File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Importance of antibody in virus infection and vaccine-mediated protection by a latency-deficient recombinant murine γ-herpesvirus-68

TitleImportance of antibody in virus infection and vaccine-mediated protection by a latency-deficient recombinant murine γ-herpesvirus-68
Authors
Issue Date2012
Citation
Journal of Immunology, 2012, v. 188, n. 3, p. 1049-1056 How to Cite?
AbstractThe human γ-herpesviruses EBV and Kaposi's sarcoma-associated herpesvirus establish lifelong latent infections, can reactivate in immunocompromised individuals, and are associated with the development of malignancies. Murine γ-herpesvirus-68 (γHV68), a rodent pathogen related to EBV and Kaposi's sarcoma-associated herpesvirus, provides an important model to dissect mechanisms of immune control and investigate vaccine strategies. Infection of mice with γHV68 elicits robust antiviral immunity, and long-term protection from γHV68 reactivation requires both cellular and humoral immune responses. Vaccination of mice with AC-replication and transcription activator (RTA), a highly lytic latency-null recombinant γHV68, results in complete protection from wild-type γHV68 infection that lasts for at least 10 mo. In this report, we examine the immune correlates of AC-RTA-mediated protection and show that sterilizing immunity requires both T cells and Ab. Importantly, Ab was also critical for mitigating viral infection in the brain, and in the absence of Ab-mediated control, amplification of the AC-RTA virus in the brain resulted in fatality. Our results highlight important considerations in the development of vaccination strategies based on live-attenuated viruses. Copyright © 2012 by The American Association of Immunologists, Inc.
Persistent Identifierhttp://hdl.handle.net/10722/285690
ISSN
2021 Impact Factor: 5.426
2020 SCImago Journal Rankings: 2.737
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorFreeman, Michael L.-
dc.contributor.authorBurkum, Claire E.-
dc.contributor.authorWoodland, David L.-
dc.contributor.authorSun, Ren-
dc.contributor.authorWu, Ting Ting-
dc.contributor.authorBlackman, Marcia A.-
dc.date.accessioned2020-08-18T04:56:23Z-
dc.date.available2020-08-18T04:56:23Z-
dc.date.issued2012-
dc.identifier.citationJournal of Immunology, 2012, v. 188, n. 3, p. 1049-1056-
dc.identifier.issn0022-1767-
dc.identifier.urihttp://hdl.handle.net/10722/285690-
dc.description.abstractThe human γ-herpesviruses EBV and Kaposi's sarcoma-associated herpesvirus establish lifelong latent infections, can reactivate in immunocompromised individuals, and are associated with the development of malignancies. Murine γ-herpesvirus-68 (γHV68), a rodent pathogen related to EBV and Kaposi's sarcoma-associated herpesvirus, provides an important model to dissect mechanisms of immune control and investigate vaccine strategies. Infection of mice with γHV68 elicits robust antiviral immunity, and long-term protection from γHV68 reactivation requires both cellular and humoral immune responses. Vaccination of mice with AC-replication and transcription activator (RTA), a highly lytic latency-null recombinant γHV68, results in complete protection from wild-type γHV68 infection that lasts for at least 10 mo. In this report, we examine the immune correlates of AC-RTA-mediated protection and show that sterilizing immunity requires both T cells and Ab. Importantly, Ab was also critical for mitigating viral infection in the brain, and in the absence of Ab-mediated control, amplification of the AC-RTA virus in the brain resulted in fatality. Our results highlight important considerations in the development of vaccination strategies based on live-attenuated viruses. Copyright © 2012 by The American Association of Immunologists, Inc.-
dc.languageeng-
dc.relation.ispartofJournal of Immunology-
dc.titleImportance of antibody in virus infection and vaccine-mediated protection by a latency-deficient recombinant murine γ-herpesvirus-68-
dc.typeArticle-
dc.description.naturelink_to_OA_fulltext-
dc.identifier.doi10.4049/jimmunol.1102621-
dc.identifier.pmid22198955-
dc.identifier.pmcidPMC3262927-
dc.identifier.scopuseid_2-s2.0-84856587352-
dc.identifier.volume188-
dc.identifier.issue3-
dc.identifier.spage1049-
dc.identifier.epage1056-
dc.identifier.eissn1550-6606-
dc.identifier.isiWOS:000299690200018-
dc.identifier.issnl0022-1767-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats