1
|
Bissa M, Forlani G, Zanotto C, Tosi G, De Giuli Morghen C, Accolla RS, Radaelli A. Fowlpoxvirus recombinants coding for the CIITA gene increase the expression of endogenous MHC-II and Fowlpox Gag/Pro and Env SIV transgenes. PLoS One 2018; 13:e0190869. [PMID: 29385169 PMCID: PMC5791965 DOI: 10.1371/journal.pone.0190869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/21/2017] [Indexed: 01/12/2023] Open
Abstract
A complete eradication of an HIV infection has never been achieved by vaccination and the search for new immunogens that can induce long-lasting protective responses is ongoing. Avipoxvirus recombinants are host-restricted for replication to avian species and they do not have the undesired side effects induced by vaccinia recombinants. In particular, Fowlpox (FP) recombinants can express transgenes over long periods and can induce protective immunity in mammals, mainly due to CD4-dependent CD8+ T cells. In this context, the class II transactivator (CIITA) has a pivotal role in triggering the adaptive immune response through induction of the expression of class-II major histocompatibility complex molecule (MHC-II), that can present antigens to CD4+ T helper cells. Here, we report on construction of novel FPgp and FPenv recombinants that express the highly immunogenic SIV Gag-pro and Env structural antigens. Several FP-based recombinants, with single or dual genes, were also developed that express CIITA, driven from H6 or SP promoters. These recombinants were used to infect CEF and Vero cells in vitro and determine transgene expression, which was evaluated by real-time PCR and Western blotting. Subcellular localisation of the different proteins was evaluated by confocal microscopy, whereas HLA-DR or MHC-II expression was measured by flow cytometry. Fowlpox recombinants were also used to infect syngeneic T/SA tumour cells, then injected into Balb/c mice to elicit MHC-II immune response and define the presentation of the SIV transgene products in the presence or absence of FPCIITA. Antibodies to Env were measured by ELISA. Our data show that the H6 promoter was more efficient than SP to drive CIITA expression and that CIITA can enhance the levels of the gag/pro and env gene products only when infection is performed by FP single recombinants. Also, CIITA expression is higher when carried by FP single recombinants than when combined with FPgp or FPenv constructs and can induce HLA-DR cell surface expression. However, in-vivo experiments did not show any significant increase in the humoral response. As CIITA already proved to elicit immunogenicity by improving antigen presentation, further in-vivo experiments should be performed to increase the immune responses. The use of prime/boost immunisation protocols and the oral administration route of the recombinants may enhance the immunogenicity of Env peptides presented by MHC-II and provide CD4+ T-cell stimulation.
Collapse
Affiliation(s)
- Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, Milan, Italy
| | - Greta Forlani
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli 32, Milan, Italy
| | - Giovanna Tosi
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Carlo De Giuli Morghen
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli 32, Milan, Italy
- Catholic University “Our Lady of Good Counsel”, Rr. Dritan Hoxha, Tirana, Albania
| | - Roberto S. Accolla
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, Milan, Italy
- CNR Institute of Neurosciences, Cellular and Molecular Pharmacology Section, University of Milan, via Vanvitelli 32, Milan, Italy
- * E-mail:
| |
Collapse
|
2
|
Nomenclature report on the major histocompatibility complex genes and alleles of Great Ape, Old and New World monkey species. Immunogenetics 2012; 64:615-31. [PMID: 22526602 DOI: 10.1007/s00251-012-0617-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 03/27/2012] [Indexed: 12/24/2022]
Abstract
The major histocompatibility complex (MHC) plays a central role in the adaptive immune response. The MHC region is characterised by a high gene density, and most of these genes display considerable polymorphism. Next to humans, non-human primates (NHP) are well studied for their MHC. The present nomenclature report provides the scientific community with the latest nomenclature guidelines/rules and current implemented nomenclature revisions for Great Ape, Old and New World monkey species. All the currently published MHC data for the different Great Ape, Old and New World monkey species are archived at the Immuno Polymorphism Database (IPD)-MHC NHP database. The curators of the IPD-MHC NHP database are, in addition, responsible for providing official designations for newly detected polymorphisms.
Collapse
|
3
|
Functional analysis of frequently expressed Chinese rhesus macaque MHC class I molecules Mamu-A1*02601 and Mamu-B*08301 reveals HLA-A2 and HLA-A3 supertypic specificities. Immunogenetics 2011; 63:275-90. [PMID: 21274527 PMCID: PMC3068250 DOI: 10.1007/s00251-010-0502-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 12/07/2010] [Indexed: 01/15/2023]
Abstract
The Simian immunodeficiency virus (SIV)-infected Indian rhesus macaque (Macaca mulatta) is the most established model of HIV infection and AIDS-related research, despite the potential that macaques of Chinese origin is a more relevant model. Ongoing efforts to further characterize the Chinese rhesus macaques' major histocompatibility complex (MHC) for composition and function should facilitate greater utilization of the species. Previous studies have demonstrated that Chinese-origin M. mulatta (Mamu) class I alleles are more polymorphic than their Indian counterparts, perhaps inferring a model more representative of human MHC, human leukocyte antigen (HLA). Furthermore, the Chinese rhesus macaque class I allele Mamu-A1*02201, the most frequent allele thus far identified, has recently been characterized and shown to be an HLA-B7 supertype analog, the most frequent supertype in human populations. In this study, we have characterized two additional alleles expressed with high frequency in Chinese rhesus macaques, Mamu-A1*02601 and Mamu-B*08301. Upon the development of MHC-peptide-binding assays and definition of their associated motifs, we reveal that these Mamu alleles share peptide-binding characteristics with the HLA-A2 and HLA-A3 supertypes, respectively, the next most frequent human supertypes after HLA-B7. These data suggest that Chinese rhesus macaques may indeed be a more representative model of HLA gene diversity and function as compared to the species of Indian origin and therefore a better model for investigating human immune responses.
Collapse
|
4
|
Transcriptionally abundant major histocompatibility complex class I alleles are fundamental to nonhuman primate simian immunodeficiency virus-specific CD8+ T cell responses. J Virol 2011; 85:3250-61. [PMID: 21270169 DOI: 10.1128/jvi.02355-10] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Simian immunodeficiency virus (SIV)-infected macaques are the preferred animal model for human immunodeficiency virus (HIV) vaccines that elicit CD8(+) T cell responses. Unlike humans, whose CD8(+) T cell responses are restricted by a maximum of six HLA class I alleles, macaques express up to 20 distinct major histocompatibility complex class I (MHC-I) sequences. Interestingly, only a subset of macaque MHC-I sequences are transcriptionally abundant in peripheral blood lymphocytes. We hypothesized that highly transcribed MHC-I sequences are principally responsible for restricting SIV-specific CD8(+) T cell responses. To examine this hypothesis, we measured SIV-specific CD8(+) T cell responses in MHC-I homozygous Mauritian cynomolgus macaques. Each of eight CD8(+) T cell responses defined by full-proteome gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assay were restricted by four of the five transcripts that are transcriptionally abundant (>1% of total MHC-I transcripts in peripheral blood lymphocytes). The five transcriptionally rare transcripts shared by these animals did not restrict any detectable CD8(+) T cell responses. Further, seven CD8(+) T cell responses were defined by identifying peptide binding motifs of the three most frequent MHC-I transcripts on the M3 haplotype. Combined, these results suggest that transcriptionally abundant MHC-I transcripts are principally responsible for restricting SIV-specific CD8(+) T cell responses. Thus, only a subset of the thousands of known MHC-I alleles in macaques should be prioritized for CD8(+) T cell epitope characterization.
Collapse
|
5
|
Solomon C, Southwood S, Hoof I, Rudersdorf R, Peters B, Sidney J, Pinilla C, Marcondes MCG, Ling B, Marx P, Sette A, Mothé BR. The most common Chinese rhesus macaque MHC class I molecule shares peptide binding repertoire with the HLA-B7 supertype. Immunogenetics 2010; 62:451-64. [PMID: 20480161 PMCID: PMC2890073 DOI: 10.1007/s00251-010-0450-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 04/19/2010] [Indexed: 01/30/2023]
Abstract
Of the two rhesus macaque subspecies used for AIDS studies, the Simian immunodeficiency virus-infected Indian rhesus macaque (Macaca mulatta) is the most established model of HIV infection, providing both insight into pathogenesis and a system for testing novel vaccines. Despite the Chinese rhesus macaque potentially being a more relevant model for AIDS outcomes than the Indian rhesus macaque, the Chinese-origin rhesus macaques have not been well-characterized for their major histocompatibility complex (MHC) composition and function, reducing their greater utilization. In this study, we characterized a total of 50 unique Chinese rhesus macaques from several varying origins for their entire MHC class I allele composition and identified a total of 58 unique complete MHC class I sequences. Only nine of the sequences had been associated with Indian rhesus macaques, and 28/58 (48.3%) of the sequences identified were novel. From all MHC alleles detected, we prioritized Mamu-A1*02201 for functional characterization based on its higher frequency of expression. Upon the development of MHC/peptide binding assays and definition of its associated motif, we revealed that this allele shares peptide binding characteristics with the HLA-B7 supertype, the most frequent supertype in human populations. These studies provide the first functional characterization of an MHC class I molecule in the context of Chinese rhesus macaques and the first instance of HLA-B7 analogy for rhesus macaques.
Collapse
Affiliation(s)
- Christopher Solomon
- Department of Biological Sciences, California State University - San Marcos, San Marcos, CA 92096 USA
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 USA
| | - Scott Southwood
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 USA
| | - Ilka Hoof
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Richard Rudersdorf
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Bjoern Peters
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 USA
| | - John Sidney
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 USA
| | - Clemencia Pinilla
- Torrey Pines Institute for Molecular Studies, La Jolla, CA 92037 USA
| | | | - Binhua Ling
- Department of Tropical Medicine, School of Public Health, Tulane University, New Orleans, LA 70112 USA
| | - Preston Marx
- Department of Tropical Medicine, School of Public Health, Tulane University, New Orleans, LA 70112 USA
| | - Alessandro Sette
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 USA
| | - Bianca R. Mothé
- Department of Biological Sciences, California State University - San Marcos, San Marcos, CA 92096 USA
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 USA
| |
Collapse
|
6
|
Rosner C, Kruse PH, Lübke T, Walter L. Erratum to: rhesus macaque MHC class I molecules show differential subcellular localizations. Immunogenetics 2010; 62:409-18. [PMID: 20445972 PMCID: PMC3128699 DOI: 10.1007/s00251-010-0447-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The MHC class I gene family of rhesus macaques is characterised by considerable gene duplications. While a HLA-C-orthologous gene is absent, the Mamu-A and in particular the Mamu-B genes have expanded, giving rise to plastic haplotypes with differential gene content. Although some of the rhesus macaque MHC class I genes are known to be associated with susceptibility/resistance to infectious diseases, the functional significance of duplicated Mamu-A and Mamu-B genes and the expression pattern of their encoded proteins are largely unknown. Here, we present data of the subcellular localization of AcGFP-tagged Mamu-A and Mamu-B molecules. We found strong cell surface and low intracellular expression for Mamu-A1, Mamu-A2 and Mamu-A3-encoded molecules as well as for Mamu-B*01704, Mamu-B*02101, Mamu-B*04801, Mamu-B*06002 and Mamu-B*13401. In contrast, weak cell surface and strong intracellular expression was seen for Mamu-A4*1403, Mamu-B*01202, Mamu-B*02804, Mamu-B*03002, Mamu-B*05704, Mamu-I*010201 and Mamu-I*0121. The different expression patterns were assigned to the antigen-binding alpha1 and alpha2 domains, suggesting failure of peptide binding is responsible for retaining 'intracellular' Mamu class I molecules in the endoplasmic reticulum. These findings indicate a diverse functional role of the duplicated rhesus macaque MHC class I genes.
Collapse
Affiliation(s)
- Cornelia Rosner
- Abteilung Primatengenetik, Deutsches Primatenzentrum-Leibniz, Institut für Primatenforschung, Kellnerweg 4, 37077, Göttingen, Germany
| | | | | | | |
Collapse
|
7
|
Rosner C, Kruse PH, Lübke T, Walter L. Rhesus macaque MHC class I molecules show differential subcellular localizations. Immunogenetics 2010; 62:149-58. [PMID: 20151120 PMCID: PMC2827799 DOI: 10.1007/s00251-010-0424-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 01/18/2010] [Indexed: 11/16/2022]
Abstract
The MHC class I gene family of rhesus macaques is characterised by considerable gene duplications. While a HLA-C-orthologous gene is absent, the Mamu-A and in particular the Mamu-B genes have expanded, giving rise to plastic haplotypes with differential gene content. Although some of the rhesus macaque MHC class I genes are known to be associated with susceptibility/resistance to infectious diseases, the functional significance of duplicated Mamu-A and Mamu-B genes and the expression pattern of their encoded proteins are largely unknown. Here, we present data of the subcellular localization of AcGFP-tagged Mamu-A and Mamu-B molecules. We found strong cell surface and low intracellular expression for Mamu-A1, Mamu-A2 and Mamu-A3-encoded molecules as well as for Mamu-B*01704, Mamu-B*02101, Mamu-B*04801, Mamu-B*06002 and Mamu-B*13401. In contrast, weak cell surface and strong intracellular expression was seen for Mamu-A4*1403, Mamu-B*01202, Mamu-B*02804, Mamu-B*03002, Mamu-B*05704, Mamu-I*010201 and Mamu-I*0121. The different expression patterns were assigned to the antigen-binding α1 and α2 domains, suggesting failure of peptide binding is responsible for retaining ‘intracellular’ Mamu class I molecules in the endoplasmic reticulum. These findings indicate a diverse functional role of the duplicated rhesus macaque MHC class I genes.
Collapse
Affiliation(s)
- Cornelia Rosner
- Abteilung Primatengenetik, Deutsches Primatenzentrum--Leibniz Institut für Primatenforschung, Kellnerweg 4, 37077, Göttingen, Germany
| | | | | | | |
Collapse
|
8
|
Elimination of helminth infection restores HIV-1C vaccine-specific T cell responses independent of helminth-induced IL-10. Vaccine 2009; 28:1310-7. [PMID: 19941988 DOI: 10.1016/j.vaccine.2009.11.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 11/02/2009] [Accepted: 11/06/2009] [Indexed: 12/18/2022]
Abstract
HIV-1 prevalence is highest in developing countries; similarly helminth parasites are often highly endemic in these same areas. Helminths are strong immune modulators, and negatively impact the ability of the infected hosts to mount protective vaccine-specific T cell immune responses for HIV-1 and other pathogens. Indeed, previously we found that Schistosoma mansoni infected mice had significantly impaired HIV-1C vaccine-specific T cell responses. Anthelminthics are available and inexpensive; therefore, in this study, we evaluated whether elimination of schistosome infection prior to vaccination with an HIV-1C DNA vaccine would increase recipients vaccine-specific responses. As expected, splenocytes from S. mansoni infected mice produced significantly elevated amounts of interleukin (IL)-4 and IL-10, and significantly lower amounts of interferon (IFN)-gamma than splenocytes from naïve mice. Following elimination of parasites by praziquantel (PZQ) treatment, splenomegaly was significantly reduced, though splenocytes produced similar or higher levels of IL-10 than splenocytes from infected mice. However, we found that PZQ treatment significantly increased levels of IFN-gamma in response to concanavalin A or SEA compared to splenocytes from untreated mice. Importantly, PZQ treatment resulted in complete restoration of HIV-1C vaccine-specific T cell responses at 8 weeks post-PZQ treatment. Restoration of HIV-1C vaccine-specific T cell responses following elimination of helminth infection was time dependent, but surprisingly independent of the levels of IL-4 and IL-10 induced by parasite antigens. Our study shows that elimination of worms offers an affordable and a simple means to restore immune responsiveness to T cell based vaccines for HIV-1 and other infectious diseases in helminth endemic settings.
Collapse
|
9
|
Broadly neutralizing monoclonal antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian-human immunodeficiency virus SHIVBa-L. J Virol 2009; 84:1302-13. [PMID: 19906907 DOI: 10.1128/jvi.01272-09] [Citation(s) in RCA: 270] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The membrane-proximal external region (MPER) of HIV-1, located at the C terminus of the gp41 ectodomain, is conserved and crucial for viral fusion. Three broadly neutralizing monoclonal antibodies (bnMAbs), 2F5, 4E10, and Z13e1, are directed against linear epitopes mapped to the MPER, making this conserved region an important potential vaccine target. However, no MPER antibodies have been definitively shown to provide protection against HIV challenge. Here, we show that both MAbs 2F5 and 4E10 can provide complete protection against mucosal simian-human immunodeficiency virus (SHIV) challenge in macaques. MAb 2F5 or 4E10 was administered intravenously at 50 mg/kg to groups of six male Indian rhesus macaques 1 day prior to and again 1 day following intrarectal challenge with SHIV(Ba-L). In both groups, five out of six animals showed complete protection and sterilizing immunity, while for one animal in each group a low level of viral replication following challenge could not be ruled out. The study confirms the protective potential of 2F5 and 4E10 and supports emphasis on HIV immunogen design based on the MPER region of gp41.
Collapse
|
10
|
Selective downregulation of rhesus macaque and sooty mangabey major histocompatibility complex class I molecules by Nef alleles of simian immunodeficiency virus and human immunodeficiency virus type 2. J Virol 2008; 82:3139-46. [PMID: 18199657 DOI: 10.1128/jvi.02102-07] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) Nef downregulates HLA-A and -B molecules, but not HLA-C or -E molecules, based on amino acid differences in their cytoplasmic domains to simultaneously evade cytotoxic T lymphocyte (CTL) and natural killer cell surveillance. Rhesus macaques and sooty mangabeys express orthologues of HLA-A, -B, and -E, but not HLA-C, and many of these molecules have unique amino acid differences in their cytoplasmic tails. We found that these differences also resulted in differential downregulation by primary simian immunodeficiency virus (SIV) SIV(smm/mac) and HIV-2 Nef alleles. Thus, selective major histocompatibility complex class I downregulation is a conserved mechanism of immune evasion for pathogenic SIV infection of rhesus macaques and nonpathogenic SIV infection of sooty mangabeys.
Collapse
|
11
|
Karl JA, Wiseman RW, Campbell KJ, Blasky AJ, Hughes AL, Ferguson B, Read DS, O’Connor DH. Identification of MHC class I sequences in Chinese-origin rhesus macaques. Immunogenetics 2008; 60:37-46. [PMID: 18097659 PMCID: PMC2830873 DOI: 10.1007/s00251-007-0267-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Accepted: 12/03/2007] [Indexed: 10/22/2022]
Abstract
The rhesus macaque (Macaca mulatta) is an excellent model for human disease and vaccine research. Two populations exhibiting distinctive morphological and physiological characteristics, Indian- and Chinese-origin rhesus macaques, are commonly used in research. Genetic analysis has focused on the Indian macaque population, but the accessibility of these animals for research is limited. Due to their greater availability, Chinese rhesus macaques are now being used more frequently, particularly in vaccine and biodefense studies, although relatively little is known about their immunogenetics. In this study, we discovered major histocompatibility complex (MHC) class I cDNAs in 12 Chinese rhesus macaques and detected 41 distinct Mamu-A and Mamu-B sequences. Twenty-seven of these class I cDNAs were novel, while six and eight of these sequences were previously reported in Chinese and Indian rhesus macaques, respectively. We then performed microsatellite analysis on DNA from these 12 animals, as well as an additional 18 animals, and developed sequence specific primer PCR (PCR-SSP) assays for eight cDNAs found in multiple animals. We also examined our cohort for potential admixture of Chinese and Indian origin animals using a recently developed panel of single nucleotide polymorphisms (SNPs). The discovery of 27 novel MHC class I sequences in this analysis underscores the genetic diversity of Chinese rhesus macaques and contributes reagents that will be valuable for studying cellular immunology in this population.
Collapse
Affiliation(s)
- Julie A. Karl
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - Roger W. Wiseman
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - Kevin J. Campbell
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - Alex J. Blasky
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - Austin L. Hughes
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - Betsy Ferguson
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - Daniel S. Read
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| | - David H. O’Connor
- J. A. Karl · D. H. O’Connor, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- R. W. Wiseman · K. J. Campbell · A. J. Blasky · D. H. O’Connor, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- A. L. Hughes, Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- B. Ferguson, Genetics Research and Informatics Program, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97006, USA
- B. Ferguson, Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
- D. S. Read, Battelle Biomedical Research Center, Columbus, OH 43201-2693, USA
- D. H. O’Connor, University of Wisconsin-Madison, 555 Science Drive, Madison, Wisconsin 53711, USA,
| |
Collapse
|
12
|
Kaizu M, Borchardt GJ, Glidden CE, Fisk DL, Loffredo JT, Watkins DI, Rehrauer WM. Molecular typing of major histocompatibility complex class I alleles in the Indian rhesus macaque which restrict SIV CD8+ T cell epitopes. Immunogenetics 2007; 59:693-703. [PMID: 17641886 DOI: 10.1007/s00251-007-0233-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 05/21/2007] [Indexed: 01/09/2023]
Abstract
The utility of the rhesus macaque as an animal model in both HIV vaccine development and pathogenesis studies necessitates the development of accurate and efficient major histocompatibility complex (MHC) genotyping technologies. In this paper, we describe the development and application of allele-specific polymerase chain reaction (PCR) amplification for the simultaneous detection of eight MHC class I alleles from the rhesus macaque (Macaca mulatta) of Indian descent. These alleles were selected, as they have been implicated in the restriction of CD8(+) T cell epitopes of simian immunodeficiency virus (SIV). Molecular typing of Mamu-A 01, Mamu-A 02, Mamu-A 08, Mamu-A 11, Mamu-B 01, Mamu-B 03, Mamu-B 04, and Mamu-B 17 was conducted in a high throughput fashion using genomic DNA. Our amplification strategy included a conserved internal control target to minimize false negative results and can be completed in less than 5 h. We have genotyped over 4,000 animals to establish allele frequencies from colonies all over the western hemisphere. The ability to identify MHC-defined rhesus macaques will greatly enhance investigation of the immune responses, which are responsible for the control of viral replication. Furthermore, application of this technically simple and accurate typing method should facilitate selection, utilization, and breeding of rhesus macaques for AIDS virus pathogenesis and vaccine studies.
Collapse
Affiliation(s)
- Masahiko Kaizu
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Lafont BAP, McGraw CM, Stukes SA, Buckler-White A, Plishka RJ, Byrum RA, Hirsch VM, Martin MA. The locus encoding an oligomorphic family of MHC-A alleles (Mane-A*06/Mamu-A*05) is present at high frequency in several macaque species. Immunogenetics 2007; 59:211-23. [PMID: 17256149 DOI: 10.1007/s00251-007-0190-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 12/11/2006] [Indexed: 10/23/2022]
Abstract
Several macaques species are used for HIV pathogenesis and vaccine studies, and the characterization of their major histocompatibility complex (MHC) class I genes is required to rigorously evaluate the cellular immune responses induced after immunization and/or infection. In this study, we demonstrate that the gene expressing the Mane-A*06 allele of pig-tailed macaques is an orthologue of the locus encoding the Mamu-A*05 allele family in rhesus macaques. Analysis of the distribution of this locus in a cohort of 63 pig-tailed macaques revealed that it encodes an oligomorphic family of alleles, highly prevalent (90%) in the pig-tailed macaque population. Similarly, this locus was very frequently found (62%) in a cohort of 80 Indian rhesus macaques. An orthologous gene was also detected in cynomolgus monkeys originating from four different geographical locations, but was absent in two African monkey species. Expression analysis in pig-tailed macaques revealed that the Mane-A*06 alleles encoded by this locus are transcribed at 10- to 20-fold lower levels than other MHC-A alleles (Mane-A*03 or Mane-A*10). Despite their conservation and high prevalence among Asian macaque species, the alleles of the Mane-A*06 family and, by extension their orthologues in rhesus and cynomolgus monkeys, may only modestly contribute to cellular immune responses in macaques because of their low level of expression.
Collapse
Affiliation(s)
- Bernard A P Lafont
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Chu F, Lou Z, Chen YW, Liu Y, Gao B, Zong L, Khan AH, Bell JI, Rao Z, Gao GF. First glimpse of the peptide presentation by rhesus macaque MHC class I: crystal structures of Mamu-A*01 complexed with two immunogenic SIV epitopes and insights into CTL escape. THE JOURNAL OF IMMUNOLOGY 2007; 178:944-52. [PMID: 17202356 DOI: 10.4049/jimmunol.178.2.944] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The infection of rhesus macaques (Macaca mulatta) by the SIV is the best animal model for studying HIV infection and for AIDS vaccine development. A prevalent MHC class I allele, Mamu-A*01, is known to correlate with containment of SIV, which has been extensively explored in studies of CTL-based vaccination concepts. We determined the crystal structures of Mamu-A*01 complexed with two immunodominant SIV epitopes: the nonamer CM9 of group-specific Ag (Gag, 181-189; CTPYDINQM) and the octamer TL8 of transcription activator (Tat, 28-35; TTPESANL). The overall structures of the two Mamu-A*01 complexes are similar to other MHC class I molecules. Both structures confirm the presence of an absolutely conserved proline anchor residue in the P3 position of the Ag, bound to a D pocket of the Mamu-A*01 H chain with optimal surface complementarity. Like other MHC/peptide complex structures, the P2 and C-terminal residues of the epitopes are also important for anchoring to the MHC molecule, whereas the middle residues form an arch and their side chains are directed into solvent. These two structures reveal details of how Mamu-A*01 interacts with two well-studied epitopes at the atomic level. We discuss the structural basis of CTL escape, based on molecular models made possible by these two structures. The results we present in this study are most relevant for the rational design of Mamu-A*01-restricted CTL epitopes with improved binding, as a step toward development of AIDS vaccines.
Collapse
Affiliation(s)
- Fuliang Chu
- Center for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, 13 Beiyitiao, Zhongguancun, Beijing 100080, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Radaelli A, Bonduelle O, Beggio P, Mahe B, Pozzi E, Elli V, Paganini M, Zanotto C, De Giuli Morghen C, Combadière B. Prime-boost immunization with DNA, recombinant fowlpox virus and VLP(SHIV) elicit both neutralizing antibodies and IFNgamma-producing T cells against the HIV-envelope protein in mice that control env-bearing tumour cells. Vaccine 2006; 25:2128-38. [PMID: 17241705 DOI: 10.1016/j.vaccine.2006.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 10/05/2006] [Accepted: 11/03/2006] [Indexed: 10/23/2022]
Abstract
Different primings with DNA and fowlpox virus (FP) recombinants or FP alone were used in a pre-clinical trial to evaluate and compare immunogenicity and efficacy against HIV/SHIV. Three immunization regimens were tested in three groups of mice in which the SIV gag/pol and HIV-1 env transgenes were separately expressed by DNA and FP vectors, followed by VLP(SHIV) boosting. All of the protocols were effective in eliciting homologous neutralizing antibodies, although the mice immunized with DNA followed by FP recombinants or DNA+FP recombinants showed both high titres of neutralizing antibodies and high frequencies of env-specific IFNgamma-producing T lymphocytes. Vaccine efficacy, as demonstrated by growth control of env-expressing tumours, was obtained in both of these two groups of mice. These results establish a preliminary profile for the combined use of these recombinant vectors in protocols to be tested in the SHIV-macaque model of HIV-1 infection.
Collapse
Affiliation(s)
- Antonia Radaelli
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Da'Dara AA, Lautsch N, Dudek T, Novitsky V, Lee TH, Essex M, Harn DA. Helminth infection suppresses T-cell immune response to HIV-DNA-based vaccine in mice. Vaccine 2006; 24:5211-9. [PMID: 16675073 DOI: 10.1016/j.vaccine.2006.03.078] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Revised: 03/21/2006] [Accepted: 03/24/2006] [Indexed: 10/24/2022]
Abstract
A number of HIV-1 vaccines are in various phases of clinical trials and many more are in the developmental pipeline. Vaccines are especially needed for developing countries where morbidity and mortality due to HIV/AIDS is most severe, the prevalence of HIV infection is highest, and its incidence is often still rising dramatically. Individuals living in these regions are often infected with one or more helminth parasites which systemically bias the immune system towards Th2-type as well as drive immune anergy. The goal of this study was to develop a multi-T-cell epitope DNA-based vaccine for HIV-1 subtype C and to determine the impact of helminth infection on the immune response to this vaccine. We found that vaccination of naïve mice with the multi-epitope vaccine, designated TD158, induced a strong HIV-1C-specific T-cell immune response, and that the addition of the Igkappa leader sequence to the TD158 vaccine construct significantly increased the frequencies of IFN-gamma secreting CD8+ T cells. However, the TD158 vaccine specific response of mice infected with the human helminth Schistosoma mansoni was significantly suppressed. The impact of schistosome infection on suppressing the virus-specific immune response was the same whether mice were vaccinated with the TD158 vaccine or with the Igkappa enhanced TD158. The results of this study suggest that helminth infection may pose a serious problem for vaccination with the DNA-based HIV-1 vaccine in developing country populations, and that the prevalence of helminth infections in the vaccine cohorts should be taken into account for HIV-1 vaccine trial design.
Collapse
Affiliation(s)
- Akram A Da'Dara
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Marsac D, Puaux AL, Rivière Y, Michel ML. In vivo induction of cellular and humoral immune responses by hybrid DNA vectors encoding simian/human immunodeficiency virus/hepatitis B surface antigen virus particles in BALB/c and HLA-A2-transgenic mice. Immunobiology 2005; 210:305-19. [PMID: 16164038 DOI: 10.1016/j.imbio.2005.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
To improve the immunogenicity of epitopes derived from Gag proteins of simian immunodeficiency virus (SIV) and from the envelope (Env) protein of human immunodeficiency virus type 1 (HIV-1), we have designed hybrid DNA vaccines by inserting sequences encoding antigenic domains of SIV and HIV-1 into the hepatitis B virus envelope gene. This gene encodes the hepatitis B surface antigen (HBsAg) capable of spontaneous assembly into virus-like particles that were used here as carrier. Injections of hybrid vectors encoding B-cell epitopes from the gp41 and the gp120 envelope proteins of HIV-1 induced specific humoral responses in BALB/c mice. Furthermore, high frequencies of IFN-gamma-secreting CD8+ T cells specific for various antigenic determinants of SIV-Gag were observed after intramuscular injections of hybrid DNA vectors in BALB/c mice. Genetic immunization of HLA-A2.1-transgenic mice with HIV-Env/HBsAg-encoding DNA generated a strong CTL response and IFN-gamma-secreting CD8+ T lymphocytes specific for HIV-1 envelope-derived peptide. H-2d-restricted HBs-specific T-cell responses dominated over SIV-Gag responses in BALB/c mice whereas HLA-A2-restricted HIV-Env response was enhanced after fusion with HBsAg. These data demonstrate that different B and T-cell epitopes of vaccine-relevant viral antigens can be expressed in vivo as fusion proteins with HBsAg but that the optimal immunogenicity may differ strikingly between individual epitopes.
Collapse
Affiliation(s)
- Delphine Marsac
- INSERM U 370 Carcinogenèse Hépatique et Virologie Moléculaire, Département de Médecine Moléculaire, Institut Pasteur, 28, rue du Docteur Roux, 75724 PARIS CEDEX 15, France
| | | | | | | |
Collapse
|
18
|
Hickman-Miller HD, Bardet W, Gilb A, Luis AD, Jackson KW, Watkins DI, Hildebrand WH. Rhesus macaque MHC class I molecules present HLA-B-like peptides. THE JOURNAL OF IMMUNOLOGY 2005; 175:367-75. [PMID: 15972670 DOI: 10.4049/jimmunol.175.1.367] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
SIV-infected Indian rhesus macaques (Macaca mulatta) are an important animal model for humans infected with HIV. Understanding macaque (M. mulatta class I (Mamu)) MHC class I-peptide binding facilitates the comparison of SIV- and HIV-specific cellular immune responses. In this study, we characterized the endogenous peptide-binding properties of three Mamu-A (A*02, A*08, A*11) and three Mamu-B (B*01, B*03, B*12) class I molecules. Motif comparisons revealed that five of the six macaque class I molecules (A*02, A*08, A*11, B*01, and B*03) have peptide-binding motifs similar to those of human class I molecules. Of the 65 macaque endogenous peptide ligands that we sequenced by tandem mass spectroscopy, 5 were previously eluted from HLA class I molecules. Nonamers predominated among the individual ligands, and both the motifs and the individual ligands indicated P2, P9, and various ancillary anchors. Interestingly, peptide binding of the Mamu-A and Mamu-B molecules exhibited cross-species peptide-presentation overlap primarily with HLA-B molecules. Indeed, all of the macaque class I molecules appeared HLA-B-like in peptide presentation. Remarkably, the overlap in macaque- and HLA-peptide presentation occurred despite divergent class I peptide-binding grooves. Macaque and human class I differing by up to 42 aa (13-23%) within the alpha-1 and alpha-2 domains, including substantial divergence within specificity pockets A-F, bound the same endogenous peptide. Therefore, endogenous peptide characterization indicates that macaque class I molecules may be the functional equivalents of HLA-B molecules.
Collapse
Affiliation(s)
- Heather D Hickman-Miller
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Otting N, Heijmans CMC, Noort RC, de Groot NG, Doxiadis GGM, van Rood JJ, Watkins DI, Bontrop RE. Unparalleled complexity of the MHC class I region in rhesus macaques. Proc Natl Acad Sci U S A 2005; 102:1626-31. [PMID: 15665097 PMCID: PMC545086 DOI: 10.1073/pnas.0409084102] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The highly polymorphic gene products of the classical MHC class I genes in humans (HLA-A, HLA-B, and HLA-C) play a critical role in the immune defense against intracellular infections. Because non-human primates are important models for AIDS vaccine research, rhesus monkeys from a thoroughly pedigreed and serotyped colony were subjected to full-length cDNA analysis of MHC class I gene transcripts. Rhesus macaques express multiple dominant Mamu-A and Mamu-B transcripts (majors) per chromosome, which are characterized by high expression levels. The presence of additional cDNAs with low levels of expression (minors) suggests evidence for transcriptional control of MHC class I genes. Moreover, phylogenetic analyses illustrate that most of the Mamu-A and Mamu-B loci/lineages identified display no or only limited levels of allelic polymorphism. Thus, MHC class I diversity in rhesus macaques is typified by the existence of an unmatched high number of Mamu-A and Mamu-B region configurations that exhibit polymorphism with regard to the number and combination of transcribed loci present per chromosome.
Collapse
Affiliation(s)
- Nel Otting
- Department of Comparative Genetics and Refinement, Biomedical Primate Research Centre, PO Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Puaux AL, Marsac D, Prost S, Singh MK, Earl P, Moss B, Le Grand R, Riviere Y, Michel ML. Efficient priming of simian/human immunodeficiency virus (SHIV)-specific T-cell responses with DNA encoding hybrid SHIV/hepatitis B surface antigen particles. Vaccine 2004; 22:3535-45. [PMID: 15315833 DOI: 10.1016/j.vaccine.2004.03.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Revised: 03/18/2004] [Accepted: 03/29/2004] [Indexed: 10/26/2022]
Abstract
Recent efforts to design an human immunodeficiency virus type 1 (HIV-1) vaccine candidate have focused on means of eliciting anti-viral T-cell responses. We tried to improve the immunogenicity of DNA vaccines by designing hybrid DNA constructs encoding hepatitis B surface antigen (HBsAg) fused to antigenic domains of simian/human immunodeficiency virus (SHIV 89.6P). Immunisation with hybrid DNA induced both effector and long-lasting precursor T-cells. Following boosting with a recombinant modified vaccinia Ankara (rMVA) producing full-length SIV and HIV antigens, it appeared that priming with hybrid DNA had increased virus-specific T-cell responses in terms of both the number of virus-specific IFN-gamma-secreting T-cells and virus-specific lymphoproliferation. After intrarectal challenge with SHIV 89.6P, immunised animals demonstrated early control of SHIV 89.6P replication and stable CD4+ T-cell counts.
Collapse
Affiliation(s)
- Anne-Laure Puaux
- INSERM U 370, Carcinogenèse Hépatique et Virologie Moléculaire, Département de Médecine Moléculaire, Institut Pasteur, 28 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Uda A, Tanabayashi K, Yamada YK, Akari H, Lee YJ, Mukai R, Terao K, Yamada A. Detection of 14 alleles derived from the MHC class I A locus in cynomolgus monkeys. Immunogenetics 2004; 56:155-63. [PMID: 15164234 DOI: 10.1007/s00251-004-0683-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2003] [Revised: 04/26/2004] [Indexed: 10/26/2022]
Abstract
A basic understanding of the major histocompatibility complex (MHC) class I, which, together with T-cell receptors, is a key player in antigen recognition by cytotoxic T lymphocytes, is necessary to study the cellular immune response to intracellular pathogens. The MHC has hardly been reported in cynomolgus monkeys ( Macaca facicularis), although cynomolgus monkeys have been frequently used as the surrogate animal model. We attempted to determine the nucleotide sequences of the MHC class I A locus of cynomolgus monkeys ( Mafa-A) and eventually 34 independent sequences of Mafa-A were obtained from 29 cynomolgus monkeys. These 34 sequences were classified into 14 Mafa-A alleles according to the results of phylogenetic analyses using the neighbor-joining method. One to three Mafa-A alleles were obtained from a single animal. We also tried to establish a multiplex PCR-SSP method for convenient typing of Mafa-A alleles. cDNA from a family of cynomolgus monkeys, which is composed of four sirs and four dams, were examined by multiplex PCR-SSP. The result of multiplex PCR-SSP showed that an individual cynomolgus monkey had two or three Mafa-A alleles, suggesting that the A locus of cynomolgus monkeys might be duplicated.
Collapse
Affiliation(s)
- Akihiko Uda
- Tsukuba Primate Center for Medical Science, National Institute of Infectious Diseases, 1 Hachimandai, Tsukuba, 305-0843, Ibaraki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Mühl T, Krawczak M, Ten Haaft P, Hunsmann G, Sauermann U. MHC class I alleles influence set-point viral load and survival time in simian immunodeficiency virus-infected rhesus monkeys. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3438-46. [PMID: 12218167 DOI: 10.4049/jimmunol.169.6.3438] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In HIV-infected humans and SIV-infected rhesus macaques, host genes influence viral containment and hence the duration of the disease-free latency period. Our knowledge of the rhesus monkey immunogenetics, however, is limited. In this study, we describe partial cDNA sequences of five newly discovered rhesus macaque (Mamu) class I alleles and PCR-based typing techniques for the novel and previously published Mhc class I alleles. Using 15 primer pairs for PCR-based typing and DNA sequence analysis, we identified at least 21 Mhc class I alleles in a cohort of 91 SIV-infected macaques. The results confirm the presence of multiple class I genes in rhesus macaques. Of these alleles, Mamu-A*01 was significantly associated with lower set-point viral load and prolonged survival time. Mamu-A*1303 was associated with longer survival and a "novel" Mhc class I allele with lower set-point viral load. The alleles are frequent in rhesus macaques of Indian origin (12-22%). In addition, survival probability of individual SIV-infected rhesus monkeys increased with their number of alleles considered to be associated with longer survival. The results contribute to improve the interpretation and quality of preclinical studies in rhesus monkeys.
Collapse
Affiliation(s)
- Thorsten Mühl
- Department of Virology and Immunology, German Primate Center, Goettingen, Germany
| | | | | | | | | |
Collapse
|
23
|
Robinson S, Charini WA, Newberg MH, Kuroda MJ, Lord CI, Letvin NL. A commonly recognized simian immunodeficiency virus Nef epitope presented to cytotoxic T lymphocytes of Indian-origin rhesus monkeys by the prevalent major histocompatibility complex class I allele Mamu-A*02. J Virol 2001; 75:10179-86. [PMID: 11581386 PMCID: PMC114592 DOI: 10.1128/jvi.75.21.10179-10186.2001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability to monitor vaccine-elicited CD8(+) cytotoxic T-lymphocyte (CTL) responses in simian immunodeficiency virus (SIV)- and simian-human immunodeficiency virus (SHIV)-infected rhesus monkeys has been limited by our knowledge of viral epitopes predictably presented to those lymphocytes by common rhesus monkey MHC class I alleles. We now define an SIV and SHIV Nef CTL epitope (YTSGPGIRY) that is presented to CD8(+) T lymphocytes by the common rhesus monkey MHC class I molecule Mamu-A*02. All seven infected Mamu-A*02(+) monkeys evaluated demonstrated this response, and peptide-stimulated interferon gamma Elispot assays indicated that the response represents a large proportion of the entire CD8(+) T-lymphocyte SIV- or SHIV-specific immune response of these animals. Knowledge of this epitope and MHC class I allele substantially increases the number of available rhesus monkeys that can be used for testing prototype HIV vaccines in this important animal model.
Collapse
Affiliation(s)
- S Robinson
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
The use of chimeric simian and human immunodeficiency viruses (SHIVs) that encode HIV-1 Env and are infectious to macaques has made it possible to analyze the pathogenicity of HIV-1 in vivo, and to evaluate the efficacy of candidate vaccines in macaques. In addition, we believe that gene-deleted SHIVs could potentially be used as anti-HIV-1 live-attenuated vaccines. Gene-deleted SHIVs replicate transiently, are non-pathogenic and induce strong protection against challenge infection. The most important advantage of gene-deleted SHIVs is that their efficacy and safety can be evaluated in macaques before they are used in humans.
Collapse
Affiliation(s)
- T Kuwata
- Institute for Virus Research, Kyoto University, Shogoin-Kawara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
25
|
Patterson LJ, Robey F, Muck A, Van Remoortere K, Aldrich K, Richardson E, Alvord WG, Markham PD, Cranage M, Robert-Guroff M. A conformational C4 peptide polymer vaccine coupled with live recombinant vector priming is immunogenic but does not protect against rectal SIV challenge. AIDS Res Hum Retroviruses 2001; 17:837-49. [PMID: 11429125 DOI: 10.1089/088922201750252034] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The conserved, immunogenic CD4 binding site on the viral envelope is an attractive HIV or SIV vaccine candidate. Polymerization of an 18 amino acid segment derived from the C4 domain of SIV gp120 produced a peptide polymer or "peptomer," having an alpha-helical conformation possibly mimicking a proposed structure of the C4 domain in the unbound native protein. The SIV peptomer and native gp120 were compared as subunit boosts following two adenovirus type 5 host range (Ad5hr)-SIVenv recombinant priming immunizations. Both vaccine regimens successfully elicited SIV-specific CTL responses in five of six immunized macaques. Peptomer-boosted macaques exhibited significantly higher envelope-specific T cell proliferative responses than either the gp120-boosted macaques or controls. Peptomer immunization also elicited peptomer and SIV gp120-specific binding antibodies, but only native gp120 boosting elicited SIV neutralizing antibodies. Upon intrarectal challenge with SIVmac32H, all nine macaques became infected. The solely envelope-based vaccine conferred no protection. However, changing the boosting immunogen to the C4 peptomer did not improve protective efficacy in spite of its elicitation of humoral and cellular immune responses, including robust T-helper activity. In spite of the peptomer's strong immunogenicity and potential for induction of broadly protective immune responses, it was not effective as a subunit vaccine.
Collapse
Affiliation(s)
- L J Patterson
- Basic Research Laboratory, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fu TM, Freed DC, Trigona WL, Guan L, Zhu L, Long R, Persaud NV, Manson K, Dubey S, Shiver JW. Evaluation of cytotoxic T-lymphocyte responses in human and nonhuman primate subjects infected with human immunodeficiency virus type 1 or simian/human immunodeficiency virus. J Virol 2001; 75:73-82. [PMID: 11119575 PMCID: PMC113899 DOI: 10.1128/jvi.75.1.73-82.2001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2000] [Accepted: 10/02/2000] [Indexed: 11/20/2022] Open
Abstract
Cytotoxic T-lymphocyte (CTL) responses have been implicated as playing an important role in control of human immunodeficiency virus (HIV) infection. However, it is technically difficult to demonstrate CTL responses consistently in nonhuman primate and human subjects using traditional cytotoxicity assay methods. In this study, we systematically evaluated culture conditions that may affect the proliferation and expansion of CTL effector cells and presented a sensitive method for detection of cytotoxicity responses with bulk CTL cultures. We confirmed the sensitivity and specificity of this method by demonstration of vigorous CTL responses in a simian-HIV (SHIV)-infected rhesus macaque. The expansion of epitope-specific CTL effector cells was also measured quantitatively by CTL epitope-major histocompatibility complex tetramer complex staining. In addition, two new T-cell determinants in the SIV gag region are identified. Last, we showed the utility of this method for studying CTL responses in chimpanzee and human subjects.
Collapse
Affiliation(s)
- T M Fu
- Department of Virus and Cell Biology, Merck Research Laboratories, West Point, Pennsylvania 19486, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Stipp HL, Kumar A, Narayan O. Characterization of immune escape viruses from a macaque immunized with live-virus vaccine and challenged with pathogenic SHIVKU-1. AIDS Res Hum Retroviruses 2000; 16:1573-80. [PMID: 11054270 DOI: 10.1089/088922200750006092] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We characterized two immune escape viruses (SHIV(KU-1/105w52) and SHIV(KU-1/105w98)) from a macaque immunized with DeltavpuDeltanef SHIV-4 and challenged with pathogenic SHIV(KU-1). This macaque developed neutralizing antibodies as well as virus-specific CTLs against the challenge virus. However, the two new viruses could not be neutralized by anti-SHIV(KU-1)-specific neutralizing antibodies and were poorly recognized by challenge virus-specific CTLs. Sequence analysis of the gene encoding gp120 revealed several mutations in the protein that might have contributed to the development of the immune-escape viruses.
Collapse
Affiliation(s)
- H L Stipp
- Merrell Dow Laboratory of Viral Pathogenesis and Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | |
Collapse
|
28
|
Lobashevsky AL, Thomas JM. Six mamu-A locus alleles defined by a polymerase chain reaction sequence specific primer method. Hum Immunol 2000; 61:1013-20. [PMID: 11082514 DOI: 10.1016/s0198-8859(00)00177-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Rhesus monkeys are relevant models for human diseases and transplantation. In each case, a complete understanding of these models requires knowledge of macaque MHC. Due to high polymorphism and multiple genes per haplotype, it has been difficult to develop a rapid typing method for rhesus monkey MHC class I. We developed a simple and rapid PCR-SSP strategy for rhesus monkey Mamu-A locus typing. Fifty-two rhesus monkeys were included in the study. Six rhesus monkey allel-specific primer pairs were designed based on published Mamu-A locus gene sequences. Allele-specific PCR products ranged in size from 346 to 788 bp; 5' and 3' Mamu-A locus allele specific primers were located in the second and third exons, respectively. Specific PCR product gel purification was followed by direct sequencing, without subcloning, in both directions. Our data showed variability in the number of Mamu-A alleles ranging from 1 to 4 per genotype. The highest frequencies were observed for Mamu-A*02, -A*04, and -A*03 alleles. Thus, we report here the first PCR-SSP typing method for Mamu-A*02, -03, -04, -05, -06, and -07 array of class I alleles. This technique appears to be a highly reproducible and discriminatory method for detecting this subset of class I A locus genes in rhesus monkeys.
Collapse
Affiliation(s)
- A L Lobashevsky
- Department of Surgery, Transplantation Immunobiology Division, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
29
|
Evans DT, Jing P, Allen TM, O'Connor DH, Horton H, Venham JE, Piekarczyk M, Dzuris J, Dykhuzen M, Mitchen J, Rudersdorf RA, Pauza CD, Sette A, Bontrop RE, DeMars R, Watkins DI. Definition of five new simian immunodeficiency virus cytotoxic T-lymphocyte epitopes and their restricting major histocompatibility complex class I molecules: evidence for an influence on disease progression. J Virol 2000; 74:7400-10. [PMID: 10906193 PMCID: PMC112260 DOI: 10.1128/jvi.74.16.7400-7410.2000] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2000] [Accepted: 05/15/2000] [Indexed: 11/20/2022] Open
Abstract
Simian immunodeficiency virus (SIV) infection of the rhesus macaque is currently the best animal model for AIDS vaccine development. One limitation of this model, however, has been the small number of cytotoxic T-lymphocyte (CTL) epitopes and restricting major histocompatibility complex (MHC) class I molecules available for investigating virus-specific CTL responses. To identify new MHC class I-restricted CTL epitopes, we infected five members of a family of MHC-defined rhesus macaques intravenously with SIV. Five new CTL epitopes bound by four different MHC class I molecules were defined. These included two Env epitopes bound by Mamu-A*11 and -B*03 and three Nef epitopes bound by Mamu-B*03, -B*04, and -B*17. All four restricting MHC class I molecules were encoded on only two haplotypes (b or c). Interestingly, resistance to disease progression within this family appeared to be associated with the inheritance of one or both of these MHC class I haplotypes. Two individuals that inherited haplotypes b and c separately survived for 299 and 511 days, respectively, while another individual that inherited both haplotypes survived for 889 days. In contrast, two MHC class I-identical individuals that did not inherit either haplotype rapidly progressed to disease (survived <80 days). Since all five offspring were identical at their Mamu-DRB loci, MHC class II differences are unlikely to account for their patterns of disease progression. These results double the number of SIV CTL epitopes defined in rhesus macaques and provide evidence that allelic differences at the MHC class I loci may influence rates of disease progression among AIDS virus-infected individuals.
Collapse
Affiliation(s)
- D T Evans
- Wisconsin Regional Primate Research Center, University of Wisconsin, Madison, Wisconsin 53715, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Urvater JA, Otting N, Loehrke JH, Rudersdorf R, Slukvin II, Piekarczyk MS, Golos TG, Hughes AL, Bontrop RE, Watkins DI. Mamu-I: a novel primate MHC class I B-related locus with unusually low variability. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1386-98. [PMID: 10640754 DOI: 10.4049/jimmunol.164.3.1386] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The rhesus macaque is an important animal model for several human diseases and organ transplantation. Therefore, definition of the MHC of this species is crucial to the development of these models. Unfortunately, unlike humans, lymphocytes from a single rhesus macaque express up to 12 different MHC class I cDNAs. From which locus these various alleles are derived is unclear. In our attempts to define the MHC class I loci of the rhesus macaque, we have identified an unusual MHC class I locus, Mamu-I. We isolated 26 I locus alleles from three different macaque species but not from three other Cercopithecine genera, suggesting that the I locus is the result of a recent duplication of the B locus occurring after the divergence of macaques from the ancestor of the other extant Cercopithecine genera. Mamu-I mRNA transcripts were detected in all tissues examined and Mamu-I protein was produced in rhesus B lymphoblastoid cell lines. Furthermore, Mamu-I protein was detected by flow cytometry on the surface of human 721.221 cells transfected with Mamu-I. In contrast to the polymorphism present at this locus, there is unusually low sequence variability, with the mean number of nucleotide differences between alleles being only 3.6 nt. Therefore, Mamu-I is less variable than any other polymorphic MHC class I locus described to date. Additionally, no evidence for positive selection on the peptide binding region was observed. Together, these results suggest that Mamu-I is an MHC class I locus in primates that has features of both classical and nonclassical loci.
Collapse
Affiliation(s)
- J A Urvater
- Wisconsin Regional Primate Research Center, Department of Genetics, University of Wisconsin, Madison, WI 53715, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ui M, Kuwata T, Igarashi T, Ibuki K, Miyazaki Y, Kozyrev IL, Enose Y, Shimada T, Uesaka H, Yamamoto H, Miura T, Hayami M. Protection of macaques against a SHIV with a homologous HIV-1 Env and a pathogenic SHIV-89.6P with a heterologous Env by vaccination with multiple gene-deleted SHIVs. Virology 1999; 265:252-63. [PMID: 10600597 DOI: 10.1006/viro.1999.0049] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To evaluate the potential of SHIVs as anti-HIV-1 live vaccines, we constructed two gene-deleted SHIVs, designated SHIV-drn and SHIV-dxrn. The former lacks vpr/nef and the latter lacks vpx/vpr/nef. Four macaques that had been vaccinated with SHIV-drn were challenged with SHIV-NM-3rN, which has an HIV-1 Env that is the same as that of SHIV-drn. No challenge virus was detected by DNA PCR in, or recovered from, two of the macaques. In the other two, challenge virus was detected once and twice, respectively. Plasma viral loads were much lower than those in unvaccinated controls. Another four macaques were vaccinated with SHIV-dxrn. These macaques showed resistance but less than that of SHIV-drn-vaccinated macaques. When the two SHIV-drn-vaccinated macaques were challenged with pathogenic SHIV-89.6P, which has an HIV-1 Env that is antigenically different from that of SHIV-drn, replication of the challenge virus was restricted, and the usual decrease in the number of CD4(+) cells was prevented. In this protection, it is noteworthy that protection involved not only neutralizing antibodies and killer cell activity, but also other unknown specific and nonspecific immunity elicited by the infection.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Antigens, Viral/immunology
- Gene Deletion
- Gene Products, nef/genetics
- Gene Products, nef/immunology
- Gene Products, vpr/genetics
- Gene Products, vpr/immunology
- HIV-1/genetics
- HIV-1/immunology
- Humans
- Macaca mulatta
- Male
- Simian Acquired Immunodeficiency Syndrome/prevention & control
- Simian Immunodeficiency Virus/genetics
- Simian Immunodeficiency Virus/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Regulatory and Accessory Proteins/genetics
- Viral Regulatory and Accessory Proteins/immunology
- nef Gene Products, Human Immunodeficiency Virus
- vpr Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- M Ui
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Furchner M, Erickson AL, Allen T, Watkins DI, Sette A, Johnson PR, Walker CM. The simian immunodeficiency virus envelope glycoprotein contains two epitopes presented by the Mamu-A*01 class I molecule. J Virol 1999; 73:8035-9. [PMID: 10482552 PMCID: PMC112819 DOI: 10.1128/jvi.73.10.8035-8039.1999] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cytotoxic T lymphocyte (CTL) responses against the simian immunodeficiency virus (SIV) envelope and Gag proteins were monitored in a Mamu-A*01-positive rhesus macaque infected with SIVsmE660. Peripheral blood mononuclear cells (PBMC) cultured with synthetic peptides spanning the entire gp160 and Gag coding region recognized a total of three epitopes. One located in Gag was identified as the previously described Mamu-A*01-restricted p11cC-->M epitope (CTPYDINQM). The other two epitopes, designated p15m and p54m, were located in the gp160 envelope protein. Both were nine amino acids in length and were predicted to bind Mamu-A*01 because they contained proline and leucine residues at positions 3 and 9, respectively. Indeed, expression of this class I major histocompatibility complex molecule was required for target cell recognition by envelope-specific CD8(+) T cells directed against both epitopes. These Mamu-A*01-restricted epitopes in the SIV envelope will be useful for monitoring immune responses in vaccinated or infected animals.
Collapse
Affiliation(s)
- M Furchner
- Children's Research Institute, Children's Hospital, Inc., Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Polacino PS, Stallard V, Klaniecki JE, Pennathur S, Montefiori DC, Langlois AJ, Richardson BA, Morton WR, Benveniste RE, Hu SL. Role of immune responses against the envelope and the core antigens of simian immunodeficiency virus SIVmne in protection against homologous cloned and uncloned virus challenge in Macaques. J Virol 1999; 73:8201-15. [PMID: 10482571 PMCID: PMC112838 DOI: 10.1128/jvi.73.10.8201-8215.1999] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously showed that envelope (gp160)-based vaccines, used in a live recombinant virus priming and subunit protein boosting regimen, protected macaques against intravenous and intrarectal challenges with the homologous simian immunodeficiency virus SIVmne clone E11S. However, the breadth of protection appears to be limited, since the vaccines were only partially effective against intravenous challenge by the uncloned SIVmne. To examine factors that could affect the breadth and the efficacy of this immunization approach, we studied (i) the effect of priming by recombinant vaccinia virus; (ii) the role of surface antigen gp130; and (iii) the role of core antigens (Gag and Pol) in eliciting protective immunity. Results indicate that (i) priming with recombinant vaccinia virus was more effective than subunit antigen in eliciting protective responses; (ii) while both gp130 and gp160 elicited similar levels of SIV-specific antibodies, gp130 was not as effective as gp160 in protection, indicating a possible role for the transmembrane protein in presenting functionally important epitopes; and (iii) although animals immunized with core antigens failed to generate any neutralizing antibody and were infected upon challenge, their virus load was 50- to 100-fold lower than that of the controls, suggesting the importance of cellular immunity or other core-specific immune responses in controlling acute infection. Complete protection against intravenous infection by the pathogenic uncloned SIVmne was achieved by immunization with both the envelope and the core antigens. These results indicate that immune responses to both antigens may contribute to protection and thus argue for the inclusion of multiple antigens in recombinant vaccine designs.
Collapse
Affiliation(s)
- P S Polacino
- Regional Primate Research Center, University of Washington, Seattle, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ui M, Kuwata T, Igarashi T, Miyazaki Y, Tamaru K, Shimada T, Nakamura M, Uesaka H, Yamamoto H, Hayami M. Protective immunity of gene-deleted SHIVs having an HIV-1 Env against challenge infection with a gene-intact SHIV. J Med Primatol 1999; 28:242-8. [PMID: 10593491 DOI: 10.1111/j.1600-0684.1999.tb00275.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We constructed three simian-human immunodeficiency viruses (SHIVs) lacking regulatory gene(s) and analyzed their induction of protective immunity against challenge infection with gene-intact SHIV in rhesus macaques. Inoculation of SHIV-dn lacking nef and SHIV-drn lacking nef and vpr induced transient viremia, while that of SHIV-dxrn lacking nef, vpr, and vpx induced no viremia. The SHIVs with fewer deletions were more effective in inducing neutralizing antibodies and cytotoxic T lymphocyte responses. When these macaques were challenged with parental gene-intact SHIV-NM-3rN, all the SHIV-dn-vaccinated macaques and two of the four SHIV-drn-vaccinated macaques showed complete resistance. The other two SHIV-drn-vaccinated macaques and all SHIV-dxrn-vaccinated macaques did not show complete resistance, but they did show suppression of replication of the challenge virus. These results suggested that as more genes were deleted, protective immunity was decreased.
Collapse
Affiliation(s)
- M Ui
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Balla-Jhagjhoorsingh SS, Koopman G, Mooij P, Haaksma TGM, Teeuwsen VJP, Bontrop RE, Heeney JL. Conserved CTL Epitopes Shared Between HIV-Infected Human Long-Term Survivors and Chimpanzees. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.4.2308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Certain HIV-1 infected humans that do not progress to AIDS have been documented to share particular MHC class I alleles that appear to correlate with long-term survival. HIV-1-infected chimpanzees are relatively resistant to progression to AIDS. Out of a group of 10 chimpanzees with CTL activity and nonprogressive HIV-1 infection, 2 animals with prominent cytolytic CD3+CD8+ T cell responses to HIV-1 Ags were studied in detail. Characterization of these CTL revealed that they contained the granzymes A and B, T cell intracellular Ag-1, and perforin and induced calcium-dependent cytolysis that correlated with the presence of apoptotic nuclei in target cells. These CTL responses were directed against two gagpeptides, which were found to be identical to previously described epitopes recognized in the context of HLA-B27 and HLA-B57 molecules. The latter two restriction elements occur with increased frequency in human long-term survivor cohorts. Phylogenetic comparisons revealed that the chimpanzee restriction elements, Patr-B*02and -B*03, described here do not show any obvious similarity with the HLA-B*27 and -B*57 alleles, suggesting that CTL responses to HIV-1 in distinct primate species may be controlled by different types of HLA-B-like molecules. The CTL responses in these two chimpanzees are directed, however, against highly conserved epitopes mapping across the majority of HIV-1 clades.
Collapse
Affiliation(s)
| | - Gerrit Koopman
- Departments of Virology and Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Petra Mooij
- Departments of Virology and Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Tom G. M. Haaksma
- Departments of Virology and Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Vera J. P. Teeuwsen
- Departments of Virology and Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Ronald E. Bontrop
- Departments of Virology and Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Jonathan L. Heeney
- Departments of Virology and Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
36
|
Mavoungou E, Toure FS, Yaba P, Délicat A, Poaty-Mavoungou V. Peptide immunization restimulates the memory CD4 T cell response but fails to induce cytotoxic T lymphocytes in cynomolgus monkeys. J Med Primatol 1998; 27:202-9. [PMID: 9879861 DOI: 10.1111/j.1600-0684.1998.tb00072.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A potential strategy to induce peptide specific CTL in vivo was investigated. A synthetic vaccine consisting of an SIV-derived, HLA-A2. 1-binding CTL epitope and a tetanus toxin-derived T helper epitope was evaluated for its capacity to induce peptide-specific CTL in monkeys. Thirteen animals were immunized and boosted twice with 150 microg of CTL plus 250 microg of the T helper peptide (p30). Peripheral blood mononuclear cells (PBMC) were regularly analysed for cytotoxic and proliferative responses before, between, and after the immunizations, and the serum was tested for anti-peptide antibodies. No unequivocal induction of SIV peptide-specific CTL in any of the monkeys was observed. However, a wide pattern of mild and transient side reactions were observed, ranging from local redness at the injection site to generalized exanthema, myalgias, arthralgias, and fever. The side-effects were related to the T helper epitope, as they were similar to the side-effects experienced after tetanus immunization, correlated to the magnitude of the p30-specific in vitro proliferative response, and occurred only if p30 was co-injected. No antibody against the SIV-derived peptides nor against p30 was detectable in the serum after repeated immunizations. The data suggest that the CTL peptide, at the concentration used in this study, failed to induce a cytotoxic immune response in vivo, although the T helper peptide seems to be capable of restimulating the specific memory T cells.
Collapse
Affiliation(s)
- E Mavoungou
- Unit of Immunology, International Center for Medical Research, Franceville, (CIRMF), Gabon.
| | | | | | | | | |
Collapse
|
37
|
Allen TM, Sidney J, del Guercio MF, Glickman RL, Lensmeyer GL, Wiebe DA, DeMars R, Pauza CD, Johnson RP, Sette A, Watkins DI. Characterization of the Peptide Binding Motif of a Rhesus MHC Class I Molecule (Mamu-A*01) That Binds an Immunodominant CTL Epitope from Simian Immunodeficiency Virus. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.12.6062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The majority of immunogenic CTL epitopes bind to MHC class I molecules with high affinity. However, peptides longer or shorter than the optimal epitope rarely bind with high affinity. Therefore, identification of optimal CTL epitopes from pathogens may ultimately be critical for inducing strong CTL responses and developing epitope-based vaccines. The SIV-infected rhesus macaque is an excellent animal model for HIV infection of humans. Although a number of CTL epitopes have been mapped in SIV-infected rhesus macaques, the optimal epitopes have not been well defined, and their anchor residues are unknown. We have now defined the optimal SIV gag CTL epitope restricted by the rhesus MHC class I molecule Mamu-A*01 and defined a general peptide binding motif for this molecule that is characterized by a dominant position 3 anchor (proline). We used peptide elution and sequencing, peptide binding assays, and bulk and clonal CTL assays to demonstrate that the optimal Mamu-A*01-restricted SIV gag CTL epitope was CTPYDINQM181–189. Mamu-A*01 is unique in that it is found at a high frequency in rhesus macaques, and all SIV-infected Mamu-A*01-positive rhesus macaques studied to date develop an immunodominant gag-specific CTL response restricted by this molecule. Identification of the optimal SIV gag CTL epitope will be critical for a variety of studies designed to induce CD8+ CTL responses specific for SIV in the rhesus macaque.
Collapse
Affiliation(s)
| | - John Sidney
- ‡Eppimune, San Diego, CA 92121; Division of Immunology, New England Regional Primate Research Center, Harvard Medical School, Southborough, MA 01772; and
| | - Marie-France del Guercio
- ‡Eppimune, San Diego, CA 92121; Division of Immunology, New England Regional Primate Research Center, Harvard Medical School, Southborough, MA 01772; and
| | - Rhona L. Glickman
- ¶Infectious Disease Unit and Partners AIDS Research Center, Massachusetts General Hospital, Charlestown, MA 02129
| | - Gary L. Lensmeyer
- †Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53715
| | - Donald A. Wiebe
- †Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53715
| | - R. DeMars
- ∥Laboratory of Genetics, University of Wisconsin, Madison, WI 53706
| | - C. David Pauza
- *Wisconsin Regional Primate Research Center and
- †Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53715
| | - R. Paul Johnson
- ¶Infectious Disease Unit and Partners AIDS Research Center, Massachusetts General Hospital, Charlestown, MA 02129
| | - Alessandro Sette
- ‡Eppimune, San Diego, CA 92121; Division of Immunology, New England Regional Primate Research Center, Harvard Medical School, Southborough, MA 01772; and
| | - David I. Watkins
- *Wisconsin Regional Primate Research Center and
- †Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53715
| |
Collapse
|
38
|
Knapp LA, Lehmann E, Piekarczyk MS, Urvater JA, Watkins DI. A high frequency of Mamu-A*01 in the rhesus macaque detected by polymerase chain reaction with sequence-specific primers and direct sequencing. TISSUE ANTIGENS 1997; 50:657-61. [PMID: 9458122 DOI: 10.1111/j.1399-0039.1997.tb02927.x] [Citation(s) in RCA: 165] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIV infection of rhesus macaques is an excellent model for HIV infection of humans. Unfortunately, it is has been difficult to identify macaques expressing particular MHC class I alleles. Here we describe the use of PCR-SSP for Mamu-A*01 typing of rhesus macaques. The Mamu-A*01 allele was amplified from genomic DNA using Mamu-A*01-specific primers and positive PCR products were directly sequenced. Our technique identified 15 Mamu-A*01-positive animals of 68 tested. We validated our molecular analysis by showing that lymphocytes from 8 Mamu-A*01-positive animals expressed Mamu-A*01 as determined by immunoprecipitation and 1-D IEF. The technical simplicity and accuracy of this typing method should facilitate selection of Mamu-A*01-positive rhesus macaques for AIDS virus pathogenesis and vaccine studies.
Collapse
Affiliation(s)
- L A Knapp
- Wisconsin Regional Primate Research Center, University of Wisconsin, Madison 53715, USA
| | | | | | | | | |
Collapse
|