1
|
Srivastava S, Verma S, Kamthania M, Agarwal D, Saxena AK, Kolbe M, Singh S, Kotnis A, Rathi B, Nayar SA, Shin HJ, Vashisht K, Pandey KC. Computationally validated SARS-CoV-2 CTL and HTL Multi-Patch vaccines, designed by reverse epitomics approach, show potential to cover large ethnically distributed human population worldwide. J Biomol Struct Dyn 2022; 40:2369-2388. [PMID: 33155524 PMCID: PMC7651196 DOI: 10.1080/07391102.2020.1838329] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is responsible for the COVID-19 outbreak. The highly contagious COVID-19 disease has spread to 216 countries in less than six months. Though several vaccine candidates are being claimed, an effective vaccine is yet to come. A novel reverse epitomics approach, 'overlapping-epitope-clusters-to-patches' method is utilized to identify the antigenic regions from the SARS-CoV-2 proteome. These antigenic regions are named as 'Ag-Patch or Ag-Patches', for Antigenic Patch or Patches. The identification of Ag-Patches is based on the clusters of overlapping epitopes rising from SARS-CoV-2 proteins. Further, we have utilized the identified Ag-Patches to design Multi-Patch Vaccines (MPVs), proposing a novel method for the vaccine design. The designed MPVs were analyzed for immunologically crucial parameters, physiochemical properties and cDNA constructs. We identified 73 CTL (Cytotoxic T-Lymphocyte) and 49 HTL (Helper T-Lymphocyte) novel Ag-Patches from the proteome of SARS-CoV-2. The identified Ag-Patches utilized to design MPVs cover 768 overlapping epitopes targeting 55 different HLA alleles leading to 99.98% of world human population coverage. The MPVs and Toll-Like Receptor ectodomain complex shows stable complex formation tendency. Further, the cDNA analysis favors high expression of the MPVs constructs in a human cell line. We identified highly immunogenic novel Ag-Patches from the entire proteome of SARS CoV-2 by a novel reverse epitomics approach and utilized them to design MPVs. We conclude that the novel MPVs could be a highly potential novel approach to combat SARS-CoV-2, with greater effectiveness, high specificity and large human population coverage worldwide. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sukrit Srivastava
- Molecular Medicine Lab., School of Life Science, Jawaharlal Nehru University, New Delhi, India
- Infection Biology Group, Indian Foundation for Fundamental Research, RaeBareli, India
| | - Sonia Verma
- Parasite-Host Biology Group, Protein Biochemistry & Engineering Lab, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Mohit Kamthania
- Infection Biology Group, Indian Foundation for Fundamental Research, RaeBareli, India
| | - Deepa Agarwal
- Infection Biology Group, Indian Foundation for Fundamental Research, RaeBareli, India
| | - Ajay Kumar Saxena
- Molecular Medicine Lab., School of Life Science, Jawaharlal Nehru University, New Delhi, India
| | - Michael Kolbe
- Department for Structural Infection Biology, Centre for Structural Systems Biology (CSSB) & Helmholtz-Centre for Infection Research, Hamburg, Germany
- Faculty of Mathematics, Informatics and Natural Sciences, University of Hamburg, Hamburg, Germany
| | - Sarman Singh
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Ashwin Kotnis
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Brijesh Rathi
- Laboratory For Translational Chemistry and Drug Discovery, Hansraj College, University of Delhi, New Delhi, India
| | - Seema A. Nayar
- Department of Microbiology, Government Medical College, Trivandrum, India
- Department of Microbiology, Sree Gokulam Medical College, Trivandrum, India
| | - Ho-Joon Shin
- Department of Microbiology, School of Medicine, Ajou University, Suwon, South Korea
| | - Kapil Vashisht
- Parasite-Host Biology Group, Protein Biochemistry & Engineering Lab, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Kailash C. Pandey
- Parasite-Host Biology Group, Protein Biochemistry & Engineering Lab, ICMR-National Institute of Malaria Research, New Delhi, India
| |
Collapse
|
2
|
Yarmarkovich M, Warrington JM, Farrel A, Maris JM. Identification of SARS-CoV-2 Vaccine Epitopes Predicted to Induce Long-Term Population-Scale Immunity. Cell Rep Med 2020; 1:100036. [PMID: 32835302 PMCID: PMC7276303 DOI: 10.1016/j.xcrm.2020.100036] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/29/2020] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
Abstract
Here we propose a SARS-CoV-2 vaccine design concept based on identification of highly conserved regions of the viral genome and newly acquired adaptations, both predicted to generate epitopes presented on major histocompatibility complex (MHC) class I and II across the vast majority of the population. We further prioritize genomic regions that generate highly dissimilar peptides from the human proteome and are also predicted to produce B cell epitopes. We propose sixty-five 33-mer peptide sequences, a subset of which can be tested using DNA or mRNA delivery strategies. These include peptides that are contained within evolutionarily divergent regions of the spike protein reported to increase infectivity through increased binding to the ACE2 receptor and within a newly evolved furin cleavage site thought to increase membrane fusion. Validation and implementation of this vaccine concept could specifically target specific vulnerabilities of SARS-CoV-2 and should engage a robust adaptive immune response in the vast majority of the population.
Collapse
Affiliation(s)
- Mark Yarmarkovich
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John M. Warrington
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alvin Farrel
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Yarmarkovich M, Warrington JM, Farrel A, Maris JM. A SARS-CoV-2 Vaccination Strategy Focused on Population-Scale Immunity. SSRN 2020:3575161. [PMID: 32714112 PMCID: PMC7366814 DOI: 10.2139/ssrn.3575161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/05/2020] [Indexed: 11/15/2022]
Abstract
Here we propose a vaccination strategy for SARS-CoV-2 based on identification of both highly conserved regions of the virus and newly acquired adaptations that are presented by MHC class I and II across the vast majority of the population, are highly dissimilar from the human proteome, and are predicted B cell epitopes. We present 65 peptide sequences that we expect to result in a safe and effective vaccine which can be rapidly tested in DNA, mRNA, or synthetic peptide constructs. These include epitopes that are contained within evolutionarily divergent regions of the spike protein reported to increase infectivity through increased binding to the ACE2 receptor, and within a novel furin cleavage site thought to increase membrane fusion. This vaccination strategy specifically targets unique vulnerabilities of SARS-CoV-2 and should engage a robust adaptive immune response in the vast majority of the human population.
Collapse
Affiliation(s)
- Mark Yarmarkovich
- Division of Oncology and Center for Childhood Cancer Research; Children’s Hospital of Philadelphia; Philadelphia, PA, 19104; USA
| | - John M. Warrington
- Division of Oncology and Center for Childhood Cancer Research; Children’s Hospital of Philadelphia; Philadelphia, PA, 19104; USA
| | - Alvin Farrel
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia; Philadelphia, PA, 19104
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research; Children’s Hospital of Philadelphia; Philadelphia, PA, 19104; USA
- Perelman School of Medicine at the University of Pennsylvania; Philadelphia, PA, 19104
| |
Collapse
|
4
|
Yarmarkovich M, Warrington JM, Farrel A, Maris JM. A SARS-CoV-2 Vaccination Strategy Focused on Population-Scale Immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.03.31.018978. [PMID: 32511347 PMCID: PMC7255782 DOI: 10.1101/2020.03.31.018978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Here we propose a vaccination strategy for SARS-CoV-2 based on identification of both highly conserved regions of the virus and newly acquired adaptations that are presented by MHC class I and II across the vast majority of the population, are highly dissimilar from the human proteome, and are predicted B cell epitopes. We present 65 peptide sequences that we expect to result in a safe and effective vaccine which can be rapidly tested in DNA, mRNA, or synthetic peptide constructs. These include epitopes that are contained within evolutionarily divergent regions of the spike protein reported to increase infectivity through increased binding to the ACE2 receptor, and within a novel furin cleavage site thought to increase membrane fusion. This vaccination strategy specifically targets unique vulnerabilities of SARS-CoV-2 and should engage a robust adaptive immune response in the vast majority of the human population.
Collapse
|
5
|
Terry FE, Moise L, Martin RF, Torres M, Pilotte N, Williams SA, De Groot AS. Time for T? Immunoinformatics addresses vaccine design for neglected tropical and emerging infectious diseases. Expert Rev Vaccines 2014; 14:21-35. [PMID: 25193104 PMCID: PMC4743591 DOI: 10.1586/14760584.2015.955478] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Vaccines have been invaluable for global health, saving lives and reducing healthcare costs, while also raising the quality of human life. However, newly emerging infectious diseases (EID) and more well-established tropical disease pathogens present complex challenges to vaccine developers; in particular, neglected tropical diseases, which are most prevalent among the world's poorest, include many pathogens with large sizes, multistage life cycles and a variety of nonhuman vectors. EID such as MERS-CoV and H7N9 are highly pathogenic for humans. For many of these pathogens, while their genomes are available, immune correlates of protection are currently unknown. These complexities make developing vaccines for EID and neglected tropical diseases all the more difficult. In this review, we describe the implementation of an immunoinformatics-driven approach to systematically search for key determinants of immunity in newly available genome sequence data and design vaccines. This approach holds promise for the development of 21st century vaccines, improving human health everywhere.
Collapse
|
6
|
Chen F, Zhai MX, Zhu YH, Qi YM, Zhai WJ, Gao YF. In vitro and in vivo identification of a novel cytotoxic T lymphocyte epitope from Rv3425 of Mycobacterium tuberculosis. Microbiol Immunol 2012; 56:548-53. [PMID: 22537173 PMCID: PMC7168511 DOI: 10.1111/j.1348-0421.2012.00470.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The identification of novel cytotoxic T lymphocyte (CTL) epitopes is important to analysis of the involvement of CD8+ T cells in Mycobacterium tuberculosis infection as well as to the development of peptide vaccines. In this study, a novel CTL epitope from region of difference 11 encoded antigen Rv3425 was identified. Epitopes were predicted by the reversal immunology approach. Rv3425‐p118 (LIASNVAGV) was identified as having relatively strong binding affinity and stability towards the HLA‐A*0201 molecule. Peripheral blood mononuclear cells pulsed by this peptide were able to release interferon‐γ in healthy donors (HLA‐A*02+ purified protein derivative+). In cytotoxicity assays in vitro and in vivo, Rv3425‐p118 induced CTLs to specifically lyse the target cells. Therefore, this epitope could provide a subunit component for designing vaccines against Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Fei Chen
- Department of Bioengineering, Zhengzhou University, Zhengzhou 450001, China
| | | | | | | | | | | |
Collapse
|
7
|
The signal peptide sequence impacts the immune response elicited by a DNA epitope vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1776-80. [PMID: 21832097 DOI: 10.1128/cvi.05179-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the effect of two leader sequences, one from a transmembrane molecule (H2-L(d)) and another from a secreted molecule (rat KC chemokine), on the immunogenicity of DNA epitope vaccines. The chemokine leader enhanced vaccine immunogenicity, thus underscoring the importance of the leader sequence in DNA epitope vaccine design.
Collapse
|
8
|
Shan S, Jiang Y, Bu Z, Ellis T, Zeng X, Edwards J, Tian G, Li Y, Ge J, Chen H, Fenwick S. Strategies for improving the efficacy of a H6 subtype avian influenza DNA vaccine in chickens. J Virol Methods 2011; 173:220-6. [PMID: 21333689 DOI: 10.1016/j.jviromet.2011.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 02/10/2011] [Accepted: 02/10/2011] [Indexed: 10/18/2022]
Abstract
A low-pathogenicity avian influenza H6N2 virus was used to investigate approaches to improve DNA vaccine efficacy. The viral hemagglutinin (HA) gene or its chicken biased HA gene, incorporating a Kozak sequence, was cloned into a pCAGGS vector to produce the pCAG-HAk and pCAG-optiHAk constructs. Following two intramuscular injections, the seroconversion rate in vaccinated chickens with 10, 100 or 300 μg pCAG-HAk were 87.5%, 75% and 75%, respectively. The profile of H6 hemagglutination inhibition (HI) antibodies induced by different doses of pCAG-HAk during the 8-week study period was similar. The HI titer rose significantly in the three different dose groups following the booster and reached a plateau 2-3 weeks post-booster. In a single dose vaccination group with 100 μg pCAG-HAk, a maximum seroconversion rate reached 53.3% at 5 weeks post-vaccination. The earliest time of seroconversion appeared two weeks after DNA immunization. Following two electroporation (EP) vaccinations with 100 μg pCAG-HAk, all birds seroconverted and the HI antibody titers were significantly higher than those using intramuscular immunization, suggesting that EP was more efficient than intramuscular delivery of the DNA vaccines. In comparison, chickens immunized with 10 or 100 μg pCAG-optiHAk showed 37.5% and 87.5% seroconversion rates, respectively, at 3 weeks following the booster. The pCAG-HAk was not significantly different from the pCAG-optiHAk in either the seroconversion rate or H6 HI titer, suggesting that the codon-optimized HA DNA vaccine did not achieve significantly better immunogenicity than the pCAG-HAk vaccine.
Collapse
Affiliation(s)
- Songhua Shan
- School of Veterinary and Biomedical Sciences, Murdoch University, Perth, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Epitope-Based Immunome-Derived Vaccines: A Strategy for Improved Design and Safety. CLINICAL APPLICATIONS OF IMMUNOMICS 2008. [PMCID: PMC7122239 DOI: 10.1007/978-0-387-79208-8_3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vaccine science has extended beyond genomics to proteomics and has come to also encompass ‘immunomics,’ the study of the universe of pathogen-derived or neoplasm-derived peptides that interface with B and T cells of the host immune system. It has been theorized that effective vaccines can be developed using the minimum essential subset of T cell and B cell epitopes that comprise the ‘immunome.’ Researchers are therefore using bioinformatics sequence analysis tools, epitope-mapping tools, microarrays, and high-throughput immunology assays to discover the minimal essential components of the immunome. When these minimal components, or epitopes, are packaged with adjuvants in an appropriate delivery vehicle, the complete package comprises an epitope-based immunome-derived vaccine. Such vaccines may have a significant advantage over conventional vaccines, as the careful selection of the components may diminish undesired side effects such as have been observed with whole pathogen and protein subunit vaccines. This chapter will review the pre-clinical and anticipated clinical development of computer-driven vaccine design and the validation of epitope-based immunome-derived vaccines in animal models; it will also include an overview of heterologous immunity and other emerging issues that will need to be addressed by vaccines of all types in the future.
Collapse
|
10
|
Chen L, Gao T, Yang N, Huang J, Chen Y, Gao T, Li Q, Ren D. Immunization with a Synthetic Multiepitope Antigen Induces Humoral and Cellular Immune Responses to Hepatitis C Virus in Mice. Viral Immunol 2007; 20:170-9. [PMID: 17425431 DOI: 10.1089/vim.2006.0067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The immunogenicity of a synthetic multiepitope PCX3 antigen, which contains triple tandem repeats of five conserved epitopes from hepatitis C virus (HCV) polyprotein, was studied in BALB/c mice given three intraperitoneal injections of antigen with Freund's adjuvant. Both a strong antibody response and specific cytotoxic T lymphocytes were induced. The specific anti-PCX3 IgG was able to bind HCV particles from hepatitis C patient sera by incubation overnight. In particular, in transgenic mice with chimeric human livers, anti-PCX3 antibody was able to lower the viral load in two of five mice and to eliminate HCV infection in three of five mice by 2 wk after inoculation with HCV-positive serum from patients. These results indicated that the synthetic multiepitope PCX3 antigen elicits a potent humoral and cellular immune response against HCV.
Collapse
Affiliation(s)
- Lishan Chen
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
The precise mechanisms that govern memory T-cell lineage commitment during an immune response continue to be the subject of intense scrutiny. The existence of memory T-cell subsets defined by location, function, and phenotype adds an additional layer of complexity to the overall memory T-cell population. In this review, the integration of memory subset development and migration and the functional consequences of specific tissue localization are discussed.
Collapse
Affiliation(s)
- Leo Lefrançois
- University of Connecticut Health Center, Department of Immunology, Farmington, CT 06107, USA.
| |
Collapse
|
12
|
Borrego B, Fernandez-Pacheco P, Ganges L, Domenech N, Fernandez-Borges N, Sobrino F, Rodríguez F. DNA vaccines expressing B and T cell epitopes can protect mice from FMDV infection in the absence of specific humoral responses. Vaccine 2006; 24:3889-99. [PMID: 16563575 DOI: 10.1016/j.vaccine.2006.02.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2005] [Revised: 02/09/2006] [Accepted: 02/13/2006] [Indexed: 11/22/2022]
Abstract
Despite foot-and-mouth disease virus (FMDV) being responsible for one of the most devastating animal diseases, little is known about the cellular immune mechanisms involved in protection against this virus. In this work we have studied the potential of DNA vaccines based on viral minigenes corresponding to three major B and T-cell FMDV epitopes (isolate C-S8c1) originally identified in natural hosts. The BTT epitopes [VP1 (133-156)-3A (11-40)-VP4 (20-34)] were cloned into the plasmid pCMV, either alone or fused to ubiquitin, the lysosomal targeting signal from LIMPII, a soluble version of CTLA4 or a signal peptide from the human prion protein, to analyze the effect of processing through different antigenic presentation pathways on the immunogenicity of the FMDV epitopes. As a first step in the analysis of modulation exerted by these target signals, a FMDV infection inhibition assay in Swiss outbred mice was developed and used to analyze the protection conferred by the different BTT-expressing plasmids. Only one of the 37 mice immunized with minigene-bearing plasmids developed specific neutralizing antibodies prior to FMDV challenge. As expected, this single mouse that had been immunized with the BTT tandem epitopes fused to a signal peptide (pCMV-spBTT) was protected against FMDV infection. Interestingly, nine more of the animals immunized with BTT-expressing plasmids did not show viremia at 48 h post-infection (pi), even in the absence of anti-FMDV antibodies prior to challenge. The highest protection (50%, six out of 12 mice) was observed with the plasmid expressing BTT alone, indicating that the targeting strategies used did not result in an improvement of the protection conferred by BTT epitopes. Interestingly, peptide specific CD4+ T-cells were detected for some of the BTT-protected mice. Thus, a DNA vaccine based on single FMDV B and T cell epitopes can protect mice, in the absence of specific antibodies at the time of challenge. Further work must be done to elucidate the mechanisms involved in protection and to determine the protective potential of these vaccines in natural FMDV hosts.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Antigens, CD
- Antigens, Differentiation/genetics
- CD36 Antigens/genetics
- CD4-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Disease Models, Animal
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Foot-and-Mouth Disease/immunology
- Foot-and-Mouth Disease/pathology
- Foot-and-Mouth Disease/prevention & control
- Foot-and-Mouth Disease Virus/genetics
- Foot-and-Mouth Disease Virus/immunology
- Genetic Vectors
- Immunity, Cellular
- Lysosomal Membrane Proteins/genetics
- Mice
- Neutralization Tests
- Protein Transport/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Ubiquitin/genetics
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Vaccines/immunology
- Viremia
Collapse
|
13
|
|
14
|
McKinney DM, Skvoretz R, Livingston BD, Wilson CC, Anders M, Chesnut RW, Sette A, Essex M, Novitsky V, Newman MJ. Recognition of variant HIV-1 epitopes from diverse viral subtypes by vaccine-induced CTL. THE JOURNAL OF IMMUNOLOGY 2004; 173:1941-50. [PMID: 15265928 DOI: 10.4049/jimmunol.173.3.1941] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recognition by CD8(+) T lymphocytes (CTL) of epitopes that are derived from conserved gene products, such as Gag and Pol, is well documented and conceptually supports the development of epitope-based vaccines for use against diverse HIV-1 subtypes. However, many CTL epitopes from highly conserved regions within the HIV-1 genome are highly variable, when assessed by comparison of amino acid sequences. The TCR is somewhat promiscuous with respect to peptide binding, and, as such, CTL can often recognize related epitopes. In these studies, we evaluated CTL recognition of five sets of variant HIV-1 epitopes restricted to HLA-A*0201 and HLA-A*1101 using HLA transgenic mice. We found that numerous different amino acid substitutions can be introduced into epitopes without abrogating their recognition by CTL. Based on our findings, we constructed an algorithm to predict those CTL epitopes capable of inducing responses in the HLA transgenic mice to the greatest numbers of variant epitopes. Similarity of CTL specificity for variant epitopes was demonstrated for humans using PBMC from HIV-1-infected individuals and CTL lines produced in vitro using PBMC from HIV-1-uninfected donors. We believe the ability to predict CTL epitope immunogenicity and recognition patterns of variant epitopes can be useful for designing vaccines against multiple subtypes and circulating recombinant forms of HIV-1.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- Algorithms
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- Antigenic Variation/genetics
- Antigenic Variation/immunology
- Epitopes/chemistry
- Epitopes/genetics
- Epitopes/immunology
- Gene Products, env/chemistry
- Gene Products, env/immunology
- Gene Products, gag/chemistry
- Gene Products, gag/immunology
- Gene Products, pol/chemistry
- Gene Products, pol/immunology
- Genes, MHC Class I
- HIV Antigens/chemistry
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV Infections/immunology
- HIV-1/classification
- HIV-1/immunology
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- HLA-A3 Antigen/genetics
- HLA-A3 Antigen/immunology
- Humans
- Mice
- Mice, Transgenic
- Peptide Fragments/chemical synthesis
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell/immunology
- Sequence Alignment
- T-Cell Antigen Receptor Specificity
- T-Lymphocytes, Cytotoxic/immunology
Collapse
|
15
|
Baird M, Wilson R, Young L, Williman J, Young S, Wilson M, Slobbe L, Lockhart E, Buchan G. Bystander Help within a Polyepitope DNA Vaccine Improves Immune Responses to Influenza Antigens. Scand J Immunol 2004; 60:363-71. [PMID: 15379861 DOI: 10.1111/j.0300-9475.2004.01487.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A polyepitope DNA vaccine has the potential to generate protective immune responses to a range of antigens in a single construct. We investigated whether it was possible to obtain responses to individual epitopes from different antigens, directly linked in a string, and whether the response to a given epitope was enhanced by adjacent epitopes within the construct. A polyepitope plasmid was created, which included three Th epitopes (influenza haemagglutinin, moth cytochrome c and ovalbumin), a Tc epitope (ovalbumin) and two B cell epitopes (haemagglutinin and ovalbumin). Mice were immunized with DNA by using a gene gun. Responses to the polyepitope DNA vaccine were compared with those to DNA vaccine comprising only the haemagglutinin Th and B epitopes (HAT(h)B) or with responses to the recombinant protein. These experiments showed that the polyepitope DNA vaccine induced greater antigen-specific responses to HAT(h)B peptide than the HAT(h)B DNA vaccine. Antigen-specific in vivo cytotoxic responses following polyepitope DNA vaccination were also clearly demonstrable. We conclude that a 'naked DNA' polyepitope vaccine generates specific responses to constituent epitopes and that adjacent irrelevant epitopes may enhance these responses.
Collapse
Affiliation(s)
- M Baird
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Liu F, Whitton JL, Slifka MK. The rapidity with which virus-specific CD8+ T cells initiate IFN-gamma synthesis increases markedly over the course of infection and correlates with immunodominance. THE JOURNAL OF IMMUNOLOGY 2004; 173:456-62. [PMID: 15210805 DOI: 10.4049/jimmunol.173.1.456] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Primary CD8+ T cell responses play a major role in controlling infection by many viruses, and CD8+ memory T cells can confer immunity to virus challenge. In this study we report that for many epitope-specific CD8+ T cell populations, the regulation of an important effector molecule, IFN-gamma, changes dramatically over the course of infection. During the acute phase of infection, many CD8+ T cells exhibit a significant lag before producing IFN-gamma in response to Ag contact; in contrast, the onset of IFN-gamma production by memory cells of the same epitope specificity is markedly accelerated. The biological consequences of this improved responsiveness are manifold. Moreover, during the acute phase of the CD8+ T cell response when immunodominance is being established, there is a strong correlation (p = 0.0002) between the abundance of each epitope-specific T cell population and the rapidity with which it initiates IFN-gamma synthesis. Previous studies have indicated that IFN-gamma plays a critical role in determining the immunodominance hierarchy of an on-going T cell response, and in this report we present evidence for an underlying mechanism: we propose that the CD8+ T cells that most rapidly initiate IFN-gamma production may be at a selective advantage, permitting them to dominate the developing T cell response.
Collapse
Affiliation(s)
- Fei Liu
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
17
|
Leifert JA, Rodriguez-Carreno MP, Rodriguez F, Whitton JL. Targeting plasmid-encoded proteins to the antigen presentation pathways. Immunol Rev 2004; 199:40-53. [PMID: 15233725 DOI: 10.1111/j.0105-2896.2004.0135.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The antigen presentation pathways constitute a fulcrum on which adaptive immunity is balanced, and their manipulation should allow us to induce designer immune responses. The ease and rapidity with which DNA vaccines can be constructed and altered make them ideal candidates with which to test the various targeting strategies that have been conceived to date. These approaches and the mechanisms that may (or may not) underlie their success are reviewed in this article.
Collapse
Affiliation(s)
- Jens A Leifert
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
18
|
Vanniasinkam T, Barton MD, Heuzenroeder MW. The immunogenicity of Rhodococcus equi GroEL2-based vaccines in a murine model. Vet Immunol Immunopathol 2004; 98:91-100. [PMID: 15127846 DOI: 10.1016/j.vetimm.2003.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Rhodococcus equi is a significant intracellular bacterial pathogen in foals. However, at present there is no commercially available vaccine for the prevention of R. equi-induced disease in these animals. Studies have shown that GroEL based vaccines can afford protection against some intracellular pathogens. In this study, the R. equi gene encoding the heat shock protein GroEL2 was cloned and sequenced, with a view to using it as a vaccine candidate. The promoter region of the gene contained two copies of controlling inverted repeat of chaperone expression (CIRCE) motifs, which are well-recognised transcriptional regulators of bacterial heat shock proteins. The R. equi GroEL2 was expressed in E. coli BL21 DE3 with a C-terminal His-tag and sequenced to confirm its identity. The R. equi purified His-tagged GroEL2 protein and a groEL2-based DNA vaccine were used in separate experiments to immunise BALB/c mice. The recombinant protein-based vaccine elicited a mixed Th1/Th2 response whereas the DNA vaccine was found to elicit a predominantly Th1 biased immune response. However, when vaccinated mice were challenged intravenously with 1.5 x 10(7) R. equi neither vaccine elicited enhanced bacterial clearance from the spleen or liver in this model. The reasons for this apparent lack of success are discussed.
Collapse
MESH Headings
- Actinomycetales Infections/immunology
- Actinomycetales Infections/prevention & control
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/biosynthesis
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Bacterial Vaccines/pharmacology
- Base Sequence
- Chaperonin 60/genetics
- Chaperonin 60/immunology
- Cloning, Molecular
- DNA, Bacterial/genetics
- Disease Models, Animal
- Escherichia coli/genetics
- Female
- Genes, Bacterial
- Hypersensitivity, Delayed
- Mice
- Mice, Inbred BALB C
- Phylogeny
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Rhodococcus equi/genetics
- Rhodococcus equi/immunology
- Rhodococcus equi/pathogenicity
- Th1 Cells/immunology
- Th2 Cells/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/pharmacology
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Subunit/pharmacology
Collapse
Affiliation(s)
- Thiru Vanniasinkam
- School of Pharmaceutical, Molecular and Biomedical Sciences, University of South Australia, Adelaide, Australia
| | | | | |
Collapse
|
19
|
Wilson CC, McKinney D, Anders M, MaWhinney S, Forster J, Crimi C, Southwood S, Sette A, Chesnut R, Newman MJ, Livingston BD. Development of a DNA vaccine designed to induce cytotoxic T lymphocyte responses to multiple conserved epitopes in HIV-1. THE JOURNAL OF IMMUNOLOGY 2004; 171:5611-23. [PMID: 14607970 DOI: 10.4049/jimmunol.171.10.5611] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epitope-based vaccines designed to induce CTL responses specific for HIV-1 are being developed as a means for addressing vaccine potency and viral heterogeneity. We identified a set of 21 HLA-A2, HLA-A3, and HLA-B7 restricted supertype epitopes from conserved regions of HIV-1 to develop such a vaccine. Based on peptide-binding studies and phenotypic frequencies of HLA-A2, HLA-A3, and HLA-B7 allelic variants, these epitopes are predicted to be immunogenic in greater than 85% of individuals. Immunological recognition of all but one of the vaccine candidate epitopes was demonstrated by IFN-gamma ELISPOT assays in PBMC from HIV-1-infected subjects. The HLA supertypes of the subjects was a very strong predictor of epitope-specific responses, but some subjects responded to epitopes outside of the predicted HLA type. A DNA plasmid vaccine, EP HIV-1090, was designed to express the 21 CTL epitopes as a single Ag and tested for immunogenicity using HLA transgenic mice. Immunization of HLA transgenic mice with this vaccine was sufficient to induce CTL responses to multiple HIV-1 epitopes, comparable in magnitude to those induced by immunization with peptides. The CTL induced by the vaccine recognized target cells pulsed with peptide or cells transfected with HIV-1 env or gag genes. There was no indication of immunodominance, as the vaccine induced CTL responses specific for multiple epitopes in individual mice. These data indicate that the EP HIV-1090 DNA vaccine may be suitable for inducing relevant HIV-1-specific CTL responses in humans.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/chemical synthesis
- AIDS Vaccines/immunology
- Adult
- Amino Acid Motifs/immunology
- Animals
- Cell Line, Transformed
- Conserved Sequence/immunology
- Cytotoxicity Tests, Immunologic/methods
- Drug Evaluation, Preclinical
- Enzyme-Linked Immunosorbent Assay
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/isolation & purification
- HIV Infections/immunology
- HIV-1/immunology
- HIV-1/isolation & purification
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- HLA-A3 Antigen/genetics
- HLA-A3 Antigen/immunology
- HLA-B7 Antigen/genetics
- HLA-B7 Antigen/immunology
- Histocompatibility Testing
- Humans
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/virology
- Mice
- Mice, Transgenic
- Predictive Value of Tests
- Superantigens/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/virology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/chemical synthesis
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Cara C Wilson
- University of Colorado Health Sciences Center, Denver, CO 80262, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Nagata T, Aoshi T, Uchijima M, Suzuki M, Koide Y. Cytotoxic T-Lymphocyte-, and Helper T-Lymphocyte-Oriented DNA Vaccination. DNA Cell Biol 2004; 23:93-106. [PMID: 15000749 DOI: 10.1089/104454904322759902] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
DNA vaccines have advantages over other types of vaccines in that they can induce strong cellular immune responses, namely cytotoxic T lymphocytes (CTL) and helper T lymphocytes (Th). DNA vaccines are therefore considered a promising alternative to attenuated live vaccines in the field of infectious diseases. So far, various DNA vaccines have been generated and tried to induce a particular cellular immune response by virtue of recombinant DNA technology. DNA vaccines have been designed for efficient transcription and translation of target genes by a variety of strategies. Also, various DNA vaccine strategies for induction of specific CTL and Th have been reported by taking into consideration antigen presentation pathways and the strategies have been shown to be effective to elicit particular T-cell responses. In this paper, we have reviewed these strategies, including our study on epitope-specific T-cell induction by DNA vaccination against Listeria monocytogenes infection. From this review, it has been surmised that, to induce strong immune responses by DNA vaccines, the immunization route and the immunization regimen, such as heterologous "prime-boost" regimen, should also be considered.
Collapse
Affiliation(s)
- Toshi Nagata
- Department of Microbiology and Immunology, Hamamatsu University School of Medicine, Japan.
| | | | | | | | | |
Collapse
|
21
|
Lefrançois L, Marzo A, Williams K. Sustained response initiation is required for T cell clonal expansion but not for effector or memory development in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2832-9. [PMID: 12960304 DOI: 10.4049/jimmunol.171.6.2832] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The factors determining whether an immune response is productive are poorly understood. To understand the circumstances affecting the early stage of the immune response which determine whether memory is generated, the CD8 T cell response was mapped in detail following immunization with live or heat-killed bacteria. Our results demonstrate that even in response to a weak immunogen, functional memory cell development is linked to effector cell induction in lymphoid and nonlymphoid tissues. The main defect in the response to killed microorganisms is inefficient induction of clonal expansion. This failure is due to a contracted, but costimulation-dependent activation phase in the lymphoid tissues, resulting in rapid but abortive growth. Conversely, the response to live bacteria is characterized by protracted early T cell sequestration in lymphoid tissues. Thus, memory development requires effector induction, while optimal clonal expansion is regulated by the duration of response initiation.
Collapse
Affiliation(s)
- Leo Lefrançois
- Department of Medicine, Division of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | | | | |
Collapse
|
22
|
Rodríguez EG, Vázquez DM, Herrera AM, Duarte CA. Enhanced cell-mediated IFN-gamma-secreting activity against the HIV-1IIIB V3 peptide of the TAB9 multiepitope after DNA vaccine backbone engineering. Biochem Biophys Res Commun 2003; 308:713-8. [PMID: 12927777 DOI: 10.1016/s0006-291x(03)01462-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The SV40t polyadenylation and splicing signals of the pAEC plasmid vectors were replaced by synthetic intron and synthetic rabbit beta globin-based termination/polyadenylation sequences, and 5, 10, and 20 copies of the 5'-AACGTT-3' CpG motif were inserted. Balb/c mice were immunized by intramuscular injection of 200 microg of each plasmid, coding for the HIV-1 multiepitope TAB9, under the control of the human cytomegalovirus promoter. After three doses of DNA, a fourth boost with plasmid DNA or a TAB9-expressing recombinant fowlpox virus rFPTAB9LZ was administered. ELISA and ELISPOT assays were conducted for antibody and IFN-gamma-secreting cell-mediated responses' evaluation against the whole TAB9 and the TAB9's IIIB V3 peptide, respectively. Serum IgG antibodies were not detected. Effector IFN-gamma-secreting responses were only detected on the animals receiving the new set of DNA constructs, alone or in combination with a recombinant virus boost, with or without in vitro re-stimulation. The response was dependent on the new transcriptional unit and influenced by the number of CpG motifs. We showed that plasmid backbone optimization based on these two factors could enhance the response against a multiepitope-based DNA vaccine. A new family of plasmid vectors is also available for evaluation with desired antigens.
Collapse
Affiliation(s)
- Ernesto G Rodríguez
- Vaccine Division, Center for Genetic Engineering and Biotechnology of Havana, P.O. Box 6162, Havana 10600, Cuba.
| | | | | | | |
Collapse
|
23
|
Guo CC, Ding J, Pan BR, Yu ZC, Han QL, Meng FP, Liu N, Fan DM. Development of an oral DNA vaccine against MG7-Ag of gastric cancer using attenuated salmonella typhimurium as carrier. World J Gastroenterol 2003; 9:1191-5. [PMID: 12800222 PMCID: PMC4611782 DOI: 10.3748/wjg.v9.i6.1191] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop an oral DNA vaccine against gastric cancer and evaluate its efficacy in mice.
METHODS: The genes of the MG7-Ag mimotope and a universal Th epitope (Pan-DR epitope, PADRE) were included in the PCR primers. By PCR, the fusion gene of the two epitopes was amplified. The fusion gene was confirmed by sequencing and was then cloned into pcDNA3.1 (+) plasmid. The pcDNA3.1 (+)-MG7/PADRE was used to transfect an attenuated Salmonella typhimurium. C57BL/6 mice were orally immunized with 1 × 108 cfu Salmonella transfectants. Salmonella harboring the empty pcDNA3.1 (+) plasmid and phosphate buffer saline (PBS) were used as negative controls. At the 6th week, serum titer of MG7-Ag specific antibody was detected by ELISA. At the 8th week cellular immunity was detected by an unprimed proliferation test of the spleenocytes by using a [3H]-thymidine incorporation assay. Ehrlich ascites carcinoma cells expressing MG7-Ag were used as a model in tumor challenge assay to evaluate the protective effect of the vaccine.
RESULTS: Serum titer of antibody against MG7-Ag was significantly higher in mice immunized with the vaccine than that in control groups (0.841 vs 0.347, P < 0.01; 0.841 vs 0.298, P < 0.01), while in vitro unprimed proliferation assay of the spleenocytes showed no statistical difference between those three groups. Two weeks after tumor challenge, 2 in 7 immunized mice were tumor free, while all the mice in the control groups showed tumor formation.
CONCLUSION: Oral DNA vaccine against the MG7-Ag momitope of gastric cancer is immunogenic. It can induce significant humoral immunity against tumor in mice, and the vaccine has partially protective effects.
Collapse
Affiliation(s)
- Chang-Cun Guo
- Institute of Digestive Disease, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Lásaro MO, Alves AMB, Botosso VF, Durigon EL, Ferreira LCS. Antibody-inducing properties of a prototype bivalent herpes simplex virus/enterotoxigenic Escherichia coli DNA vaccine. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2003; 35:25-31. [PMID: 12589954 DOI: 10.1111/j.1574-695x.2003.tb00645.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The antibody-inducing properties of a bacterial/viral bivalent DNA vaccine (pRECFA), expressing a peptide composed of N- and C-terminal amino acid sequences of the herpes simplex virus type 1 (HSV-1) glycoprotein D (gD) fused with an inner segment encoding the major structural subunit of enterotoxigenic Escherichia coli (ETEC) CFA/I fimbriae (CFA/I), was evaluated in BALB/c mice following intramuscular immunization. The bivalent pRECFA vaccine elicited serum antibody responses, belonging mainly to the IgG2a subclass, against both CFA/I and HSV gD proteins. pRECFA-elicited antibody responses cross-reacted with homologous and heterologous ETEC fimbrial antigens as well as with type 1 and type 2 HSV gD proteins, which could bind and inactivate intact HSV-2 particles. On the other hand, CFA/I-specific antibodies could bind but did not neutralize the adhesive functions of the bacterial CFA/I fimbriae. In spite of the functional restriction of the antibodies targeting the bacterial antigen, the present evidence suggests that fusion of heterologous peptides to the HSV gD protein represents an alternative for the design of bivalent DNA vaccines able to elicit serum antibody responses.
Collapse
Affiliation(s)
- Marcio O Lásaro
- Federal University of Rio de Janeiro, Carlos Chagas Filho Biophysics Institute, Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
25
|
Leifert JA, Harkins S, Whitton JL. Full-length proteins attached to the HIV tat protein transduction domain are neither transduced between cells, nor exhibit enhanced immunogenicity. Gene Ther 2002; 9:1422-8. [PMID: 12378404 DOI: 10.1038/sj.gt.3301819] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2002] [Accepted: 05/23/2002] [Indexed: 01/08/2023]
Abstract
Several proteins have been accorded the unusual ability to translocate across cell membranes in a receptor-independent and temperature-independent manner, and this activity has been mapped to a highly basic series of residues currently termed a 'protein transduction domain' (PTD). This translocatory attribute, if authentic, would be valuable for purposes of gene therapy and vaccination. We have evaluated the PTD from the human immunodeficiency virus type 1 (HIV) tat protein and we conclude that, when synthesized de novo, (1) the HIV tat PTD does not enhance the immunogenicity of a full-length protein to which it is tethered; and (2) the HIV tat PTD does not cause intercellular transfer of an attached marker protein, as judged by careful quantitative analyses. From our data, and from a review of published materials, we suggest that contrary to current dogma there is little evidence that these supposedly translocatory proteins can move between live cells. Furthermore, we suggest that PTDs do not act to enhance translocation, but instead merely to increase binding to the cell surface; in which case, the term 'protein transduction domain', and the related acronym, are misnomers which should be abandoned. Our conclusions explain why the most dramatic demonstrations of PTD efficacy have been obtained using fixed cells and/or denatured proteins, and have obvious implications for gene therapy and vaccination.
Collapse
Affiliation(s)
- J A Leifert
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
26
|
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, CVN-9, Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
27
|
Alexander J, Oseroff C, Dahlberg C, Qin M, Ishioka G, Beebe M, Fikes J, Newman M, Chesnut RW, Morton PA, Fok K, Appella E, Sette A. A decaepitope polypeptide primes for multiple CD8+ IFN-gamma and Th lymphocyte responses: evaluation of multiepitope polypeptides as a mode for vaccine delivery. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:6189-98. [PMID: 12055232 DOI: 10.4049/jimmunol.168.12.6189] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Proteins are generally regarded as ineffective immunogens for CTL responses. We synthesized a 100-mer decaepitope polypeptide and tested its capacity to induce multiple CD8(+) IFN-gamma and Th lymphocyte (HTL) responses in HLA transgenic mice. Following a single immunization in the absence of adjuvant, significant IFN-gamma in vitro recall responses were detected for all epitopes included in the construct (six A2.1-, three A11-restricted CTL epitopes, and one universal HTL epitope). Immunization with truncated forms of the decaepitope polypeptide was used to demonstrate that optimal immunogenicity was associated with a size of at least 30-40 residues (3-4 epitopes). Solubility analyses of the truncated constructs were used to identify a correlation between immunogenicity for IFN-gamma responses and the propensity of these constructs to form particulate aggregates. Although the decaepitope polypeptide and a pool of epitopes emulsified in IFA elicited similar levels of CD8(+) responses using fresh splenocytes, we found that the decaepitope polypeptide more effectively primed for in vitro recall CD8(+) T cell responses. Finally, immunogenicity comparisons were also made between the decaepitope polypeptide and a corresponding gene encoding the same polypeptide delivered by naked DNA immunization. Although naked DNA immunization induced somewhat greater direct ex vivo and in vitro recall responses 2 wk after a single immunization, only the polypeptide induced significant in vitro recall responses 6 wk following the priming immunization. These studies support further evaluation of multiepitope polypeptide vaccines for induction of CD8(+) IFN-gamma and HTL responses.
Collapse
MESH Headings
- Animals
- Buffers
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- DNA/administration & dosage
- DNA/immunology
- Drug Contamination
- Emulsions
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Freund's Adjuvant/immunology
- HLA Antigens/genetics
- HLA Antigens/immunology
- Humans
- Injections, Intramuscular
- Injections, Subcutaneous
- Interferon-gamma/biosynthesis
- Jurkat Cells
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Peptide Fragments/administration & dosage
- Peptide Fragments/chemical synthesis
- Peptide Fragments/immunology
- Solubility
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Transgenes/immunology
- Vaccination/methods
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
|
28
|
Wolkers MC, Toebes M, Okabe M, Haanen JBAG, Schumacher TNM. Optimizing the efficacy of epitope-directed DNA vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4998-5004. [PMID: 11994451 DOI: 10.4049/jimmunol.168.10.4998] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
An increasing number of clinical trials has been initiated to test the potential of prophylactic or curative vaccination with tumor Ag-encoding DNA vaccines. However, in the past years it has become apparent that for many Ags and in particular for tumor Ags the intracellular processing and presentation are suboptimal. To improve epitope-directed DNA vaccines we have developed a murine model system in which epitope-specific, DNA vaccine-induced T cell immunity can be followed by MHC tetramer technology directly ex vivo. We have used this well-defined model to dissect the parameters that are crucial for the induction of strong cytotoxic T cell immunity using two independent model Ags. These experiments have led to a set of five guidelines for the design of epitope-directed DNA vaccines, indicating that carboxyl-terminal fusion of the epitope to a carrier protein of foreign origin is the most favorable strategy. DNA vaccines that are based on these guidelines induce high-magnitude CD8(+) T cell responses in >95% of vaccinated animals. Moreover, T cell immunity induced by this type of optimized DNA vaccine provides long-term protection against otherwise lethal tumor challenges.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Artificial Gene Fusion
- Carrier Proteins/administration & dosage
- Carrier Proteins/genetics
- Carrier Proteins/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Green Fluorescent Proteins
- Immunity, Active/genetics
- Luminescent Proteins/administration & dosage
- Luminescent Proteins/genetics
- Luminescent Proteins/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation/immunology
- Nucleocapsid Proteins
- Nucleoproteins/administration & dosage
- Nucleoproteins/genetics
- Nucleoproteins/immunology
- Papillomaviridae/immunology
- Papillomavirus Infections/genetics
- Papillomavirus Infections/prevention & control
- Peptide Fragments/administration & dosage
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- RNA-Binding Proteins
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
- Tumor Cells, Cultured/transplantation
- Tumor Virus Infections/genetics
- Tumor Virus Infections/prevention & control
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Core Proteins/administration & dosage
- Viral Core Proteins/genetics
- Viral Core Proteins/immunology
Collapse
Affiliation(s)
- Monika C Wolkers
- Department of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
Rodriguez F, Harkins S, Slifka MK, Whitton JL. Immunodominance in virus-induced CD8(+) T-cell responses is dramatically modified by DNA immunization and is regulated by gamma interferon. J Virol 2002; 76:4251-9. [PMID: 11932390 PMCID: PMC155093 DOI: 10.1128/jvi.76.9.4251-4259.2002] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The phenomenon whereby the host immune system responds to only a few of the many possible epitopes in a foreign protein is termed immunodominance. Immunodominance occurs not only during microbial infection but also following vaccination, and clarification of the underlying mechanism may permit the rational design of vaccines which can circumvent immunodominance, thereby inducing responses to all epitopes, dominant and subdominant. Here, we show that immunodominance affects DNA vaccines and that the effects can be avoided by the simple expedient of epitope separation. DNA vaccines encoding isolated dominant and subdominant epitopes induce equivalent responses, confirming a previous demonstration that coexpression of dominant and subdominant epitopes on the same antigen-presenting cell (APC) is central to immunodominance. We conclude that multiepitope DNA vaccines should comprise a cocktail of plasmids, each with its own epitope, to allow maximal epitope dispersal among APCs. In addition, we demonstrate that subdominant responses are actively suppressed by dominant CD8(+) T-cell responses and that gamma interferon (IFN-gamma) is required for this suppression. Furthermore, priming of CD8(+) T cells to a single dominant epitope results in strong suppression of responses to other normally dominant epitopes in immunocompetent mice, in effect rendering these epitopes subdominant; however, responses to these epitopes are increased 6- to 20-fold in mice lacking IFN-gamma. We suggest that, in agreement with our previous observations, IFN-gamma secretion by CD8(+) T cells is highly localized, and we propose that its immunosuppressive effect is focused on the APC with which the dominant CD8(+) T cell is in contact.
Collapse
Affiliation(s)
- Fernando Rodriguez
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
30
|
Delogu G, Li A, Repique C, Collins F, Morris SL. DNA vaccine combinations expressing either tissue plasminogen activator signal sequence fusion proteins or ubiquitin-conjugated antigens induce sustained protective immunity in a mouse model of pulmonary tuberculosis. Infect Immun 2002; 70:292-302. [PMID: 11748195 PMCID: PMC127618 DOI: 10.1128/iai.70.1.292-302.2002] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
DNA vaccination has emerged as a powerful approach in the search for a more efficacious vaccine against tuberculosis. In this study, we evaluated the effectiveness of immunizing with combinations of 10 different tuberculosis DNA vaccines that expressed mycobacterial proteins fused at the N terminus to eukaryotic intracellular targeting sequences. In one vaccine combination, the genes were fused to the tissue plasminogen activator signal sequence (TPA), while in a second combination the same 10 genes were expressed as ubiquitin (Ub)-conjugated proteins. In ex vivo studies in which the secretion of gamma interferon was measured, cellular immune responses were detected in mice vaccinated with either the TPA DNA vaccine combination or the Ub DNA vaccine combination at 7 and 14 days following a low-dose Mycobacterium tuberculosis challenge. Moreover, mice vaccinated with the TPA combination, the Ub combination, and Mycobacterium bovis BCG were able to limit the growth of tubercle bacilli in the lung and spleen after a virulent tuberculous aerosol challenge. Histopathological analyses also showed that mice immunized with the DNA vaccine combinations had substantially improved postinfection lung pathology relative to the naïve controls. Finally, in three different long-term experiments, the survival periods following aerogenic challenge were extended as much as sevenfold for vaccinated mice compared to naïve controls. Interestingly, in all three experiments, no significant differences were detected in the mean times to death for mice immunized with the TPA combination or the Ub combination relative to the BCG controls. In conclusion, these studies demonstrate the effectiveness of immunization with DNA vaccine combinations against tuberculosis and suggest that further testing of these plasmid cocktails is warranted.
Collapse
Affiliation(s)
- Giovanni Delogu
- Laboratory of Mycobacterial Diseases and Cellular Immunology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
31
|
Talaat AM, Lyons R, Johnston SA. A combination vaccine confers full protection against co-infections with influenza, herpes simplex and respiratory syncytial viruses. Vaccine 2001; 20:538-44. [PMID: 11672920 DOI: 10.1016/s0264-410x(01)00352-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Combined/composite vaccines should be useful in reducing the number of vaccinations and provide more flexibility in confronting biological warfare scenarios. We tested the effectiveness of a composite genetic vaccine designed from previously known protective antigens directed against influenza A virus (INF-A), herpes simplex virus type-1 (HSV-1) and respiratory syncytial virus (RSV) in a mouse-based challenge. Immunizing mice with a pool of four plasmids; INF-A haemagglutinin (HA), INF-A nucleoprotein (NP), HSV-1 glycoprotein D (gD) and RSV glycoprotein F, against the three pathogens provided full protection when mice were challenged with each pathogen. Remarkably, mice challenged with all three pathogens at once were also fully protected, even when a bacterial pathogen, Mycoplasma pulmonis, was included in the challenge. If these results are extendable to other combinations of vaccines in other hosts, it would support the development of gene vaccines as multi-component, combination vaccines.
Collapse
Affiliation(s)
- A M Talaat
- Department of Internal Medicine, Center for Biomedical Inventions, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., TX 75390-8573, USA
| | | | | |
Collapse
|
32
|
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
|
33
|
Leifert JA, Lindencrona JA, Charo J, Whitton JL. Enhancing T cell activation and antiviral protection by introducing the HIV-1 protein transduction domain into a DNA vaccine. Hum Gene Ther 2001; 12:1881-92. [PMID: 11589830 DOI: 10.1089/104303401753153938] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Protein transduction domains (PTD), which can transport proteins or peptides across biological membranes, have been identified in several proteins of viral, invertebrate, and vertebrate origin. Here, we evaluate the immunological and biological consequences of including PTD in synthetic peptides and in DNA vaccines that contain CD8(+) T cell epitopes from lymphocytic choriomeningitis virus (LCMV). Synthetic PTD-peptides did not induce detectable CD8(+) T cell responses. However, fusion of an open reading frame encoding a PTD to an epitope minigene caused transfected tissue culture cells to stimulate epitope-specific T cells much more effectively. Kinetic studies indicated that the epitope reached the surface of transfected cells more rapidly and that the number of transfected cells needed to stimulate T cell responses was reduced by 35- to 50-fold when compared to cells transfected with a standard minigene plasmid. The mechanism underlying the effect of PTD linkage is not clear, but transit of the PTD-attached epitope from transfected cells to nontransfected cells (cross presentation) seemed to play, at most, a minimal role. Mice immunized once with the plasmid encoding the PTD-linked epitope showed a markedly accelerated CD8(+) T cell response and, unlike mice immunized with a standard plasmid, were completely protected against a normally lethal LCMV challenge administered only 8 days post-immunization.
Collapse
Affiliation(s)
- J A Leifert
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
34
|
Fujimura AE, Kinoshita SS, Pereira-Chioccola VL, Rodrigues MM. DNA sequences encoding CD4+ and CD8+ T-cell epitopes are important for efficient protective immunity induced by DNA vaccination with a Trypanosoma cruzi gene. Infect Immun 2001; 69:5477-86. [PMID: 11500420 PMCID: PMC98660 DOI: 10.1128/iai.69.9.5477-5486.2001] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunization of BALB/c mice with a plasmid containing the gene for Trypanosoma cruzi trans-sialidase (TS) induced antibodies that inhibited TS enzymatic activity, CD4+ Th1 and CD8+ Tc1 cells, and protective immunity against infection. We used this model to obtain basic information on the requirement of CD4 or CD8 or B-cell epitopes for an effective DNA-induced immunity against T. cruzi infection. For that purpose, mice were immunized with plasmids containing DNA sequences encoding (i) the entire TS protein, (ii) the TS enzymatic domain, (iii) the TS CD4+ T-cell epitopes, (iv) the TS CD8+ T-cell epitope, or (v) TS CD4+ and CD8+ T-cell epitopes. Plasmids expressing the entire TS or its enzymatic domain elicited similar levels of TS-inhibitory antibodies, gamma interferon (IFN-gamma)-producing T cells, and protective immunity against infection. Although the plasmid expressing TS CD4 epitopes was immunogenic, its protective efficacy against experimental infection was limited. The plasmid expressing the CD8 epitope was poorly immunogenic and provided little protective immunity. The reason for the limited priming of CD8+ T cells was due to a requirement for CD4+ T cells. To circumvent this problem, a plasmid expressing both CD4+ and CD8+ T-cell epitopes was produced. This plasmid generated levels of IFN-gamma-producing T cells and protective immunity comparable to that of the plasmid expressing the entire catalytic domain of TS. Our observations suggest that plasmids expressing epitopes recognized by CD4+ and CD8+ T cells may have a better protective potential against infection with T. cruzi.
Collapse
Affiliation(s)
- A E Fujimura
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, Brazil
| | | | | | | |
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
|
Musacchio A, Rodriguez EG, Herrera AM, Quintana D, Muzio V. Multivalent DNA-based immunization against hepatitis B virus with plasmids encoding surface and core antigens. Biochem Biophys Res Commun 2001; 282:442-6. [PMID: 11401479 DOI: 10.1006/bbrc.2001.4580] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The immune response against hepatitis B surface and core antigens was evaluated by either coinoculation or independent intramuscular administration of pAEC compact DNA immunization vectors carrying their genes. The pAEC vectors bear just the essential elements for mammalian expression and bacterial amplification. Balb/c mice were immunized with 100 microg of each construct, either alone or in combination. In spite of lacking known immunostimulatory sequences (e.g., AACGTT), significant cellular (proliferative) and humoral immune responses were raised against both antigens. Coadministration of both plasmids maintained the immune response against the two antigens, without interference between them. Modulation of the antigen expression and further immune response, by using the Kozak's translation initiation sequence, was also analyzed. No differences due to its presence or absence were observed.
Collapse
Affiliation(s)
- A Musacchio
- Vaccine Division, Center for Genetic Engineering and Biotechnology of Havana, Havana, 10 600, Cuba.
| | | | | | | | | |
Collapse
|
37
|
Slifka MK, Pagarigan R, Mena I, Feuer R, Whitton JL. Using recombinant coxsackievirus B3 to evaluate the induction and protective efficacy of CD8+ T cells during picornavirus infection. J Virol 2001; 75:2377-87. [PMID: 11160741 PMCID: PMC114821 DOI: 10.1128/jvi.75.5.2377-2387.2001] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Coxsackievirus B3 (CVB3) is a common human pathogen that has been associated with serious diseases including myocarditis and pancreatitis. To better understand the effect of cytotoxic T-lymphocyte (CTL) responses in controlling CVB3 infection, we have inserted well-characterized CTL epitopes into the CVB3 genome. Constructs were made by placing the epitope of interest upstream of the open reading frame encoding the CVB3 polyprotein, separated by a poly-glycine linker and an artificial 3Cpro/3CDpro cleavage site. This strategy results in the foreign protein being translated at the amino- terminus of the viral polyprotein, from which it is cleaved prior to viral assembly. In this study, we cloned major histocompatibility complex class I-restricted CTL epitopes from lymphocytic choriomeningitis virus (LCMV) into recombinant CVB3 (rCVB3). In vitro, rCVB3 growth kinetics showed a 1- to 2-h lag period before exponential growth was initiated, and peak titers were approximately 1 log unit lower than for wild-type virus. rCVB3 replicated to high titers in vivo and caused severe pancreatitis but minimal myocarditis. Despite the high virus titers, rCVB3 infection of naive mice failed to induce a strong CD8+ T-cell response to the encoded epitope; this has implications for the proposed role of "cross-priming" during virus infection and for the utility of recombinant picornaviruses as vaccine vectors. In contrast, rCVB3 infection of LCMV-immune mice resulted in direct ex vivo cytotoxic activity against target cells coated with the epitope peptide, demonstrating that the rCVB3-encoded LCMV-specific epitope was expressed and presented in vivo. The preexisting CD8+ memory T cells could limit rCVB replication; compared to naive mice, infection of LCMV-immune mice with rCVB3 resulted in approximately 50-fold-lower virus titers in the heart and approximately 6-fold-lower virus titers in the pancreas. Although the inserted CTL epitope was retained by rCVB3 through several passages in tissue culture, it was lost in an organ-specific manner in vivo; a substantial proportion of viruses from the pancreas retained the insert, compared to only 0 to 1.8% of myocardial viruses. Together, these results show that expression of heterologous viral proteins by recombinant CVB3 provides a useful model for determining the mechanisms underlying the immune response to this viral pathogen.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral
- CD8-Positive T-Lymphocytes/immunology
- Coxsackievirus Infections/immunology
- Coxsackievirus Infections/virology
- Enterovirus B, Human/genetics
- Enterovirus B, Human/growth & development
- Enterovirus B, Human/immunology
- Enterovirus B, Human/pathogenicity
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Humans
- Immunologic Memory
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Recombination, Genetic
- Transfection
- Viral Proteins
- Virus Replication
Collapse
Affiliation(s)
- M K Slifka
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- F Rodriguez
- Department of Neuropharmacology, CVN-9, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, 92037, USA
| | | |
Collapse
|