Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
J Med Virol ; 95(10): e29183, 2023 01.
Article in English | MEDLINE | ID: mdl-37861466

ABSTRACT

Life-long persistent herpesviruses carry "trans-inducers" to overcome the unusual codon usage of their glycoproteins for efficient expression. Strikingly, this "trans-inducibility" can be achieved by simply changing the codon-usage of acute virus glycoproteins to that of persistent herpesvirus glycoproteins with herpesviral trans-inducer. Here, we apply the "persistent viral codon-usage-trans-inducer" principle to SARS-CoV-2 Spike mRNA vaccine platform, in which the codon-usage of Spike is changed to that of Herpes Simplex Virus-1 (HSV-1) glycoprotein B (gB) with its "trans-inducer" ICP27. The HSVgB-ICP27-codon-optimized Spike mRNA vaccine induced markedly high antigen expression and stability, total IgG, neutralizing antibody, and T cell response, ultimately enhancing protection against lethal SARS-CoV-2 challenge. Moreover, the HSVgB- codon-optimized Delta (B.1.617.2) strain Spike mRNA vaccine provided significant enhancements in antigen expression and long-term protection against SARS-CoV-2 challenge. Thus, we report a novel persistent viral codon-usage-trans-inducer mRNA vaccine platform for enhanced antigen expression and long-term protection against lethal viral infection.


Subject(s)
COVID-19 Vaccines , COVID-19 , Codon , Spike Glycoprotein, Coronavirus , Humans , Antibodies, Neutralizing , Antibodies, Viral , Codon/genetics , Codon/immunology , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Glycoproteins , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines
2.
J Med Virol ; 93(6): 3549-3556, 2021 06.
Article in English | MEDLINE | ID: mdl-32940917

ABSTRACT

Rotavirus is the important etiological agents of infectious diarrhea among children under 5 years old. Rotaviruses are divided into 10 serogroups (A-J) and each group is based on genetic properties of major structural protein VP6. We designed a novel VP6 sequence optimization to increase the expression level of this protein. Numerous factors such as codon adaptation index, codon pair bias, and guanine-cytosine content were adapted based on Escherichiacoli codon usage. In addition, the ribosome binding site (RBS) of pET-15b was redesigned by the RBS calculator and the secondary structure of VP6 messenger RNA was optimized in the whole length of the coding sequence. Various factors including isopropyl beta- d-thiogalactoside (IPTG) concentration, temperature, and induction time were analyzed for the optimization of the best expression in E. coli by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blotting. The recombinant VP6 (rVP6) protein was purified by the Ni-sepharose and then the hyperimmune sera were generated against rVP6 in rabbits. Among three different temperatures, IPTG concentrations, and postinductions, the level of rVP6 was higher at 37°C, 1 mM of IPTG, and 8 h, respectively. Also, the high expression level of rVP6 was obtained in the insoluble aggregate form (43.8 g/L). After purification, the yield of rVP6 was 10.83 g/L. The rVP6 specific antiserum was confirmed by both immunofluorescent and western blotting. The versatile sequence optimization was the reason to produce a high level of rVP6 compared to other reports and can potentially apply to produce cheaper commercial kits to diagnose serological tests and new rotavirus vaccines.


Subject(s)
Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Escherichia coli/genetics , Rotavirus Vaccines/immunology , Rotavirus/genetics , Rotavirus/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/isolation & purification , Capsid Proteins/isolation & purification , Codon/genetics , Codon/immunology , Female , Humans , Immunization/methods , Immunization, Secondary , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Rotavirus/chemistry , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Vaccines, Synthetic/administration & dosage
3.
Malar J ; 18(1): 394, 2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31796032

ABSTRACT

BACKGROUND: Despite the extensive endeavours, developing an effective malaria vaccine remains as a great challenge. Apical membrane antigen 1 (AMA-1) located on the merozoite surface of parasites belonging to the genus Plasmodium is involved in red blood cell invasion. METHODS: Influenza virus-like particle (VLP) vaccines containing codon-optimized or native (non-codon optimized) AMA-1 from Plasmodium berghei were generated. VLP-induced protective immunity was evaluated in a mouse model. RESULTS: Mice immunized with VLP vaccine containing the codon-optimized AMA-1 elicited higher levels of P. berghei-specific IgG and IgG2a antibody responses compared to VLPs containing non-codon optimized AMA-1 before and after challenge infection. Codon-optimized AMA-1 VLP vaccination induced higher levels of CD4+ T cells, CD8+ T cells, B cells, and germinal centre cell responses compared to non-codon optimized AMA-1 VLPs. Importantly, the codon-optimized AMA-1 VLP vaccination showed lower body weight loss, longer survival and a significant decrease in parasitaemia compared to non-codon optimized VLP vaccination. CONCLUSION: Overall, VLP vaccine expressing codon-optimized AMA-1 induced better protective efficacy than VLPs expressing the non-codon optimized AMA-1. Current findings highlight the importance of codon-optimization for vaccine use and its potential involvement in future malaria vaccine design strategies.


Subject(s)
Antigens, Protozoan/therapeutic use , Malaria Vaccines/pharmacology , Malaria/prevention & control , Membrane Proteins/therapeutic use , Plasmodium berghei/immunology , Protozoan Proteins/therapeutic use , Vaccines, Virus-Like Particle/pharmacology , Animals , Codon/immunology , Female , Mice , Mice, Inbred BALB C
4.
Hum Immunol ; 80(7): 493-502, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30769032

ABSTRACT

Enterobacterial pathogens that have acquired antibiotic resistance genes are a leading cause of community and hospital acquired infections. In such a situation vaccination is considered as a better option to prevent such infections. In the current study reverse vaccinology approach has been used to select peptides from already known immunogenic proteins to design a chimeric construct. We selected Yersiniabactin receptor of Escherichia coli UMN026 and Flagellin of Stenotrophomonas maltophila. B-cell linear epitopes were predicted using Bepipred prediction tool. Peptide binding with reference sets of 27 alleles of MHC class I and class II was also analyzed. The predicted peptides-MHC complexes were further validated using simulation dynamics. The in-silico construction of chimera was done by restriction mapping and codon optimization. Chimera was evaluated using the immunoinformatic approach as done for the selected proteins. From the 673 amino acids of FyuA protein, a region from 1 to 492 was selected for containing more linear epitopes and the processing scores obtained were significant for MHC class I and class II binding. Similarly, from Flagellin, a region between 60 and 328 amino acids was selected and the peptides present in the selected region showed lower percentile ranks for binding with MHC molecules. The simulation studies validated the predictions of peptide-MHC complexes. The selected gene fragments accommodating maximum part of these peptides were used to design a chimaeric construct of 2454 bp. From the immunoinformatic analysis, the chimera was found to be more immunogenic in terms of increased number of B-cell and T-cell epitopes along with increased coverage of global populations with allelic variability.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Escherichia coli/immunology , Molecular Docking Simulation/methods , Vaccines/immunology , Alleles , Amino Acid Sequence , Antigens, Bacterial/immunology , Chimera/immunology , Codon/immunology , Epitopes, T-Lymphocyte/immunology , Flagellin/genetics , Flagellin/immunology , Genes, MHC Class I/immunology , Genes, MHC Class II/immunology , Humans , Phenols/immunology , Protein Binding , Protein Structure, Secondary , Sequence Analysis, Protein , Stenotrophomonas/immunology , Thiazoles/immunology
5.
Vaccine ; 35(44): 6007-6014, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28629922

ABSTRACT

Smallpox is a disease caused by Variola virus (VARV). Although eradicated by WHO in 1980, the threat of using VARV on a bioterror attack has increased. The current smallpox vaccine ACAM2000, which consists of live vaccinia virus (VACV), causes complications in individuals with a compromised immune system or with previously reported skin diseases. Thus, a safer and efficacious vaccine needs to be developed. Previously, we reported that our virus-free DNA vaccine formulation, a pVAX1 plasmid encoding codon-optimized VACV A27L gene (pA27LOPT) with and without Imiquimod adjuvant, stimulates A27L-specific production of IFN-γ and increases humoral immunity 7days post-vaccination. Here, we investigated the immune response of our novel vaccine by measuring the frequency of splenocytes producing IFN-γ by ELISPOT, the TH1 and TH2 cytokine profiles, and humoral immune responses two weeks post-vaccination, when animals were challenged with VACV. In all assays, the A27-based DNA vaccine conferred protective immune responses. Specifically, two weeks after vaccination, mice were challenged intranasally with vaccinia virus, and viral titers in mouse lungs and ovaries were significantly lower in groups immunized with pA27LOPT and pA27LOPT+Imiquimod. These results demonstrate that our vaccine formulation decreases viral replication and dissemination in a virus-free DNA vaccine platform, and provides an alternative towards a safer an efficacious vaccine.


Subject(s)
Codon/immunology , Plasmids/immunology , Vaccinia virus/immunology , Viral Proteins/immunology , Virus Replication/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/immunology , Enzyme-Linked Immunospot Assay/methods , Female , Immunity, Humoral/immunology , Mice , Mice, Inbred BALB C , Smallpox/immunology , Smallpox/prevention & control , Smallpox Vaccine/immunology , Vaccination/methods , Vaccines, DNA/immunology , Variola virus/immunology
6.
Genetics ; 206(1): 417-427, 2017 05.
Article in English | MEDLINE | ID: mdl-28315836

ABSTRACT

Phylogenetic methods have shown promise in understanding the development of broadly neutralizing antibody lineages (bNAbs). However, the mutational process that generates these lineages, somatic hypermutation, is biased by hotspot motifs which violates important assumptions in most phylogenetic substitution models. Here, we develop a modified GY94-type substitution model that partially accounts for this context dependency while preserving independence of sites during calculation. This model shows a substantially better fit to three well-characterized bNAb lineages than the standard GY94 model. We also demonstrate how our model can be used to test hypotheses concerning the roles of different hotspot and coldspot motifs in the evolution of B-cell lineages. Further, we explore the consequences of the idea that the number of hotspot motifs, and perhaps the mutation rate in general, is expected to decay over time in individual bNAb lineages.


Subject(s)
Antibodies, Neutralizing/genetics , Cell Lineage/genetics , Evolution, Molecular , Phylogeny , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Cell Lineage/immunology , Codon/genetics , Codon/immunology , Humans , Models, Genetic , Mutation
7.
Virology ; 501: 35-46, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27855284

ABSTRACT

Several arenaviruses, chiefly Lassa (LASV) in West Africa, cause hemorrhagic fever (HF) disease in humans and pose important public health problems in their endemic regions. To date, there are no FDA-approved arenavirus vaccines and current anti-arenaviral therapy is limited to the use of ribavirin that has very limited efficacy. In this work we document that a recombinant prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) with a codon deoptimized (CD) surface glycoprotein (GP), rLCMV/CD, exhibited wild type (WT)-like growth properties in cultured cells despite barely detectable GP expression levels in rLCMV/CD-infected cells. Importantly, rLCMV/CD was highly attenuated in vivo but able to induce complete protection against a subsequent lethal challenge with rLCMV/WT. Our findings support the feasibility of implementing an arenavirus GP CD-based approach for the development of safe and effective live-attenuated vaccines (LAVs) to combat diseases caused by human pathogenic arenaviruses.


Subject(s)
Arenaviridae Infections/virology , Arenavirus/genetics , Codon/genetics , Glycoproteins/genetics , Vaccines, Attenuated/genetics , Viral Proteins/genetics , Viral Vaccines/genetics , Amino Acid Sequence , Animals , Arenaviridae Infections/immunology , Arenavirus/immunology , Codon/immunology , Glycoproteins/administration & dosage , Glycoproteins/immunology , Humans , Mice , Molecular Sequence Data , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Proteins/administration & dosage , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
8.
Bioengineered ; 7(3): 155-65, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27246656

ABSTRACT

Recombinant protein subunit vaccines are formulated using defined protein antigens that can be produced in heterologous expression systems. The methylotrophic yeast Pichia pastoris has become an important host system for the production of recombinant subunit vaccines. Although many basic elements of P. pastoris expression system are now well developed, there is still room for further optimization of protein production. Codon bias, gene dosage, endoplasmic reticulum protein folding and culture condition are important considerations for improved production of recombinant vaccine antigens. Here we comment on current advances in the application of P. pastoris for the synthesis of recombinant subunit vaccines.


Subject(s)
Antigens, Viral/immunology , Pichia/genetics , Protein Engineering/methods , Vaccines, Virus-Like Particle/biosynthesis , Viral Vaccines/biosynthesis , Antigens, Viral/genetics , Cloning, Molecular , Codon/chemistry , Codon/immunology , Culture Media/chemistry , Gene Dosage , Gene Expression , Genetic Vectors , Humans , Mating Factor/genetics , Mating Factor/metabolism , Pichia/metabolism , Protein Folding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, Subunit , Vaccines, Virus-Like Particle/genetics , Viral Vaccines/genetics
9.
Virology ; 485: 135-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26247624

ABSTRACT

There is an urgent need to develop new vaccines against highly pathogenic PRRS virus (HP-PRRSV) variant in China. The actual use of each codon pairs is more or less frequent than that of the statistical prediction and codon pair bias (CPB) usage affects gene translation. We "shuffled" the existing codons in HP-PRRSV genes GP5, M, nsp2 and nsp9, so that the CPB of these genes could be more negative. De-optimization of nsp9, the RNA-dependent RNA polymerase, significantly decreased PRRSV replication in porcine alveolar macrophages (PAMs). In vitro study showed that HV-nsp9(min) and HV-nsp29(min) were remarkably attenuated in PAMs, and inoculation of pigs with 2 ml⁎10(5.0) TCID50/ml of HV-nsp9(min) or HV-nsp29(min) did not cause PRRS. Importantly, pigs immunized with HV-nsp29(min) were fully protected against different HP-PRRSV strains׳ lethal challenges. Our results imply that the CPB de-optimized HV-nsp29(min) has the potential to be used as a live vaccine candidate against HP-PRRSV.


Subject(s)
Codon/chemistry , Macrophages, Alveolar/drug effects , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/drug effects , RNA-Dependent RNA Polymerase/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Codon/immunology , Genetic Engineering , Immunization , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/mortality , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , Protein Biosynthesis , RNA-Dependent RNA Polymerase/administration & dosage , RNA-Dependent RNA Polymerase/genetics , Survival Analysis , Swine , Vaccines, Attenuated , Viral Load , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Replication/drug effects
10.
Mol Med Rep ; 12(1): 281-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25738605

ABSTRACT

Cysticercosis due to larval cysts of Taenia solium, is a serious public health problem affecting humans in numerous regions worldwide. The oncospheral stage-specific TSOL18 antigen is a promising candidate for an anti-cysticercosis vaccine. It has been reported that the immunogenicity of the DNA vaccine may be enhanced through codon optimization of candidate genes. The aim of the present study was to further increase the efficacy of the cysticercosis DNA vaccine; therefore, a codon optimized recombinant expression plasmid pVAX1/TSOL18 was developed in order to enhance expression and immunogenicity of TSOL18. The gene encoding TSOL18 of Taenia solium was optimized, and the resulting opt-TSOL18 gene was amplified and expressed. The results of the present study showed that the codon-optimized TSOL18 gene was successfully expressed in CHO-K1 cells, and immunized mice vaccinated with opt-TSOL18 recombinant expression plasmids demonstrated opt­TSOL18 expression in muscle fibers, as determined by immunohistochemistry. In addition, the codon-optimized TSOL18 gene produced a significantly greater effect compared with that of TSOL18 and active spleen cells were markedly stimulated in vaccinated mice. 3H-thymidine incorporation was significantly greater in the opt-TSOL18 group compared with that of the TSOL18, pVAX and blank control groups (P<0.01). In conclusion, the eukaryotic expression vector containing the codon-optimized TSOL18 gene was successfully constructed and was confirmed to be expressed in vivo and in vitro. The expression and immunogenicity of the codon-optimized TSOL18 gene were markedly greater compared with that of the un-optimized gene. Therefore, these results may provide the basis for an optimized TSOL18 gene vaccine against cysticercosis.


Subject(s)
Antigens, Helminth/immunology , Codon/immunology , Cysticercosis/prevention & control , Plasmids/immunology , Taenia solium/immunology , Vaccines, DNA/immunology , Vaccines/immunology , Animals , Antigens, Helminth/genetics , Base Sequence , Biological Transport , CHO Cells , Codon/chemistry , Cricetulus , Cysticercosis/immunology , Cysticercosis/parasitology , Female , Gene Expression/immunology , Genetic Engineering , Immunization , Mice , Molecular Sequence Data , Muscle, Skeletal/immunology , Plasmids/administration & dosage , Plasmids/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Spleen/immunology , Thymidine/metabolism , Vaccines/biosynthesis , Vaccines/genetics , Vaccines, DNA/biosynthesis , Vaccines, DNA/genetics
11.
Immunogenetics ; 67(1): 15-24, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25376343

ABSTRACT

Viral epitopes have a distinct codon usage that reflects their dual role in infection and immunity. On the one hand, epitopes are part of proteins important to viral function; on the other hand, they are targets of the immune response. Studies of selection are most commonly based on changes of amino acid and seen through the accumulation of non-synonymous mutations. An independent measure of selection is the codon usage and underlying changeability of the nucleotide sequences. We here use multiple tools and a large-scale analysis of viral genomes to demonstrate that viral epitopes have a distinct codon usage and that this codon usage reflects distinct short- and long-term types of selection during viral evolution. We show that CD8(+) T cell epitopes are encoded by codons more distant from stop codons and more changeable than codons outside epitopes. This biased codon usage reflects the viral population toggling back and forth from a wild-type sequence to an escape mode, which enable them to avoid immune detection when needed, and go back to the functionally favorable form when the threat is removed (i.e., in a new host).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Evolution, Molecular , Selection, Genetic , Codon/genetics , Codon/immunology , Computational Biology , Epitopes, T-Lymphocyte/immunology , Humans , Nucleotides/genetics , Nucleotides/immunology , Phylogeny , Viruses/genetics , Viruses/immunology
12.
Hum Immunol ; 74(5): 506-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23376456

ABSTRACT

Monocyte derived dendritic cells (moDC) electroporated with tumor associated antigen derived mRNA can elicit specific T cells against tumor cells in vivo. IL21 has been shown to enhance activation and cytotoxicity in CD8+ T cells. We therefore investigated in vitro effects on human CD8+ T-cells after stimulation with IL21 mRNA electroporated moDC. Codon modification of the IL21 gene significantly enhanced IL21 production upon electroporation of moDC. Tumor associated antigen specific CTL induction efficiency was significantly enhanced when codon modified IL21 mRNA was co-electroporated with tumor associated antigen mRNA. Tumor associated antigen specific T cells induced by codon modified IL21-DC demonstrated increased cytotoxic capacity and killing compared to control cultures. In conclusion, ectopic expression of codon modified IL21 by moDC enhances the priming efficiency of the DC as well as the cytotoxic potential of the induced CTL.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Interleukins/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cells, Cultured , Coculture Techniques , Codon/genetics , Codon/immunology , Cytotoxicity Tests, Immunologic/methods , Dendritic Cells/cytology , Dendritic Cells/metabolism , Electroporation , Granzymes/immunology , Granzymes/metabolism , Humans , Interleukins/genetics , Interleukins/metabolism , K562 Cells , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transfection/methods
13.
Nature ; 491(7422): 125-8, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23000900

ABSTRACT

In mammals, one of the most pronounced consequences of viral infection is the induction of type I interferons, cytokines with potent antiviral activity. Schlafen (Slfn) genes are a subset of interferon-stimulated early response genes (ISGs) that are also induced directly by pathogens via the interferon regulatory factor 3 (IRF3) pathway. However, many ISGs are of unknown or incompletely understood function. Here we show that human SLFN11 potently and specifically abrogates the production of retroviruses such as human immunodeficiency virus 1 (HIV-1). Our study revealed that SLFN11 has no effect on the early steps of the retroviral infection cycle, including reverse transcription, integration and transcription. Rather, SLFN11 acts at the late stage of virus production by selectively inhibiting the expression of viral proteins in a codon-usage-dependent manner. We further find that SLFN11 binds transfer RNA, and counteracts changes in the tRNA pool elicited by the presence of HIV. Our studies identified a novel antiviral mechanism within the innate immune response, in which SLFN11 selectively inhibits viral protein synthesis in HIV-infected cells by means of codon-bias discrimination.


Subject(s)
Codon/genetics , Gene Expression Regulation, Viral/genetics , HIV-1/genetics , Nuclear Proteins/metabolism , Protein Biosynthesis/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics , Cell Line , Cells, Cultured , Codon/immunology , HEK293 Cells , HIV-1/growth & development , HIV-1/immunology , HIV-1/metabolism , Humans , Immunity, Innate , Nuclear Proteins/immunology , Protein Biosynthesis/immunology , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Reverse Transcription , Species Specificity , Substrate Specificity , Virus Integration
14.
J Immunol ; 186(6): 3572-80, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21321105

ABSTRACT

IL-12 and IL-23 are heterodimeric cytokines involved in the induction of Th1 and Th17 immune responses. Previous work indicated that a region on chromosome 11 encoding the IL-12p40 subunit regulates strain differences in susceptibility to murine trinitrobenzene sulfonic acid-induced colitis. In addition, this region determines strain differences in LPS-induced IL-12 responses. In this study, we investigated how polymorphisms in the coding region of murine Il12b influence IL-12 and IL-23 heterodimer formation. Transfection studies using constructs containing IL-12p35 linked to IL-12p40 from the colitis-resistant C57BL/6 strain or to the polymorphic p40 variant from the colitis-susceptible SJL/J strain demonstrated that SJL/J-derived p40 constructs synthesized significantly more IL-12p70 than did constructs harboring the C57BL/6-p40 variant. This could not be attributed to differences in synthesis rate or secretion, implicating a greater affinity of SJL/J-derived IL-12p40 for its IL-12p35 subunit. This greater affinity is also associated with increased IL-23 synthesis. In addition, C57BL/6 mice transgenic for the SJL/J 40 variant synthesized significantly more IL-12p70 upon LPS challenge and were more prone to develop colonic inflammation than did C57BL/6 mice transgenic for the C57BL/6-p40 variant. The more efficient binding of the polymorphic Il12b variant to p35 and p19 is most likely due to conformational changes following differential glycosylation as a consequence of the polymorphism. The high synthesis rate of the mature cytokines resulting from this efficient binding can lead to rapid proinflammatory skewing of immune responses and distortion of the homeostatic balance underlying the greater susceptibility for colitis.


Subject(s)
Codon/immunology , Colitis/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-23/genetics , Polymorphism, Genetic/immunology , Protein Multimerization/genetics , Protein Multimerization/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Cell Line, Tumor , Colitis/genetics , Colitis/metabolism , Disease Models, Animal , Female , Genetic Predisposition to Disease , Genetic Variation/immunology , Immunity, Innate/genetics , Interleukin-12 Subunit p40/chemistry , Interleukin-12 Subunit p40/metabolism , Interleukin-23/biosynthesis , Interleukin-23/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/immunology
15.
J Immunol ; 186(1): 479-88, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21115730

ABSTRACT

The CTL response in HLA-B*27(+) HIV-infected individuals is characterized by an immunodominant response to a conserved epitope in gag p24 (aa 263-272, KRWIILGLNK; KK10). Mutations resulting in substitution of the arginine (R264) at position 2 of this epitope have been identified as escape mutations. Nineteen HLA-B*27(+) long-term nonprogressors were identified from an Australian cohort with an average follow-up of 16 y following infection. Viral and host genetic factors impacting on disease progression were determined at multiple time points. Twelve of 19 had wild-type sequences at codon 264 at all time points; 7 of 19 carried CTL escape variants. Median viral load and CD4(+) T cell counts were not significantly different between these groups at enrollment. Viral load, as judged by levels at their last visit (1,700 and 21,000 RNA copies/ml, respectively; p = 0.01) or by time-weighted area under the curve was higher in the escape group (p = 0.02). Escape mutants at other HLA-B*27-restricted epitopes were uncommon. Moreover, host polymorphisms, such as CCR5Δ32, CCR2-64I, and SDF1-3'A, or breadth of TCR repertoire responding to KK10 did not segregate to wild-type or escape groups. Host and viral factors were examined for a relationship to viral load. The only factor to affect viral load was the presence of the R264 escape mutations at the immunodominant epitope. CTL escape at R264 in the KK10 epitope is a major determinant of subsequent viral load in these HLA-B*27(+) individuals.


Subject(s)
HIV Core Protein p24/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HLA-B Antigens/biosynthesis , Immune Evasion/immunology , Immunodominant Epitopes/immunology , Viral Load/immunology , Adult , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Arginine/genetics , Codon/immunology , Conserved Sequence/genetics , Conserved Sequence/immunology , Disease Progression , HIV Core Protein p24/genetics , HIV Infections/pathology , HIV-1/genetics , HIV-1/pathogenicity , HLA-B Antigens/genetics , HLA-B Antigens/immunology , Humans , Immune Evasion/genetics , Immunodominant Epitopes/genetics , Male , Middle Aged , Predictive Value of Tests , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology , Viral Load/genetics
16.
Vaccine ; 26(48): 6083-91, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18805452

ABSTRACT

We previously demonstrated the ability of an orally administered attenuated Salmonella enterica serovar Typhimurium strain expressing the protective antigen (PA) of Bacillus anthracis to confer protection against lethal anthrax aerosol spore challenge [Stokes MG, Titball RW, Neeson BN, et al. Oral administration of a Salmonella enterica-based vaccine expressing Bacillus anthracis protective antigen confers protection against aerosolized B. anthracis. Infect Immun 2007;75(April (4)):1827-34]. To extend the utility of this approach to humans we constructed variants of S. enterica serovar Typhi Ty21a, an attenuated typhoid vaccine strain licensed for human use, which expressed and exported PA via two distinct plasmid-based transport systems: the Escherichia coli HlyA haemolysin and the S. Typhi ClyA export apparatus. Murine immunogenicity studies confirmed the ability of these constructs, especially Ty21a expressing the ClyA-PA fusion protein, to stimulate strong PA-specific immune responses following intranasal immunization. These responses were further enhanced by a subsequent boost with either parenterally delivered recombinant PA or the licensed US human alum-adsorbed anthrax vaccine (AVA). Anthrax toxin neutralizing antibody responses using this prime-boost regimen were rapid, vigorous and broad in nature. The results of this study demonstrate the feasibility of employing a mucosal prime with a licensed Salmonella Typhi vaccine strain followed by a parenteral protein boost to stimulate rapid protective immunity against anthrax.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Anthrax/prevention & control , Immunization/methods , Salmonella typhi/genetics , Salmonella typhi/immunology , Adjuvants, Immunologic , Administration, Intranasal , Administration, Oral , Alum Compounds , Animals , Anthrax/immunology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Bacillus anthracis/immunology , Codon/immunology , Female , Humans , Immunization Schedule , Immunization, Secondary , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred BALB C , Neutralization Tests , Spores, Bacterial/immunology
17.
Cancer Res ; 68(8): 2961-71, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413766

ABSTRACT

Genetic modification of clinical-grade T cells is undertaken to augment function, including redirecting specificity for desired antigen. We and others have introduced a chimeric antigen receptor (CAR) to enable T cells to recognize lineage-specific tumor antigen, such as CD19, and early-phase human trials are currently assessing safety and feasibility. However, a significant barrier to next-generation clinical studies is developing a suitable CAR expression vector capable of genetically modifying a broad population of T cells. Transduction of T cells is relatively efficient but it requires specialized manufacture of expensive clinical grade recombinant virus. Electrotransfer of naked DNA plasmid offers a cost-effective alternative approach, but the inefficiency of transgene integration mandates ex vivo selection under cytocidal concentrations of drug to enforce expression of selection genes to achieve clinically meaningful numbers of CAR(+) T cells. We report a new approach to efficiently generating T cells with redirected specificity, introducing DNA plasmids from the Sleeping Beauty transposon/transposase system to directly express a CD19-specific CAR in memory and effector T cells without drug selection. When coupled with numerical expansion on CD19(+) artificial antigen-presenting cells, this gene transfer method results in rapid outgrowth of CD4(+) and CD8(+) T cells expressing CAR to redirect specificity for CD19(+) tumor cells.


Subject(s)
Antigens, CD19/immunology , Antigens, Neoplasm/immunology , Neoplasms/immunology , Stem Cell Transplantation , T-Lymphocytes/immunology , Transposases/immunology , Burkitt Lymphoma/immunology , Cell Line, Tumor/immunology , Coculture Techniques , Codon/immunology , Hematopoietic Stem Cells/immunology , Humans , K562 Cells/immunology , Lymphocyte Transfusion , T-Lymphocytes/cytology , Transplantation, Homologous
18.
Biochem Cell Biol ; 85(2): 218-26, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17534403

ABSTRACT

Chlamydia trachomatis is a kind of obligate intracellular bacterial pathogen that causes ocular and sexually transmitted diseases. In this study, we analyzed the codon usage patterns of the C. trachomatis mouse pneumonitis biovar (MoPn) and Homo sapiens. We found large differences between MoPn and human codon usages. To enhance the expression of Chlamydia protein in mammalian cells, the DNA sequence encoding the major outer-membrane protein (MOMP) of MoPn was modified to substitute the human-preferred codons for rarely used codons. The huma-optimized MOMP gene was synthesized and cloned into the pcDNA3 vector, as was the wild-type MOMP gene. The protein expression levels of the human-optimized MOMP and wild-type MOMP genes were compared. The experiments showed that the human-optimized MOMP gene produced significantly higher levels of MOMP protein than the wild-type MOMP, both in vitro and in vivo, but no obvious difference was observed in the levels of modified and native MOMP mRNA expression. The immunogenicity of the 2 constructs was examined using BALB/c mice following intramuscular immunization. The results showed that the mice immunized with the human-optimized MOMP produced higher levels of antigen-specific IgG antibody and showed stronger delayed-type hypersensitivity reactions and proliferative T cell responses than those immunized with the wild-type MOMP. Antigen-specific stimulation of spleen cells obtained from human MOMP DNA immunized mice produced higher levels of interferon-gamma than those obtained from wild-type MOMP DNA immunized mice. Taken together, the data show that human-optimized codon optimization can significantly enhance the gene expression and immunogenicity of the C. trachomatis MOMP DNA vaccine.


Subject(s)
Antibody Formation/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Bacterial/immunology , Antibody Formation/drug effects , Antibody Specificity/genetics , Antibody Specificity/immunology , Chlamydia Infections/genetics , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Codon/genetics , Codon/immunology , Female , Genes, Bacterial/immunology , Humans , Immunization , Mice , Mice, Inbred BALB C , Vaccines, DNA/genetics , Vaccines, DNA/pharmacokinetics
19.
Vaccine ; 25(26): 4949-59, 2007 Jun 21.
Article in English | MEDLINE | ID: mdl-17350736

ABSTRACT

HIV-1 pandemic posed an unprecedented challenge to the global health and it is believed that an effective vaccine will be the final solution against HIV-1. HIV-1 envelope is the primary immunogen in developing neutralization antibody based HIV vaccine. To define the suitable Env derived immunogen, we systemically compared the immunogenicity of gp140 and gp145 in a DNA vaccination alone and a prime-boost modalities. Two DNA vaccines and two recombinant Tiantan vaccinia vaccines (rTTV) were constructed for vaccination of female Balb/c mice. Elispot assay was used to read out the T cell immunity and ELISA assay was used to quantify antibody immunity. PLL (poly-L-leucine)-ELISA assay was used in linear antibody epitope mapping. Mice primed with gp145 tended to elicit more Env-specific T cells responses than those primed with gp140, significant difference was observed in DNA immunization alone. The ultimate T cell responses in prime-boost regimen tend to be determined mainly by the priming efficacy. Linear antibody epitope mapping displayed that sera raised by gp145 priming were vigorously reactive to more peptides than that by gp140. Our data demonstrated HIV-1 Thailand B-derived gp145 may raise higher T-cell responses and broader linear peptide-specific antibody responses than gp140 does. However, it remains to be determined that how these observations are relevant to the neutralization of antibody activities.


Subject(s)
AIDS Vaccines/genetics , AIDS Vaccines/immunology , HIV-1/genetics , HIV-1/immunology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Codon/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Female , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Vaccination , Vaccines, DNA/immunology , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
20.
J Immunol ; 177(10): 7462-71, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082666

ABSTRACT

Induction of multispecific, functional CD4+ and CD8+ T cells is the immunological hallmark of acute self-limiting hepatitis C virus (HCV) infection in humans. In the present study, we showed that gene electrotransfer (GET) of a novel candidate DNA vaccine encoding an optimized version of the nonstructural region of HCV (from NS3 to NS5B) induced substantially more potent, broad, and long-lasting CD4+ and CD8+ cellular immunity than naked DNA injection in mice and in rhesus macaques as measured by a combination of assays, including IFN-gamma ELISPOT, intracellular cytokine staining, and cytotoxic T cell assays. A protocol based on three injections of DNA with GET induced a substantially higher CD4+ T cell response than an adenovirus 6-based viral vector encoding the same Ag. To better evaluate the immunological potency and probability of success of this vaccine, we have immunized two chimpanzees and have compared vaccine-induced cell-mediated immunity to that measured in acute self-limiting infection in humans. GET of the candidate HCV vaccine led to vigorous, multispecific IFN-gamma+CD8+ and CD4+ T lymphocyte responses in chimpanzees, which were comparable to those measured in five individuals that cleared spontaneously HCV infection. These data support the hypothesis that T cell responses elicited by the present strategy could be beneficial in prophylactic vaccine approaches against HCV.


Subject(s)
Electroporation , Gene Transfer Techniques , Hepacivirus/genetics , Hepacivirus/immunology , Vaccines, DNA/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line , Codon/administration & dosage , Codon/immunology , Female , Humans , Immunity, Cellular/genetics , Macaca mulatta , Mice , Mice, Inbred BALB C , Pan troglodytes , Plasmids/administration & dosage , Plasmids/immunology , Vaccines, DNA/administration & dosage , Viral Nonstructural Proteins/administration & dosage , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...