Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Exp Med ; 214(8): 2243-2255, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28666979

ABSTRACT

Immunotherapy using checkpoint-blocking antibodies against targets such as CTLA-4 and PD-1 can cure melanoma and non-small cell lung cancer in a subset of patients. The presence of CD8 T cells in the tumor correlates with improved survival. We show that immuno-positron emission tomography (immuno-PET) can visualize tumors by detecting infiltrating lymphocytes and, through longitudinal observation of individual animals, distinguish responding tumors from those that do not respond to therapy. We used 89Zr-labeled PEGylated single-domain antibody fragments (VHHs) specific for CD8 to track the presence of intratumoral CD8+ T cells in the immunotherapy-susceptible B16 melanoma model in response to checkpoint blockade. A 89Zr-labeled PEGylated anti-CD8 VHH detected thymus and secondary lymphoid structures as well as intratumoral CD8 T cells. Animals that responded to CTLA-4 therapy showed a homogeneous distribution of the anti-CD8 PET signal throughout the tumor, whereas more heterogeneous infiltration of CD8 T cells correlated with faster tumor growth and worse responses. To support the validity of these observations, we used two different transplantable breast cancer models, yielding results that conformed with predictions based on the antimelanoma response. It may thus be possible to use immuno-PET and monitor antitumor immune responses as a prognostic tool to predict patient responses to checkpoint therapies.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , CTLA-4 Antigen/antagonists & inhibitors , Mammary Neoplasms, Experimental/therapy , Animals , CTLA-4 Antigen/physiology , Female , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/therapeutic use , Immunotherapy/methods , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/immunology , Mice , Neoplasm Transplantation , Positron-Emission Tomography/methods , Treatment Outcome
2.
Immunology ; 152(2): 344-355, 2017 10.
Article in English | MEDLINE | ID: mdl-28581024

ABSTRACT

Blomia tropicalis is the major asthma allergen in the tropics comparable to Dermatophagoides pteronyssinus. However, little is known about the B. tropicalis epitopes recognized by T cells. Our aim was to identify the T-cell epitopes in the major B. tropicalis allergen, Blo t 5, and investigate the potential of the corresponding peptides to inhibit the allergic inflammatory lung response. C57BL/6 mice were immunized with plasmid DNA encoding Blo t 5 and T-cell epitopes identified using the interferon-γ ELISPOT assay with 15-mer overlapping peptides. C57BL/6 mice were sensitized with bone-marrow-derived dendritic cells (BMDC) pulsed with Blo t 5 allergen followed by intranasal Blo t 5 challenge. Two H-2b restricted epitopes (Bt576-90 and Bt5106-115 ) were recognized by CD4 T cells specific for Blo t 5, but no CD8 epitopes were identified. In mice sensitized with Blo t 5-pulsed BMDC and challenged with intranasal Blo t 5 Bt576-90 and Bt5106-115 , peptide-specific CD4 T cells were found to secrete the T helper type 2 cytokines interleukin-5 and interleukin-13. Intradermal administration of synthetic peptides encoding the identified T-cell epitopes suppressed allergic airway inflammation to further allergen challenges. Hence, we have identified novel CD4 T-cell epitopes specific for Blo t 5 and demonstrated that these peptides could be employed therapeutically to suppress the T-cell response in a murine model of allergic airway inflammation.


Subject(s)
Allergens/immunology , Anti-Asthmatic Agents/immunology , Asthma/prevention & control , CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , Mites/immunology , Peptides/immunology , Pneumonia/prevention & control , Vaccines, DNA/immunology , Allergens/administration & dosage , Allergens/genetics , Animals , Anti-Asthmatic Agents/administration & dosage , Asthma/immunology , Asthma/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Enzyme-Linked Immunospot Assay , Epitope Mapping , Immunization , Injections, Intradermal , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interferon-gamma Release Tests , Lymphocyte Activation , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/administration & dosage , Peptides/genetics , Pneumonia/immunology , Pneumonia/metabolism , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Pulmonary Eosinophilia/prevention & control , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
3.
Elife ; 42015 Aug 05.
Article in English | MEDLINE | ID: mdl-26244629

ABSTRACT

The proinflammatory cytokine IL-12 drives the generation of terminally differentiated KLRG1(+) effector CD8(+) T cells. Using a Toxoplasma vaccination model, we delineate the sequence of events that naïve CD8(+) T cells undergo to become terminal effectors and the differentiation steps controlled by IL-12. We demonstrate that direct IL-12 signaling on CD8(+) T cells is essential for the induction of KLRG1 and IFN-γ, but the subsequent downregulation of CXCR3 is controlled by IL-12 indirectly through the actions of IFN-γ and IFN-γ-inducible chemokines. Differentiation of nascent effectors occurs in an extrafollicular splenic compartment and is driven by late IL-12 production by DCs distinct from the classical CD8α(+) DC. Unexpectedly, we also found extensive proliferation of both KLRG1(-) and KLRG1(+) CD8(+) T cells in the marginal zone and red pulp, which ceases prior to the final KLRG1(Hi) CXCR3(Lo) stage. Our findings highlight the notion of an extrafollicular pathway for effector T cell generation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-12/metabolism , Protozoan Vaccines/immunology , Toxoplasma/immunology , Animals , Interferon-gamma , Lectins, C-Type , Male , Mice, Inbred C57BL , Protozoan Vaccines/administration & dosage , Receptors, CXCR3/metabolism , Receptors, Immunologic/metabolism
4.
Cytometry A ; 87(10): 967-75, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26033882

ABSTRACT

We designed conditional ligands restricted to HLA-B*08:01, -B*35:01, and -B*44:05 and proved the use of a conditional ligand previously designed for HLA-B*15:02 together with HLA-B*15:01. Furthermore, we compared the detection capabilities of specific HLA-B*15:01-restricted T cells using the HLA-B*15:01 and HLA-B*15:02 major histocompatibility complex (MHC) multimers and found remarkable differences in the staining patterns detected by flow cytometry. These new conditional ligands greatly add to the application of MHC-based technologies in the analyses of T-cell recognition as they represent frequently expressed HLA-B molecules. This expansion of conditional ligands is important to allow T-cell detection over a wide range of HLA restrictions, and provide comprehensive understanding of the T-cell recognition in a given context.


Subject(s)
HLA-B35 Antigen/immunology , HLA-B8 Antigen/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , CD8-Positive T-Lymphocytes/immunology , Humans , Ligands , Peptides/immunology
5.
Immunity ; 42(5): 929-41, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25992863

ABSTRACT

It has long been thought that clonal deletion efficiently removes almost all self-specific T cells from the peripheral repertoire. We found that self-peptide MHC-specific CD8(+) T cells in the blood of healthy humans were present in frequencies similar to those specific for non-self antigens. For the Y chromosome-encoded SMCY antigen, self-specific T cells exhibited only a 3-fold lower average frequency in males versus females and were anergic with respect to peptide activation, although this inhibition could be overcome by a stronger stimulus. We conclude that clonal deletion prunes but does not eliminate self-specific T cells and suggest that to do so would create holes in the repertoire that pathogens could readily exploit. In support of this hypothesis, we detected T cells specific for all 20 amino acid variants at the p5 position of a hepatitis C virus epitope in a random group of blood donors.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Clonal Deletion , Animals , Antigenic Variation , Female , Flow Cytometry , Humans , Male , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Self Tolerance/immunology
6.
mBio ; 6(2): e02280, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25714710

ABSTRACT

UNLABELLED: The intracellular parasite Toxoplasma gondii infects a wide variety of vertebrate species globally. Infection in most hosts causes a lifelong chronic infection and generates immunological memory responses that protect the host against new infections. In regions where the organism is endemic, multiple exposures to T. gondii likely occur with great frequency, yet little is known about the interaction between a chronically infected host and the parasite strains from these areas. A widely used model to explore secondary infection entails challenge of chronically infected or vaccinated mice with the highly virulent type I RH strain. Here, we show that although vaccinated or chronically infected C57BL/6 mice are protected against the type I RH strain, they are not protected against challenge with most strains prevalent in South America or another type I strain, GT1. Genetic and genomic analyses implicated the parasite-secreted rhoptry effectors ROP5 and ROP18, which antagonize the host's gamma interferon-induced immunity-regulated GTPases (IRGs), as primary requirements for virulence during secondary infection. ROP5 and ROP18 promoted parasite superinfection in the brains of challenged survivors. We hypothesize that superinfection may be an important mechanism to generate T. gondii strain diversity, simply because two parasite strains would be present in a single meal consumed by the feline definitive host. Superinfection may drive the genetic diversity of Toxoplasma strains in South America, where most isolates are IRG resistant, compared to North America, where most strains are IRG susceptible and are derived from a few clonal lineages. In summary, ROP5 and ROP18 promote Toxoplasma virulence during reinfection. IMPORTANCE: Toxoplasma gondii is a widespread parasite of warm-blooded animals and currently infects one-third of the human population. A long-standing assumption in the field is that prior exposure to this parasite protects the host from subsequent reexposure, due to the generation of protective immunological memory. However, this assumption is based on clinical data and mouse models that analyze infections with strains common to Europe infections with strains common to Europe and North America. In contrast, we found that the majority of strains sampled from around the world, in particular those from South America, were able to kill or reinfect the brains of hosts previously exposed to T. gondii. The T. gondii virulence factors ROP5 and ROP18, which inhibit key host effectors that mediate parasite killing, were required for these phenotypes. We speculate that these results underpin clinical observations that pregnant women previously exposed to Toxoplasma can develop congenital infection upon reexposure to South American strains.


Subject(s)
Alleles , Coinfection , Protozoan Proteins/genetics , Superinfection , Toxoplasma/genetics , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/parasitology , Animals , Mice, Inbred C57BL , North America , South America , Virulence
7.
Angew Chem Int Ed Engl ; 53(49): 13390-4, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25348595

ABSTRACT

Bioorthogonal cleavable linkers are attractive building blocks for compounds that can be manipulated to study biological and cellular processes. Sodium dithionite sensitive azobenzene-containing (Abc) peptides were applied for the temporary stabilization of recombinant MHC complexes, which can then be employed to generate libraries of MHC tetramers after exchange with a novel epitope. This technology represents an important tool for high-throughput studies of disease-specific T cell responses.


Subject(s)
Azo Compounds/chemistry , HLA-A Antigens/chemistry , Peptides/chemistry , Amino Acid Sequence , Azo Compounds/immunology , Dithionite/chemistry , Epitopes/chemistry , Epitopes/immunology , HLA-A Antigens/immunology , Humans , Ligands , Models, Molecular , Peptides/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
8.
Infect Immun ; 82(11): 4854-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25156726

ABSTRACT

CD8(+) T cells play a pathogenic role in the development of murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection in C57BL/6 mice. Only a limited number of CD8(+) epitopes have been described. Here, we report the identification of a new epitope from the bergheilysin protein recognized by PbA-specific CD8(+) T cells. Induction and functionality of these specific CD8(+) T cells were investigated in parallel with previously reported epitopes, using new tools such as tetramers and reporter cell lines that were developed for this study. We demonstrate that CD8(+) T cells of diverse specificities induced during PbA infection share many characteristics. They express cytolytic markers (gamma interferon [IFN-γ], granzyme B) and chemokine receptors (CXCR3, CCR5) and damage the blood-brain barrier in vivo. Our earlier finding that brain microvessels in mice infected with PbA, but not with non-ECM-causing strains, cross-presented a shared epitope was generalizable to these additional epitopes. Suppressing the induction of specific CD8(+) T cells through tolerization with a high-dose peptide injection was unable to confer protection against ECM, suggesting that CD8(+) T cells of other specificities participate in this process. The tools that we developed can be used to further investigate the heterogeneity of CD8(+) T cell responses that are involved in ECM.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Malaria, Cerebral/complications , T-Cell Antigen Receptor Specificity/immunology , Amino Acid Sequence , Animals , Blood-Brain Barrier , Epitopes , Gene Expression Regulation/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology
9.
J Virol ; 88(18): 10613-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24990997

ABSTRACT

UNLABELLED: Cytotoxic T lymphocytes recognizing conserved peptide epitopes are crucial for protection against influenza A virus (IAV) infection. The CD8 T cell response against the M158-66 (GILGFVFTL) matrix protein epitope is immunodominant when restricted by HLA-A*02, a major histocompatibility complex (MHC) molecule expressed by approximately half of the human population. Here we report that the GILGFVFTL peptide is restricted by multiple HLA-C*08 alleles as well. We observed that M158-66 was able to elicit cytotoxic T lymphocyte (CTL) responses in both HLA-A*02- and HLA-C*08-positive individuals and that GILGFVFTL-specific CTLs in individuals expressing both restriction elements were distinct and not cross-reactive. The crystal structure of GILGFVFTL-HLA-C*08:01 was solved at 1.84 Å, and comparison with the known GILGFVFTL-HLA-A*02:01 structure revealed that the antigen bound both complexes in near-identical conformations, accommodated by binding pockets shaped from shared as well as unique residues. This discovery of degenerate peptide presentation by both HLA-A and HLA-C allelic variants eliciting unique CTL responses to IAV infection contributes fundamental knowledge with important implications for vaccine development strategies. IMPORTANCE: The presentation of influenza A virus peptides to elicit immunity is thought to be narrowly restricted, with a single peptide presented by a specific HLA molecule. In this study, we show that the same influenza A virus peptide can be more broadly presented by both HLA-A and HLA-C molecules. This discovery may help to explain the differences in immunity to influenza A virus between individuals and populations and may also aid in the design of vaccines.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/immunology , HLA-C Antigens/immunology , Influenza A virus/immunology , Influenza, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Matrix Proteins/immunology , Amino Acid Sequence , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , HLA-A Antigens/chemistry , HLA-A Antigens/genetics , HLA-C Antigens/chemistry , HLA-C Antigens/genetics , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Influenza, Human/virology , Interferon-gamma/immunology , Molecular Sequence Data , Sequence Alignment , T-Lymphocytes, Cytotoxic/virology , Viral Matrix Proteins/genetics
10.
Sci Rep ; 4: 4166, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24566718

ABSTRACT

Restoration of antigen-specific T cell immunity has the potential to clear persistent viral infection. T cell receptor (TCR) gene therapy can reconstitute CD8 T cell immunity in chronic patients. We cloned 10 virus-specific TCRs targeting 5 different viruses, causing chronic and acute infection. All 10 TCR genetic constructs were optimized for expression using a P2A sequence, codon optimization and the addition of a non-native disulfide bond. However, maximum TCR expression was only achieved after establishing the optimal orientation of the alpha and beta chains in the expression cassette; 9/10 TCRs favored the beta-P2A-alpha orientation over alpha-P2A-beta. Optimal TCR expression was associated with a significant increase in the frequency of IFN-gamma+ T cells. In addition, activating cells for transduction in the presence of Toll-like receptor ligands further enhanced IFN-gamma production. Thus, we have built a virus-specific TCR library that has potential for therapeutic intervention in chronic viral infection or virus-related cancers.


Subject(s)
Immunotherapy , Receptors, Antigen, T-Cell/genetics , Virus Diseases/genetics , Cell Line , Genetic Therapy , Genetic Vectors , Humans , Molecular Targeted Therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transduction, Genetic , Virus Diseases/immunology , Virus Diseases/therapy
11.
J Immunol ; 192(2): 641-8, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24342804

ABSTRACT

Peptide-MHC (pMHC) multimers have become one of the most widely used tools to measure Ag-specific T cell responses in humans. With the aim of understanding the requirements for pMHC-based personalized immunomonitoring, in which individuals expressing subtypes of the commonly studied HLA alleles are encountered, we assessed how the ability to detect Ag-specific T cells for a given peptide is affected by micropolymorphic differences between HLA subtypes. First, analysis of a set of 10 HLA-A*02:01-restricted T cell clones demonstrated that staining with pMHC multimers of seven distinct subtypes of the HLA-A*02 allele group was highly variable and not predicted by sequence homology. Second, to analyze the effect of minor sequence variation in a clinical setting, we screened tumor-infiltrating lymphocytes of an HLA-A*02:06 melanoma patient with either subtype-matched or HLA-A*02:01 multimers loaded with 145 different melanoma-associated Ags. This revealed that of the four HLA-A*02:06-restricted melanoma-associated T cell responses observed in this patient, two responses were underestimated and one was overlooked when using subtype-mismatched pMHC multimer collections. To our knowledge, these data provide the first demonstration of the strong effect of minor sequence variation on pMHC-based personalized immunomonitoring, and they provide tools to prevent this issue for common variants within the HLA-A*02 allele group.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Major Histocompatibility Complex/immunology , Peptides/immunology , Polymorphism, Genetic/genetics , Alleles , Amino Acid Sequence , Antigens, Neoplasm/genetics , Clone Cells/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Major Histocompatibility Complex/genetics , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Polymorphism, Genetic/immunology , Sequence Alignment
12.
Cell Rep ; 5(5): 1204-13, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24316071

ABSTRACT

Malaria is a highly prevalent disease caused by infection by Plasmodium spp., which infect hepatocytes and erythrocytes. Blood-stage infections cause devastating symptoms and can persist for years. Antibodies and CD4(+) T cells are thought to protect against blood-stage infections. However, there has been considerable difficulty in developing an efficacious malaria vaccine, highlighting our incomplete understanding of immunity against this disease. Here, we used an experimental rodent malaria model to show that PD-1 mediates up to a 95% reduction in numbers and functional capacity of parasite-specific CD8(+) T cells. Furthermore, in contrast to widely held views, parasite-specific CD8(+) T cells are required to control both acute and chronic blood-stage disease even when parasite-specific antibodies and CD4(+) T cells are present. Our findings provide a molecular explanation for chronic malaria that will be relevant to future malaria-vaccine design and may need consideration when vaccine development for other infections is problematic.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Malaria/blood , Programmed Cell Death 1 Receptor/metabolism , Animals , CD8-Positive T-Lymphocytes/pathology , Malaria/immunology , Malaria/metabolism , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics
13.
J Immunol ; 191(8): 4010-9, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24058176

ABSTRACT

The identification of virus-specific CD8(+) T cell determinants is a fundamental requirement for our understanding of viral disease pathogenesis. T cell epitope mapping strategies increasingly rely on algorithms that predict the binding of peptides to MHC molecules. There is, however, little information on the reliability of predictive algorithms in the context of human populations, in particular, for those expressing HLA class I molecules for which there are limited experimental data available. In this study, we evaluate the ability of NetMHCpan to predict antiviral CD8(+) T cell epitopes that we identified with a traditional approach in patients of Asian ethnicity infected with Dengue virus, hepatitis B virus, or severe acute respiratory syndrome coronavirus. We experimentally demonstrate that the predictive power of algorithms defining peptide-MHC interaction directly correlates with the amount of training data on which the predictive algorithm has been constructed. These results highlight the limited applicability of the NetMHCpan algorithm for populations expressing HLA molecules for which there are little or no experimental binding data, such as those of Asian ethnicity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dengue/immunology , Hepatitis B/immunology , Histocompatibility Antigens Class I/immunology , Severe Acute Respiratory Syndrome/immunology , Algorithms , Coronavirus/immunology , Dengue/virology , Dengue Virus/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A11 Antigen/immunology , HLA-A24 Antigen/immunology , Hepatitis B/virology , Hepatitis B virus/immunology , Humans , Severe Acute Respiratory Syndrome/virology , Singapore
14.
EMBO Mol Med ; 5(7): 984-99, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23681698

ABSTRACT

Cerebral malaria is a devastating complication of Plasmodium falciparum infection. Its pathogenesis is complex, involving both parasite- and immune-mediated events. CD8(+) T cells play an effector role in murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection. We have identified a highly immunogenic CD8 epitope in glideosome-associated protein 50 that is conserved across rodent malaria species. Epitope-specific CD8(+) T cells are induced during PbA infection, migrating to the brain just before neurological signs manifest. They are functional, cytotoxic and can damage the blood-brain barrier in vivo. Such CD8(+) T cells are also found in the brain during infection with parasite strains/species that do not induce neuropathology. We demonstrate here that PbA infection causes brain microvessels to cross-present parasite antigen, while non-ECM-causing parasites do not. Further, treatment with fast-acting anti-malarial drugs before the onset of ECM reduces parasite load and thus antigen presentation in the brain, preventing ECM death. Thus our data suggest that combined therapies targeting both the parasite and host antigen-presenting cells may improve the outcome of CM patients.


Subject(s)
Antigens, Protozoan/immunology , Brain/blood supply , Brain/parasitology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/analysis , Antimalarials/therapeutic use , Blood-Brain Barrier/immunology , Blood-Brain Barrier/parasitology , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/parasitology , Cross-Priming/drug effects , Female , Humans , Malaria, Cerebral/drug therapy , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Microvessels/immunology , Microvessels/parasitology , Microvessels/pathology , Molecular Sequence Data , Parasite Load , Plasmodium berghei/drug effects , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/immunology
15.
Eur J Immunol ; 43(4): 1109-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23280567

ABSTRACT

Conditional ligands have enabled the high-throughput production of human leukocyte antigen (HLA) libraries that present defined peptides. Immunomonitoring platforms typically concentrate on restriction elements associated with European ancestry, and such tools are scarce for Asian HLA variants. We report 30 novel irradiation-sensitive ligands, specifically targeting South East Asian populations, which provide 93, 63, and 79% coverage for HLA-A, -B, and -C, respectively. Unique ligands for all 16 HLA types were constructed to provide the desired soluble HLA product in sufficient yield. Peptide exchange was accomplished for all variants as demonstrated by an ELISA-based MHC stability assay. HLA tetramers with redirected specificity could detect antigen-specific CD8(+) T-cell responses against human cytomegalovirus, hepatitis B (HBV), dengue virus (DENV), and Epstein-Barr virus (EBV) infections. The potential of this population-centric HLA library was demonstrated with the characterization of seven novel T-cell epitopes from severe acute respiratory syndrome coronavirus, HBV, and DENV. Posthoc analysis revealed that the majority of responses would be more readily identified by our unbiased discovery approach than through the application of state-of-the-art epitope prediction. This flow cytometry-based technology therefore holds considerable promise for monitoring clinically relevant antigen-specific T-cell responses in populations of distinct ethnicity.


Subject(s)
Asian People , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Virus Diseases/immunology , Amino Acid Sequence , Epitopes, T-Lymphocyte/chemistry , HLA Antigens/chemistry , HLA Antigens/genetics , Humans , Ligands , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Protein Multimerization , Protein Stability
16.
PLoS One ; 7(12): e51397, 2012.
Article in English | MEDLINE | ID: mdl-23251518

ABSTRACT

Class I Major Histocompatibility Complex (MHC) molecules evolved to sample degraded protein fragments from the interior of the cell, and to display them at the surface for immune surveillance by CD8(+) T cells. The ability of these lymphocytes to identify immunogenic peptide-MHC (pMHC) products on, for example, infected hepatocytes, and to subsequently eliminate those cells, is crucial for the control of hepatitis B virus (HBV). Various protein scaffolds have been designed to recapitulate the specific recognition of presented antigens with the aim to be exploited both diagnostically (e.g. to visualize cells exposed to infectious agents or cellular transformation) and therapeutically (e.g. for the delivery of drugs to compromised cells). In line with this, we report the construction of a soluble tetrameric form of an αß T cell receptor (TCR) specific for the HBV epitope Env(183-191) restricted by HLA-A*02:01, and compare its avidity and fine-specificity with a TCR-like monoclonal antibody generated against the same HLA target. A flow cytometry-based assay with streptavidin-coated beads loaded with Env(183-191)/HLA-A*02:01 complexes at high surface density, enabled us to probe the specific interaction of these molecules with their cognate pMHC. We demonstrate that the TCR tetramer has similar avidity for the pMHC as the antibody, but they differ in their fine-specificity, with only the TCR tetramer being capable of binding both natural variants of the Env(183-191) epitope found in HBV genotypes A/C/D (187Arg) and genotype B (187Lys). Collectively, the results highlight the promiscuity of our soluble TCR, which could be an advantageous feature when targeting cells infected with a mutation-prone virus, but that binding of the soluble oligomeric TCR relies considerably on the surface density of the presented antigen.


Subject(s)
Autoantibodies/immunology , HLA Antigens/metabolism , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Cell Line , Cloning, Molecular , Flow Cytometry , HLA Antigens/immunology , Humans , Limit of Detection , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sequence Homology, Amino Acid , Surface Plasmon Resonance
17.
Proc Natl Acad Sci U S A ; 109(34): 13739-44, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22869725

ABSTRACT

We used somatic cell nuclear transfer (SCNT) to generate a mouse from the nucleus of an IgG1(+) ovalbumin-specific B cell. The resulting OBI mice show generally normal B-cell development, with elevated percentages of marginal zone B cells and a reduction in B-1 B cells. Whereas OBI RAG1(-/-) mice have exclusively IgG1 anti-ovalbumin in their serum, OBI mice show elevated levels of anti-ovalbumin of nearly all isotypes 3' of the γ1 constant region in the IgH locus, indicating that class switch recombination (CSR) occurs in the absence of immunization with ovalbumin. This CSR is associated with the presence of IgM(+)IgG1(+) double producer B cells that represent <1% of total B cells, accumulate in the peritoneal cavity, and account for near-normal levels of serum IgM and IgG3.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Nucleus/metabolism , Immunoglobulin G/biosynthesis , Alleles , Animals , Cell Separation , Crosses, Genetic , Female , Immunoglobulin Class Switching , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Male , Mice , Mice, Inbred BALB C , Nuclear Transfer Techniques , Recombination, Genetic
18.
Virus Res ; 168(1-2): 64-72, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22728446

ABSTRACT

Human enterovirus 71 (EV71) has become a major public health threat across Asia Pacific. The virus causes hand, foot, and mouth disease which can lead to neurological complications in young children. There are no specific antivirals or vaccines against EV71 infection. The major neutralizing epitope of EV71 is located in the carboxy-terminal half of the VP1 protein at amino acid positions 215-219 (Lim et al., 2012). To study the immunogenicity of VP1 we have developed a baculovirus vector which displays VP1 as a type II transmembrane protein, providing an accessible C-terminus. Immunization of mice with this recombinant baculovirus elicited neutralizing antibodies against heterologous EV71 in an in vitro microneutralization assay. Passive protection of neonatal mice confirmed the prophylactic efficacy of the antisera. Additionally, EV71 specific T cell responses were stimulated. Taken together, our results demonstrate that the display of VP1 as a type II transmembrane protein efficiently stimulated both humoral and cellular immunities.


Subject(s)
B-Lymphocytes/immunology , Capsid Proteins/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Enterovirus Infections/prevention & control , T-Lymphocytes/immunology , Animals , Antibodies, Viral/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Enterovirus A, Human/chemistry , Enterovirus A, Human/genetics , Enterovirus Infections/virology , Humans , Immunity , Immunization , Male , Mice , Mice, Inbred BALB C , Neutralization Tests , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
19.
J Virol ; 85(20): 10464-71, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21813600

ABSTRACT

Severe acute respiratory syndrome (SARS) is a highly contagious and life threatening disease, with a fatality rate of almost 10%. The etiologic agent is a novel coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV), with animal reservoirs found in bats and other wild animals and thus the possibility of reemergence. In this study, we first investigated at 6 years postinfection whether SARS-specific memory T cells persist in SARS-recovered individuals, demonstrating that these subjects still possess polyfunctional SARS-specific memory CD4+ and CD8+ T cells. A dominant memory CD8+ T cell response against SARS-CoV nucleocaspid protein (NP; amino acids 216 to 225) was then defined in SARS-recovered individuals carrying HLA-B*40:01, a HLA-B molecule present in approximately one-quarter of subjects of Asian ethnicities. To reconstitute such a CD8+ T cell response, we isolated the alpha and beta T cell receptors of the HLA-B*40:01-restricted SARS-specific CD8+ T cells. Using T cell receptor gene transfer, we generated SARS-specific redirected T cells from the lymphocytes of normal individuals. These engineered CD8+ T cells displayed avidity and functionality similar to that of natural SARS-specific memory CD8+ T cells. They were able to degranulate and produce gamma interferon, tumor necrosis factor alpha, and macrophage inflammatory proteins 1α and 1ß after antigenic stimulation. Since there is no effective treatment against SARS, these transduced T cells specific for an immunodominant SARS epitope may provide a new avenue for treatment during a SARS outbreak.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Degranulation , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Genetic Engineering , Humans , Immunologic Memory , Interferon-gamma/metabolism , Receptors, Antigen, T-Cell/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
J Biol Chem ; 286(32): 28466-75, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21680737

ABSTRACT

The binding and stabilization capacity of potential T cell epitopes to class I MHC molecules form the basis for their immunogenicity and provide fundamental insight into factors that dictate cellular immune responses. We have developed a versatile high throughput cell-free method to measure MHC stability by capturing a variety of MHC products on the surface of streptavidin-coated particles followed by flow cytometry analysis. Arrays of peptide-MHC combinations, generated by exchanging conditional ligand-loaded MHC, could be probed in a single experiment, thus combining the molecular precision of biochemically purified MHCs with high content multiparametric flow cytometry-based assays. Semiquantitative determination of the peptide affinity for the restriction element could also be accomplished through competition experiments using this bead-based assay. Furthermore, the generated peptide-MHC reagents could directly be applied to antigen-specific CD8(+) T lymphocyte analysis. The combinatorial labeling of beads allowed straightforward identification by their unique fluorescent signatures and provided a convenient means for extended assay multiplexing.


Subject(s)
CD8-Positive T-Lymphocytes/chemistry , Epitopes, T-Lymphocyte/chemistry , Flow Cytometry/methods , Histocompatibility Antigens Class I/chemistry , Peptide Library , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell-Free System/chemistry , Cell-Free System/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...