Your browser doesn't support javascript.
loading
Aggregate complexes of HIV-1 induced by multimeric antibodies.
Stieh, Daniel J; King, Deborah F; Klein, Katja; Liu, Pinghuang; Shen, Xiaoying; Hwang, Kwan Ki; Ferrari, Guido; Montefiori, David C; Haynes, Barton; Pitisuttithum, Punnee; Kaewkungwal, Jaranit; Nitayaphan, Sorachai; Rerks-Ngarm, Supachai; Michael, Nelson L; Robb, Merlin L; Kim, Jerome H; Denny, Thomas N; Tomaras, Georgia D; Shattock, Robin J.
  • Stieh DJ; Center for Infection, Department of Cellular and Molecular Medicine, St George's, University of London, London, SW17 0RE, UK. dstieh@northwestern.edu.
  • King DF; Current address: Department of Cellular and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA. dstieh@northwestern.edu.
  • Klein K; Mucosal Infection & Immunity Group, Section of Infectious Diseases, Imperial College London, St Mary's Campus, London, W2 1PG, UK. d.king@imperial.ac.uk.
  • Liu P; Mucosal Infection & Immunity Group, Section of Infectious Diseases, Imperial College London, St Mary's Campus, London, W2 1PG, UK. k.klein11@imperial.ac.uk.
  • Shen X; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. ping.liu@duke.edu.
  • Hwang KK; Current address: Division of Swine Infectious Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China. ping.liu@duke.edu.
  • Ferrari G; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. sxshen@duke.edu.
  • Montefiori DC; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. kkhwang@duke.edu.
  • Haynes B; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. gflmp@duke.edu.
  • Pitisuttithum P; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. id.montefiori@duke.edu.
  • Kaewkungwal J; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. hayne002@mc.duke.edu.
  • Nitayaphan S; Faculty of Tropical Medicine, Mahidol, Thailand. punnee.pit@mahidol.ac.th.
  • Rerks-Ngarm S; Faculty of Tropical Medicine, Mahidol, Thailand. tmjkk@mahidol.ac.th.
  • Michael NL; Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand. Sorachain@afrims.org.
  • Robb ML; Ministry of Public Health, Bangkok, Thailand. supachai@health.moph.go.th.
  • Kim JH; Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America. nmichael@hivresearch.org.
  • Denny TN; Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America. mrobb@hivresearch.org.
  • Tomaras GD; Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America. jkim@hivresearch.org.
  • Shattock RJ; Duke Human Vaccine Center, Duke University Medical Center, Durham, NC, 27710, USA. thomas.denny@duke.edu.
Retrovirology ; 11: 78, 2014 Oct 02.
Article en En | MEDLINE | ID: mdl-25274446
ABSTRACT

BACKGROUND:

Antibody mediated viral aggregation may impede viral transfer across mucosal surfaces by hindering viral movement in mucus, preventing transcytosis, or reducing inter-cellular penetration of epithelia thereby limiting access to susceptible mucosal CD4 T cells and dendritic cells. These functions may work together to provide effective immune exclusion of virus from mucosal tissue; however little is known about the antibody characteristics required to induce HIV aggregation. Such knowledge may be critical to the design of successful immunization strategies to facilitate viral immune exclusion at the mucosal portals of entry.

RESULTS:

The potential of neutralizing and non-neutralizing IgG and IgA monoclonals (mAbs) to induce HIV-1 aggregation was assessed by Dynamic light scattering (DLS). Although neutralizing and non-neutralizing IgG mAbs and polyclonal HIV-Ig efficiently aggregated soluble Env trimers, they were not capable of forming viral aggregates. In contrast, dimeric (but not monomeric) IgA mAbs induced stable viral aggregate populations that could be separated from uncomplexed virions. Epitope specificity influenced both the degree of aggregation and formation of higher order complexes by dIgA. IgA purified from serum of uninfected RV144 vaccine trial responders were able to efficiently opsonize viral particles in the absence of significant aggregation, reflective of monomeric IgA.

CONCLUSIONS:

These results collectively demonstrate that dIgA is capable of forming stable viral aggregates providing a plausible basis for testing the effectiveness of aggregation as a potential protection mechanism at the mucosal portals of viral entry.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Anticuerpos Anti-VIH / VIH-1 Límite: Humans Idioma: En Año: 2014 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Anticuerpos Anti-VIH / VIH-1 Límite: Humans Idioma: En Año: 2014 Tipo del documento: Article