains of HIV-1 bind to discrete regions of the envelope glycoproteins, including the gp41 MPER. We engineered recombinant reoviruses that displayed MPER epitopes in attachment protein σ1 (REO-MPER vectors). The REO-MPER vectors replicated with wild-type efficiency, were genetically stable, and retained native antigenicity. However, we did not detect HIV-1-specific immune responses following inoculation of the REO-MPER vectors into small animals. This work provides proof of principle for engineering reovirus to express antigenic epitopes and illustrates the difficulty in eliciting MPER-specific immune responses. American Society for Microbiology 2016-05-18 /pmc/articles/PMC4888892/ /pubmed/27303748 http://dx.doi.org/10.1128/mSphere.00086-16 Text en Copyright © 2016 Boehme et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license (http://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Boehme, Karl W.
Ikizler, Mine'
Iskarpatyoti, Jason A.
Wetzel, J. Denise
Willis, Jordan
Crowe, James E.
LaBranche, Celia C.
Montefiori, David C.
Wilson, Gregory J.
Dermody, Terence S.
Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1
title Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1
title_full Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1
title_fullStr Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1
title_full_unstemmed Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1
title_short Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1
title_sort engineering recombinant reoviruses to display gp41 membrane-proximal external-region epitopes from hiv-1
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4888892/
https://www.ncbi.nlm.nih.gov/pubmed/27303748
http://dx.doi.org/10.1128/mSphere.00086-16
work_keys_str_mv AT boehmekarlw engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT ikizlermine engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT iskarpatyotijasona engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT wetzeljdenise engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT willisjordan engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT crowejamese engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT labrancheceliac engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT montefioridavidc engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT wilsongregoryj engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1
AT dermodyterences engineeringrecombinantreovirusestodisplaygp41membraneproximalexternalregionepitopesfromhiv1