Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Immunol ; 304-305: 35-43, 2016.
Article in English | MEDLINE | ID: mdl-27212461

ABSTRACT

The recent increase in cases of whooping cough among teenagers in the US suggests that the acellular Bordetella pertussis vaccine (aP) that became standard in the mid 1990s might be relatively less effective than the whole-bacteria formulation (wP) previously used since the 1950s. To understand this effect, we compared antibody and T cell responses to a booster immunization in subjects who received either the wP or aP vaccine as their initial priming dose in childhood. Antibody responses in wP- and aP-primed donors were similar. Magnitude of T cell responses was higher in aP-primed individuals. Epitope mapping revealed the T cell immunodominance patterns were similar for both vaccines. Further comparison of the ratios of IFNγ and IL-5 revealed that IFNγ strongly dominates the T cell response in wP-primed donors, while IL-5 is dominant in aP primed individuals. Surprisingly, this differential pattern is maintained after booster vaccination, at times from eighteen years to several decades after the original aP/wP priming. These findings suggest that childhood aP versus wP vaccination induces functionally different T cell responses to pertussis that become fixed and are unchanged even upon boosting.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Bordetella pertussis/immunology , Th1 Cells/immunology , Th1-Th2 Balance , Th2 Cells/immunology , Vaccines, Acellular/immunology , Whooping Cough/prevention & control , Adolescent , Adult , Age Factors , Antibody Formation , Cells, Cultured , Child , Child, Preschool , Humans , Immunization, Secondary/methods , Interferon-gamma/metabolism , Interleukin-5/metabolism , Th1 Cells/microbiology , Th2 Cells/microbiology , Whooping Cough/epidemiology , Whooping Cough/immunology , Young Adult
2.
Tuberculosis (Edinb) ; 95(6): 722-735, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26526557

ABSTRACT

Non-human primate (NHP) models of tuberculosis (TB) immunity and pathogenesis, especially rhesus and cynomolgus macaques, are particularly attractive because of the high similarity of the human and macaque immune systems. However, little is known about the MHC class II epitopes recognized in macaques, thus hindering the establishment of immune correlates of immunopathology and protective vaccination. We characterized immune responses in rhesus macaques vaccinated against and/or infected with Mycobacterium tuberculosis (Mtb), to a panel of antigens currently in human vaccine trials. We defined 54 new immunodominant CD4(+) T cell epitopes, and noted that antigens immunodominant in humans are also immunodominant in rhesus macaques, including Rv3875 (ESAT-6) and Rv3874 (CFP10). Pedigree and inferred restriction analysis demonstrated that this phenomenon was not due to common ancestry or inbreeding, but rather presentation by common alleles, as well as, promiscuous binding. Experiments using a second cohort of rhesus macaques demonstrated that a pool of epitopes defined in the previous experiments can be used to detect T cell responses in over 75% of individual monkeys. Additionally, 100% of cynomolgus macaques, irrespective of their latent or active TB status, responded to rhesus and human defined epitope pools. Thus, these findings reveal an unexpected general repertoire overlap between MHC class II epitopes recognized in both species of macaques and in humans, showing that epitope pools defined in humans can also be used to characterize macaque responses, despite differences in species and antigen exposure. The results have general implications for the evaluation of new vaccines and diagnostics in NHPs, and immediate applicability in the setting of macaque models of TB.


Subject(s)
Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Immunodominant Epitopes , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/microbiology , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Epitope Mapping , Histocompatibility Antigens Class II/immunology , Host-Pathogen Interactions , Humans , Interferon-gamma Release Tests , Macaca fascicularis , Macaca mulatta , Mycobacterium tuberculosis/pathogenicity , Species Specificity , Tuberculosis/microbiology , Tuberculosis/prevention & control
3.
J Immunol ; 194(12): 6164-6176, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25948811

ABSTRACT

Identification of the specific HLA locus and allele presenting an epitope for recognition by specific TCRs (HLA restriction) is necessary to fully characterize the immune response to Ags. Experimental determination of HLA restriction is complex and technically challenging. As an alternative, the restricting HLA locus and allele can be inferred by genetic association, using response data in an HLA-typed population. However, simple odds ratio (OR) calculations can be problematic when dealing with large numbers of subjects and Ags, and because the same epitope can be presented by multiple alleles (epitope promiscuity). In this study, we develop a tool, denominated Restrictor Analysis Tool for Epitopes, to extract inferred restriction from HLA class II-typed epitope responses. This automated method infers HLA class II restriction from large datasets of T cell responses in HLA class II-typed subjects by calculating ORs and relative frequencies from simple data tables. The program is validated by: 1) analyzing data of previously determined HLA restrictions; 2) experimentally determining in selected individuals new HLA restrictions using HLA-transfected cell lines; and 3) predicting HLA restriction of particular peptides and showing that corresponding HLA class II tetramers efficiently bind to epitope-specific T cells. We further design a specific iterative algorithm to account for promiscuous recognition by calculation of OR values for combinations of different HLA molecules while incorporating predicted HLA binding affinity. The Restrictor Analysis Tool for Epitopes program streamlines the prediction of HLA class II restriction across multiple T cell epitopes and HLA types.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Models, Immunological , Receptors, Antigen, T-Cell/metabolism , Software , T-Lymphocyte Subsets/immunology , Algorithms , Alleles , Computational Biology/methods , Datasets as Topic , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Testing , Humans , Internet , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Protein Binding , Receptors, Antigen, T-Cell/genetics , Reproducibility of Results , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/metabolism
4.
J Immunol Methods ; 422: 28-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25862607

ABSTRACT

Computational prediction of HLA class II restricted T cell epitopes has great significance in many immunological studies including vaccine discovery. In recent years, prediction of HLA class II binding has improved significantly but a strategy to globally predict the most dominant epitopes has not been rigorously defined. Using human immunogenicity data associated with sets of 15-mer peptides overlapping by 10 residues spanning over 30 different allergens and bacterial antigens, and HLA class II binding prediction tools from the Immune Epitope Database and Analysis Resource (IEDB), we optimized a strategy to predict the top epitopes recognized by human populations. The most effective strategy was to select peptides based on predicted median binding percentiles for a set of seven DRB1 and DRB3/4/5 alleles. These results were validated with predictions on a blind set of 15 new allergens and bacterial antigens. We found that the top 21% predicted peptides (based on the predicted binding to seven DRB1 and DRB3/4/5 alleles) were required to capture 50% of the immune response. This corresponded to an IEDB consensus percentile rank of 20.0, which could be used as a universal prediction threshold. Utilizing actual binding data (as opposed to predicted binding data) did not appreciably change the efficacy of global predictions, suggesting that the imperfect predictive capacity is not due to poor algorithm performance, but intrinsic limitations of HLA class II epitope prediction schema based on HLA binding in genetically diverse human populations.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Protein Binding/immunology , Algorithms , Epitope Mapping , HLA-DRB1 Chains/immunology , HLA-DRB3 Chains/immunology , HLA-DRB4 Chains/immunology , HLA-DRB5 Chains/immunology , Humans , Peptides/immunology
5.
J Biol Chem ; 288(39): 27872-80, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23946480

ABSTRACT

Protein arginine methyltransferase (PRMT) 8 is unique among the PRMTs, as it has a highly restricted tissue expression pattern and an N terminus that contains two automethylation sites and a myristoylation site. PRMTs catalyze the transfer of a methyl group from S-adenosylmethionine (AdoMet) to a peptidylarginine on a protein substrate. Currently, the physiological roles, regulation, and cellular substrates of PRMT8 are poorly understood. However, a thorough understanding of PRMT8 kinetics should provide insights into each of these areas, thereby enhancing our understanding of this unique enzyme. In this study, we determined how automethylation regulates the enzymatic activity of PRMT8. We found that preventing automethylation with lysine mutations (preserving the positive charge of the residue) increased the turnover rate and decreased the Km of AdoMet but did not affect the Km of the protein substrate. In contrast, mimicking automethylation with phenylalanine (i.e. mimicking the increased hydrophobicity) decreased the turnover rate. The inhibitory effect of the PRMT8 N terminus could be transferred to PRMT1 by creating a chimeric protein containing the N terminus of PRMT8 fused to PRMT1. Thus, automethylation of the N terminus likely regulates PRMT8 activity by decreasing the affinity of the enzyme for AdoMet.


Subject(s)
Membrane Proteins/metabolism , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , S-Adenosylmethionine/metabolism , Catalysis , HeLa Cells , Humans , Lysine/metabolism , Methylation , Mutagenesis, Site-Directed , Mutation , Phenylalanine/metabolism , Protein Binding , Protein Structure, Tertiary
6.
ACS Chem Biol ; 7(7): 1198-204, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22506763

ABSTRACT

Protein arginine methyltransferases (PRMTs) catalyze the posttranslational methylation of arginine using S-adenosylmethionine (SAM) as a methyl-donor. The PRMT family is widely expressed and has been implicated in biological functions such as RNA splicing, transcriptional control, signal transduction, and DNA repair. Therefore, specific inhibitors of individual PRMTs have potentially significant research and therapeutic value. In particular, PRMT1 is responsible for >85% of arginine methyltransferase activity, but currently available inhibitors of PRMT1 lack specificity, efficacy, and bioavailability. To address this limitation, we developed a high-throughput screening assay for PRMT1 that utilizes a hyper-reactive cysteine within the active site, which is lacking in almost all other PRMTs. This assay, which monitors the kinetics of the fluorescence polarization signal increase upon PRMT1 labeling by a rhodamine-containing cysteine-reactive probe, successfully identified two novel inhibitors selective for PRMT1 over other SAM-dependent methyltransferases.


Subject(s)
Drug Discovery/methods , Fluorescence Polarization/methods , High-Throughput Screening Assays/methods , Maleimides/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Maleimides/chemistry , Maleimides/metabolism , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...