1
|
Ampie L, McGavern DB. Immunological defense of CNS barriers against infections. Immunity 2022; 55:781-799. [PMID: 35545028 PMCID: PMC9087878 DOI: 10.1016/j.immuni.2022.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022]
Abstract
Neuroanatomical barriers with physical, chemical, and immunological properties play an essential role in preventing the spread of peripheral infections into the CNS. A failure to contain pathogens within these barriers can result in very serious CNS diseases. CNS barriers are inhabited by an elaborate conglomerate of innate and adaptive immune cells that are highly responsive to environmental challenges. The CNS and its barriers can also be protected by memory T and B cells elicited by prior infection or vaccination. Here, we discuss the different CNS barriers from a developmental, anatomical, and immunological standpoint and summarize our current understanding of how memory cells protect the CNS compartment. We then discuss a contemporary challenge to CNS-barrier system (SARS-CoV-2 infection) and highlight approaches to promote immunological protection of the CNS via vaccination.
Collapse
Affiliation(s)
- Leonel Ampie
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Department of Surgical Neurology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Computational design of a potential multi-epitope subunit vaccine using immunoinformatics to fight Ebola virus. INFECTION GENETICS AND EVOLUTION 2020; 85:104464. [PMID: 32681997 DOI: 10.1016/j.meegid.2020.104464] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/23/2020] [Accepted: 07/12/2020] [Indexed: 01/11/2023]
Abstract
Ebola virus (EBOV) is a rare but fatal disease that has been a burden to mankind for over 40 years. EBOV exhibits several symptoms including severe bleeding, organ failure and if left untreated causes death. It is assumed that fruit bats of the Pteropodidae family are natural hosts for the virus. Over the years, there has been no effective vaccine that can confer immunity to this virus. Considering the necessity of a vaccine against EBOV, this study to develop a multi-epitope subunit vaccine for the EBOV using the immunoinformatics approach was conducted. The construct was designed using structural and non-structural proteins of EBOV. Class I and Class II MHC epitopes were predicted and linked along with β defensin and compatible linkers. B-cell linear epitopes were also assessed and the physiological parameters of the vaccine were determined. The vaccine was capable of administration to humans and also is capable of an immune response. The vaccine was modeled further and affinity towards the TLR4 receptor was studied by docking and simulation for 20 ns. The trajectory analysis high affinity between the vaccine and the construct with an average hydrogen bond of 18. For ease of purification, the vaccine construct was ligated into pET28a(+) vector with His-tag. Concluding from the results, the vaccine construct has the potentiality to help develop immunity against the Ebola virus. Furthermore, experimental and immunological investigations will be required to verify the feasibility of the multi-epitope subunit construct as a commercial vaccine.
Collapse
|
3
|
Abdulla F, Adhikari UK, Uddin MK. Exploring T & B-cell epitopes and designing multi-epitope subunit vaccine targeting integration step of HIV-1 lifecycle using immunoinformatics approach. Microb Pathog 2019; 137:103791. [PMID: 31606417 DOI: 10.1016/j.micpath.2019.103791] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Abstract
Till now, AIDS, caused by the human immunodeficiency virus (HIV) is still a severe health problem worldwide. It weakens the immune system by targeting the T-helper cells. Specifically, the severity of the pandemic HIV-1 makes the emergence of an enduring effective vaccine against HIV-1. Therefore, we have applied a series of immunoinformatics approaches to the four conserved domains of HIV-1 integrase (IN) proteins to design an effective multi-epitope based subunit vaccine which might induce a competent immunity against HIV-1. Therefore, we have selected three peptide fragments that contained all overlapping epitopes (35 CD4+, 8 CD8+ T-cell epitopes, and 3 B-cell epitopes) where the epitopes had a high conservancy score. The cumulative population coverage for combined CD8+ and CD4+ T-cell epitopes and their respective HLA-alleles were found as 98.03% in the world which is also followed by East Asia (96.24%), South Asia (96.31%), North Africa (96.14%), North America (98.99%), and Europe (98.80%). The proposed vaccine composed by an adjuvant (β-defensin) at the N-terminal site of the vaccine constructs and three peptide fragments where the adjuvant was fused by EAAAK linker and the peptide fragments were fused by GPGPG linker. The designed final vaccine construct (length: 159 amino acid) was found to be antigenic and non-allergic, which indicates its safety. The vaccine construct was found as good antigenic, stable, higher thermostable, and hydrophilic in nature. The codon adaptation and in silico cloning ensured the high expression rate of the vaccine constructs in E. coli K12 with CAI value of 1.0. Finally, the binding affinity of the vaccine constructs with the immune receptor TLR3 was confirmed by the lowest energy score of -1026.8 evaluated by molecular docking. However, the proposed in silico vaccine construct needs experimental validation for assuring the safety and immunogenicity profile which will ensure an active immunity against HIV-1.
Collapse
Affiliation(s)
- Faruq Abdulla
- Department of Statistics, Islamic University, Kushtia-7003, Bangladesh
| | - Utpal Kumar Adhikari
- School of Medicine, Western Sydney University, Campbelltown, NSW-2560, Australia
| | - M Kamal Uddin
- Department of Statistics, Islamic University, Kushtia-7003, Bangladesh.
| |
Collapse
|
4
|
Pichugin A, Zarling S, Perazzo L, Duffy PE, Ploegh HL, Krzych U. Identification of a Novel CD8 T Cell Epitope Derived from Plasmodium berghei Protective Liver-Stage Antigen. Front Immunol 2018; 9:91. [PMID: 29434602 PMCID: PMC5796907 DOI: 10.3389/fimmu.2018.00091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/12/2018] [Indexed: 11/13/2022] Open
Abstract
We recently identified novel Plasmodium berghei (Pb) liver stage (LS) genes that as DNA vaccines significantly reduce Pb LS parasite burden (LPB) in C57Bl/6 (B6) mice through a mechanism mediated, in part, by CD8 T cells. In this study, we sought to determine fine antigen (Ag) specificities of CD8 T cells that target LS malaria parasites. Guided by algorithms for predicting MHC class I-restricted epitopes, we ranked sequences of 32 Pb LS Ags and selected ~400 peptides restricted by mouse H-2Kb and H-2Db alleles for analysis in the high-throughput method of caged MHC class I-tetramer technology. We identified a 9-mer H-2Kb restricted CD8 T cell epitope, Kb-17, which specifically recognized and activated CD8 T cell responses in B6 mice immunized with Pb radiation-attenuated sporozoites (RAS) and challenged with infectious sporozoites (spz). The Kb-17 peptide is derived from the recently described novel protective Pb LS Ag, PBANKA_1031000 (MIF4G-like protein). Notably, immunization with the Kb-17 epitope delivered in the form of a minigene in the adenovirus serotype 5 vector reduced LPB in mice infected with spz. On the basis of our results, Kb-17 peptide was available for CD8 T cell activation and recall following immunization with Pb RAS and challenge with infectious spz. The identification of a novel MHC class I-restricted epitope from the protective Pb LS Ag, MIF4G-like protein, is crucial for advancing our understanding of immune responses to Plasmodium and by extension, toward vaccine development against malaria.
Collapse
Affiliation(s)
- Alexander Pichugin
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Stasya Zarling
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Leah Perazzo
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Patrick Emmet Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, (NIH), Rockville, MD, United States
| | - Hidde Lolke Ploegh
- Program in Cellular and Molecular Medicine, Division of Molecular Biology, Department of Medicine, Boston Children's Hospital, Boston, MD, United States
| | - Urszula Krzych
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
5
|
Khan MA, Hossain MU, Rakib-Uz-Zaman SM, Morshed MN. Epitope-based peptide vaccine design and target site depiction against Ebola viruses: an immunoinformatics study. Scand J Immunol 2015; 82:25-34. [PMID: 25857850 PMCID: PMC7169600 DOI: 10.1111/sji.12302] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/21/2015] [Indexed: 12/28/2022]
Abstract
Ebola viruses (EBOVs) have been identified as an emerging threat in recent year as it causes severe haemorrhagic fever in human. Epitope‐based vaccine design for EBOVs remains a top priority because a mere progress has been made in this regard. Another reason is the lack of antiviral drug and licensed vaccine although there is a severe outbreak in Central Africa. In this study, we aimed to design an epitope‐based vaccine that can trigger a significant immune response as well as to prognosticate inhibitor that can bind with potential drug target sites using various immunoinformatics and docking simulation tools. The capacity to induce both humoral and cell‐mediated immunity by T cell and B cell was checked for the selected protein. The peptide region spanning 9 amino acids from 42 to 50 and the sequence TLASIGTAF were found as the most potential B and T cell epitopes, respectively. This peptide could interact with 12 HLAs and showed high population coverage up to 80.99%. Using molecular docking, the epitope was further appraised for binding against HLA molecules to verify the binding cleft interaction. In addition with this, the allergenicity of the epitopes was also evaluated. In the post‐therapeutic strategy, docking study of predicted 3D structure identified suitable therapeutic inhibitor against targeted protein. However, this computational epitope‐based peptide vaccine designing and target site prediction against EBOVs open up a new horizon which may be the prospective way in Ebola viruses research; the results require validation by in vitro and in vivo experiments.
Collapse
Affiliation(s)
- M A Khan
- Department of Science and Humanities, Military Institute of Science and Technology (MIST), Mirpur Cantonment, Bangladesh
| | - M U Hossain
- Department of Biotechnology and Genetic Engineering, Life Science Faculty, Mawlana Bhashani Science and Technology University, Santosh, Bangladesh
| | - S M Rakib-Uz-Zaman
- Department of Genetic Engineering and Biotechnology, Life Science Faculty, Shahjalal University of Science and Technology, Kumargaon, Bangladesh
| | - M N Morshed
- Department of Science and Humanities, Military Institute of Science and Technology (MIST), Mirpur Cantonment, Bangladesh
| |
Collapse
|
6
|
Villarreal DO, Svoronos N, Wise MC, Shedlock DJ, Morrow MP, Conejo-Garcia JR, Weiner DB. Molecular adjuvant IL-33 enhances the potency of a DNA vaccine in a lethal challenge model. Vaccine 2015; 33:4313-20. [PMID: 25887087 DOI: 10.1016/j.vaccine.2015.03.086] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/26/2015] [Accepted: 03/25/2015] [Indexed: 12/25/2022]
Abstract
Identifying new molecular adjuvants that elicit effective vaccine-induced CD8(+) T cell immunity may be critical for the elimination of many challenging diseases including Tuberculosis, HIV and cancer. Here, we report that co-administration of molecular adjuvant IL-33 during vaccination enhanced the magnitude and function of antigen (Ag)-specific CD8(+) T cells against a model Ag, LCMV NP target protein. These enhanced responses were characterized by higher frequencies of Ag-specific, polyfunctional CD8(+) T cells exhibiting cytotoxic characteristics. Importantly, these cells were capable of robust expansion upon Ag-specific restimulation in vivo and conferred remarkable protection against a high dose lethal LCMV challenge. In addition, we demonstrate the ability of IL-33 to amplifying the frequency of Ag-specific KLRG1(+) effector CD8(+) T cells. These data show that IL-33 is a promising immunoadjuvant at improving T cell immunity in a vaccine setting and suggest further development and understanding of this molecular adjuvant for strategies against many obstinate infectious diseases and cancer.
Collapse
Affiliation(s)
- Daniel O Villarreal
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nikolaos Svoronos
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Tumor Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Megan C Wise
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Devon J Shedlock
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew P Morrow
- Inovio Pharmaceuticals, Inc., 660 West Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - David B Weiner
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
7
|
A comprehensive immunoinformatics and target site study revealed the corner-stone toward Chikungunya virus treatment. Mol Immunol 2015; 65:189-204. [PMID: 25682054 PMCID: PMC7172456 DOI: 10.1016/j.molimm.2014.12.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 01/07/2023]
Abstract
Recent concerning facts of Chikungunya virus (CHIKV); a Togaviridae family alphavirus has proved this as a worldwide emerging threat which causes Chikungunya fever and devitalizing arthritis. Despite severe outbreaks and lack of antiviral drug, a mere progress has been made regarding to an epitope-based vaccine designed for CHIKV. In this study, we aimed to design an epitope-based vaccine that can trigger a significant immune response as well as to prognosticate inhibitor that can bind with potential drug target sites by using various immunoinformatics and docking simulation tools. Initially, whole proteome of CHIKV was retrieved from database and perused to identify the most immunogenic protein. Structural properties of the selected protein were analyzed. The capacity to induce both humoral and cell-mediated immunity by T cell and B cell were checked for the selected protein. The peptide region spanning 9 amino acids from 397 to 405 and the sequence YYYELYPTM were found as the most potential B cell and T cell epitopes respectively. This peptide could interact with as many as 19 HLAs and showed high population coverage ranging from 69.50% to 84.94%. By using in silico docking techniques the epitope was further assessed for binding against HLA molecules to verify the binding cleft interaction. In addition with this, the allergenicity of the epitopes was also evaluated. In the post therapeutic strategy, three dimensional structure was predicted along with validation and verification that resulted in molecular docking study to identify the potential drug binding sites and suitable therapeutic inhibitor against targeted protein. Finally, pharmacophore study was also performed in quest of seeing potent drug activity. However, this computational epitope-based peptide vaccine designing and target site prediction against CHIKV opens up a new horizon which may be the prospective way in Chikungunya virus research; the results require validation by in vitro and in vivo experiments.
Collapse
|
8
|
Hasan MA, Hossain M, Alam MJ. A computational assay to design an epitope-based Peptide vaccine against saint louis encephalitis virus. Bioinform Biol Insights 2013; 7:347-55. [PMID: 24324329 PMCID: PMC3855041 DOI: 10.4137/bbi.s13402] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Saint Louis encephalitis virus, a member of the flaviviridae subgroup, is a culex mosquito-borne pathogen. Despite severe epidemic outbreaks on several occasions, not much progress has been made with regard to an epitope-based vaccine designed for Saint Louis encephalitis virus. The envelope proteins were collected from a protein database and analyzed with an in silico tool to identify the most immunogenic protein. The protein was then verified through several parameters to predict the T-cell and B-cell epitopes. Both T-cell and B-cell immunity were assessed to determine that the protein can induce humoral as well as cell-mediated immunity. The peptide sequence from 330-336 amino acids and the sequence REYCYEATL from the position 57 were found as the most potential B-cell and T-cell epitopes, respectively. Furthermore, as an RNA virus, one important thing was to establish the epitope as a conserved one; this was also done by in silico tools, showing 63.51% conservancy. The epitope was further tested for binding against the HLA molecule by computational docking techniques to verify the binding cleft epitope interaction. However, this is a preliminary study of designing an epitope-based peptide vaccine against Saint Louis encephalitis virus; the results awaits validation by in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md Anayet Hasan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | | | | |
Collapse
|
9
|
Hosking MP, Flynn CT, Botten J, Whitton JL. CD8+ memory T cells appear exhausted within hours of acute virus infection. THE JOURNAL OF IMMUNOLOGY 2013; 191:4211-22. [PMID: 24026080 DOI: 10.4049/jimmunol.1300920] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CD8(+) memory T cells are abundant and are activated in a near-synchronous manner by infection, thereby providing a unique opportunity to evaluate the coordinate functional and phenotypic changes that occur in vivo within hours of viral challenge. Using two disparate virus challenges of mice, we show that splenic CD8(+) memory T cells rapidly produced IFN-γ in vivo; however, within 18-24 h, IFN-γ synthesis was terminated and remained undetectable for ≥ 48 h. A similar on/off response was observed in CD8(+) memory T cells in the peritoneal cavity. Cessation of IFN-γ production in vivo occurred despite the continued presence of immunostimulatory viral Ag, indicating that the initial IFN-γ response had been actively downregulated and that the cells had been rendered refractory to subsequent in vivo Ag contact. Downregulation of IFN-γ synthesis was accompanied by the upregulation of inhibitory receptor expression on the T cells, and ex vivo analyses using synthetic peptides revealed a concurrent hierarchical loss of cytokine responsiveness (IL-2, then TNF, then IFN-γ) taking place during the first 24 h following Ag contact. Thus, within hours of virus challenge, CD8(+) memory T cells display the standard hallmarks of T cell exhaustion, a phenotype that previously was associated only with chronic diseases and that is generally viewed as a gradually developing and pathological change in T cell function. Our data suggest that, instead, the "exhaustion" phenotype is a rapid and normal physiological T cell response.
Collapse
Affiliation(s)
- Martin P Hosking
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | | | | | | |
Collapse
|
10
|
Islam R, Sakib MS, Zaman A. A computational assay to design an epitope-based peptide vaccine against chikungunya virus. Future Virol 2012. [DOI: 10.2217/fvl.12.95] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Chikungunya virus, an arthropod-borne alphavirus, belongs to the Togavirus family. Despite severe epidemic outbreaks on several occasions, not much progress has been made with regard to epitope-based drug design for chikungunya virus. In this study we performed a proteome-wide search to look for a conserved region among the available viral proteins, one which has the capacity to trigger a significant immune response. Materials & methods: The conserved region was analyzed by performing an alignment of sequences collected from sources from varied geographic locations and time periods. Subsequently, the immune parameters for the peptide sequences were determined using several in silico tools and immune databases. Results: Both T-cell immunity and B-cell immunity were checked for the peptides to ensure that they had the capacity to induce both humoral and cell-based immunity. Our study reveals a stretch of conserved region in glycoprotein E2; yet this peptide sequence could interact with as many as seven HLAs and showed population coverage as high as 73.46%. The epitope was further tested for binding against the HLA structure using in silico docking techniques to validate the binding cleft epitope interaction in detail. Conclusion: Although the study requires further in vivo screening, keeping in mind the consistency and reproducibility of the immune system at selecting and reacting to peptide epitopes, this study allows us to claim a novel peptide antigen target in E2 protein with good confidence.
Collapse
Affiliation(s)
- Rezaul Islam
- Department of Biochemistry & Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - M Sadman Sakib
- Department of Biochemistry & Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Aubhishek Zaman
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
11
|
Memory CD8+ T cells specific for a single immunodominant or subdominant determinant induced by peptide-dendritic cell immunization protect from an acute lethal viral disease. J Virol 2012; 86:9748-59. [PMID: 22740418 DOI: 10.1128/jvi.00981-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The antigens recognized by individual CD8(+) T cells are small peptides bound to major histocompatibility complex (MHC) class I molecules. The CD8(+) T cell response to a virus is restricted to several peptides, and the magnitudes of the effector as well as memory phases of the response to the individual peptides are generally hierarchical. The peptide eliciting a stronger response is called immunodominant (ID), and those with smaller-magnitude responses are termed subdominant (SD). The relative importance of ID and SD determinants in protective immunity remains to be fully elucidated. We previously showed that multispecific memory CD8(+) T cells can protect susceptible mice from mousepox, an acute lethal viral disease. It remained unknown, however, whether CD8(+) T cells specific for single ID or SD peptides could be protective. Here, we demonstrate that immunization with dendritic cells pulsed with ID and some but not all SD peptides induces memory CD8(+) T cells that are fully capable of protecting susceptible mice from mousepox. Additionally, while natural killer (NK) cells are essential for the natural resistance of nonimmune C57BL/6 (B6) to mousepox, we show that memory CD8(+) T cells of single specificity also protect B6 mice depleted of NK cells. This suggests it is feasible to produce effective antiviral CD8(+) T cell vaccines using single CD8(+) T cell determinants and that NK cells are no longer essential when memory CD8(+) T cells are present.
Collapse
|
12
|
A highly optimized DNA vaccine confers complete protective immunity against high-dose lethal lymphocytic choriomeningitis virus challenge. Vaccine 2011; 29:6755-62. [PMID: 21238574 DOI: 10.1016/j.vaccine.2010.12.064] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protection against infection is the hallmark of immunity and the basis of effective vaccination. For a variety of reasons there is a great demand to develop new, safer and more effective vaccine platforms. In this regard, while 'first-generation' DNA vaccines were poorly immunogenic, new genetic 'optimization' strategies and the application of in vivo electroporation (EP) have dramatically boosted their potency. We developed a highly optimized plasmid DNA vaccine that expresses the lymphocytic choriomeningitis virus (LCMV) nucleocapsid protein (NP) and evaluated it using the LCMV challenge model, a gold standard for studying infection and immunity. When administered intramuscularly with EP, robust NP-specific cellular and humoral immune responses were elicited, the magnitudes of which approached those following acute LCMV infection. Furthermore, these responses were capable of providing 100% protection against a high-dose, normally lethal virus challenge. This is the first non-infectious vaccine conferring complete protective immunity up to 8 weeks after vaccination and demonstrates the potential of 'next-generation' DNA vaccines.
Collapse
|
13
|
A multivalent vaccination strategy for the prevention of Old World arenavirus infection in humans. J Virol 2010; 84:9947-56. [PMID: 20668086 DOI: 10.1128/jvi.00672-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Arenaviruses cause severe human disease ranging from aseptic meningitis following lymphocytic choriomeningitis virus (LCMV) infection to hemorrhagic fever syndromes following infection with Guanarito virus (GTOV), Junin virus (JUNV), Lassa virus (LASV), Machupo virus (MACV), Sabia virus (SABV), or Whitewater Arroyo virus (WWAV). Cellular immunity, chiefly the CD8(+) T-cell response, plays a critical role in providing protective immunity following infection with the Old World arenaviruses LASV and LCMV. In the current study, we evaluated whether HLA class I-restricted epitopes that are cross-reactive among pathogenic arenaviruses could be identified for the purpose of developing an epitope-based vaccination approach that would cross-protect against multiple arenaviruses. We were able to identify a panel of HLA-A*0201-restricted peptides derived from the same region of the glycoprotein precursor (GPC) of LASV (GPC spanning residues 441 to 449 [GPC(441-449)]), LCMV (GPC(447-455)), JUNV (GPC(429-437)), MACV (GPC(444-452)), GTOV (GPC(427-435)), and WWAV (GPC(428-436)) that displayed high-affinity binding to HLA-A*0201 and were recognized by CD8(+) T cells in a cross-reactive manner following LCMV infection or peptide immunization of HLA-A*0201 transgenic mice. Immunization of HLA-A*0201 mice with the Old World peptide LASV GPC(441-449) or LCMV GPC(447-455) induced high-avidity CD8(+) T-cell responses that were able to kill syngeneic target cells pulsed with either LASV GPC(441-449) or LCMV GPC(447-455) in vivo and provided significant protection against viral challenge with LCMV. Through this study, we have demonstrated that HLA class I-restricted, cross-reactive epitopes exist among diverse arenaviruses and that individual epitopes can be utilized as effective vaccine determinants for multiple pathogenic arenaviruses.
Collapse
|
14
|
Kang YJ, Wang X, Lin SJ, Hsu YM, Chang HC. An active CD8alpha/pMHCI interaction is required for CD8 single positive thymocyte differentiation. Eur J Immunol 2010; 40:836-48. [PMID: 19950178 DOI: 10.1002/eji.200939663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recognition of viral antigenic peptides bound to major histocompatibility complex class I molecules (MHCI) by TCR is critical for initiating the responses of CD8(+) T cells that ultimately lead to elimination of virus-infected cells. This antigen recognition is enhanced by the CD8 coreceptor through its interaction with the peptide-MHCI complexes (pMHCI). Mouse CD8alphabeta can form two different complexes with pMHCI via either the CD8alpha- or CD8beta-dominated interaction. To understand the functional significance of these complexes in vivo, we generated Tg mice carrying a variant CD8alphabeta (CD8alpha(m3)beta) capable of forming only the CD8beta-dominated CD8alphabeta/pMHCI complex. These mice show sub-optimal thymic differentiation with reduced populations of CD8(+) single-positive thymocytes. Tg CD8(+) T cells exhibit a compromised developmental capacity when competing with CD8(+) T cells from B6 mice in mixed bone marrow chimera experiments. However, once these CD8(+) T cells have emigrated to the peripheral lymphoid organs, they exhibit normal effector function against viral infection. Our observations indicate that, in addition to the CD8 activity conferred by CD8beta-dominated CD8alphabeta/pMHCI complexes, full thymocyte differentiation requires additional coreceptor activities conferred by CD8alphaalpha and/or CD8alphabeta with CD8alpha-dominated CD8/pMHCI complexes.
Collapse
Affiliation(s)
- Yoon-Joong Kang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | | | | | | | | |
Collapse
|
15
|
Highly efficient antiviral CD8+ T-cell induction by peptides coupled to the surfaces of liposomes. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1383-92. [PMID: 19675224 DOI: 10.1128/cvi.00116-09] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In previous studies, we have demonstrated that liposomes with differential lipid components display differential adjuvant effects when antigens (Ags) are chemically coupled to their surfaces. When ovalbumin was coupled to liposomes made by using unsaturated fatty acids, it was found to be presented not only to CD4(+) T cells but also to CD8(+) T cells and induced cytotoxic T lymphocytes (CTLs) which effectively eradicated the tumor from mice. In this study, we coupled liposomes to immunodominant CTL epitope peptides derived from lymphocytic choriomeningitis virus (LCMV) and evaluated its potency as an antiviral vaccine. The intramuscular immunization of mice with the peptide-liposome conjugates along with CpG resulted in the efficient induction of antiviral CD8(+) T-cell responses which conferred complete protection against not only LCMV Armstrong but also a highly virulent mutant strain, clone 13, that establishes persistent infections in immunocompetent mice. The intranasal vaccination induced mucosal immunity effective enough to protect mice from the virus challenge via the same route. Complete protection was achieved in mice even when the Ag dose was reduced to as low as 280 ng of liposomal peptide. This form of vaccination with a single CTL epitope induced Ag-specific memory CD8(+) T cells in the absence of CD4(+) T-cell help, which could be shown by the complete protection of CD4-knockout mice in 10 weeks as well as by the analysis of recall responses. Thus, surface-linked liposomal peptide might have a potential advantage for the induction of antiviral immunity.
Collapse
|
16
|
Immune system derived from homeostatic proliferation generates normal CD8 T-cell memory but altered repertoires and diminished heterologous immune responses. Blood 2008; 112:680-9. [PMID: 18509089 DOI: 10.1182/blood-2008-01-132464] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The host responds to lymphopenic environments by acute homeostatic proliferation of T lymphocytes, which acquire phenotypes similar to memory cells. Using T-cell knockout (KO) mice adoptively reconstituted with splenocytes from immunologically naive mice, we examined the immune responses of an immune system derived from homeostatically proliferating (HP) T cells. HP cells mounted relatively normal acute CD8 T-cell responses to lymphocytic choriomeningitis virus (LCMV), but with altered T-cell receptor (TCR) repertoires, and they became functional memory cells capable of recall responses. Although homeostatic proliferation does not normally fully restore T-cell numbers, the CD8(+) T-cell pool was completely restored in T-cell KO mice after LCMV infection. CD4 T-cell responses were lower and not fully restored but seemed sufficient to allow for complete differentiation of CD8 memory T cells. The LCMV-immune HP mouse had an immune repertoire heavily biased with LCMV epitope-specific T cells with oligoclonal expansions. LCMV-immune HP mice had reduced cross-reactive and non-cross-reactive CD8 T-cell responses when challenged with a T cell-cross-reactive virus. Thus, whereas an HP immune system is capable of mounting relatively normal acute and memory CD8 T-cell responses, the narrowing of the T-cell repertoire may reduce immune responses to subsequently encountered pathogens.
Collapse
|
17
|
Heiny AT, Miotto O, Srinivasan KN, Khan AM, Zhang GL, Brusic V, Tan TW, August JT. Evolutionarily conserved protein sequences of influenza a viruses, avian and human, as vaccine targets. PLoS One 2007; 2:e1190. [PMID: 18030326 PMCID: PMC2065905 DOI: 10.1371/journal.pone.0001190] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 10/17/2007] [Indexed: 01/16/2023] Open
Abstract
Background Influenza A viruses generate an extreme genetic diversity through point mutation and gene segment exchange, resulting in many new strains that emerge from the animal reservoirs, among which was the recent highly pathogenic H5N1 virus. This genetic diversity also endows these viruses with a dynamic adaptability to their habitats, one result being the rapid selection of genomic variants that resist the immune responses of infected hosts. With the possibility of an influenza A pandemic, a critical need is a vaccine that will recognize and protect against any influenza A pathogen. One feasible approach is a vaccine containing conserved immunogenic protein sequences that represent the genotypic diversity of all current and future avian and human influenza viruses as an alternative to current vaccines that address only the known circulating virus strains. Methodology/Principal Findings Methodologies for large-scale analysis of the evolutionary variability of the influenza A virus proteins recorded in public databases were developed and used to elucidate the amino acid sequence diversity and conservation of 36,343 sequences of the 11 viral proteins of the recorded virus isolates of the past 30 years. Technologies were also applied to identify the conserved amino acid sequences from isolates of the past decade, and to evaluate the predicted human lymphocyte antigen (HLA) supertype-restricted class I and II T-cell epitopes of the conserved sequences. Fifty-five (55) sequences of 9 or more amino acids of the polymerases (PB2, PB1, and PA), nucleoprotein (NP), and matrix 1 (M1) proteins were completely conserved in at least 80%, many in 95 to 100%, of the avian and human influenza A virus isolates despite the marked evolutionary variability of the viruses. Almost all (50) of these conserved sequences contained putative supertype HLA class I or class II epitopes as predicted by 4 peptide-HLA binding algorithms. Additionally, data of the Immune Epitope Database (IEDB) include 29 experimentally identified HLA class I and II T-cell epitopes present in 14 of the conserved sequences. Conclusions/Significance This study of all reported influenza A virus protein sequences, avian and human, has identified 55 highly conserved sequences, most of which are predicted to have immune relevance as T-cell epitopes. This is a necessary first step in the design and analysis of a polyepitope, pan-influenza A vaccine. In addition to the application described herein, these technologies can be applied to other pathogens and to other therapeutic modalities designed to attack DNA, RNA, or protein sequences critical to pathogen function.
Collapse
Affiliation(s)
- A. T. Heiny
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Olivo Miotto
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Systems Science, National University of Singapore, Singapore, Singapore
| | - Kellathur N. Srinivasan
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Maryland, United States of America
- Product Evaluation and Registration Division, Centre for Drug Administration, Health Sciences Authority, Singapore, Singapore
| | - Asif M. Khan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - G. L. Zhang
- Institute for Infocomm Research, Singapore, Singapore
| | - Vladimir Brusic
- Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Tin Wee Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - J. Thomas August
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Maryland, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
18
|
Doherty PC, Tripp RA, Sixbey JW. Evasion of host immune responses by tumours and viruses. CIBA FOUNDATION SYMPOSIUM 2007; 187:245-56; discussion 256-60. [PMID: 7796674 DOI: 10.1002/9780470514672.ch16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Viruses and tumours use various mechanisms to avoid immune surveillance. Oncogenic viruses have achieved a balance with the immune system through evolutionary time to ensure long-term persistence. Mutations that promote escape mechanisms favouring tumour growth to the detriment of host survival through reproductive age offer no selective advantage and will not generally be maintained in the viral genome that persists in nature. Conventional (non-oncogenic) and tumour viruses interact with various immune mediators and T cells in different ways. Oncogenic viruses cannot operate solely in the context of a lytic cycle, though this may be characteristic of the initial phase of infection that is limited by the acute immune response. Some oncogenic viruses interact with normal cellular growth control and signalling mechanisms. Synthesis of key viral proteins may be tightly controlled in replicating cells that are subject to T cell surveillance, such as basal epithelia, while productive infection occurs in non-proliferating progeny that are lost under normal physiological conditions, such as desquamating epithelia. Tumorigenesis may be an aberrant consequence of the molecular mechanisms needed to maintain this pattern of viral growth regulation in the context of the cell cycle. Vaccines designed to limit the acute phase of infection with cell-free oncogenic viruses should be as effective as those for conventional viruses.
Collapse
Affiliation(s)
- P C Doherty
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38104, USA
| | | | | |
Collapse
|
19
|
Botten JW, Kotturi MF. Adaptive immunity to Lymphocytic choriomeningitis virus: new insights into antigenic determinants. Future Virol 2007. [DOI: 10.2217/17460794.2.5.495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lymphocytic choriomeningitis virus (LCMV) is one of the most studied infectious disease models in mice. Human infection with LCMV can result in severe disease, ranging from aseptic meningitis in immunocompetent individuals, hydrocephalus, chorioretinitis or microcephaly in fetal infection, or to a highly lethal outcome in immunosuppressed individuals. This review examines recent advances in our understanding of the adaptive immune response to LCMV and how the cell-mediated and humoral immune responses contribute to protective immunity. New insights into the antigenicity of the LCMV proteome and the complexity of the cell-mediated immune response are addressed. We also discuss state-of-the-art approaches for T-cell epitope discovery in murine and human backgrounds and their recent application to LCMV. New findings regarding CD4+ T-cell dysregulation during chronic LCMV infection, and potential avenues for the treatment of chronic viral infection through modulation of the programmed cell death-1 receptor and/or IL-10 signaling pathways, are also evaluated.
Collapse
Affiliation(s)
- Jason W Botten
- The Scripps Research Institute, Molecular & Integrative Neurosciences Department, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maya F Kotturi
- La Jolla Institute for Allergy & Immunology, Division of Vaccine Discovery, 9420 Athena Circle, La Jolla, CA 92037, USA
| |
Collapse
|
20
|
Holst PJ, Bartholdy C, Stryhn A, Thomsen AR, Christensen JP. Rapid and sustained CD4+ T-cell-independent immunity from adenovirus-encoded vaccine antigens. J Gen Virol 2007; 88:1708-1716. [PMID: 17485530 DOI: 10.1099/vir.0.82727-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Many novel vaccine strategies rely on recombinant viral vectors for antigen delivery, and adenovirus vectors have emerged among the most potent of these. In this report, we have compared the immune response induced through priming with adenovirus vector-encoded full-length viral protein to that elicited with an adenovirus-encoded minimal epitope covalently linked to β
2-microglobulin. We demonstrate that the β
2-microglobulin-linked epitope induced an accelerated and augmented CD8+ T-cell response. Furthermore, the immunity conferred by vaccination with β
2-microglobulin-linked lymphocytic choriomeningitis virus (LCMV)-derived epitopes was long-lived and protective. Notably, in contrast to full-length protein, the response elicited with the β
2-microglobulin-linked LCMV-derived epitope was CD4+ T-cell independent. Furthermore, virus-specific CD8+ T cells primed in the absence of CD4+ T-cell help were sustained in the long term and able to expand and control a secondary challenge with LCMV. Our results demonstrate that modifications to the antigen used in adenovirus vaccines may be used to improve the induced T-cell response. Such a strategy for CD4+ T-cell-independent immunity from adenovirus vectors offers prospects for vaccination against opportunistic pathogens in AIDS patients and possibly immunotherapy in chronic virus infections.
Collapse
Affiliation(s)
- Peter J Holst
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Christina Bartholdy
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Annette Stryhn
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Allan R Thomsen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | - Jan P Christensen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, The Panum Institute Building 22.5, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
21
|
Harrison JM, Bertram EM, Ramshaw IA. Exploiting 4-1BB Costimulation for Enhancing Antiviral Vaccination. Viral Immunol 2006; 19:593-601. [PMID: 17201654 DOI: 10.1089/vim.2006.19.593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
4-1BB, a member of the tumor necrosis factor receptor (TNFR) superfamily, is emerging as an important costimulatory molecule, particularly in the regulation of CD8(+) T cell responses. Costimulation through 4-1BB, such as by utilizing agonistic anti-4-1BB monoclonal antibodies, has been well studied in various tumor models. However, 4-1BB is also an important regulator of antiviral CD8(+) T cell responses. This review summarizes these findings and describes how 4-1BB is beginning to be exploited in terms of boosting antiviral vaccine responses.
Collapse
Affiliation(s)
- Jodie M Harrison
- Department of Immunology and Genetics, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | | | | |
Collapse
|
22
|
Tang J, Murtadha M, Schnell M, Eisenlohr LC, Hooper J, Flomenberg P. Human T-cell responses to vaccinia virus envelope proteins. J Virol 2006; 80:10010-20. [PMID: 17005679 PMCID: PMC1617304 DOI: 10.1128/jvi.00601-06] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
One approach for a safer smallpox vaccine is to utilize recombinant subunits rather than live vaccinia virus (VACV). The products of the VACV envelope genes A27L, L1R, B5R, and A33R induce protective antibodies in animal models. We propose that proteins that elicit T-cell responses, as well as neutralizing antibodies, will be important to include in a molecular vaccine. To evaluate VACV-specific memory T-cell responses, peripheral blood mononuclear cells (PBMC) from four VACV vaccinees were tested against whole VACV and the individual envelope proteins A27, B5, L1, and A33, using gamma interferon enzyme-linked immunospot and cytokine flow cytometry assays. PBMC were stimulated with autologous dendritic cells infected with VACV or electroporated with individual VACV protein mRNAs. T-cell lines from all donors, vaccinated from 1 month to over 20 years ago, recognized all four VACV envelope proteins. Both CD4(+) and CD8(+) T-cell responses to each protein were detected. Further analysis focused on representative proteins B5 and A27. PBMC from a recent vaccinee exhibited high frequencies of CD4(+) and CD8(+) T-cell precursors to both B5 (19.8 and 20%, respectively) and A27 (6.8 and 3.7%). In comparison, B5- and A27-specific T-cell frequencies ranged from 0.4 to 1.3% in a donor vaccinated 3 years ago. Multiple CD4(+) and CD8(+) T-cell epitopes were identified from both A27 and B5, using overlapping 15-mer peptides. These data suggest that all four VACV envelope proteins may contribute to protective immunity, not only by inducing antibody responses, but also by eliciting T-cell responses.
Collapse
Affiliation(s)
- Jie Tang
- Thomas Jefferson University, 1020 Locust Street, Rm. 329, Philadelphia, PA 19107, USA
| | | | | | | | | | | |
Collapse
|
23
|
Mandal M, Kawamura KS, Wherry EJ, Ahmed R, Lee KD. Cytosolic delivery of viral nucleoprotein by listeriolysin O-liposome induces enhanced specific cytotoxic T lymphocyte response and protective immunity. Mol Pharm 2005; 1:2-8. [PMID: 15832496 DOI: 10.1021/mp034021m] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cytotoxic T lymphocytes (CTLs) are capable of conferring protection against intracellular pathogens and tumor. Protective antiviral immunity, mediated by the activation of antigenic epitope-specific CTL, can be achieved by delivering exogenous antigen into the cytosol of antigen-presenting cells. Cytosolic introduction of vaccine antigen, however, requires a specialized delivery strategy due to the membrane barrier limiting the access of macromolecules to the cytosol. In this study, we have investigated the potential ability of listeriolysin O-containing liposomes (LLO-liposomes) to deliver lymphocytic choriomeningitis virus (LCMV) nucleoprotein (NP), harnessing the intracellular invasion mechanism of Listeria monocytogenes, to stimulate a NP-specific CTL response. We have analyzed the ability of LLO-liposomes to induce an enhanced CTL response and determined the extent of CTL-mediated protection using an in vivo infection model. Mice immunized with LLO-liposomes containing NP generated a higher frequency of NP-specific CD8+ T cells with greater effector activity than the control groups immunized with either non-LLO-liposomal NP or LLO-liposomes containing control protein. Moreover, LLO-liposomal NP-immunized mice were completely protected against a lethal intracerebral challenge with a virulent strain of LCMV and were capable of clearing a chronic LCMV infection. Our study demonstrates that LLO-liposomes can be used as an efficient vaccine delivery system carrying a viral antigenic protein to generate protective antiviral immunity.
Collapse
Affiliation(s)
- Manas Mandal
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
24
|
Rodriguez-Carreno MP, Nelson MS, Botten J, Smith-Nixon K, Buchmeier MJ, Whitton JL. Evaluating the immunogenicity and protective efficacy of a DNA vaccine encoding Lassa virus nucleoprotein. Virology 2005; 335:87-98. [PMID: 15823608 DOI: 10.1016/j.virol.2005.01.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2004] [Revised: 01/04/2005] [Accepted: 01/14/2005] [Indexed: 10/25/2022]
Abstract
Several viruses in the Arenavirus genus of the family Arenaviridae cause severe, often fatal, hemorrhagic fever. One such virus, Lassa virus (LV), is a frequent cause of disease in Africa, and survivors often are left with substantial neurological impairment. The feasibility of protective immunization against LV infection, and the associated disease, has been demonstrated in animal models, using recombinant vaccinia viruses to deliver Lassa proteins. Circumstantial evidence implicates cellular immunity in this Lassa-induced protection, but this has not been confirmed. Here, we describe DNA vaccines that encode LV proteins. A single inoculation of a plasmid encoding full-length Lassa nucleoprotein (LNP) can induce CD8(+) T cell responses in mice and can protect against challenge with two arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Pichinde virus (PV). A DNA minigene vaccine encoding a 9 amino acid sequence from LNP also induces CD8(+) T cells and protects against arenavirus challenge, thus confirming prior speculation that protective cellular immunity is induced by LV proteins.
Collapse
Affiliation(s)
- Maria P Rodriguez-Carreno
- Department of Neuropharmacology, CVN-9, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Epitope based minigenes (epigenes) have been under investigation for several years as an experimental approach to vaccination against infectious diseases. The essence of this technology is that short DNA sequences, encoding well-defined cytotoxic T-lymphocyte- (CTL), antibody- (Ab) or helper T-lymphocyte- (HTL) specific epitopes are used as immunogens. Compared to other vaccine strategies, several potential advantages are apparent. These include the increased 'safety' of an immunisation strategy that mimics antigen processing and presentation during natural infections, without actually causing disease, and the 'flexibility' in epitope selection, which allows induction and optimisation of the desired type of immunity. In addition, the 'high immunogenicity' of epitope based constructs relative to constructs based on whole antigenic proteins is an important factor. This paper presents and discusses recent developments in the use of minigenes or multiple epitope genes that allow vaccines to be designed. The preclinical studies available to date clearly demonstrate the great potential of this vaccine approach, in terms of both prophylaxis and therapy.
Collapse
Affiliation(s)
- L L An
- Ixsys, Inc., 3520 Dunhill Street, San Diego, CA, 92121, USA.
| | | |
Collapse
|
26
|
Smith MZ, Dale CJ, De Rose R, Stratov I, Fernandez CS, Brooks AG, Weinfurter J, Krebs K, Riek C, Watkins DI, O'connor DH, Kent SJ. Analysis of pigtail macaque major histocompatibility complex class I molecules presenting immunodominant simian immunodeficiency virus epitopes. J Virol 2005; 79:684-95. [PMID: 15613296 PMCID: PMC538543 DOI: 10.1128/jvi.79.2.684-695.2005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Accepted: 09/03/2004] [Indexed: 11/20/2022] Open
Abstract
Successful human immunodeficiency virus (HIV) vaccines will need to induce effective T-cell immunity. We studied immunodominant simian immunodeficiency virus (SIV) Gag-specific T-cell responses and their restricting major histocompatibility complex (MHC) class I alleles in pigtail macaques (Macaca nemestrina), an increasingly common primate model for the study of HIV infection of humans. CD8+ T-cell responses to an SIV epitope, Gag164-172KP9, were present in at least 15 of 36 outbred pigtail macaques. The immunodominant KP9-specific response accounted for the majority (mean, 63%) of the SIV Gag response. Sequencing from six macaques identified 7 new Mane-A and 13 new Mane-B MHC class I alleles. One new allele, Mane-A*10, was common to four macaques that responded to the KP9 epitope. We adapted reference strand-mediated conformational analysis (RSCA) to MHC class I genotype M. nemestrina. Mane-A*10 was detected in macaques presenting KP9 studied by RSCA but was absent from non-KP9-presenting macaques. Expressed on class I-deficient cells, Mane-A*10, but not other pigtail macaque MHC class I molecules, efficiently presented KP9 to responder T cells, confirming that Mane-A*10 restricts the KP9 epitope. Importantly, naive pigtail macaques infected with SIVmac251 that respond to KP9 had significantly reduced plasma SIV viral levels (log10 0.87 copies/ml; P=0.025) compared to those of macaques not responding to KP9. The identification of this common M. nemestrina MHC class I allele restricting a functionally important immunodominant SIV Gag epitope establishes a basis for studying CD8+ T-cell responses against AIDS in an important, widely available nonhuman primate species.
Collapse
Affiliation(s)
- Miranda Z Smith
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sullivan BM, Juedes A, Szabo SJ, von Herrath M, Glimcher LH. Antigen-driven effector CD8 T cell function regulated by T-bet. Proc Natl Acad Sci U S A 2003; 100:15818-23. [PMID: 14673093 PMCID: PMC307651 DOI: 10.1073/pnas.2636938100] [Citation(s) in RCA: 328] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Type 1 immunity relies on the differentiation of two major subsets of T lymphocytes, the CD4+ T helper (Th) cell and the CD8+ cytotoxic T cell, that direct inflammatory and cytotoxic responses essential for the destruction of intracellular and extracellular pathogens. In contrast to CD4 cells, little is known about transcription factors that control the transition from the CD8 naïve to effector cell stage. Here, we report that the transcription factor T-bet, known to regulate Th cell differentiation, also controls the generation of the CD8+ cytotoxic effector cell. Antigen-driven generation of effector CD8+ cells was impaired in OT-I T cell receptor transgenic mice lacking T-bet, resulting in diminished cytotoxicity and a marked shift in cytokine secretion profiles. Furthermore, mice lacking T-bet responded poorly to infection with lymphocytic choriomeningitis virus. T-bet is a key player in the generation of type 1 immunity, in both Th and T cytotoxic cells.
Collapse
Affiliation(s)
- Brandon M Sullivan
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
The conventional, currently available vaccines, though quite successful, suffer from a few shortcomings which hamper future vaccine development. We present herewith some of the new approaches that are presently being pursued, including (1) the development of recombinant, or genetically engineered, vaccines which are based either on the expression of the relevant protective antigen and its formulation into vaccine, or the production of live vaccines, where an appropriate live vector (virus or bacterium) presents the foreign antigen. (2) The development of naked DNA vaccines that include the gene(s) coding for the relevant protective antigen(s). (3) Peptide vaccines that include defined B cell and T cell epitopes, either in a chemically synthesized molecule or in a synthetic recombinant construct. The efficacy of such vaccines is usually dependent on adequate presentation and delivery, namely, carrier/adjuvant technology. (4) Therapeutic vaccines, based on all of the above approaches, may be applied for chronic or long-term infections, or for noninfectious diseases including autoimmune diseases, various neurological disorders, allergy and cancer.
Collapse
Affiliation(s)
- Ruth Arnon
- Department of Immunology, The Weizmann Institute, PO Box 26, 76100 Rehovot, Israel.
| | | |
Collapse
|
29
|
Arribillaga L, de Cerio ALD, Sarobe P, Casares N, Gorraiz M, Vales A, Bruna-Romero O, Borrás-Cuesta F, Paranhos-Baccala G, Prieto J, Ruiz J, Lasarte JJ. Vaccination with an adenoviral vector encoding hepatitis C virus (HCV) NS3 protein protects against infection with HCV-recombinant vaccinia virus. Vaccine 2002; 21:202-10. [PMID: 12450695 DOI: 10.1016/s0264-410x(02)00456-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular immune response plays an important role in the clearance of hepatitis C virus (HCV). Thus, development of efficient ways to induce anti-viral cellular immune responses is an important step toward prevention and/or treatment of HCV infection. With this aim, we have constructed a replication-deficient recombinant adenovirus expressing HCV NS3 protein (RAdNS3). The efficacy of RAdNS3 was tested in vivo by measuring the protection against infection with a recombinant vaccinia virus expressing HCV-polyprotein (vHCV1-3011). Immunisation with 10(9)pfu of RAdNS3 induced anti-NS3 humoral, T helper and T cytotoxic responses. We identified eight epitopes recognised by IFN-gamma producing cells, five of them exhibiting lytic activity. Moreover, we show that RAdNS3 immunised mice were protected against challenge with vHCV1-3011 and that this protection was mediated by CD8(+) cells. In conclusion, our results suggest that adenoviral vectors encoding NS3 might be useful for the induction of prophylactic and/or therapeutic anti-HCV immunity.
Collapse
Affiliation(s)
- Laura Arribillaga
- Department of Internal Medicine, Centro de Investigaciones Médicas Aplicadas (CIMA), University of Navarra, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Oldstone MBA. Biology and pathogenesis of lymphocytic choriomeningitis virus infection. Curr Top Microbiol Immunol 2002; 263:83-117. [PMID: 11987822 DOI: 10.1007/978-3-642-56055-2_6] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- M B A Oldstone
- Division of Virology, Department of Neuropharmacology, Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
31
|
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, CVN-9, Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
32
|
Abstract
Listeria monocytogenes (Lm) is an attractive vector to elicit T cell immunity because it infects antigen-presenting cells and because infection originates at the mucosa. Lm expressing HIV gag elicits sustained high levels of gag-specific CTL in mice. Since Lm causes disease in immunocompromised hosts, a highly attenuated strain of Lm that requires D-Ala for viability was produced. Attenuated bacteria expressing HIV-1 gag (Lmdd-gag) are as efficient as wild-type recombinants at stimulating gag-specific murine CTL when administered with D-Ala and at boosting human CTL in vitro. Lmdd-gag immunization protects mice from vaccinia-gag challenge and induces mucosal CTL, even after systemic immunization.
Collapse
Affiliation(s)
- Judy Lieberman
- The Center for Blood Research and Department of Pediatrics, Harvard Medical School, 800 Huntington Avenue, Boston, MA 02115, USA.
| | | |
Collapse
|
33
|
Shams H, Poblete F, Rüssmann H, Galán JE, Donis RO. Induction of specific CD8+ memory T cells and long lasting protection following immunization with Salmonella typhimurium expressing a lymphocytic choriomeningitis MHC class I-restricted epitope. Vaccine 2001; 20:577-85. [PMID: 11672924 DOI: 10.1016/s0264-410x(01)00363-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Numerous studies have shown the potential of Salmonella typhimurium as a vector for delivery of heterologous proteins for vaccination against other pathogens. Earlier studies showed that the inefficient elicitation of MHC class I-restricted responses could limit the use of S. typhimurium as a heterologous antigen delivery vector for vaccination. We recently developed an approach to overcome this limitation by using a bacterial-encoded specialized protein secretion system, termed type III, to deliver proteins into the class I antigen presenting pathways. Thus, peptides of interest fused to proteins bearing the type III secretion signal, which can elicit protective CTL responses. Because protective immunity is usually assessed a few weeks after vaccination, there is a paucity of information regarding duration of protective immunity induced by this system. We show here that mice immunized orally with S. typhimurium vectors expressing a MHC class I-restricted epitope of the lymphocytic choriomeningitis virus (LCMV) nucleoprotein developed specific antiviral CTL responses. CD8+ T cells were found to be necessary for this CTL activity against targets presenting the LCMV epitope. The survival of mice challenged with lethal doses of LCMV 60 or 135 days after vaccination was as complete as the survival of mice challenged 2 weeks after immunization with the same vectors. By demonstrating their ability to induce prolonged protective immunity after oral delivery, S. typhimurium vectors have met an essential requirement in support of their development as vectors for heterologous vaccination.
Collapse
Affiliation(s)
- H Shams
- Department of Veterinary and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | | | | | | | | |
Collapse
|
34
|
Rodriguez F, Harkins S, Redwine JM, de Pereda JM, Whitton JL. CD4(+) T cells induced by a DNA vaccine: immunological consequences of epitope-specific lysosomal targeting. J Virol 2001; 75:10421-30. [PMID: 11581410 PMCID: PMC114616 DOI: 10.1128/jvi.75.21.10421-10430.2001] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2001] [Accepted: 07/19/2001] [Indexed: 11/20/2022] Open
Abstract
Our previous studies have shown that targeting DNA vaccine-encoded major histocompatibility complex class I epitopes to the proteasome enhanced CD8(+) T-cell induction and protection against lymphocytic choriomeningitis virus (LCMV) challenge. Here, we expand these studies to evaluate CD4(+) T-cell responses induced by DNA immunization and describe a system for targeting proteins and minigenes to lysosomes. Full-length proteins can be targeted to the lysosomal compartment by covalent attachment to the 20-amino-acid C-terminal tail of lysosomal integral membrane protein-II (LIMP-II). Using minigenes encoding defined T-helper epitopes from lymphocytic choriomeningitis virus, we show that the CD4(+) T-cell response induced by the NP(309-328) epitope of LCMV was greatly enhanced by addition of the LIMP-II tail. However, the immunological consequence of lysosomal targeting is not invariably positive; the CD4(+) T-cell response induced by the GP(61-80) epitope was almost abolished when attached to the LIMP-II tail. We identify the mechanism which underlies this marked difference in outcome. The GP(61-80) epitope is highly susceptible to cleavage by cathepsin D, an aspartic endopeptidase found almost exclusively in lysosomes. We show, using mass spectrometry, that the GP(61-80) peptide is cleaved between residues F(74) and K(75) and that this destroys its ability to stimulate virus-specific CD4(+) T cells. Thus, the immunological result of lysosomal targeting varies, depending upon the primary sequence of the encoded antigen. We analyze the effects of CD4(+) T-cell priming on the virus-specific antibody and CD8(+) T-cell responses which are mounted after virus infection and show that neither response appears to be accelerated or enhanced. Finally, we evaluate the protective benefits of CD4(+) T-cell vaccination in the LCMV model system; in contrast to DNA vaccine-induced CD8(+) T cells, which can confer solid protection against LCMV challenge, DNA vaccine-mediated priming of CD4(+) T cells does not appear to enhance the vaccinee's ability to combat viral challenge.
Collapse
Affiliation(s)
- F Rodriguez
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
35
|
Rodriguez F, Slifka MK, Harkins S, Whitton JL. Two overlapping subdominant epitopes identified by DNA immunization induce protective CD8(+) T-cell populations with differing cytolytic activities. J Virol 2001; 75:7399-409. [PMID: 11462012 PMCID: PMC114975 DOI: 10.1128/jvi.75.16.7399-7409.2001] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Subdominant CD8(+) T-cell responses contribute to control of several viral infections and to vaccine-induced immunity. Here, using the lymphocytic choriomeningitis virus model, we demonstrate that subdominant epitopes can be more reliably identified by DNA immunization than by other methods, permitting the identification, in the virus nucleoprotein, of two overlapping subdominant epitopes: one presented by L(d) and the other presented by K(d). This subdominant sequence confers immunity as effective as that induced by the dominant epitope, against which >90% of the antiviral CD8(+) T cells are normally directed. We compare the kinetics of the dominant and subdominant responses after vaccination with those following subsequent viral infection. The dominant CD8(+) response expands more rapidly than the subdominant responses, but after virus infection is cleared, mice which had been immunized with the "dominant" vaccine have a pool of memory T cells focused almost entirely upon the dominant epitope. In contrast, after virus infection, mice which had been immunized with the "subdominant" vaccine retain both dominant and subdominant memory cells. During the acute phase of the immune response, the acquisition of cytokine responsiveness by subdominant CD8(+) T cells precedes their development of lytic activity. Furthermore, in both dominant and subdominant populations, lytic activity declines more rapidly than cytokine responsiveness. Thus, the lysis(low)-cytokine(competent) phenotype associated with most memory CD8(+) T cells appears to develop soon after antigen clearance. Finally, lytic activity differs among CD8(+) T-cell populations with different epitope specificities, suggesting that vaccines can be designed to selectively induce CD8(+) T cells with distinct functional attributes.
Collapse
Affiliation(s)
- F Rodriguez
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
36
|
von Herrath MG, Coon B, Wolfe T. Tolerance induction with agonist peptides recognized by autoaggressive lymphocytes is transient: therapeutic potential for type 1 diabetes is limited and depends on time-point of administration, choice of epitope and adjuvant. J Autoimmun 2001; 16:193-9. [PMID: 11334483 DOI: 10.1006/jaut.2000.0497] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Immunization with agonist peptides recognized by autoaggressive lymphocytes has been used successfully in several animal models for type 1 diabetes (T1D) or multiple sclerosis (MS) to prevent disease. Depending on the timing of immunization, use of adjuvant and route of administration either elimination of autoaggressive T cells or induction of regulation reflected by cytokine shifts were described. Since it was also reported that such agonist peptides could enhance autoimmunity by activating aggressive lymphocytes, our goal was to re-evaluate their efficacy in an antigen-specific model of virally-induced T1D that allowed us to precisely track the autoaggressive response. We find that rather than the route of administration (oral versus sc) the precise timing is important for inducing tolerance to self-antigens. Tolerance is transient and only immunization during a susceptible phase 10 to 20 days prior to the induction of disease but not in prediabetic mice resulted in protection. Further, use of a stronger adjuvant (CFA) compared to IFA enhanced the protective effect. Mechanistically, a transient loss of autoaggressive T cells was responsible for preventing disease, the effect was quantitative and no regulatory lymphocytes or cytokine shifts were induced by any of our treatments. Thus, MHC class I-restricted agonist peptides might only find a limited use in treating autoimmune disorders, because tolerance induction is transient and treatment has to be given very early, ideally prior to activation of the aggressive response.
Collapse
Affiliation(s)
- M G von Herrath
- Depts. of Immunology and Neuropharmacology, IMM6, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
37
|
Abstract
This chapter discusses the virus infections of the central nervous system (CNS) and DNA vaccines. Mild central nervous system (CNS) symptoms, such as headache and drowsiness, can result from systemically elevated cytokine levels and therefore are common in many virus infections, even in the absence of the infection of the CNS. CNS infection is quite unusual and is initiated either as a result of the viremia or, more rarely, as a result of neural spread. The poliovirus infects the anterior horn motor neurons of the spinal cord, causing poliomyelitis, the disease for which the virus is named. DNA vaccination is a relatively new entrant in the vaccine sweepstakes, but is viewed with optimism, for a number of reasons. DNA vaccines encoding the nucleoprotein from lymphocytic choriomeningitis virus can confer protection against the normally lethal intracranial challenge. In rabies, in a mouse model, immunization with plasmids encoding the rabies glycoprotein conferred complete protection against subsequent viral challenge. Several virus-induced CNS diseases may be explained by their triggering of autoimmunity. Experimental autoimmune encephalomyelitis is a well-characterized CNS disease induced by the administration of certain CNS proteins.
Collapse
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, CVN-9, Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
38
|
Djavani M, Yin C, Lukashevich IS, Rodas J, Rai SK, Salvato MS. Mucosal immunization with Salmonella typhimurium expressing Lassa virus nucleocapsid protein cross-protects mice from lethal challenge with lymphocytic choriomeningitis virus. JOURNAL OF HUMAN VIROLOGY 2001; 4:103-8. [PMID: 11437313 PMCID: PMC2391007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
OBJECTIVES Lassa fever virus (LAS) is transmitted to man by rodent carriers and is fatal in a third of untreated cases. Our goal is to provide immune protection from Lassa fever by mucosal vaccination. STUDY DESIGN/METHODS Mice were vaccinated intragastrically with control vectors or with vectors (vaccinia or Salmonella) expressing LAS nucleocapsid protein (NP). Mice were challenged intracranially with a lethal dose of the related arenavirus, lymphocytic choriomeningitis virus (LCMV), as a measure of the vaccine's ability to elicit cross-protection. RESULTS Salmonella and vaccinia vectors expressing LAS NP each protected a third of the mice from lethal challenge with LCMV. All mice vaccinated with a vector expressing LCMV NP were protected as expected. CONCLUSIONS The LAS recombinant Salmonella vector is comparable to the LAS recombinant vaccinia vector in its ability to cross-protect mice from lethal challenge. Nucleocapsid protein is an inadequate immunogen on its own, but provides sufficient cross-protection to make it a useful component of a broadly reactive arenavirus vaccine.
Collapse
Affiliation(s)
- Mahmoud Djavani
- Institute of Human Virology, University of Maryland Biotechnology Center, Baltimore, Maryland, USA
| | - Cheng Yin
- Department of Pathology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Igor S. Lukashevich
- Institute of Human Virology, University of Maryland Biotechnology Center, Baltimore, Maryland, USA
| | - Juan Rodas
- Institute of Human Virology, University of Maryland Biotechnology Center, Baltimore, Maryland, USA
| | - Sharath K. Rai
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Maria S. Salvato
- Institute of Human Virology, University of Maryland Biotechnology Center, Baltimore, Maryland, USA
| |
Collapse
|
39
|
Sewell AK, Price DA, Oxenius A, Kelleher AD, Phillips RE. Cytotoxic T lymphocyte responses to human immunodeficiency virus: control and escape. Stem Cells 2000; 18:230-44. [PMID: 10924089 DOI: 10.1634/stemcells.18-4-230] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Effective preventive and therapeutic intervention in individuals exposed to or infected with human immunodeficiency virus (HIV) depends, in part, on a clear understanding of the interactions between the virus and those elements of the host immune response which control viral replication. Recent advances have provided compelling evidence that cytotoxic T lymphocytes (CTLs) constitute an essential component of protective antiretroviral immunity. Here, we review briefly the significance of this work in the context of previous studies, and outline the mechanisms through which HIV evades CTL activity.
Collapse
Affiliation(s)
- A K Sewell
- The Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, UK.
| | | | | | | | | |
Collapse
|
40
|
Hassett DE, Zhang J, Slifka M, Whitton JL. Immune responses following neonatal DNA vaccination are long-lived, abundant, and qualitatively similar to those induced by conventional immunization. J Virol 2000; 74:2620-7. [PMID: 10684276 PMCID: PMC111750 DOI: 10.1128/jvi.74.6.2620-2627.2000] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/1999] [Accepted: 12/17/1999] [Indexed: 11/20/2022] Open
Abstract
Virus infections are devastating to neonates, and the induction of active antiviral immunity in this age group is an important goal. Here, we show that a single neonatal DNA vaccination induces cellular and humoral immune responses which are maintained for a significant part of the animal's life span. We employ a sensitive technique which permits the first demonstration and quantitation, directly ex vivo, of virus-specific CD8(+) T cells induced by DNA immunization. One year postvaccination, antigen-specific CD8(+) T cells were readily detectable and constituted 0.5 to 1% of all CD8(+) T cells. By several criteria-including cytokine production, perforin content, development of lytic ability, and protective capacity-DNA vaccine-induced CD8(+) memory T cells were indistinguishable from memory cells induced by immunization with a conventional (live-virus) vaccine. Analyses of long-term humoral immune responses revealed that, in contrast to the strong immunoglobulin G2a (IgG2a) skewing of the humoral response seen after conventional vaccination, IgG1 and IgG2a levels were similar in DNA-vaccinated neonatal and adult animals, indicating a balanced T helper response. Collectively, these results show that a single DNA vaccination within hours or days of birth can induce long-lasting CD8(+) T- and B-cell responses; there is no need for secondary immunization (boosting). Furthermore, the observed immune responses induced in neonates and in adults are indistinguishable by several criteria, including protection against virus challenge.
Collapse
Affiliation(s)
- D E Hassett
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
41
|
Djavani M, Yin C, Xia L, Lukashevich IS, Pauza CD, Salvato MS. Murine immune responses to mucosally delivered Salmonella expressing Lassa fever virus nucleoprotein. Vaccine 2000; 18:1543-54. [PMID: 10618553 DOI: 10.1016/s0264-410x(99)00439-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Arenaviruses are emerging pathogens known to infect via the mucosa, however no formal attempts to make mucosal vaccines have been undertaken. Here we describe a recombinant aroA attenuated Salmonella typhimurium that expresses the nucleoprotein (NP) gene of Lassa fever virus (LAS). The complete NP gene was cloned downstream of the bacterial groEL promotor and integrated into the aroA locus of S. typhimurium. Lassa NP protein was detected in whole cell extracts from the recombinant Salmonella by immunoblot analysis with serum from Lassa-infected people. Mice were inoculated by intragastric intubation with 5 x 10(9) S. typhimurium and boosted with the same recombinant Salmonella 21 days after the primary inoculation. Both local mucosal IgA and serum immunoglobulins against Lassa NP were observed. Splenic cytotoxic T-lymphocyte responses to LAS NP were detected after the boost and they cross-reacted with target cells infected with the related arenavirus, lymphocytic choriomeningitis virus. Recombinant Salmonella elicits humoral and cell mediated immune responses against Lassa fever virus in mice and should be considered as a potential vaccine strategy in man.
Collapse
Affiliation(s)
- M Djavani
- Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
DNA immunization works, as has been amply demonstrated in a variety of microbial and tumor models. However, the mechanisms which underpin its success remain unclear. Using intramuscular delivery of DNA, we wish to precisely define how DNA-encoded antigens induce CD8+ T-cells (most cytotoxic T-cells; CTL), CD4+ T-cells (mostly helper cells) and antibodies; and to use the accrued knowledge to rationally manipulate DNA vaccines, thus enabling us to optimize each of the above three types of immune response. We consider it likely that different mechanisms operate in each case. We have designed a DNA vaccine which induces CTL, but not antibodies. We will present evidence that CTL are induced by endogenously-synthesized protein, not by protein released from cells; and that in the absence of release of intact protein, antibodies are not induced, while CTL induction remains strong. We have used plasmid-encoded minigenes and have found that these short sequences also induce CTL; this, too, argues that CTL are induced by antigens presented following endogenous synthesis. We are attempting to determine how antigens are released from transfected cells, to interact with B-cells and induce antibodies, and are currently evaluating the CD4 responses induced by DNA vaccines.
Collapse
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
43
|
Kang CY, Luo L, Wainberg MA, Li Y. Development of HIV/AIDS vaccine using chimeric gag-env virus-like particles. Biol Chem 1999; 380:353-64. [PMID: 10223338 DOI: 10.1515/bc.1999.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We attempted to develop a candidate HIV/AIDS vaccine, by using unprocessed HIV-2 gag pr45 precursor protein. We found that a 45 kDa unprocessed HIV-2 gag precursor protein (pr45), with a deletion of a portion of the viral protease, assembles as virus-like particles (VLP). We mapped the functional domain of HIV-2 gag VLP formation in order to find the minimum length of gag protein to form VLP. A series of deletion mutants was constructed by sequentially removing the C-terminal region of HIV-2 gag precursor protein and expressed truncated genes in Spodoptera frugiperda (SF) cells by infecting recombinant baculoviruses. We found that deletion of up to 143 amino acids at the C-terminus of HIV-2 gag, leaving 376 amino acids at the N-terminus of the protein, did not affect VLP formation. There is a proline-rich region at the amino acid positions 373 to 377 of HIV-2 gag, and replacement of these proline residues by site-directed mutagenesis completely abolished VLP assembly. Our data demonstrate that the C-terminal p12 region of HIV-2 gag precursor protein, and zinc finger domains, are dispensable for gag VLP assembly, but the presence of at least one of the three prolines at amino acid positions 373, 375 or 377 of HIV-2NIH-Z is required for VLP formation. Animals immunized with these gag particles produced high titer antibodies and Western blot analyses showed that anti-gag pr45 rabbit sera react with p17, p24 and p55 gag proteins of HIV-1. We then constructed chimeric gag genes, which carry the hypervariable V3 region of HIV-1 gp120, because the V3 loop is known to interact with chemokine receptor as a coreceptor, and known to induce the major neutralizing antibodies and stimulate the cytoxic T lymphocyte responses in humans and mice. We expressed chimeric fusion protein of HIV-2 gag with 3 tandem copies of consensus V3 domain that were derived from 245 different isolates of HIV-1. In addition, we also constructed and expressed chimeric fusion protein that contains HIV-2 gag with V3 domains of HIV-1IIIB, HIV-1MN, HIV-1SF2 and HIV-1RF. The chimeric gag-env particles had a spherical morphology, and the size was slightly larger than that of a gag particle. Immunoprecipitation and Western blot analyses show that these chimeric proteins were recognized by HIV-1 positive human sera and antisera raised against V3 peptides, as well as by rabbit anti-gp120 serum. We obtained virus neutralizing antibodies in rabbits by immunizing these gag-env VLPs. In addition, we found that gag-env chimeric VLPs induce a strong CTL activity against V3 peptide-treated target cells. Our results indicate that V3 peptides from all major clades of HIV-1 carried by HIV-2 gag can be used as a potential HIV/AIDS vaccine.
Collapse
Affiliation(s)
- C Y Kang
- Western Science Centre, The University of Western Ontario, London, Canada
| | | | | | | |
Collapse
|
44
|
Abstract
Borna disease virus (BDV) is a neurotropic virus with a broad host and geographic range. Lewis rats were immunized against BDV with a recombinant vaccinia virus expressing the BDV nucleoprotein and were later infected with BDV to evaluate protection against Borna disease (BD). Relative to animals that were not immunized, immunized animals had a decreased viral burden after challenge with infectious virus, more marked inflammation, and aggravated clinical disease. These data suggest that a more robust immune response in Borna disease can reduce viral load at the expense of increased morbidity.
Collapse
Affiliation(s)
- A J Lewis
- Department of Neurology, University of California, Irvine, California 92697-4292, USA
| | | | | | | | | |
Collapse
|
45
|
Carbonetti NH, Irish TJ, Chen CH, O'Connell CB, Hadley GA, McNamara U, Tuskan RG, Lewis GK. Intracellular delivery of a cytolytic T-lymphocyte epitope peptide by pertussis toxin to major histocompatibility complex class I without involvement of the cytosolic class I antigen processing pathway. Infect Immun 1999; 67:602-7. [PMID: 9916065 PMCID: PMC96361 DOI: 10.1128/iai.67.2.602-607.1999] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A CD8(+) cytolytic T-lymphocyte (CTL) response to antigen-presenting cells generally requires intracellular delivery or synthesis of antigens in order to access the major histocompatibility complex (MHC) class I processing and presentation pathway. To test the ability of pertussis toxin (PT) to deliver peptides to the class I pathway for CTL recognition, we constructed fusions of CTL epitope peptides with a genetically detoxified derivative of PT (PT9K/129G). Two sites on the A (S1) subunit of PT9K/129G tolerated the insertion of peptides, allowing efficient assembly and secretion of the holotoxin fusion by Bordetella pertussis. Target cells incubated with these fusion proteins were specifically lysed by CTLs in vitro, and this activity was shown to be MHC class I restricted. The activity was inhibited by brefeldin A, suggesting a dependence on intracellular trafficking events, but was not inhibited by the proteasome inhibitors lactacystin and N-acetyl-L-leucyl-L-leucyl-L-norleucinal (LLnL). Furthermore, the activity was present in mutant antigen-presenting cells lacking the transporter associated with antigen processing, which transports peptides from the cytosol to the endoplasmic reticulum for association with MHC class I molecules. PT may therefore bypass the proteasome-dependent cytosolic pathway for antigen presentation and deliver epitopes to class I molecules via an alternative route.
Collapse
Affiliation(s)
- N H Carbonetti
- Departments of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Henke A, Wagner E, Whitton JL, Zell R, Stelzner A. Protection of mice against lethal coxsackievirus B3 infection by using DNA immunization. J Virol 1998; 72:8327-31. [PMID: 9733878 PMCID: PMC110201 DOI: 10.1128/jvi.72.10.8327-8331.1998] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccination with DNA and recombinant vaccinia viruses (rec.VV) has been studied with the coxsackievirus B3 (CVB3) model system. Plasmids encoding all structural proteins of CVB3, when injected intramuscularly, induced only low levels of virus-specific antibodies. However, DNA vaccination with the major structural protein VP1 protected 72.2% of mice from lethal challenge, whereas VP1 expressed by rec.VV was much less efficient.
Collapse
Affiliation(s)
- A Henke
- Institute of Virology, Medical Center, Friedrich Schiller University, 07745 Jena, Germany
| | | | | | | | | |
Collapse
|
47
|
Abstract
How the immune response matures from recognizing a single or a few structures of the antigen to many is an obviously important process. Models of B-cell epitope spreading have been developed in a variety of systems. For example, immunization of animals with PPPGMRPP, one of the earliest B-cell epitopes in the anti-Sm response found in human lupus, leads to antispliceosomal autoimmunity and features of lupus. The humoral immune response spreads from PPPGMRPP to other structures of the spliceosome in an apparently reproducible sequence. B-cell epitope spreading has provided the experimental basis from which a relationship between lupus and Epstein-Barr virus was suspected. An understanding of B-cell epitope spreading is likely to lead to important principles in basic immunology and to answers to human disease problems.
Collapse
Affiliation(s)
- J A James
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma Medical Research Foundation, Oklahoma City 73104, USA
| | | |
Collapse
|
48
|
Whitton JL. An overview of antigen presentation and its central role in the immune response. Curr Top Microbiol Immunol 1998; 232:1-13. [PMID: 9557391 DOI: 10.1007/978-3-642-72045-1_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
49
|
Warner JF, Jolly DJ, Merritt J. Human immunodeficiency virus immunotherapy using a retroviral vector. Curr Top Microbiol Immunol 1998; 226:145-60. [PMID: 9479840 DOI: 10.1007/978-3-642-80475-5_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- J F Warner
- Chiron Viagene, San Diego, CA 92121, USA
| | | | | |
Collapse
|
50
|
Luo L, Li Y, Chang JS, Cho SY, Kim TY, Choi MJ, Cheong HS, Kim HJ, Ahn HJ, Min MK, Chun BH, Jung SM, Woo SG, Park SY, Kang CY. Induction of V3-specific cytotoxic T lymphocyte responses by HIV gag particles carrying multiple immunodominant V3 epitopes of gp120. Virology 1998; 240:316-25. [PMID: 9454705 DOI: 10.1006/viro.1997.8922] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Effort to develop a vaccine to prevent infection of human immunodeficiency virus (HIV) have focused on the induction of neutralizing antibodies. In our previous study, we reported that chimeric gag-env virus-like particles (VLPs) induce neutralizing antibodies which block HIV infection. In addition to the neutralizing antibodies, the cytotoxic T-lymphocyte (CTL) response is considered to be another major immune defense mechanism required for recovery from many different viral infections. In the present study, we have constructed chimeric fusion proteins using HIV-2 gag precursor protein with (1) four neutralizing epitopes from HIV-1 gp160; (2) three tandem copies of consensus V3 domain, which have been derived from 245 different isolates of HIV-1 and carries both the principal neutralizing determinant (PND) and CTL epitopes; and (3) V3 domains from HIV-1IIIB, HIV-1MN, HIV-1RF, and HIV-1SF2. These chimeric fusion proteins were expressed in a large quantity within insect cells, and released as VLPs into the cell culture medium. The purified gag-env VLPs from all three constructs appear to be spherical particles similar to immature HIV but slightly larger than the gag VLPs. Immunoprecipitation analysis showed that the chimeric proteins were recognized not only by HIV-1 positive patient sera, but also by monoclonal and polyclonal antisera raised against V3 peptides of HIV-1IIIB, HIV-1MN, HIV-1RF, and the gp120 antiserum against HIV-1SF2. Balb/C mice immunized with these chimeric VLPs successfully induced CTL activity against V3 peptide-stimulated target cells. In addition, a high degree of cross-reactivity was observed among the four different strains of HIV-1 V3 domain, indicating that the tandem multiple consensus V3 peptide sequence carried by HIV-2 gag can be used as a potential HIV vaccine against various HIVs.
Collapse
Affiliation(s)
- L Luo
- Western Science Centre, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|